Download as pdf or txt
Download as pdf or txt
You are on page 1of 171

SECTION

VI Pain

Overview and Controversies


Kim J. Burchiel, Feridun Acar, and Andrew C. Zacest

This chapter is divided into several components: an initial over- molecular basis of nociception has also made great strides. To
view of the topics contained in the pain section, a history of the some degree, this progress contrasts with the relatively static
neurosurgical management of pain, and a series of reflections on condition of, perhaps even a retreat from, surgical management
controversies and unresolved questions related to pain. of pain.
Pain, originating from the Greek poine, which means “penalty”
or “punishment,” has been described since the dawn of writing. HISTORICAL OVERVIEW OF NEUROSURGICAL
In Greek civilization, pain secondary to disease was believed to
be caused by supernatural forces. According to a Latin inscription MANAGEMENT OF PAIN
attributed to Hippocrates of Kos (460-370 bc), alleviating pain The modern era in the neurosurgical management of pain began
was the work of the divine: sedare dolorem opus divinum est. In at the start of the 19th century. In 1809, Walker2 proposed that
Greek mythology, drugs (including opium, hashish, and man- the anterior and posterior spinal roots serve distinct motor and
drake), magic, and even surgery were used by the gods to produce sensory functions. Bell3 subsequently expanded this idea, and
hypnosis and amnesia and to heal pain. However, as the myth of Magendie4 in 1822 provided evidence of the role of the posterior
the wounded centaur Chiron shows, even an immortal would give spinal roots in the transmission of pain. However, it was almost
up divinity to avoid a life of unrelieved pain.1 100 years later that Spiller5 and Schüller6 described the pain
conduction pathways in the anterolateral columns of the spinal
cord. Although debate concerning the anatomic and physiologic
OVERVIEW OF PAIN SECTION substrate serving pain continued, evidence for the specificity
The “Pain” section of this book is composed of 18 chapters, theory of pain accumulated, and neurosurgeons exploited the
divided into the following four subsections: existing knowledge of pain pathways to perform pain surgery,
often with great success. Neurosurgeons were also quick to adapt
“Basic Science,” which comprises Chapters 166 (“Anatomy
new technology—most notably, stereotaxy, radiofrequency gen-
and Physiology of Pain”) and 167 (“Molecular Basis of
erators, and the operating microscope,—for pain procedures.
Nociception”)
A chronology of neurosurgical management of pain is presented
“Nonsurgical Therapy,” which comprises Chapters 168
in Table VI-1, which covers techniques introduced from 1873
(“Approach to the Patient with Chronic Pain”) and 169
to 1991.
(“Pharmacologic Treatment of Pain”)
A procedure for the relief of pain was probably first reported
“Treatment of Trigeminal Neuralgia,” which comprises Chapters
in 1873, when Létiévant7 published his description of peripheral
170 (“Evidence-Based Approach to the Treatment of Facial
and cranial nerve rhizotomies. Sectioning of the posterior spinal
Pain”), 171 (“Trigeminal Neuralgia: Diagnosis and Nonop-
roots for the relief of pain in humans was first proposed by Dana8
erative Management”), 172 (“Percutaneous Procedures for
in 1887 and performed by Bennet9 in 1889 and also by Abbe10,11
Trigeminal Neuralgia”), 173 (“Stereotactic Radiosurgery for
in four patients. Otrid Foerster of Breslau, Germany, a neurolo-
Trigeminal Neuralgia”), and 174 (“Microvascular Decom-
gist by training, obtained enough experience with the operation
pression for Trigeminal Neuralgia”)
of posterior rhizotomy to perform it himself. Foerster12 later
“Surgical Procedures for Nontrigeminal Pain,” which begins
presented his landmark paper mapping the dermatomes in
with Chapter 175, an initial overview (“Neurosurgical Man-
humans.
agement of Intractable Pain”), and then is divided into two
Neurosurgical management of pain has been focused primar-
parts: “Neuromodulation,” which comprises Chapters 176
ily in six areas: trigeminal neuralgia, cordotomy, stereotaxy,
(“Evidence-Based Neurostimulation for Pain”), 177 (“Periph-
gate theory of pain, intraspinal opioids, and evidence-based
eral Nerve Stimulation for Neuropathic Pain”), and 178
medicine.
(“Spinal Cord Stimulation”); and “Destructive Procedures,”
which comprises Chapters 179 (“Evidence Base for Destruc-
tive Procedures”), 180 (“Dorsal Rhizotomy and Dorsal Root Trigeminal Neuralgia
Ganglionectomy”), 181 (“Diagnosis and Management of
Early pain surgeons were also interested in cranial nerve patho-
Painful Neuromas”), 182 (“Dorsal Root Entry Zone Lesions
logic conditions, especially tic douloureux, a condition that had
for Pain”), and 183 (“Percutaneous Cordotomy and Trigemi-
vexed patients and physicians alike for more than two centuries.13
nal Tractotomy-Nucleotomy”)
Bell14 had established that the trigeminal nerve was the sensory
In summary, content of the “Pain” section focuses on the nerve for the face in 1844. Victor Horsley is credited with the
general principles of pain and nociception, with special emphasis first gasserian ganglionectomy and retrogasserian neurotomy in
on the role that neurosurgeons may play in this subspecialty. We 1891, but in truth, the contributory efforts of several other neu-
believe that substantial progress has been made in understanding rosurgical pioneers helped transform the management of tri-
the physiologic features of pain perception and its distributed geminal neuralgia. Horsley and colleagues15 used the extradural
nature in the central nervous system. The understanding of the temporal approach described by Hartley16 and Krause,17 but
1373
1374 SECTION 6  Pain

TABLE VI-1  Chronology of Neurosurgical Management of Pain Cordotomy


Year Investigator Technique
Bell,14 with the aid solely of the naked eye and the scalpel, had
1873 Létiévant7 Neurotomies for facial and extremity also traced the posterior spinal roots up the spinal cord into the
neuralgias brainstem and cerebrum. Accurately filling in the details would
1889 Abbe10 and Bennet9 Spinal dorsal rhizotomy take the rest of the 19th century. Both Moritz Schiff and Charles
1891 Horsley et al15 Gasserian ganglionectomy for trigeminal Edouard Brown-Séquard had performed numerous experiments
neuralgia
trying to locate the sensory tracts in the spinal cord35 when, in
1899 François-Franck84 Conception of the potential of
sympathectomy
1871, Müller36 cited a case of a stab wound involving half of the
1901 Spiller and Frazier19 Open trigeminal rhizotomy (middle fossa) spinal cord and the opposite dorsal column that produced bilat-
1905 Spiller5 Spinothalamic tract in the anterolateral eral anesthesia for touch but caused analgesia only on the side
cord opposite the lesion. Gowers37 later reported a case that he had
1912 Spiller and Martin39 First cordotomy seen in 1876 of a student who had shot himself through the
1913 Leriche85 Sympathectomy for pain in extremities mouth. The patient had intact tactile sensibility in his left limbs,
1916 Jonesco86 Sympathectomy for angina pectoris but pain sensation was abolished. Postmortem examination
1922 Läwen87 Diagnostic nerve blocks revealed that the injury to the spinal cord was a spicule of bone
1925 Dandy20 Trigeminal rhizotomy (posterior fossa) that had effectively caused unilateral sectioning of the cervical
1927 Armour88 Midline commissural myelotomy
1933 Foerster12 Mapping of the spinal dermatomes in
cord and destroyed the continuity of the anterior and lateral
humans columns on the right side. Gowers concluded that this part of the
1938 Sjöqvist89 Trigeminal tractotomy cord carries the fibers for the transmission of contralateral pain
1941 Schwartz and Open medullary spinothalamic impulses. Edinger38 in 1889 demonstrated the existence of the
O’Leary45 and tractotomy spinothalamic tract in newborn cats and amphibians. It remained
White47 for Spiller, however, to prove conclusively that the spinothalamic
1942 Walker46 Open mesencephalic tractotomy tract carries pain and temperature impulses. In 1905, Spiller5
1953 Spiegel and Wycis48 Stereotactic thalamotomy and described a patient with pain and temperature sensory loss in the
mesencephalotomy lower part of the body who at autopsy was confirmed to have
1960 Heath and Mickle61 Deep brain (septal) stimulation for pain
Mazars et al62 Thalamic (VPL) stimulation for pain
bilateral tuberculomas involving the lower thoracic anterolateral
1962 Foltz and White90 Bilateral cingulotomy for pain tracts. Schüller6 in 1910 sectioned the anterolateral tract in
1963 Mullan et al41 First percutaneous cordotomy monkeys. At the instigation of Spiller in 1912, Martin performed
1965 Melzack and Wall57 Gate theory of pain the first “cordotomy” in another patient with a tuberculoma of
1966 Sano et al54 Posteromedial hypothalamotomy for pain the cord.39 The short- and long-term results were encouraging
Kudo et al56 Pulvinotomy and established the technique for treating intractable pain. In
1967 Jannetta25,26 Trigeminal microvascular decompression 1931, Stookey40 was probably the first to perform a high cervical
Shealy et al59 Spinal cord stimulation cordotomy for pain in the chest and upper extremity. Mullan
1969 Kapur and Dalton91 Hypophysectomy for cancer pain and colleagues introduced a technique for percutaneous cordot-
1970 Hitchcock49 Extralemniscal myelotomy
omy at the C1-C2 level that involved use of a radioactive needle
1971 Leksell51 Gamma Knife for trigeminal neuralgia
1972 Sindou68 DREZ lesions tip and reported it in 1963.41 The utility of radiofrequency
1973 Hitchcock92 Stereotactic pontine spinothalamic thermocoagulation for creating lesions was applied to the tech-
tractotomy nique by Rosomoff and coworkers,42 and cord penetration
1974 Sweet and Wepsic93 Radiofrequency trigeminal rhizolysis as determined by electrical impedance was described by Gilden-
1976 Sweet33 and Peripheral nerve stimulation berg and associates.43 Kanpolat and colleagues44 described a com-
Nashold et al69 puted tomography–guided percutaneous procedure. Despite the
1979 Wang et al73 Intrathecal morphine obvious efficacy of this procedure, particularly for malignant
Behar et al72 Epidural morphine disease and with low rates of morbidity when performed with the
1981 Hakanson32 Glycerol trigeminal chemoneurolysis
most recent advances, cordotomy is unfortunately being used less
1983 Mullan and Lichtor34 Trigeminal balloon microcompression
1991 Tsubokawa et al67 Motor cortex stimulation
and less, in part because of advances in pain management, lack of
referral, and patient unwillingness to undergo ablative and poten-
DREZ, dorsal root entry zone; VPL, nucleus ventroposterolateralis. tially irreversible procedures.

Stereotaxy
Pain affecting the face, head, and shoulder could not be managed
because the patient died, retrogasserian neurotomy was aban- effectively by cordotomy, so open brainstem spinothalamic trac-
doned until it was revived by Tiffany18 in 1896 and by Spiller and totomies were attempted.45-47 Unfortunately, the analgesic effects
Frazier19 in 1901. Dandy20 described the posterior fossa approach were short-lived, and morbidity and mortality rates were high.
for retrogasserian neurotomy in 1925, although this was probably Spiegel and Wycis,48 the great pioneers of human stereotaxy,
not his own original idea.21 His experience led him to conclude applied their technique to perform mesencephalotomy and
that subtotal section spared significant sensation without a major thalamotomy with greater accuracy and success. Subsequently,
increase in pain recurrence.22 Although Dandy frequently Hitchcock49,50 introduced a stereotactic technique for pontine
observed vascular loops compressing the trigeminal nerve and, spinothalamic and trigeminal tractotomy.
like Gardner and Miklos23 more than 30 years later, suspected Stereotaxy not only enabled more accurate localization of
this to be the cause of neuralgia, Dandy did not conceive of targets51 but also generated important insight into the patho-
decompression as a solution. With the introduction of better physiologic mechanisms of chronic denervation pain.52 Lesions
medical therapy and percutaneous procedures with the use of the in the thalamic somatosensory ventrocaudal nuclei (which receive
Härtel approach to the foramen ovale,24 open surgical procedures input from the neospinothalamic system) were not very successful
for trigeminal neuralgia fell into decline until Jannetta25 in 1967 in relieving chronic pain and were often associated with postop-
reported his experience with microvascular decompression, a erative ataxia and dysesthesias.53 Attention was focused on lesion-
procedure that has stood the test of time.26-34 ing targets of the paleoreticulospinothalamic system, especially
SECTION 6  Pain 1375

the nonspecific intralaminar nuclei54,55 and the pulvinar nuclei,56 designated an experimental procedure for pain relief. In a similar
with greater success. manner, insurance approval in the United States for motor cortex 
stimulation for relief of pain has become increasingly difficult to
obtain, in part because of the lack of trials demonstrating clear
Gate Theory of Pain efficacy.
A major milestone in the neurosurgical management of pain came Against this backdrop, higher levels of medical evidence of
with the publication in 1965 of Melzack and Wall’s57 gate theory. efficacy have been demanded in the pain medicine literature, and
Their proposal that afferent pain transmission might be modu- the results have been sobering. In a 2008 review79 of ablative
lated by a spinal gating mechanism introduced the possibility of neurosurgical procedures published in the previous three decades,
pain management by neuromodulation. This motivated Wall and level I evidence from randomized clinical studies could be estab-
Sweet58 in 1967 to perform peripheral nerve stimulation, the first lished only for rhizotomy for trigeminal neuralgia and facet syn-
augmentative procedure for pain relief. In 1967, Shealy and dromes. Using a similar method for neuromodulatory therapies
coworkers59 performed the first trial of spinal dorsal column to relieve non–cancer-related pain, Coffey and Lozano80 con-
stimulation. Heath, after observing pain relief in psychiatric cluded that there has been no successful clinical study focused on
patients with septal stimulation,60 repeated these results in non- establishing the efficacy of neurostimulation for pain, despite
psychiatric patients in 1960.61 That same year, Mazars and associ- randomized trials in which spinal cord stimulation is compared
ates62 showed that thalamic nucleus ventroposterolateralis with best medical management and repeated surgery. Although
stimulation was also effective for chronic pain. As stimulation an inherent difficulty of trials of spinal cord stimulation is the
technology improved, other groups confirmed these results.63,64 issue of sham stimulation, most of the criticisms of trial design
Encouraged by the phenomenon of stimulation-induced analge- from this report are valid and must be addressed in future work.
sia in animals after electrical stimulation of the periaqueductal These reviews have raised the benchmark for future studies of
gray matter and the suggestion that this was mediated by endog- pain relief efficacy, thus underscoring the need for investigators
enous opioids, Richardson and Akil65,66 implanted a stimulating to provide unambiguous entry diagnosis, suitable controls that
electrode in the periventricular gray matter in humans and receive sham treatment, long-term follow-up, and randomization
reported effective pain relief. Years later, Tsubokawa and col- and establishment of blind conditions of patients, investigators,
leagues67 described the efficacy of motor cortex stimulation for and device programmers.
deafferentation pain of thalamic origin.
Many ablative techniques have since been supplanted by aug- CURRENT CONTROVERSIES AND UNRESOLVED
mentative techniques. One notable exception was the introduc-
tion by Sindou68 in 1972 of dorsal root entry zone (DREZ) QUESTIONS RELATED TO NEUROSURGICAL
ablation for deafferentation pain associated with brachial plexus MANAGEMENT OF PAIN
avulsion and traumatic paraplegia. This pain had remained The status of surgical procedures for pain control has always
intractable to all previously attempted ablative and augmenta- lagged behind the understanding of the “pain matrix.” The role
tive techniques. Nashold and coworkers69 further popularized of top-down modulation of pain circuits is still not well under-
this procedure by using a radiofrequency thermocoagulation stood and is the subject of intense continuing basic research.
technique. Furthermore, the multiple molecules that have been implicated
in pain and chronic pain contribute to the difficulty in finding
effective therapeutic agents. Alternative strategies now include
Intraspinal Opioids the idea of medicines individualized on the basis of genetic pro-
The discoveries of morphine receptors in the central nervous files and neurostimulation of pain circuits. An emerging idea is
system in 197370 and in the spinal cord in 197771 were soon fol- that there are common brain mechanisms involved in reward
lowed by the display of their utility for analgesia in the spinal processing, addiction, and chronic pain.81 Thus the basic science
fluid and epidural space.72,73 Controlled opiate delivery from of nociception and chronic pain continues to support multidisci-
implanted pumps was introduced as an analgesic technique in the plinary integrated approaches to treat chronic pain.
late 1970s for a variety of neuropathic pain syndromes and has Outlined as follows are several areas of ongoing controversies
become a mainstay neuromodulatory procedure. Pump technol- and unresolved questions related to the neurosurgical manage-
ogy has become more sophisticated, and drugs other than opioids ment of pain.
have been delivered to the spinal fluid. Despite initial enthusiasm
for this therapy, prospective studies have shown modest analgesic
gains (36% of patients reported better than 50% improvement
Spinal Cord Stimulation
at 2 years), and the complications of therapy—including catheter Spinal cord stimulation (SCS) is one of the foundations of neu-
malfunction, catheter granulomas,74,75 and hypogonadism76-78— romodulation therapy for pain. In the past, the colocalization of
have become more greatly appreciated. The result has been a evoked paresthesias encompassing the painful area has been a
steady decline in the number of devices implanted for intrathecal dictum of SCS; however, results of more recent studies indicate
opioid delivery. The expertise acquired from this complex therapy that high-frequency stimulation appears to be associated with
may be realized in the future with novel intrathecal drugs cur- pain relief without the need for stimulation-induced paresthesias.
rently under investigation (e.g., gabapentin). High-frequency stimulation results in physiologic changes in
cells that appear unrelated to the membrane depolarization block
that had been previously considered one of the potential mecha-
Evidence-Based Medicine nisms of SCS. Spinal cord stimulator leads can be implanted
Today, as in other branches of medicine, neurosurgical treatments while the patient is under general anesthesia, but it remains
of pain are increasingly scrutinized by regulatory authorities, unclear whether the lack of patient feedback might compromise
insurance providers, and the medical profession itself. This the pain relief obtained postoperatively. There is also experimen-
was demonstrated most dramatically when the Food and Drug tal evidence that SCS can be used for the treatment of ischemia,
Administration (FDA) demanded evidence for the efficacy of but the results of prospective clinical studies have been mixed. A
deep brain stimulation for the treatment of pain. Two industry- closed-loop design for SCS might potentially improve the effi-
sponsored trials failed to show a significant benefit of this therapy; cacy and efficiency of SCS, and a new generation of sensing
FDA approval was thus denied, and deep brain stimulation was technologies that can, in turn, modulate stimulation is now
1376 SECTION 6  Pain

coming to market. This could allow SCS to be a true closed-loop not applicable. Despite these limitations, image-guided cordot-
therapy—a potentially transformational development. omy is reemerging as a viable strategy for managing certain types
of pain related to cancer, particularly in patients with a limited
life expectancy. However, the practice of image-guided cordot-
Nerve Grafts and Conduits omy will remain limited, in part, by the numbers of neurosur-
The surgical treatment of neuromas now includes, in addition to geons who are trained to perform the procedure.
neuroma excision and burying into muscle or bone, the use of
nerve grafts and conduits that provide an environment that can
help prevent neuroma re-formation. The outcomes data to
CONCLUSION
support any minimally invasive techniques (e.g., radiofrequency We offer some predictions for the future, as well as some con-
ablation, chemical neurolysis) or open techniques (e.g., excision, cerns about the future in relation to neurosurgeons’ involvement
burying into muscle, conduit to nowhere) are lacking. It is diffi- in pain management. If history is to repeat, we expect that out of
cult to recommend any one therapy over another on the basis of new knowledge regarding pain transmission, representation, and
actual evidence. regulation, new formulations of surgical pain management will
emerge. The surgical procedures of the future will probably
involve reconstruction of the spinal dorsal horn, and possibly sub-
Limitations of Destructive Techniques cortical structures, that have been fundamentally altered by
The reader should be aware that classic neuroma pain (confined peripheral or central nervous system injury. Our prediction is
to the distribution of the injured nerve, reproduced as Tinel’s that although neuromodulator and neurodestruction will con-
sign, and temporarily abolished by a nerve block) is appropriately tinue to play an important role in surgical pain management,
treated with a destructive technique. Centralized pain (spread restorative neurosurgery will become a third aspect of the treat-
beyond the confines of the injured nerve, associated with complex ment armamentarium.
findings outside the nerve distribution such as allodynia, color Training in the fundamentals of pain management must also
changes, and trophic changes) often responds poorly to destruc- continue. Few neurosurgeons devote themselves to this, and the
tive techniques and may in fact worsen. lack of expertise has hampered progress in this area. Further-
more, procedures that have proved effective, such as percutane-
ous gangliolysis for trigeminal neuralgia or DREZ lesions for
Genetics and Pain Syndromes brachial plexus avulsion pain, will fall into disuse if the current
One of the more intriguing questions in the field of nerve injury generation of neurosurgical residents has no experience with
is why only a small fraction of patients who sustain a nerve injury these procedures. Similarly, if clinical research into percutaneous
actually develop a pain syndrome. This variability suggests that cordotomy or trigeminal tractotomy falters, these potentially
either patient genetics or epigenetics are in play. Several studies useful and minimally invasive procedures will disappear. It would
have suggested that genetics may have a major influence on the be unfortunate if patients had to await the next historical cycle
development of neuropathic pain in general82 and phantom pain for physicians to rediscover these approaches to pain relief.
specifically.83 Considerable ongoing work is directed at genes that The current model of pain medicine is to attempt to classify
may predispose individuals to the development of neuropathic pain syndromes as nociceptive, neuropathic, or some combina-
pain. Knowledge of these genetic predispositions, if they exist, tion of the two; the goal is to choose appropriate therapy tailored
will help direct future pharmacologic and gene therapies for to the type of pain. In general, conservative, augmentative, or
neuropathic pain. modulatory techniques are preferred over ablative approaches. In
part, this is because of the lack of contemporary data on destruc-
tive techniques, the dearth of preoperative predictors that a
Lesions in the Dorsal Root Entry Zone destructive procedure will be effective, the possibility of worsen-
DREZ lesions for nerve root avulsion pain remain one of the ing the pain, and the irreversibility of the approaches. Challenges
most effective, and probably underutilized, therapies for chronic for the future of pain medicine are twofold: first, to discover new
deafferentation pain. It is the first choice of therapy for brachial therapies that work at the molecular or genetic level to target the
plexus avulsion pain, a condition in which SCS is uniformly inef- pathophysiologic mechanisms of pain and, second, to design
fective. DREZ lesions should also be considered in cases of seg- studies that conform to the requirements for level I or II evidence
mental pain after spinal cord or cauda equina injury, in which so that these benefits may be realized for patients. In doing so,
SCS is of marginal benefit, if any. The ineffectiveness of SCS in physicians should take advantage of the wisdom gleaned from
these instances is related to the degeneration of dorsal column more than a century of discovery.
fibers up to the level of the dorsal column nuclei within the
medulla, leaving no spinal cord substrate (dorsal column) for
stimulation at the segmental level of the spinal cord. Until phar- SUGGESTED READINGS
macologic interventions can be devised to create selective inhibi- Barker FG 2nd, Jannetta PJ, Bissonette DJ, et al. The long-term outcome
of microvascular decompression for trigeminal neuralgia. N Engl J
tion of deafferentation hyperactivity within the pain transmission Med. 1996;334:1077-1083.
neurons of the spinal cord segment (nucleus proprius), DREZ Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment
lesions will remain an important treatment modality for certain of nonmalignant pain: a structured literature review. J Neurosurg.
types of posttraumatic deafferentation pain. 2008;109:389-404.
Coffey RJ, Lozano AM. Neurostimulation for chronic noncancer pain:
an evaluation of the clinical evidence and recommendations for future
Limitations of Cordotomy trial designs. J Neurosurg. 2006;105:175-189.
Cordotomy was devised as a result of the discovery of the func- Finger S. Origins of Neuroscience: A History of Explorations into Brain Func-
tion of the anterolateral spinothalamic tracts, and spinal cord tion. New York: Oxford University Press; 2001.
Hakanson S. Trigeminal neuralgia treated by the injection of glycerol into
stimulation emerged from the gate control theory. If anything, the trigeminal cistern. Neurosurgery. 1981;9:638-646.
the concept that pain is represented in a complex set of central Härtel F. Ueber die intracranielle Injektionsbehandlung der tri-
nervous system centers poses a new challenge to the surgical geminusneuralgie. Med Klin (Munich). 1914;10:582.
amelioration of pain. The “labeled line” theory of Descartes—a Leksell L. Sterotaxic radiosurgery in trigeminal neuralgia. Acta Chir
line that could simply be disrupted by surgical intervention—is Scand. 1971;137:311-314.
SECTION 6  Pain 1377

Melzack R, Wall PD. Pain mechanisms: a new theory. Science. 1965; Sweet WH. Controlled thermocoagulation of trigeminal ganglion and
150:971-979. rootlets for differential destruction of pain fibers: facial pain other than 
Miller J, Acar F, Hamilton B, et al. Preoperative visualization of neuro- trigeminal neuralgia. Clin Neurosurg. 1976;23:96-102.
vascular anatomy in trigeminal neuralgia. J Neurosurg. 2008;108: Ture H, Ture U, Gogus FY, et al. The art of alleviating pain in Greek
477-482. mythology. Neurosurgery. 2005;56:178-185.
Sindou M, Leston J, Howeidy T, et al. Micro-vascular decompression for
primary trigeminal neuralgia (typical or atypical). Long-term effective-
ness on pain; prospective study with survival analysis in a consecutive
series of 362 patients. Acta Neurochir (Wien). 2006;148:1235-1245. See a full reference list on ExpertConsult.com
SECTION 6  Pain 1377.e1

REFERENCES vascular decompression in trigeminal neuralgia (350 cases). Stereotact


1. Ture H, Ture U, Gogus FY, et al. The art of alleviating pain in Greek Funct Neurosurg. 1994;63:203-207. 
mythology. Neurosurgery. 2005;56:178-185. 31. Miller J, Acar F, Hamilton B, et al. Preoperative visualization of
2. Walker A. New anatomy and physiology of the brain in particu- neurovascular anatomy in trigeminal neuralgia. J Neurosurg. 2008;
lar and of the nervous system in general. Arch Univ Sci. 1809;3: 108:477-482.
173-174. 32. Hakanson S. Trigeminal neuralgia treated by the injection of glycerol
3. Bell C. Idea of a New Anatomy of the Brain, Submitted for the Observation into the trigeminal cistern. Neurosurgery. 1981;9:638-646.
of His Friends. London: Strahan & Preston; 1811. 33. Sweet WH. Controlled thermocoagulation of trigeminal ganglion
4. Magendie F. Experiences sur les fonctíons des racines des nerfs rachi- and rootlets for differential destruction of pain fibers: facial pain
diens. J Physiol Exp Pathol. 1822;2:336. other than trigeminal neuralgia. Clin Neurosurg. 1976;23:96-102.
5. Spiller W. The location within the spinal cord of the fibers for 34. Mullan S, Lichtor T. Percutaneous microcompression of the trigemi-
temperature and pain sensations. J Nervous Mental Dis. 1905;32: nal ganglion for trigeminal neuralgia. J Neurosurg. 1983;59:1007-
318-320. 1012.
6. Schüller A. Ueber operative Durchtrennung der Rücken- 35. Finger S. Origins of Neuroscience: A History of Explorations into Brain
markesstränge (Chordotomie). Wien Med Wochenschr. 1910;60:2292- Function. New York: Oxford University Press; 2001.
2296. 36. Müller W. Beiträge zur patholocischen Anatomie und Physiologie des men-
7. Létiévant E. Traité des Sections Nerveuses. Paris: JB Balliere et Fils; schlichen Rückenmarks. Leipzig: L Voss; 1871.
1873. 37. Gowers W. A case of unilateral gunshot injury to the spinal cord.
8. Dana C. A clinical study of neuralgias and of the origin of reflex or Trans Clin Soc Lond. 1878;11:24-32.
transferred pains. N Y Med J. 1887;66:121-127. 38. Edinger L. Vorlesungen über den Bau der nervösen Centralorgane des
9. Bennet W. A case in which acute spasmodic pain in the left lower Menschen und der Thiere für Aerzte und Studirende. Leipzig: FCW
extremity was completely relieved by subdural division of the poste- Vogel; 1900-1904.
rior roots of certain spinal nerves. Med Chir Soc Lond Trans. 1889; 39. Spiller W, Martin E. The treatment of persistent pain of organic
72:329-348. origin in the lower part of the body by division of the anterolateral
10. Abbe R. A contribution to the surgery of the spine. Med Rec. 1889; column of the spinal cord. JAMA. 1912;58:1489-1490.
35:149-152. 40. Stookey B. Chordotomy of the second cervical segment for relief
11. Abbe R. Intradural section of the spinal nerves for neuralgia. Boston from pain due to recurrent carcinoma of the breast. Arch Neurol
Med Surg J. 1896;135:329-335. Psychiatry. 1931;26:443.
12. Foerster O. The dermatomes in man. Brain. 1933;56:1-39. 41. Mullan S, Harper PV, Hekmatpanah J, et al. Percutaneous interrup-
13. Lewy F. The first authentic case of major trigeminal neuralgia and tion of spinal-pain tracts by means of a strontium 90 needle.
some comments on the history of the disease. Ann Med Hist. 1938; J Neurosurg. 1963;20:931-939.
10:247-250. 42. Rosomoff HL, Brown CJ, Sheptak P. Percutaneous radiofrequency
14. Bell C. The Nervous System. 3rd ed. London: Spottiswode; 1844. cervical cordotomy: technique. J Neurosurg. 1965;23:639-644.
15. Horsley V, Taylor J, Colman W. Remarks on the various surgical 43. Gildenberg PL, Zanes C, Flitter M, et al. Impedance measuring
procedures devised for the relief or cure of trigeminal neuralgia (tic device for detection of penetration of the spinal cord in anterior
douloureux). Br Med J. 1891;2:1139-1143, 1191-1193, 1249-1252. percutaneous cervical cordotomy. Technical note. J Neurosurg. 1969;
16. Hartley F. Intracranial neurectomy of the second and third divisions 30:87-92.
of the fifth nerve: a new method. N Y Med J. 1892;55:317-319. 44. Kanpolat Y, Deda H, Akyar S, et al. CT-guided percutaneous cor-
17. Krause F. Resection des Trigeminus innerhalb der Schädelhöhle. dotomy. Acta Neurochir Suppl. 1989;46:67-68.
Arch Klin Chir. 1892;44:821-832. 45. Schwartz H, O’Leary J. Section of the spinothalamic tract in the
18. Tiffany L. Intracranial operations for the cure of facial neuralgia. Ann medulla with observations on the pathway for pain. Surgery. 1941;
Surg. 1896;24:575-619, 736-748. 9:183-193.
19. Spiller W, Frazier C. The division of the sensory root of the trigemi- 46. Walker A. Relief of pain by mesencephalic tractotomy. Arch Neurol
nus for the relief of tic douloureux: an experimental, pathological and Psychiatry. 1942;43:284-298.
clinical study, with a preliminary report of one surgically successful 47. White J. Spinothalamic tractotomy in the medulla oblongata. Arch
case. Univ Pa Med Bull. 1901;14:341-352. Surg. 1941;43:113-127.
20. Dandy W. Section of the sensory root of the trigeminal nerve at the 48. Spiegel EA, Wycis HT. Mesencephalotomy in treatment of intrac-
pons. Bull Johns Hopkins Hosp. 1925;36:105-106. table facial pain. AMA Arch Neurol Psychiatry. 1953;69:1-13.
21. Stookey B, Ransohoff J. Trigeminal Neuralgia: Its History and Treat- 49. Hitchcock E. Stereotactic trigeminal tractotomy. Ann Clin Res. 1970;
ment. Springfield, IL: Charles C Thomas; 1959. 2:131-135.
22. Dandy W. An operation for the cure of tic douloureux: partial section 50. Hitchcock E, Sotelo MG, Kim MC. Analgesic levels and technical
of the sensory root at the pons. Arch Surg. 1929;18:687-734. method in stereotactic pontine spinothalamic tractotomy. Acta Neu-
23. Gardner WJ, Miklos MV. Response of trigeminal neuralgia to rochir (Wien). 1985;77:29-36.
decompression of sensory root; discussion of cause of trigeminal 51. Leksell L. Sterotaxic radiosurgery in trigeminal neuralgia. Acta Chir
neuralgia. JAMA. 1959;170:1773-1776. Scand. 1971;137:311-314.
24. Härtel F. Ueber die intracranielle injektionsbehandlung der tri- 52. Tasker RR, Kiss ZH. The role of the thalamus in functional neuro-
geminusneuralgie. Med Klin (Munich). 1914;10:582-584. surgery. Neurosurg Clin N Am. 1995;6:73-104.
25. Jannetta PJ. Arterial compression of the trigeminal nerve at the pons 53. Tasker R, Organ L, Hawrylshyn P. The Thalamus and Midbrain of
in patients with trigeminal neuralgia. J Neurosurg. 1967;26(suppl): Man: A Physiological Atlas Using Electrical Stimulation. Springfield, IL:
159-162. Charles C Thomas; 1982.
26. Jannetta PJ. Arterial compression of the trigeminal nerve at the pons 54. Sano K, Yoshioka M, Ogashiwa M, et al. Thalamolaminotomy. A
in patients with trigeminal neuralgia 1967. J Neurosurg. 2007;107: new operation for relief of intractable pain. Confin Neurol. 1966;27:
216-219. 63-66.
27. Barker FG 2nd, Jannetta PJ, Bissonette DJ, et al. The long-term 55. Spiegel EA, Wycis HT, Szekely EG, et al. Combined dorsomedial,
outcome of microvascular decompression for trigeminal neuralgia. intralaminar and basal thalamotomy for relief of so-called intractable
N Engl J Med. 1996;334:1077-1083. pain. J Int Coll Surg. 1964;42:160-168.
28. Pollock BE, Phuong LK, Gorman DA, et al. Stereotactic radiosur- 56. Kudo T, Yoshii N, Shimizu S, et al. Effects of stereotaxic thalamot-
gery for idiopathic trigeminal neuralgia. J Neurosurg. 2002;97:347- omy on intractable pain and numbness. Preliminary report. Keio J
353. Med. 1966;15:191-195.
29. Sindou M, Leston J, Howeidy T, et al. Micro-vascular decompression 57. Melzack R, Wall PD. Pain mechanisms: a new theory. Science. 1965;
for primary trigeminal neuralgia (typical or atypical). Long-term 150:971-979.
effectiveness on pain; prospective study with survival analysis in a 58. Wall PD, Sweet WH. Temporary abolition of pain in man. Science.
consecutive series of 362 patients. Acta Neurochir (Wien). 2006; 1967;155:108-109.
148:1235-1245. 59. Shealy CN, Mortimer JT, Reswick JB. Electrical inhibition of pain
30. Sindou MP, Chiha M, Mertens P. Anatomical findings observed by stimulation of the dorsal columns: preliminary clinical report.
during microsurgical approaches of the cerebellopontine angle for Anesth Analg. 1967;46:489-491.
1377.e2 SECTION 6  Pain

60. Heath R. Studies in Schizophrenia. Cambridge, MA: Harvard Univer- 77. Finch PM, Roberts LJ, Price L, et al. Hypogonadism in patients
sity Press; 1954. treated with intrathecal morphine. Clin J Pain. 2000;16:251-254.
61. Heath R, Mickle W. Evaluation of seven years experience with depth 78. Schneider J. Hypogonadism in men treated with chronic opioids.
electrode studies in human patients. In: Ramey E, O’Doherty D, eds. Arch Phys Med Rehabil. 2008;89:1414.
Electrical Studies in the Anesthetized Brain. New York: Harper & Row; 79. Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treat-
1960:214-247. ment of nonmalignant pain: a structured literature review. J Neuro-
62. Mazars G, Roge R, Mazars Y. [Results of the stimulation of the surg. 2008;109:389-404.
spinothalamic fasciculus and their bearing on the physiopathology of 80. Coffey RJ, Lozano AM. Neurostimulation for chronic noncancer
pain]. Rev Prat. 1960;103:136-138. pain: an evaluation of the clinical evidence and recommendations for
63. Adams JE, Hosobuchi Y, Fields HL. Stimulation of internal capsule future trial designs. J Neurosurg. 2006;105:175-189.
for relief of chronic pain. J Neurosurg. 1974;41:740-744. 81. Elman I, Borsook D. Common mechanisms of chronic pain and
64. Hosobuchi Y, Adams JE, Rutkin B. Chronic thalamic stimulation addiction. Neuron. 2016;89:11-36.
for the control of facial anesthesia dolorosa. Arch Neurol. 1973;29: 82. Clarke H, Katz J, Flor H, et al. Genetics of chronic post-surgical
158-161. pain: a crucial step toward personal pain medicine. Can J Anaesth.
65. Richardson DE, Akil H. Pain reduction by electrical brain stimula- 2015;62:294-303.
tion in man. Part 2: chronic self-administration in the periventricular 83. Costigan M, Belfer I, Griffin RS, et al. Multiple chronic pain states
gray matter. J Neurosurg. 1977;47:184-194. are associated with a common amino acid-changing allele in KCNS1.
66. Richardson DE, Akil H. Pain reduction by electrical brain stimula- Brain. 2010;133:2519-2527.
tion in man. Part 1: acute administration in periaqueductal and peri- 84. François-Franck CE. Signification physiologique de la résection du
ventricular sites. J Neurosurg. 1977;47:178-183. sympathique dans la maladie de Basendour, l’épilepsie, l’idiotie et de
67. Tsubokawa T, Katayama Y, Yamamoto T, et al. Chronic motor cortex glaucome. Bull Acad Natl Med. 1899;41:565-568.
stimulation for the treatment of central pain. Acta Neurochir Suppl. 85. Leriche R. De l’élongation et de la section des nerfs périvasculaires
1991;52:137-139. dans certains syndromes douloureux d’origine artérielle et dans
68. Sindou M. Étude de la jonction radiculo-médullaire postérieure. La radicel- quelques troubles trophiques. Lyon Chir. 1913;10:378-382.
lotomie postérieure sélective dans la chirurgie de la doleur. Villeurbanne, 86. Jonesco T. Angine de poitrine guérie par la résection du sympathique
France: University of Lyon; 1972. das la maladie de Basendour, l’épilepsie, l’idiote, et de glaucome
69. Nashold B, Urban B, Zorub D. Phantom pain relief by focal destruc- [Cardiac chest pain alleviated by resection of the sympathetic
tion of the substantia gelatinosa of Rolando. Adv Pain Res Ther. system, as used in the treatment of Basendour’s disease, epilepsy,
1976;1:950-963. mental retardation and glaucoma]. Bull Acad Natl Med. 1920;84:
70. Pert CB, Snyder SH. Opiate receptor: demonstration in nervous 93-102.
tissue. Science. 1973;179:1011-1014. 87. Läwen A. Ueber segmentäre Schmerzaufhebung durch papaverte-
71. Atweh SF, Kuhar MJ. Autoradiographic localization of opiate recep- brale Novokaininjektionen zur Differentialdiagnose intra-
tors in rat brain. I. Spinal cord and lower medulla. Brain Res. 1977; adbominaler Erkrankungen. Med Wochenschr. 1922;69:1423.
124:53-67. 88. Armour D. Surgery of the spinal cord and its membranes. Lancet.
72. Behar M, Magora F, Olshwang D, et al. Epidural morphine in treat- 1927;1:691-697.
ment of pain. Lancet. 1979;1:527-529. 89. Sjöqvist O. Studies on pain conduction in the trigeminal nerve. Acta
73. Wang JK, Nauss LA, Thomas JE. Pain relief by intrathecally applied Psychiatr Neurol Scand Suppl. 1938;17:1-139.
morphine in man. Anesthesiology. 1979;50:149-151. 90. Foltz EL, White LE Jr. Pain “relief” by frontal cingulumotomy.
74. Cabbell KL, Taren JA, Sagher O. Spinal cord compression by cath- J Neurosurg. 1962;19:89-100.
eter granulomas in high-dose intrathecal morphine therapy: case 91. Kapur TR, Dalton GA. Trans-sphenoidal hypophysectomy for meta-
report. Neurosurgery. 1998;42:1176-1180. static carcinoma of the breast. Br J Surg. 1969;56:332-337.
75. Langsam A. Spinal cord compression by catheter granulomas in 92. Hitchcock ER. Stereotaxic pontine spinothalamic tractotomy. J Neu-
high-dose intrathecal morphine therapy: case report. Neurosurgery. rosurg. 1973;39:746-752.
1999;44:689-691. 93. Sweet WH, Wepsic JG. Controlled thermocoagulation of trigeminal
76. Daniell HW. Hypogonadism in men consuming sustained-action ganglion and rootlets for differential destruction of pain fibers. 1.
oral opioids. J Pain. 2002;3:377-384. Trigeminal neuralgia. J Neurosurg. 1974;40:143-156.
PART 1 Basic Science

166 Anatomy and Physiology of Pain


Mary M. Heinricher and Daniel R. Cleary

SENSORY TRANSMISSION OF stimulus (i.e., tissue damage) and the ensuing sensory experience
is rarely straightforward. This is true even in the simplest situa-
PAIN-RELATED INFORMATION tion in which a reasonably uniform sensory experience might be
Nociceptive information is conveyed from the periphery to the expected, such as acute injury to healthy tissue.
spinal cord by small myelinated and unmyelinated primary affer- Cognitive and emotional factors, including attention, learn-
ents that travel through the dorsal root to synapse in the dorsal ing, and mood, can all influence pain perception in situations
horn. These afferents enter the dorsal horn and terminate both nowhere nearly as extreme as the battlefield. The brain is not a
superficially (in laminae I and II) and more deeply (in laminae V, passive receiver of information about noxious stimuli but actively
VI, and VII), as well as around the central canal. Second-order regulates its own input. Indeed, the coding of afferent informa-
neurons in the nociceptive pathways are found in the same layers tion is now known to be shaped dynamically by descending
and are usually divided into two classes: wide dynamic range modulatory systems that influence nociceptive processing at the
(WDR) and nociceptive-specific. WDR neurons receive conver- first central relay, the dorsal horn. These descending control
gent input from both nociceptive and nonnociceptive primary systems are themselves regulated by afferent sensory input but
afferents, which can allow normally nonpainful stimuli such as are also recruited by higher centers to modulate spinal nocicep-
touch to give rise to a sensation of pain under certain conditions. tive processing in accord with behavioral priorities. The most
Nociceptive information is conveyed to the brain via the important descending control system has links in the midbrain
spinoreticular, spinomesencephalic, spinoparabrachial, and spi- PAG and to the rostral ventromedial medulla (RVM), which
nothalamic tracts, all of which ascend through the anterolateral constitute the PAG/RVM system. Electrical stimulation at either
quadrant. Direct spinothalamic projections terminate in both the site can enhance or inhibit pain, depending on the exact site,
medial and lateral thalamus, and these two targets are considered current level, and the behavioral context. These effects are medi-
more in the affective-motivational and sensory-discriminative ated by modulation of nociceptive activity in the dorsal horn. The
aspects of pain, respectively. Parallel spinoreticular and spinomes- anatomic substrate for this descending control is a significant
encephalic pathways may contribute to conscious sensation, but projection from the PAG to the RVM, which in turn projects via
these pathways may be more important for arousal, autonomic the dorsolateral funiculus to the dorsal horn to influence sensory
and motor responses to noxious input and for recruitment of processing. The PAG/RVM system is well situated to mediate the
descending control systems. The spinoparabrachial pathway con- effects of cognitive and emotional variables on pain transmission.
sists of projections of neurons primarily in lamina I and relays It also functions as a positive-feedback loop, so that discrete
information to the amygdala and hypothalamus, as well as to the noxious stimuli or more prolonged inflammatory events activate
midbrain periaqueductal gray matter (PAG) and caudal ventro- descending facilitation, thereby amplifying responses to subse-
lateral medulla. quent input
The advent of functional imaging approaches has stimulated Much information about primary afferents and nociceptive
interest in the cortical representation of pain, and it is now appre- signal transduction has become available, but it must be empha-
ciated that no single cortical “pain center” mediates all aspects sized that pain as a sensory experience requires transmission and
of the complex sensation that is pain. Rather, a reasonably well- integration of sensory input within the central nervous system. It
defined cortical network is recruited as a result of acute noxious is now recognized that pain reflects activation of parallel ascend-
stimulation. Although no lesion at a single site can eliminate ing pathways that engage a distributed thalamocortical network,
the perception of pain, stimulation of any one of many sites a network sometimes referred to as a “pain matrix.” This network
can elicit painful perceptions, in conjunction with or indepen- is not a passive receiver, and input in the ascending pain system
dent of other somatosensory sensations. Experiments involving is dynamically regulated in accord with behavioral priorities by a
such stimulation, along with advances in both noninvasive and descending modulatory system. Finally, there is increasing evi-
invasive techniques, have shed light on the contributions that dence that as pain becomes chronic, the systems engaged shift to
the numerous cortical regions make to the “pain matrix,” which reflect the growing importance of conditioning and other higher
includes somatosensory cortex, insula, and anterior cingulate order processes. Understanding and manipulating these dynamic
cortex (ACC). Of importance is that the cortical representation aspects of pain sensing and modulation is crucial for future
of longstanding chronic pain adds new layers of complexity to advances in therapeutic approaches to pain.
this picture.

DESCENDING MODULATION OF PAIN


A recurrent theme in clinical and experimental approaches to
pain has been that the relationship between the presumed Full text of this chapter is available online at ExpertConsult.com

1378
PART 1 Basic Science 166

166 Anatomy and Physiology of Pain


Mary M. Heinricher and Daniel R. Cleary

This chapter outlines current thinking concerning mechanisms noxious range. WDR neurons are distributed somatotopically
and pathways for transmission and modulation of pain, with a within the dorsal horn, and although receptive fields are rela-
focus on central nervous system mechanisms. Dorsal horn pro- tively large, they are not so large as to preclude a contribution
cessing of nociceptive information, and ascending pathways car- to stimulus localization. WDR neurons are more common in
rying the nociceptive signal to the brain are described, as are the deeper dorsal horn but are also present more superficially,
thalamocortical circuits that play important roles in pain. Next, in laminae I and II. Nociceptive-specific neurons do not receive
we discuss the malleability of pain. We consider how the nocicep- nonnociceptive input and respond exclusively to noxious stimuli,
tive transmission pathways are sensitized after injury to either the carried either by A delta mechanoreceptors or by both A delta and
tissue or to the central nervous system itself, which leads to C nociceptors. Their receptive fields are small, which indicates an
hyperalgesia and allodynia (i.e., pain evoked by innocuous stimu- important role in stimulus localization. They are concentrated in
lation). Finally, we focus on descending modulatory systems that the more superficial layers.
regulate transmission pathways, as part of positive and negative- Nociceptive information is conveyed to the brain primarily
feedback loops and through top-down control mechanisms. via the spinoreticular, spinomesencephalic, spinoparabrachial,
These systems can enhance or suppress the nociceptive signal in and spinothalamic tracts, all of which ascend through the antero-
accord with behavioral priorities, during stress, or after adminis- lateral quadrant (Fig. 166-1). The importance of the anterolateral
tration of analgesic drugs, including opioids. systems in pain (and temperature) sensibility is confirmed by the
ability of anterolateral cordotomy to relieve pain, at least in
the short term, in both patients and experimental studies.1,5,6
PRIMARY AFFERENT NOCICEPTORS The spinothalamic tract also carries nonnociceptive information
Nociceptive information is conveyed from the periphery to the (innocuous warming and cooling, innocuous tactile information).
spinal cord by small myelinated and unmyelinated primary affer- Tactile information is in addition conveyed by the spinocervical
ents that travel through the dorsal root to synapse in the dorsal tract and through the dorsal columns (which includes ascending
horn. As described in detail in Chapter 167 (focused on periph- branches of large-diameter, low-threshold primary afferents,
eral transduction and properties of nociceptive primary affer- as well as the postsynaptic dorsal column system, which com-
ents), information about the mechanisms of nociceptive signal prises second-order projections of low-threshold dorsal horn
transduction and the properties of primary afferents has become neurons).
plentiful. Individual afferents manifest a high degree of specific- Direct spinothalamic projections terminate in both the medial
ity; nociceptive neurons are different from low-threshold affer- and lateral thalamus, and these two targets can be considered
ents in terms of physiology, morphology, and neurochemistry. more important in the affective-motivational and sensory-
The primary afferent fibers also exhibit significant plasticity in discriminative aspects of pain, respectively.7,8 Parallel spinoreticu-
response to tissue conditions, with alterations of neuronal phe- lar and spinomesencephalic pathways may contribute to conscious
notype and enhanced responsiveness during inflammation or in sensation, but these pathways may be more important for arousal,
response to damage to the nerve itself. Such primary afferent for autonomic and motor responses to noxious inputs, and for
sensitization is now recognized to be an important contributor recruitment of descending control systems (see “Descending
to hyperalgesia and abnormal pain states. Modulatory Systems” section). The spinoparabrachial pathway
comprises projections of neurons primarily in lamina I and relays
information to the amygdala and hypothalamus, as well as mid-
DORSAL HORN AND ASCENDING PATHWAYS brain PAG and caudal ventrolateral medulla. The spinoparabra-
Nociceptive afferents enter the dorsal horn and terminate both chial pathway is considered particularly important in emotional
superficially (laminae I and II) and more deeply (in laminae V, and autonomic aspects of pain.9
VI, and VII), as well as around the central canal. Low-threshold
tactile afferents, in contrast, spare the superficial laminae and
send their terminals primarily to laminae III and IV (sometimes
Role of Dorsal Column Pathway in Visceral Pain
called the nucleus proprius).1,2 The different terminations of noci- The classic view that ascending pathways through the dorsal
ceptive and low-threshold inputs to the dorsal horn highlights the columns are involved exclusively in transmission of nonnoxious
link between anatomic and functional organization of nocicep- information has been called into question by evidence that a
tive and nonnociceptive somatosensory pathways and extends the postsynaptic dorsal column component contributes to visceral
specificity at the level of primary afferents to the central nervous pain sensibility.10 These studies were motivated by the clinical
system. Second-order neurons in the nociceptive pathways are observation that midline myelotomy relieved pain in patients
present primarily in the superficial layers and in laminae V and with cancer involving pelvic visceral structures. Experimental
VI of the dorsal horn and are usually divided into two classes: studies subsequently revealed a significant projection ascending
WDR and nociceptive-specific.3,4 WDR neurons receive conver- ipsilaterally through the dorsal columns and transmitting infor-
gent inputs from both nociceptive and nonnociceptive primary mation to the ventroposterolateral nucleus of the thalamus. Of
afferents. They consequently exhibit low thresholds, within the importance is that this pathway may not contribute to pain sensa-
innocuous range, but unlike low-threshold tactile dorsal horn tion under normal conditions, but it could become sensitized by
neurons, WDR neurons code stimulus intensity through the visceral inflammation.11,12
e593
e594 SECTION 6  Pain

information related to pain, whereas the medial system is more


PFC closely associated with emotional aspects of pain. The lateral
system encompasses the ventral and posterior nuclei of the thala-
mus and their linkages with lateral somatosensory cortices. The
S-I lateral thalamic nuclei receive direct spinothalamic input from
ACC both superficial and deep layers of the dorsal horn.20,21 Thalamic
nuclei assigned to the lateral system include the ventral posterior
M-T l medial (VPM) nucleus and the ventral posterior lateral (VPL)
S-II nucleus, as well as the ventral posterior inferior (VPI) nucleus.
L-T

Insula Together, the VPM and VPL nuclei in humans are referred to as
hal
ha

the ventrocaudal (Vc) nucleus. Medial and intralaminar nuclei,


Hyp along with their targets in the ACC and medial prefrontal corti-
PB ces, have been considered to constitute the medial pathway. In a
Amg still-controversial remapping of these systems, Dostrovsky and
Craig16 argued that a novel, primate-specific nucleus, the poste-
rior part of the ventromedial nucleus (VMpo) represents the main
thalamic relay for sensory-discriminative information, with the
insula as the principal cortical target. The validity of this frame-
Reticular area

work remains a topic of debate,20-24 and the controversy probably


reflects the somewhat artificial nature of the concept of medial/
lateral pain system. Certainly it should be recognized that these
divisions are not as well demarcated as was once thought,
and overlap and interconnections must exist in function and
structure.

Lateral Thalamic Nuclei


Ventral Caudal Nucleus
The structures of the lateral thalamus primarily associated with
nociception are the three nuclei that make up the Vc nucleus,
Figure 166-1. Nociceptive transmission pathways. This schematic
also known as the principal sensory nucleus of the thalamus in
highlights some components of the anterolateral system. Axons
humans and as the ventral posterior complex in primates. In both
ascending from the dorsal horn target the brainstem (spinoreticular,
humans and primates, the constituent nuclei are the VPM and
spinomesencephalic, and spinoparabrachial), as well as the
VPL nuclei, which are functionally matched. Spinothalamic pro-
spinothalamic tracts: the medial thalamus (M-Thal) and lateral
jections to the VPL nucleus arise from the spinal dorsal horn,
thalamus (L-Thal). Spinoreticular information can be carried
whereas inputs to the VPM nucleus are from the spinal trigemi-
over multisynaptic pathways to the thalamus (not shown).
nal nucleus (i.e., the “medullary dorsal horn”).25-27 Spinothalamic/
Spinoparabrachial information is relayed directly to the amygdala
trigeminothalamic terminations are interdigitated with terminals
(Amg) and hypothalamus (Hyp). ACC, anterior cingulate cortex;
of the medial lemniscal pathway, wherein somatotopically orga-
PB, parabrachial nuclei; PFC, prefrontal cortex; S-I, somatosensory
nized tactile information is carried through the dorsal columns
cortex; S-II, secondary somatosensory cortex.
and relayed through the dorsal column nuclei.28,29 The spinotha-
lamic terminations in the VPL nucleus arrive in clusters and are
organized, approximately somatotopically, in concordance with
the same medial-lateral somatotopic organization as the lemnis-
SUPRASPINAL NOCICEPTIVE TARGETS cal pathway.30-32
The outputs of the Vc nucleus are primarily to sensory cortical
Thalamus areas, most notably to the primary somatosensory cortex (SI).
Using retrograde tracing in the primate, Gingold and associates33
The thalamus was proposed to be involved in pain since as early found that Vc nucleus provides the majority of thalamic input to
as 1911. Indeed, pain was at that time considered uniquely primi- SI. Of importance is that as many as 90% of identified nociceptive
tive among sensory systems as being perceived at the level of the neurons of the VPL nucleus could be activated antidromically
thalamus, without important cortical involvement.13 It is now from the SI.34 Other identified targets of Vc nucleus projections
recognized that reentrant interconnections between different include the secondary somatosensory cortex (SII), insula, and
thalamic nuclei and cortex areas set the stage for conscious per- posterior parietal cortex.35-38
ception of pain.14-19 In addition to anatomic approaches, recording and stimulation
Much of the understanding of the human thalamus is based of Vc neurons provide evidence for the involvement of the region
on comparative physiology, especially retrograde and anterograde in sensory and discriminative aspects of pain. The majority of
tracing studies in rats, cats, and nonhuman primates. Naming of neurons in this region respond to innocuous or low-threshold
functionally matched areas differs among many species, and there mechanical stimuli, but as many as 10% are activated by noxious
are significant species-specific differences in structure and orga- stimuli or temperature changes.39-42 Clusters of these nocire-
nization, particularly between rodents and primates. In this dis- sponding neurons can be identified most consistently near the
cussion, we therefore present data primarily from humans and posterior and inferior aspect.28,31,34,43,44 Neurons analogous to the
nonhuman primates. nociceptive-specific and WDR classes first described in the dorsal
In the study of nociception, the thalamus is generally sepa- horn can be identified.45 The Vc nucleus receives visceral, as well
rated into medial and lateral aspects. Although this division is as cutaneous, input and neurons in this nucleus have been shown
no longer as clear-cut as was once thought, the lateral system is to respond to noxious visceral stimuli, although the organization
most strongly linked to the processing of sensory-discriminative of responsive neurons is not apparently viscerotopic.46,47
CHAPTER 166  Anatomy and Physiology of Pain e595

Stimulation of Vc neurons in humans most often produces thermoreceptive-specific neurons and the lack of low-threshold
contralateral, nonpainful paresthesias even at high intensities, at neurons. The recorded neurons have an anterior-posterior topo- 166
least in patients without a chronic pain complaint, although tem- graphic organization and small receptive fields.60 Subsequent
perature and pain sensations can be evoked from some sites at electrophysiologic recordings in awake human patients demon-
the lowest stimulus intensity.48,49 In one study, stimulation sites strated that neurons in a region tentatively identified as the
near the posterior and inferior border were significantly more VMpo nucleus responded to innocuous or noxious cooling of the
likely to evoke painful or thermal sensations than those in the skin and that stimulation of the region elicited perception of cold
core region.48 Bagley and associates50 and Lenz and colleagues51 or cooling.69 These findings suggest that the VMpo nucleus could
were able to distinguish stimulation sites at which binary sensa- be important, not specifically as a nociceptive relay, but rather for
tions (pain versus no pain) can be elicited and other sites at which cooling/cold information.
an analogue sensation, in which the sensory magnitude is related The location, identification, and characterization of the
to stimulus intensity, is produced. They suggested that the infor- primate and human VMpo nuclei have become a subject of
mation conveyed at sites associated with binary signaling are debate. Arguments regarding the projection of lamina I neurons
related to an “alarm” aspect of pain processing, whereas process- in the VMpo nucleus are based on questions identifying the
ing at analogue signaling sites is more important for coding terminal versus fibers of passage. Using a different antibody to
stimulus intensity. the same antigen, another group failed to replicate the staining
The effects of lesions and inactivation of the Vc nucleus results that first identified the VMpo nucleus in primates, and the
further support the idea that the Vc nucleus is a functional relay human VMpo nucleus was identified primarily on the basis of
for nociceptive information. Focal application of lidocaine in this antibody staining against the same antigen.21,22,24,58 In a study of
region in nonhuman primates results in reduced detection of poststroke lesions in humans, Kim and colleagues57 found that
small changes in skin temperature in the noxious range.52 In poststroke pain and changes in temperature perception still
humans, lesions of the Vc nucleus and, more broadly, the lateral occurred in the absence of VMpo damage, although lesions that
thalamus have been attempted for the treatment of neuropathic did not include a significant part of the Vc nucleus did not cause
pain, with resulting decreases in contralateral detection of thermal these symptoms. Further research is needed to confirm previous
and mechanical pain, as well as in touch and proprioception.53-55 findings on the boundaries and connectivity of the VMpo nucleus
However, such surgeries are necessarily conducted with caution in primates and to characterize the physiology and responsiveness
because of the risk of triggering iatrogenic central pain.56,57 of VMpo cells in humans and primates.

Ventralis Caudalis Parvocellularis Medial Thalamic Nuclei


The human ventralis caudalis parvocellularis is thought to be
analogous to the region referred to as the VPI nucleus in the
Intralaminar Nuclei
primate.58 Although not as well studied as the Vc nucleus, this The intralaminar nuclei have long been considered part of a
region is strongly associated with nociception, and one of its “nonspecific” medial complex that sends diffuse projections to
functions is as a relay nucleus for nociceptive signals. Primate the entirety of cerebral cortex.70 Intralaminar nuclei postulated
studies show inputs to VPI traveling via the spinothalamic tract, to be involved in nociception include the central lateral, center
arising from neurons in laminae I, IV, and V.30,59,60 The VPI differs median, and parafascicular nuclei, with the parafascicular and
from the neighboring ventral posterior complex in that the center median nuclei are sometimes considered together as
former projects primarily to the SII and insular cortex, both of the center median–parafascicular complex. Nuclear boundaries
which are involved in perception of pain.61 Although VPI is ana- appear to be well matched between species, with only few signifi-
tomically contiguous with the VPL and VPM regions, it is func- cant differences. One of the primary interspecies differences in
tionally distinct and has been linked to both the sensory and the intralaminar nuclei is in their inputs. In humans, the central
affective components of pain.62 Recording studies in both humans lateral nucleus is the only one of the three nuclei that receives
and primates have revealed numerous neurons in this region that spinothalamic projections, whereas in primates, there are addi-
respond specifically to the application of noxious stimuli.41,63,64 In tional spinothalamic connections to the center median and para-
humans, stimulation of the ventralis caudalis parvocellularis fascicular nuclei.26,27,71-73 Inputs to the central lateral nucleus arise
apparently elicits painful sensations more reliably than does stim- from deeper aspects of the spinal gray matter and laminae V and
ulation of the Vc nucleus itself.48,65 VII.25,74,75 Outputs from the intralaminar nuclei go diffusely to
the cortex, including the posterior parietal and motor cortex.
However, organized connections with striatal structures suggest
Posterior Part of the Ventral Medial Nucleus that these nuclei produce important motor responses to noxious
The VMpo nucleus is the most recently identified of the pain- inputs.76-80
related nuclei of the lateral thalamus, and it remains a topic of sig- Recordings made in intralaminar nuclei support the idea
nificant debate.21-24 First identified by Craig and colleagues60,66,67 that they are involved in processing of nociceptive information,
in primates on the basis of direct spinothalamic inputs from although their specific role remains unclear. Neurons responding
lamina I, the VMpo nucleus is in the caudal aspect of the only to stimuli of noxious intensity have been identified in
thalamus in a location that was formerly considered part of humans and primates in all three nuclei and in general have large,
the posterior complex. A similar region posteromedial to the bilateral receptive fields.81,82 Recordings from the center median
Vc nucleus has since been identified in humans.68 No analo- and parafascicular nuclei have demonstrated that a large propor-
gous site has been identified in nonprimate species. The VMpo tion of neurons responded to pinprick or noxious heat, but none
nucleus projects primarily to the insula, with additional projec- responded to nonnoxious stimuli.83,84
tions to the SI.60 Lesions of the medial thalamus have been created for intrac-
This area is of particular interest because according to current table or neuropathic pain and have produced generally positive
evidence, it appears to be a specifically nociceptive region of results. In one study of 69 patients with neurogenic pain, medial
the thalamus. In initial recordings of neurons from primate thalamotomy was found to relieve the pain for 46 (67%).84,85
VMpo nuclei, 97% of the neurons characterized were found to Stimulation of the intralaminar nuclei produces pain and thermal
respond to either thermal or noxious stimuli. Further studies sensation, among unpleasant sensations such as dyspnea and
have confirmed the presence of WDR, nociceptive-specific, and dizziness.55,86,87
e596 SECTION 6  Pain

noninvasive and invasive techniques, have shed light on the con-


Ventral Caudal Part of the Medial Dorsal Nucleus tributions that the numerous cortical regions make to the “pain
This area of medial thalamus is often grouped with the intrala- matrix.” Of importance is that the cortical representation of
minar nuclei because of similarities in afferents and physiology, longstanding chronic pain adds new layers of complexity to this
although the ventral caudal part of the medial dorsal nucleus picture.
(MDvc) has received additional attention as a nociceptive relay The role of five specific cortical areas in nociception is dis-
after it was shown to receive a spinothalamic projection from cussed here: the SI, SII, insula, ACC, and prefrontal cortex. Each
lamina I of the spinal cord.21,60 The outputs from the MDvc are of these regions adds a distinct element to the experience of pain.
argued to be primarily to the ACC and frontal lobe. On the basis The SI has been proposed to be involved in sensory and discrimi-
of these projections and the identification of nociceptive-specific native aspects of pain, whereas the SII is important in pain learn-
neurons localized to MDvc, this region is proposed to be impor- ing and memory. The ACC is involved in the affective aspects of
tant in the motivational aspects of pain. pain, and the insula is proposed to be a site of integration between
the sensory and affective aspects of pain. The prefrontal cortex,
although not implicated in the direct pain pathways, plays a role
Brainstem in higher executive functions, attention, and placebo, all of which
The anterolateral system comprises spinoreticular, spinomesen- can affect the perception of pain and the detection of noxious
cephalic, spinoparabrachial, and spinothalamic pathways; indeed, stimuli.
the brainstem projections outnumber those going directly from
the dorsal horn to the thalamus.
The spinoreticular pathway arises in deeper laminae of the
Primary Somatosensory Cortex
dorsal horn and sends projections to medial and lateral brainstem The SI is part of the anterior parietal lobe and forms the post-
core areas—including the lateral reticular nucleus, dorsal reticu- central gyrus. The role of the SI in somatosensory and mecha-
lar nucleus, gigantocellular reticular nucleus, and rostral ventro- nosensory function is extensively documented.92,93 This region is
medial medulla—and to the internal parabrachial nucleus (see thus well suited for the proposed role in nociception of discrimi-
Gauriau and Bernard9 for a review). These connections are prob- nating location and intensity, although its specific role has been
ably important in somatomotor integration of nociceptive debated.36,94,95
responses, recruitment of descending modulatory systems, and The primary thalamic input to SI is via the ventral posterior
engagement of arousal mechanisms. nuclei of the thalamus, which, as noted previously, receives both
The spinoparabrachial pathway, consisting of projections spinothalamic and lemniscal projections. In monkeys, nociceptive
from lamina I to the lateral parabrachial region, is receiving input to the SI has been demonstrated,33 and studies in rats have
increasing attention for its role in pain, especially chronic pain.9 also shown clusters of nociceptive neurons in this region of the
The majority of neurons in the lateral/external parabrachial area cortex.96 Nociceptive neurons in the SI are much rarer in pri-
are strongly activated by noxious stimulation over wide areas of mates, however, and although some reports have mentioned their
the body. The nociceptive portion of the parabrachial complex existence, mechanosensory neurons are far more common.97
projects heavily to the central nucleus of the amygdala and to the Nevertheless, subdural and other recording methods in humans
ventromedial hypothalamus. The connection through the amyg- have also shown evidence for neurons specifically activated by
dala to the extended amygdala has been implicated in emotional noxious stimuli and specific activation of the SI in pain-related
reactions to painful stimuli, and this input through the spinopara- tasks.98-100
brachial system is probably reinforced by direct projections to the A majority of imaging studies have demonstrated activation of
amygdala from deeper spinal laminae that have been demon- the SI with experimental noxious stimuli, and SI activation has
strated in both rodents and primates.88,89 The connection from been linked specifically to pain intensity.101-103 Interestingly, a
the nociceptive parabrachial complex to the hypothalamus seems sustained noxious stimulation (immersion of the hand in hot water
more likely to be related to motivated behaviors triggered by for 3 minutes under laboratory conditions) has been reported to
pain, including defensive behaviors, flight, and aggression. produce a decreased cortical signal in this region.104
Spinomesencephalic pathways terminate primarily in lateral Despite the findings of nociceptive neurons and activation of
and ventrolateral PAG matter, which are thought to be important the SI in imaging studies, stimulation of the SI predominantly
in active and passive coping strategies for dealing with escapable produces contralateral sensations of temperature, paresthesias,
and inescapable pain, respectively.90,91 These inputs presumably and other innocuous sensations; few, if any, studies have demon-
also trigger descending control mechanisms (see “Descending strated sensations of pain evoked by stimulation of the human
Modulatory Systems” section). SI.105-107 Studies of surgical and ischemic lesions in the SI and
surrounding areas have also shed light on its role in pain process-
ing. In early reports of SI damage from head trauma, hypoalgesia
CORTICAL PROCESSING and deficits in spatial and temporal discrimination of noxious
There is no specific cortical location in which creating a lesion stimuli were described.108-110 In a more recent case report of
eliminates pain. This fact gave rise to an idea that pain was stroke-related damage to the right SI, the patient was reported
uniquely primitive among sensory systems in being processed to have difficulty in localizing laser-evoked noxious stimuli on the
entirely at subcortical levels.13 However, the advent of functional left side of the body; this patient was unable to discriminate the
imaging approaches reawakened interest in the cortical represen- location of the stimuli farther than to a single limb.111 However,
tation of pain, and the notion that the cerebral cortex is unin- surgical interventions in the SI for pain control have been
volved in detection of noxious stimuli has since been discounted. reported as generally ineffective.112
It is now appreciated that no single cortical “pain center” medi-
ates all aspects of the complex sensation that is pain. Rather,
a reasonably well-defined cortical network is recruited as a
Secondary Somatosensory Cortex
result of acute noxious stimulation. Although no lesion in a single The SII is located on the parietal operculum at the superior bank
site can eliminate the perception of pain, stimulation of any of the sylvian fissure, and although the functional specifics of pain
one of many sites can elicit painful perceptions, in conjunction processing are still being described, SII has an unambiguous role
with or independent of other somatosensory sensations. Experi- in cortically mediated nociception. SII is proposed to be involved
ments involving such stimulation, along with advances in both in recognition of painful and thermal stimuli, pain-related
CHAPTER 166  Anatomy and Physiology of Pain e597

learning, and integration of tactile and nociceptive information. areas such as the SII. Depth electrodes implanted in humans
Thalamic input to the SII comes largely from the VPI.113 Indi- reveal activation of insular activity from painful stimuli with 166
vidual neurons in the SII may be nociceptive specific and tend slightly longer latency (40 to 60 msec) than similarly activated
to have large receptive fields, with contralateral or bilateral suprasylvian areas, which indicates serial rather than parallel pro-
activation.114 cessing.127 Stimulation of the insula results in a range of somato-
The SII is consistently activated in imaging studies involving sensory responses (temperature, pain, and paresthesias), as well
nociception. Its proximity to the temporal lobe has made it ame- as some nonsomatosensory responses, including pharyngolaryn-
nable to clinical research involving implantable, intracortical geal constriction and interruption of speech. With results of
electrodes in patients with temporal lobe epilepsy, and a wealth stimulating the insula, Mazzola and associates107 devised a crude
of knowledge about function has been obtained from such studies. somatosensory map with much larger areas of sensation, includ-
With laser stimuli and implantable electrodes, SII responses were ing whole body, than is seen with stimulation of the SI or SII. In
shown to have gradations with stimulation intensity, from the previous work, Ostrowsky and colleagues119 devised a somato-
lowest sensory threshold to the detection of the stimuli as painful. topic pain map that overlapped with a nonpain, somatotopic map,
Of interest, however, is that the level of SII activation did not which supported the idea of integration as a key role of the insula.
increase with increasing stimulation intensity above the threshold In patients with implanted electrodes, a laser was used as the
for detection of pain.115 In another study, Frot and associates,116 noxious stimuli to evoke responses within the insula. In contrast
using depth electrodes, showed an increased latency in activation to activation in the SII, cells in the insula did not show activation
of SII from tactile input in comparison with that from noxious until the stimulus reached the threshold for pain. Increasing
stimuli, which again supports the idea of SII involvement in intensity above threshold increased the activity of cells, which
integration and learning. In imaging studies, the SII is activated indicates that one function of the insula is to code for intensity
concurrently with or even before the SI, which indicates a paral- of painful stimuli.115 With lesions localized to the insular cortex,
lel, rather than serial, relationship between these two regions.117 some patients show an increase in pain tolerance or loss of affec-
Stimulation and lesion studies are sparse because of interpre- tive quality of pain while retaining their ability to detect intensity
tative difficulties related to location and because many ischemic and heat-pain thresholds.122
lesions extend into other areas. Only a few stimulation protocols
have focused specifically on the SII. Ostrowsky and colleagues118,119
reported that stimulation of the SII in humans did not evoke any
Anterior Cingulate Cortex
painful sensations, although this work was focused primarily on The ACC commonly is functionally divided into anterior and
the insula. In a systematic exploration of stimulation of the SII, posterior segments, and the ACC, which encompasses Brodmann
Mazzola and associates107 found the responses to be a mix of areas 24 and 32, is further divided into the midcingulate cortex
somatosensory, temperature, and pain sensations; the percentage and the ACC proper. The entire ACC has been specifically impli-
of evoked pain sensations was the same as that produced by cated in the affective and motivational aspects of pain, although
stimulation of the insula. Difficulties with location have also the region has many other functions, which include overall affect,
inhibited studies of lesions of the SII, although, in general, postle- response selection, and autonomic control. Activation of the
sion deficits in the SII support the idea of its direct involvement midcingulate cortex in response to noxious stimuli has been show
in pain processing. In a case involving a pure sensory stroke of to occur with the same latency as that of SII, which suggests that
only the SII, the patient showed contralateral restricted deficits processing of affective and sensory-discriminative processing
in light touch, pain, and temperature sensation.120 In other occurs in parallel, rather than serially. The ACC has few afferents,
cases involving SII injury, researchers have reported central and most input to the ACC comes from medial thalamic nuclei,
pain syndrome, hyperalgesia, and thermal and mechanical defi- the mediodorsal nuclei, and the parafascicular nuclei.128 In con-
cits; of note, however, is that no such changes are observed with trast, the posterior cingulate cortex receives afferents from ante-
parietal lesions that do not involve SII.121,122 Despite some imped- rior thalamic nuclei and numerous other cortical regions and, of
iments to interpreting data related to SII injury, several lines of note, also has strong interconnections with the ACC.129
research unequivocally indicate the involvement of this region in Recordings from neurons in the ACC in rabbits and rats reveal
pain processing and the integration of tactile and nociceptive nociceptive-specific neurons with whole-body receptive fields
signals. and no apparent somatotopic order or mapping.130,131 In psychi-
atric patients awaiting cingulotomy, recordings made from the
ACC show clearly pain-specific neurons that respond to contra-
Insula lateral thermal, noxious thermal, and noxious mechanical stimuli.
When viewed sagittally, the insula is a triangular region of cortex Of interest, with excitatory stimulation, even with high-current
located fully inside the sylvian sulcus. Before recent advances in stimulation at the site of a nociceptive-specific neuron, painful
imaging technology, the role of the insula in nociception was sensations were not evoked.132 Earlier findings with stimulation
unclear, although now it is considered to be a central structure in the ACC in humans was consistent with these results in that
in the “pain matrix” and is the site most commonly activated in no painful responses were elicited with stimulation, although
cortical imaging studies on pain.14 Many structural subdivisions such autonomic signs as changes in blood pressure and heart rate
of the insula have been proposed, but the system most commonly occurred.133 One explanation of the discrepancy between stimula-
used entails divisions are along the anterior-posterior axis. The tion and recording results is that ACC requires coactivation of
anterior or middle/anterior portion of the insula is often impli- other pain-related areas to interpret the ACC response as painful,
cated in processing of nociceptive information, whereas the although an experiment verifying this idea would be very difficult
posterior portion of the insula is more involved in tactile to conduct.
processing.118,119,123 Functional magnetic resonance imaging indi- Lesions and selective modulation of the affective aspect of
cates a somatotopic organization.124,125 Baliki and colleagues126 pain shed additional light onto the role of the ACC in pain
provided evidence that the insula includes circuits both specific responses. An imaging study in which the unpleasantness of a
to pain per se and more generally linked to encoding the intensity painful stimulus was manipulated through hypnosis revealed con-
or magnitude of sensory stimuli, including painful stimuli. sistent correlates of perceived unpleasantness with activation of
Recording and stimulation studies confirm the role of the only the ACC, which highlights its role in the emotional and
insula in nociception and shed light on differences in function motivational aspects of pain.134 In addition, in a study of 12
and processing between it and neighboring or functionally similar patients who underwent therapeutic cingulotomy for intractable
e598 SECTION 6  Pain

pain, pain ratings were improved only modestly, but the pain was The variability in pain experience has long been recognized
considered less bothersome or distressing. On a long-term basis, and frequently underpins a characterization of pain reports as
however, the same patients also reported significant deficits in “subjective” and therefore not to be trusted. One of the first
executive functioning and intention.135 systematic scientific discussions of the variability of pain and of
According to imaging studies of activation of the cingulate the importance of cognitive and emotional factors in pain sensa-
cortex in response to noxious stimuli, the primary site of activa- tion was advanced by Beecher,149 who quantified the pain expe-
tion is the rostral ACC or midcingulate area, although different rienced by wounded soldiers according to the amount of narcotics
patterns of activation can occur through modulation of higher they required. He noted that the pain experienced by many of
cognitive function.136-138 With anticipation of pain or the early these soldiers was much less than would have been predicted
onset of a painful stimulus, the rostral segment of the ACC is on the basis of their injuries, and he argued that the apparent
primarily activated, whereas if the stimulus is maintained, the absence of pain reflected a positive cognitive appraisal of the
caudal ACC may be activated.139 Together, these data indicate that injury, which would help remove that soldier from the war, at
the ACC is a primary cortical site of the “pain matrix,” but it is least temporarily.
involved in the emotional distress of pain and in selection of Clinicians now appreciate that cognitive and emotional
responses to painful stimuli. factors—including attention, learning, and mood—can all influ-
ence pain perception in situations nowhere nearly as extreme as
the battlefield.150,151 The brain is not a passive receiver of informa-
Prefrontal Cortex tion about noxious stimuli, and it actively regulates its own inputs.
The prefrontal cortex encompasses a large part of the frontal Indeed, the coding of afferent information is shaped dynamically
cortex just anterior to the motor cortex. Although no evidence by descending modulatory systems that influence nociceptive
exists for direct nociceptive connections, the area is mentioned processing at the first central relay, the dorsal horn. These
here because of its role in higher cognitive function and endog- descending control systems are, in turn, regulated by afferent
enous modulation of pain. The prefrontal cortex is implicated in sensory input but are also recruited by higher centers to modulate
the majority of pain-related imaging studies and is even more spinal nociceptive processing in accord with behavioral priorities.
frequently involved during chronic pain.14 The medial prefrontal In this section, we consider the organization of the descending
cortex and dorsolateral prefrontal cortex are the subdivisions modulatory systems in the central core of the brainstem, with a
more commonly activated during pain, and both areas are particular focus on the PAG/RVM system.
involved in executive function, attention, and execution of high-
order tasks. The dorsolateral prefrontal cortex is specifically
involved in the placebo response, in which pain sensations are
Descending Modulation and the PAG/RVM System
modulated by expectation.140 It has been known since the work of Sherrington152 at the begin-
ning of the 20th century that spinal nocifensive reflexes are
SENSITIZATION OF ASCENDING PAIN altered in the absence of descending influences. However, the
idea that pain modulation is a specific function of the nervous
TRANSMISSION PATHWAYS system is usually traced to a report in the late 1960s that elec-
Stimulation of injured or inflamed tissue is well known to give trical stimulation in the midbrain PAG could produce potent
rise to exaggerated pain responses, with hyperalgesia (enhanced antinociception in rats.153 These original observations were rela-
pain in response to a normally painful stimulus) and allodynia tively crude, but subsequent work confirmed the antinociceptive
(pain produced by a normally innocuous stimulus). Such responses potency of PAG stimulation in rats154 and extended it to other
can be attributed at least in part to a dynamic regulation of the species, including humans.155 This phenomenon of “stimulation-
pain transmission elements in accord with stimulus history, the produced analgesia” inspired experimental studies in a number
state of the tissue (inflamed versus normal), and immune signals. of laboratories and ultimately led to definition of a brainstem
Plasticity in nociceptive processing has been documented at every pain-modulating circuit (Fig. 166-2) with critical links in the
level of processing, so that an increased signal carried by sensi- PAG and the RVM. Electrical stimulation at either site resulted
tized primary afferents is further amplified at the dorsal horn and in antinociception, an effect mediated by inhibition of nocicep-
supraspinally. Changes at the dorsal horn are probably the best tive activity in the dorsal horn. The anatomic substrate for this
studied, and the net effect at this level is often referred to as descending control is a significant projection from the PAG to the
central sensitization because molecular, cellular, and circuit-level RVM, which in turn projects via the dorsolateral funiculus to the
changes in the dorsal horn give rise to an increase in sensitivity dorsal horn to influence sensory processing. The PAG itself sends
of nociceptive neurons, with lowered thresholds, increased only minor projections to the spinal cord and receives substantial
responsiveness, and enlarged receptive fields.141-146 More recently inputs from limbic forebrain structures, including the ACC, pre-
this concept has been extended to include supraspinal systems, frontal cortex, insula, amygdala, and hypothalamus. This infor-
and there is mounting evidence of reorganization of thalamocor- mation is integrated and transmitted to the RVM.
tical and subcortical forebrain circuits in chronic pain states, not Both the RVM and, especially, the PAG also receive significant
all of which may be reversible.147,148 nociceptive inputs from the dorsal horn, via the spinoreticular
and spinomesencephalic tracts. On the basis of the connectivity
of this system alone, the PAG/RVM system appears well situated
DESCENDING MODULATORY SYSTEMS to integrate higher order influences important in cognition and
The discussion of central nociceptive transmission pathways has emotion with afferent input from the dorsal horn. Other brain-
emphasized that afferent input is processed at the level of the stem systems, such as the dorsal reticular nucleus in the caudal
dorsal horn and transmitted over parallel ascending pathways medulla (which mediates effects of counterirritation) and the
to the brainstem, thalamus, hypothalamus, and limbic fore- noradrenergic systems in the dorsolateral pontine tegmentum are
brain. However, a recurrent theme in clinical and experimental also known to modulate nociceptive transmission, but the PAG/
approaches to pain has been that the relationship between the RVM system is the best studied and probably has the greatest
presumed stimulus—tissue damage—and the ensuing sensory effect on pain.
experience is rarely straightforward. This is true even in the sim- Interest in the PAG/RVM system was heightened when it
plest situation, in which a reasonably uniform sensory experience became clear that it was an important substrate for opioid anal-
(i.e., acute injury to healthy tissue) would be expected. gesic drugs.150,156,157 It had long been recognized that morphine
CHAPTER 166  Anatomy and Physiology of Pain e599

166
P FC
m-

C
AC
RVM
ON-cell OFF-cell
Hyp

PA
Amg

G
(+) Pronocioceptive Antinocioceptive (-)

DRG Anterolateral
C-Nociceptor
RVM Að-Nociceptor system

Pain
Figure 166-2. Brainstem pain-modulating network with links in behavior
the midbrain periaqueductal gray matter (PAG) and rostral
ventromedial medulla (RVM). The RVM, which includes the nucleus Figure 166-3. Two populations of rostral ventromedial medulla (RVM)
raphe magnus and adjacent reticular formation, receives a large input neurons, ON-cells and OFF-cells, provide the neural basis for
from the PAG. The RVM in turn projects to the dorsal horn, primarily bidirectional control of spinal processing by the RVM. DRG, dorsal
to the superficial layers and lamina V, where it can facilitate or inhibit root ganglion.
processing of nociceptive information. Significant input to the PAG
from forebrain and hypothalamus provide an anatomic substrate for
top-down control of pain. ACC, anterior cingulate cortex; Amg,
amygdala; Hyp, hypothalamus; m-PFC, medial prefrontal cortex.
hyperalgesia after inflammation, with nerve-injury pain, and with
hyperalgesia induced by chronic opioid administration (opioid-
induced hyperalgesia).161,162 The current assumption is of a modu-
and other opioid agents act primarily in the brain. Using micro- latory system, rather than an “analgesia system,” with the potential
injection mapping to delineate central sites that were directly for bidirectional control.151
sensitive to opioids, Yaksh and Rudy157 found that both the
PAG and RVM could support opioid analgesia and, in fact,
that these regions are required for the analgesic actions of sys-
Neural Basis for Bidirectional Control
temically administered opioids. This system also employs endog- Functional studies thus clearly demonstrate that, as a region, the
enous opioids as neurotransmitters, and there is evidence that RVM exerts bidirectional control of nociceptive processing, with
endogenous opioids in the PAG and RVM contribute to the pain- a net pronociceptive action under some conditions and a net
modulating function of this system. antinociceptive role in others. The neural basis for the pain-
Because the direct cellular effects of opioids are inhibitory,158 inhibiting and pain-facilitating influences from the RVM is now
it may be surprising that focal opioid application and electrical recognized to be two classes of RVM neurons, termed OFF-cells
stimulation in these regions has the same net behavioral effect: and ON-cells, respectively (Fig. 166-3).150,163 OFF-cells mediate
analgesia. However, the answer to this apparent paradox lies in the analgesic actions of µ-opioid agents and are probably recruited
the circuitry within the PAG and RVM, where the opioids act in behavioral states associated with decreased pain, such as stress-
directly on inhibitory neurons that normally inhibit the pain- induced analgesia. ON-cells are activated as part of positive-
inhibiting output neurons. Opioids thus activate descending inhi- feedback loop that amplifies pain after tissue or nerve injury and
bition through disinhibition.159 during inflammation. These neurons are also activated by higher
cognitive structures (e.g., hypothalamus) in the absence of noxious
input and could thus account for increased pain sensitivity stimu-
Bidirectional Control lated by cognitive and emotional processes, as occurs during mild
Along with the evidence that this system mediates the analgesic stress.164,165
actions of exogenous and endogenous opioids, the initial discov-
ery of the PAG/RVM circuit as the basis for stimulation-produced
analgesia led to an early view of this circuit as an “analgesia
Recruitment of the PAG/RVM Modulatory System
system.” This view is now known to be incomplete; that the The discussion of descending modulatory systems has to this
brainstem exerts bidirectional control, inhibiting or facilitating point focused on anatomic and physiologic delineation of the
pain under different conditions. The bidirectional nature of PAG/RVM circuitry and the effects of experimental manipula-
descending modulation is best studied in the RVM. Results of a tions on dorsal horn processing and nociceptive behavior. This
number of early behavioral and electrophysiologic studies sug- understanding prompts the question of how these systems come
gested that electrical stimulation in the RVM could facilitate to be activated in behaving animals. Although the information is
pain, depending on the stimulating current and exact location of in many ways still incomplete, there is now strong evidence that
the electrode.160 The implications of these observations were the balance between the pain-facilitating and pain-inhibiting
unclear until researchers found evidence that the RVM contrib- outflow from the RVM is dynamic and allows the organism to
utes to increased pain in a number of paradigms. Thus, for amplify or suppress nociceptive afferent transmission in accord
example, blocking activity in the RVM interferes with secondary with behavioral priorities.150,151
e600 SECTION 6  Pain

the most intensively studied model, with coactivation of the ACC


Activation by Noxious Inputs: A Positive-Feedback Loop and PAG during both opioid- and placebo-induced analge-
An early view of descending modulatory systems focused on the sia.140,191,192 Activation of the PAG has also been inversely corre-
phenomenon of counterirritation, in which pain at one site on lated with pain rating in paradigms associated with increased
the body suppressed inputs from other regions. A reasonable pain.193-195 Although correlational, these studies confirm the
assumption was that this remote inhibition was mediated by the recruitment of brainstem core systems in humans by cognitive
PAG/RVM system as part of a negative-feedback loop. However, and emotional variables.
negative-feedback processes triggered by noxious input are medi-
ated not by the PAG/RVM system but by the dorsal reticular
nucleus in the caudal medulla.166,167 It can be argued that this
CONCLUSION
process is important as a contrast mechanism and for integration Information about primary afferents and nociceptive signal trans-
of multiple sensory inputs with motor outputs. This negative- duction has burgeoned, but pain as a sensory experience requires
feedback process is in contrast with the PAG/RVM system, which transmission and integration of sensory input within the central
mediates an acute positive-feedback process. Discrete noxious nervous system. It is now recognized that pain reflects activation
stimuli or more prolonged inflammatory events activate ON-cells, of parallel ascending pathways that engage a distributed thalamo-
amplifying responses to subsequent inputs.168,169 cortical network, a network sometimes referred to as a “pain
matrix.” This network is not a passive receiver, and input in the
ascending pain system is dynamically regulated in accord with
Stress behavioral priorities by a descending modulatory system. Finally,
A very different approach to the function of the RVM system is there is increasing evidence that as pain becomes chronic, the
to consider descending modulation within the larger context of systems engaged shift to reflect the growing importance of con-
integrating and organizing defensive responses to noxious or ditioning and other higher order processes. Understanding and
threatening aspects of the environment.150,170,171 The phenome- manipulating these dynamic aspects of pain sensing and modula-
non of stress-induced analgesia—which occurs, for example, tion will be crucial for future advances in therapeutic approaches
upon encounters with biologically relevant threat stimuli, such as to pain.
a predator, or during major trauma—is well documented. Stress-
Acknowledgments
induced analgesia presumably functions to suppress nociception
in situations in which distress or overt behaviors that would Mary M. Heinricher’s work is supported by grants from the National
Institute of Neurological Disorder and Stroke (NINDS). We thank Andy
otherwise be evoked by a noxious stimulus might interfere with Rekito, MS, for preparation of the artwork for this chapter.
effective coping.154,170,172,173 The role of the RVM in stress-induced
analgesia is well documented, and RVM OFF-cells are recruited
in a model of fear-induced analgesia, in which these neurons are SUGGESTED READINGS
activated through the amygdala.174,175 Apkarian AV, Baliki MN, Geha PY. Towards a theory of chronic pain. Prog
Stress can also give rise to stress-induced hyperalgesia, an Neurobiol. 2009;87:81-97.
effect probably more relevant than stress-induced analgesia to Apkarian AV, Bushnell MC, Treede RD, et al. Human brain mechanisms
everyday experience and to the clinical environment. In humans, of pain perception and regulation in health and disease. Eur J Pain.
2005;9:463-484.
anxiety or anticipation of pain can be shown to enhance pain Bingel U, Tracey I. Imaging CNS modulation of pain in humans. Physiol-
sensitivity,176-180 and stress is often asserted to exacerbate chronic ogy. 2008;23:371-380.
clinical pain.181-183 Hyperalgesia mediated by activation of RVM Blomqvist A, Zhang ET, Craig AD. Cytoarchitectonic and immunohisto-
ON-cells has been demonstrated in an animal model of mild or chemical characterization of a specific pain and temperature relay, the
emotional stress.164,165 posterior portion of the ventral medial nucleus, in the human thalamus.
Brain. 2000;123:601-619.
Bushnell MC, Ceko M, Low LA. Cognitive and emotional control of
Contribution to Chronic Pain States pain and its disruption in chronic pain. Nat Rev Neurosci. 2013;14:
502-511.
Descending facilitation from the PAG/RVM system has been
Coderre TJ, Katz J, Vaccarino AL, et al. Contribution of central neuro-
shown to contribute to chronic pain states. Inactivation of the plasticity to pathological pain: review of clinical and experimental
RVM attenuates or blocks allodynia and hyperalgesia in animal evidence. Pain. 1993;52:259-285.
models of inflammation and nerve-injury pain, which indicates Craig AD. Pain mechanisms: labeled lines versus convergence in central
that this region functions as part of a positive-feedback loop trig- processing. Annu Rev Neurosci. 2003;26:1-30.
gered by injury and inflammation.161,184-187 Further evidence indi- DeLeo JA, Sorkin LS, Watkins LR, eds. Immune and Glial Regulation of
cates that recruitment and sensitization of RVM ON-cells are Pain. Seattle: International Association for Study of Pain Press;
important specifically in this positive-feedback process.161,169,188 2007:443.
RVM ON-cells can be engaged by the hypothalamus and preop- Dostrovsky JO, Craig AD. The thalamus and nociceptive processing. In:
tic areas in models of stress and illness; this observation shows Basbaum AI, Bushnell MC, eds. The Senses: A Comprehensive Reference.
Vol. 5: Pain. San Diego, CA: Academic Press; 2008:635-654.
that pain processing can be facilitated by top-down influences in Fields H. State-dependent opioid control of pain. Nat Rev Neurosci.
the absence of an initial noxious trigger. Such top-down modula- 2004;5:565-575.
tion could be relevant to functional pain disorders such as fibro- Gauriau C, Bernard JF. Pain pathways and parabrachial circuits in the rat.
myalgia or irritable bowel syndrome. Exp Physiol. 2002;87:251-258.
Heinricher MM, Fields HL. Central nervous system mechanisms of pain
modulation. In: McMahon S, Koltzenburg M, Tracey I, et al., eds. Wall
Descending Modulation in Humans: Evidence from and Melzack’s Textbook of Pain. 6th ed. London: Elsevier; 2013:129-142.
Imaging Studies Heinricher MM, Ingram SL. The brainstem and nociceptive modulation.
In: Basbaum AI, Bushnell MC, eds. The Senses: A Comprehensive Refer-
The existence of the brainstem modulatory system in humans was
ence. Vol 5: Pain. San Diego, CA: Academic Press; 2008:593-626.
first demonstrated by the analgesic action of deep brain stimula- Heinricher MM, Tavares I, Leith JL, et al. Descending control of noci-
tion in the PAG/periventricular gray area in some forms of clini- ception: specificity, recruitment and plasticity. Brain Res Rev. 2009;60:
cal pain.155 Functional imaging approaches have increased the 214-225.
understanding of when brainstem pain-modulating systems are McMahon SB, Lewin GR, Wall PD. Central hyperexcitability triggered
recruited in humans.103,189,190 Placebo analgesia has probably been by noxious inputs. Curr Opin Neurobiol. 1993;3:602-610.
CHAPTER 166  Anatomy and Physiology of Pain e601

Porreca F, Ossipov MH, Gebhart GF. Chronic pain and medullary 23. Jones EG. A pain in the thalamus. J Pain. 2002;3(2):102-104.
descending facilitation. Trends Neurosci. 2002;25:319-325. 24. Graziano A, Jones EG. Widespread thalamic terminations of fibers 166
Ren K, Dubner R. Central nervous system plasticity and persistent pain. arising in the superficial medullary dorsal horn of monkeys and
J Orofac Pain. 1999;13:155-163. their relation to calbindin immunoreactivity. J Neurosci. 2004;24(1):
Todd AJ. Neuronal circuitry for pain processing in the dorsal horn. Nat 248-256.
Rev Neurosci. 2010;11:823-836. 25. Applebaum AE, Leonard RB, Kenshalo DR Jr, et al. Nuclei in which
Tracey I, Mantyh PW. The cerebral signature for pain perception and its functionally identified spinothalamic tract neurons terminate.
modulation. Neuron. 2007;55:377-391. J Comp Neurol. 1979;188:575-585.
Treede RD, Apkarian AV. Nociceptive processing in the cerebral cortex. 26. Apkarian AV, Hodge CJ. Primate spinothalamic pathways: III. Tha-
In: Basbaum AI, Bushnell MC, eds. The Senses: A Comprehensive Refer- lamic terminations of the dorsolateral and ventral spinothalamic
ence. Vol. 5: Pain. San Diego, CA: Academic Press; 2008:669-698. pathways. J Comp Neurol. 1989;288(3):493-511.
Willis WD, ed. Hyperalgesia and Allodynia. New York: Raven Press; 1992. 27. Mehler WR. The anatomy of the so-called “pain tract” in man: an
Willis WD, Westlund KN. Dorsal columns and visceral pain. In: Basbaum analysis of the course and distribution of the ascending fibers of the
AI, Bushnell MC, eds. The Senses: A Comprehensive Reference. Vol 5: Pain. fasciculus anterolateralis. In: French JD, Porter RW, eds. Basic
San Diego, CA: Academic Press; 2008:527-542. Research in Paraplegia. Springfield, IL: Charles C Thomas; 1962.
28. Kaas JH, Nelson RJ, Sur M, et al. The somatotopic organization of
the ventroposterior thalamus of the squirrel monkey, Saimiri sciu-
REFERENCES reus. J Comp Neurol. 1984;226(1):111-140.
1. Willis WD, Coggleshall RE. Sensory Mechanisms of the Spinal Cord. 29. Jones EG. Correlation and revised nomenclature of ventral nuclei
New York: Plenum Press; 1991. in the thalamus of human and monkey. Stereotact Funct Neurosurg.
2. Todd AJ. Neuronal circuitry for pain processing in the dorsal horn. 1990;54-55:1-20.
Nat Rev Neurosci. 2010;11(12):823-836. 30. Mehler WR. The posterior thalamic region in man. Confin Neurol.
3. Dubner R, Bennett GJ. Spinal and trigeminal mechanisms of noci- 1966;27(1):18-29.
ception. Annu Rev Neurosci. 1983;6:381-418. 31. Lenz FA, Dostrovsky JO, Tasker RR, et al. Single-unit analysis of
4. Price DD, Dubner R. Neurons that subserve the sensory- the human ventral thalamic nuclear group: somatosensory responses.
discriminative aspects of pain. Pain. 1977;3:307-338. J Neurophysiol. 1988;59(2):299-316.
5. Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treat- 32. Willis WD, Westlund KN. Neuroanatomy of the pain system and
ment of nonmalignant pain: a structured literature review. J Neuro- of the pathways that modulate pain. J Clin Neurophysiol. 1997;
surg. 2008;109(3):389-404. 14(1):2-31.
6. Raslan AM, Cetas JS, McCartney S, et al. Destructive procedures 33. Gingold SI, Greenspan JD, Apkarian AV. Anatomic evidence of
for control of cancer pain: the case for cordotomy. J Neurosurg. nociceptive inputs to primary somatosensory cortex: relationship
2011;114(1):155-170. between spinothalamic terminals and thalamocortical cells in squir-
7. Bushnell MC. Thalamic processing of sensory-discriminative and rel monkeys. J Comp Neurol. 1991;308(3):467-490.
affective-motivational dimensions of pain. In: Besson JM, Guilbaud 34. Kenshalo DR Jr, Giesler GJ Jr, Leonard RB, et al. Responses of
G, Ollat H, eds. Forebrain Areas Involved in Pain Processing. Paris: neurons in primate ventral posterior lateral nucleus to noxious
John Libbey Eurotext; 1985. stimuli. J Neurophysiol. 1980;43(6):1594-1614.
8. Melzack R, Casey KL. Sensory, motivational, and central control 35. Burton H, Carlson M. Second somatic sensory cortical area (SII) in
determinants of pain. In: Kenshalo DR, ed. The Skin Senses. Spring- a prosimian primate, Galago crassicaudatus. J Comp Neurol. 1986;
field, IL: Charles C Thomas; 1968:423-443. 247(2):200-220.
9. Gauriau C, Bernard JF. Pain pathways and parabrachial circuits in 36. Kenshalo DR Jr, Willis WD. The Role of the Cerebral Cortex in Pain
the rat. Exp Physiol. 2002;87(2):251-258. Sensation. New York: Plenum Press; 1991.
10. Willis WD, Westlund KN. The role of the dorsal column pathway 37. Burton H, Jones EG. The posterior thalamic region and its cortical
in visceral nociception. Curr Pain Headache Rep. 2001;5(1):20-26. projection in New World and Old World monkeys. J Comp Neurol.
11. Palecek J, Paleckova V, Willis WD. Postsynaptic dorsal column 1976;168(2):249-301.
neurons express NK1 receptors following colon inflammation. Neu- 38. Jones EG, Burton H. Areal differences in the laminar distribution
roscience. 2003;116(2):565-572. of thalamic afferents in cortical fields of the insular, parietal
12. Palecek J. The role of dorsal columns pathway in visceral pain. and temporal regions of primates. J Comp Neurol. 1976;168(2):
Physiol Res. 2004;53(suppl 1):S125-S130. 197-247.
13. Head H, Holmes G. Sensory disturbances from cerebral lesions. 39. Lenz FA, Dougherty PM. Neurons in the human thalamic somato-
Brain. 1911;34:102-254. sensory nucleus (ventralis caudalis) respond to innocuous cool and
14. Apkarian AV, Bushnell MC, Treede RD, et al. Human brain mecha- mechanical stimuli. J Neurophysiol. 1998;79(4):2227-2230.
nisms of pain perception and regulation in health and disease. Eur 40. Willis WD. Nociceptive pathways: anatomy and physiology of noci-
J Pain. 2005;9(4):463-484. ceptive ascending pathways. Philos Trans R Soc Lond B Biol Sci.
15. Ralston HJ 3rd. Pain and the primate thalamus. Prog Brain Res. 1985;308(1136):253-270.
2005;149:1-10. 41. Apkarian AV, Shi T. Squirrel monkey lateral thalamus. I. Somatic
16. Dostrovsky JO, Craig AD. The thalamus and nociceptive processing. nociresponsive neurons and their relation to spinothalamic termi-
In: Basbaum AI, Bushnell MC, eds. The Senses: A Comprehensive Ref- nals. J Neurosci. 1994;14(11 Pt 2):6779-6795.
erence. Vol. 5: Pain. San Diego, CA: Academic Press; 2008:635-654. 42. Bushnell MC, Duncan GH, Tremblay N. Thalamic VPM nucleus
17. Willis WD Jr. Pain pathways in the primate. Prog Clin Biol Res. in the behaving monkey. I. Multimodal and discriminative proper-
1985;176:117-133. ties of thermosensitive neurons. J Neurophysiol. 1993;69:739-752.
18. Iannetti GD, Mouraux A. From the neuromatrix to the pain matrix 43. Lee J, Dougherty PM, Antezana D, et al. Responses of neurons in
(and back). Exp Brain Res. 2010;205(1):1-12. the region of human thalamic principal somatic sensory nucleus to
19. Legrain V, Iannetti GD, Plaghki L, et al. The pain matrix reloaded: mechanical and thermal stimuli graded into the painful range.
a salience detection system for the body. Prog Neurobiol. 2011; J Comp Neurol. 1999;410(4):541-555.
93(1):111-124. 44. Lenz FA, Garonzik IM, Zirh TA, et al. Neuronal activity in the
20. Willis WD, Zhang X, Honda CN, et al. Projections from the mar- region of the thalamic principal sensory nucleus (ventralis caudalis)
ginal zone and deep dorsal horn to the ventrobasal nuclei of the in patients with pain following amputations. Neuroscience. 1998;86(4):
primate thalamus. Pain. 2001;92(1–2):267-276. 1065-1081.
21. Willis WD Jr, Zhang X, Honda CN, et al. A critical review of 45. Lenz FA, Gracely RH, Rowland LH, et al. A population of cells in
the role of the proposed VMpo nucleus in pain. J Pain. 2002;3(2): the human thalamic principal sensory nucleus respond to painful
79-94. mechanical stimuli. Neurosci Lett. 1994;180:46-50.
22. Jones EG, Lensky KM, Chan VH. Delineation of thalamic nuclei 46. Bruggemann J, Shi T, Apkarian AV. Squirrel monkey lateral thala-
immunoreactive for calcium-binding proteins in and around the mus. II. Viscerosomatic convergent representation of urinary
posterior pole of the ventral posterior complex. Thalamus & Related bladder, colon, and esophagus. J Neurosci. 1994;14(11 Pt 2):
Systems. 2001;1(3):213-224. 6796-6814.
e602 SECTION 6  Pain

47. Chandler MJ, Hobbs SF, Fu QG, et al. Responses of neurons in 72. Bowsher D. Some afferent and efferent connections of the
ventroposterolateral nucleus of primate thalamus to urinary bladder parafascicular-center median complex. In: Purpura D, Yahr M, eds.
distension. Brain Res. 1992;571:26-34. The Thalamus. Columbia University Press; 1966:99-108.
48. Lenz FA, Seike M, Richardson RT, et al. Thermal and pain sensa- 73. Kerr FW. The ventral spinothalamic tract and other ascending
tions evoked by microstimulation in the area of human ventrocaudal systems of the ventral funiculus of the spinal cord. J Comp Neurol.
nucleus. J Neurophysiol. 1993;70(1):200-212. 1975;159(3):335-356.
49. Davis KD, Kiss ZH, Tasker RR, et al. Thalamic stimulation-evoked 74. Craig AD Jr, Linington AJ, Kniffki KD. Cells of origin of spinotha-
sensations in chronic pain patients and in nonpain (movement dis- lamic tract projections to the medial and lateral thalamus in the cat.
order) patients. J Neurophysiol. 1996;75(3):1026-1037. J Comp Neurol. 1989;289(4):568-585.
50. Bagley CA, Ohara S, Lawson HC, et al. Psychophysics of CNS 75. Giesler GJ Jr, Yezierski RP, Gerhart KD, et al. Spinothalamic tract
pain-related activity: binary and analog channels and memory neurons that project to medial and/or lateral thalamic nuclei: evi-
encoding. Neuroscientist. 2006;12(1):29-42. dence for a physiologically novel population of spinal cord neurons.
51. Lenz FA, Ohara S, Gracely RH, et al. Pain encoding in the human J Neurophysiol. 1981;46:1285-1308.
forebrain: binary and analog exteroceptive channels. J Neurosci. 76. Sadikot AF, Parent A, Francois C. The centre median and parafas-
2004;24(29):6540-6544. cicular thalamic nuclei project respectively to the sensorimotor and
52. Duncan GH, Bushnell MC, Oliveras JL, et al. Thalamic VPM associative-limbic striatal territories in the squirrel monkey. Brain
nucleus in the behaving monkey. III. Effects of reversible inactiva- Res. 1990;510(1):161-165.
tion by lidocaine on thermal and mechanical discrimination. J Neu- 77. Casey KL, Minoshima S, Morrow TJ, et al. Comparison of human
rophysiol. 1993;70:2086-2096. cerebral activation pattern during cutaneous warmth, heat pain, and
53. Albe-Fessard D, Dondey M, Nicolaidis S, et al. Remarks concerning deep cold pain. J Neurophysiol. 1996;76(1):571-581.
the effect of diencephalic lesions on pain and sensitivity with special 78. Sadikot AF, Parent A, Francois C. Efferent connections of the cen-
reference to lemniscally mediated control of noxious afferences. tromedian and parafascicular thalamic nuclei in the squirrel monkey:
Confin Neurol. 1970;32(2):174-184. a PHA-L study of subcortical projections. J Comp Neurol. 1992;
54. Mark VH, Ervin FR, Yakovlev PI. Correlation of pain relief, sensory 315(2):137-159.
loss, and anatomical lesion sites in pain patients treated with stereo- 79. Powell TP, Cowan WM. The interpretation of the degenerative
tactic thalamotomy. Trans Am Neurol Assoc. 1961;86:86-90. changes in the intralaminar nuclei of the thalamus. J Neurol Neuro-
55. Richardson DE. Thalamotomy for intractable pain. Confin Neurol. surg Psychiatry. 1967;30(2):140-153.
1967;29(2):139-145. 80. Groenewegen HJ, Berendse HW. The specificity of the “nonspe-
56. Tasker RR, Kiss ZH. The role of the thalamus in functional neuro- cific” midline and intralaminar thalamic nuclei. Trends Neurosci.
surgery. Neurosurg Clin N Am. 1995;6(1):73-104. 1994;17:52-57.
57. Kim JH, Greenspan JD, Coghill RC, et al. Lesions limited to the 81. Albe-Fessard D, Berkley KJ, Kruger L, et al. Diencephalic mecha-
human thalamic principal somatosensory nucleus (ventral caudal) nisms of pain sensation. Brain Res. 1985;356(3):217-296.
are associated with loss of cold sensations and central pain. J Neu- 82. Bushnell MC, Duncan GH. Sensory and affective aspects of pain
rosci. 2007;27(18):4995-5004. perception: is medial thalamus restricted to emotional issues? Exp
58. Treede RD. Spinothalamic and thalamocortical nociceptive path- Brain Res. 1989;78:415-418.
ways. J Pain. 2002;3(2):109-112. 83. Ishijima B, Yoshimasu N, Fukushima T, et al. Nociceptive neurons
59. Ralston HJ 3rd, Ralston DD. The primate dorsal spinothalamic in the human thalamus. Confin Neurol. 1975;37(1–3):99-106.
tract: evidence for a specific termination in the posterior nuclei (Po/ 84. Jeanmonod D, Magnin M, Morel A. Thalamus and neurogenic pain:
SG) of the thalamus. Pain. 1992;48(1):107-118. physiological, anatomical and clinical data. Neuroreport. 1993;4(5):
60. Craig AD, Bushnell MC, Zhang ET, et al. A thalamic nucleus spe- 475-478.
cific for pain and temperature sensation. Nature. 1994;372(6508): 85. Tsubokawa T, Moriyasu N. Follow-up results of centre median
770-773. thalamotomy for relief of intractable pain. A method of evaluating
61. Friedman DP, Murray EA. Thalamic connectivity of the second the effectiveness during operation. Confin Neurol. 1975;37(1–3):
somatosensory area and neighboring somatosensory fields of 280-284.
the lateral sulcus of the macaque. J Comp Neurol. 1986;252(3): 86. Richardson DE. Thalamotomy for control of chronic pain. Acta
348-373. Neurochir (Wien). 1974;(suppl 21):77-88.
62. Millan MJ. The induction of pain: an integrative review. Prog Neu- 87. Sano K. Intralaminar thalamotomy (thalamolaminotomy) and
robiol. 1999;57(1):1-164. postero-medial hypothalamotomy in the treatment of intractable
63. Pollin B, Albe-Fessard D. Organization of somatic thalamus in pain. Progress in Neurology and Surgery. 1977;8:50-103.
monkeys with and without section of dorsal spinal tracts. Brain Res. 88. Newman HM, Stevens RT, Apkarian AV. Direct spinal projections
1979;173(3):431-449. to limbic and striatal areas: anterograde transport studies from the
64. Casey KL, Morrow TJ. Nociceptive neurons in the ventral posterior upper cervical spinal cord and the cervical enlargement in squirrel
thalamus of the awake squirrel monkey: observations on identifica- monkey and rat. J Comp Neurol. 1996;365(4):640-658.
tion, modulation, and drug effects. In: Besson JM, Guilbaud G, 89. Burstein R, Potrebic S. Retrograde labeling of neurons in the spinal
Peschanski M, eds. Thalamus and Pain. Amsterdam: Elsevier; 1987. cord that project directly to the amygdala or the orbital cortex in
65. Halliday AM, Logue V. Painful sensations evoked by electrical the rat. J Comp Neurol. 1993;335:469-485.
stimulation in the thalamus. In: Somjen GG, ed. Neurophysiology 90. Keay KA, Feil K, Gordon BD, et al. Spinal afferents to functionally
studied in man. Amsterdam: Excerpta Medica; 1972:221-230. distinct periaqueductal gray columns in the rat: an anterograde and
66. Craig AD. Distribution of brainstem projections from spinal lamina retrograde tracing study. J Comp Neurol. 1997;385(2):207-229.
I neurons in the cat and the monkey. J Comp Neurol. 1995;361(2): 91. Lumb BM. Inescapable and escapable pain is represented in distinct
225-248. hypothalamic-midbrain circuits: specific roles for Adelta- and
67. Han ZS, Zhang ET, Craig AD. Nociceptive and thermoreceptive C-nociceptors. Exp Physiol. 2002;87(2):281-286.
lamina I neurons are anatomically distinct. Nat Neurosci. 1998;1(3): 92. Kaas JH, Nelson RJ, Sur M, et al. Multiple representations of the
218-225. body within the primary somatosensory cortex of primates. Science.
68. Blomqvist A, Zhang ET, Craig AD. Cytoarchitectonic and immu- 1979;204(4392):521-523.
nohistochemical characterization of a specific pain and temperature 93. Ploner M, Schmitz F, Freund HJ, et al. Differential organization of
relay, the posterior portion of the ventral medial nucleus, in the touch and pain in human primary somatosensory cortex. J Neuro-
human thalamus. Brain. 2000;123(Pt 3):601-619. physiol. 2000;83(3):1770-1776.
69. Davis KD, Lozano RM, Manduch M, et al. Thalamic relay site for 94. Bushnell MC, Duncan GH, Hofbauer RK, et al. Pain perception:
cold perception in humans. J Neurophysiol. 1999;81(4):1970-1973. is there a role for primary somatosensory cortex? Proc Natl Acad Sci
70. Jones EG. A new view of specific and nonspecific thalamocortical U S A. 1999;96(14):7705-7709.
connections. Adv Neurol. 1998;77:49-71. 95. Treede RD, Kenshalo DR, Gracely RH, et al. The cortical repre-
71. Apkarian AV, Hodge CJ. Primate spinothalamic pathways: I. A sentation of pain. Pain. 1999;79(2–3):105-111.
quantitative study of the cells of origin of the spinothalamic pathway. 96. Lamour Y, Willer JC, Guilbaud G. Rat somatosensory (SmI) cortex:
J Comp Neurol. 1989;288(3):447-473. I. Characteristics of neuronal responses to noxious stimulation and
CHAPTER 166  Anatomy and Physiology of Pain e603

comparison with responses to non-noxious stimulation. Exp Brain 122. Greenspan JD, Lee RR, Lenz FA. Pain sensitivity alterations as a
Res. 1983;49:35-45. function of lesion location in the parasylvian cortex. Pain. 1999; 166
97. Kenshalo DR Jr, Isensee O. Responses of primate SI cortical 81(3):273-282.
neurons to noxious stimuli. J Neurophysiol. 1983;50(6):1479-1496. 123. Treede RD, Apkarian AV, Bromm B, et al. Cortical representation
98. Kanda M, Nagamine T, Ikeda A, et al. Primary somatosensory of pain: functional characterization of nociceptive areas near the
cortex is actively involved in pain processing in human. Brain Res. lateral sulcus. Pain. 2000;87(2):113-119.
2000;853(2):282-289. 124. Brooks JC, Zambreanu L, Godinez A, et al. Somatotopic organisa-
99. Timmermann L, Ploner M, Haucke K, et al. Differential coding of tion of the human insula to painful heat studied with high resolution
pain intensity in the human primary and secondary somatosensory functional imaging. Neuroimage. 2005;27(1):201-209.
cortex. J Neurophysiol. 2001;86(3):1499-1503. 125. Henderson LA, Gandevia SC, Macefield VG. Somatotopic organi-
100. Ohara S, Crone NE, Weiss N, et al. Amplitudes of laser evoked zation of the processing of muscle and cutaneous pain in the
potential recorded from primary somatosensory, parasylvian and left and right insula cortex: a single-trial fMRI study. Pain. 2007;
medial frontal cortex are graded with stimulus intensity. Pain. 128(1–2):20-30.
2004;110(1–2):318-328. 126. Baliki MN, Geha PY, Apkarian AV, et al. Beyond feeling: chronic
101. Rainville P, Carrier B, Hofbauer RK, et al. Dissociation of sensory pain hurts the brain, disrupting the default-mode network dynam-
and affective dimensions of pain using hypnotic modulation. Pain. ics. J Neurosci. 2008;28(6):1398-1403.
1999;82(2):159-171. 127. Frot M, Mauguiere F. Dual representation of pain in the operculo-
102. Tracey I. Nociceptive processing in the human brain. Curr Opin insular cortex in humans. Brain. 2003;126(Pt 2):438-450.
Neurobiol. 2005;15(4):478-487. 128. Vogt BA, Nimchinsky EA, Vogt LJ, et al. Human cingulate cortex:
103. Tracey I, Mantyh PW. The cerebral signature for pain perception surface features, flat maps, and cytoarchitecture. J Comp Neurol.
and its modulation. Neuron. 2007;55(3):377-391. 1995;359(3):490-506.
104. Apkarian AV, Stea RA, Manglos SH, et al. Persistent pain inhibits 129. Vogt BA, Rosene DL, Pandya DN. Thalamic and cortical afferents
contralateral somatosensory cortical activity in humans. Neurosci differentiate anterior from posterior cingulate cortex in the monkey.
Lett. 1992;140(2):141-147. Science. 1979;204(4389):205-207.
105. Penfield WB. E. Somatic motor and sensory representation in the 130. Sikes RW, Vogt BA. Nociceptive neurons in area 24 of rabbit cin-
cerebral cortex of man as studied by electrical stimulation. Brain. gulate cortex. J Neurophysiol. 1992;68(5):1720-1732.
1937;60:389-443. 131. Yamamura H, Iwata K, Tsuboi Y, et al. Morphological and electro-
106. Coghill RC, Talbot JD, Evans AC, et al. Distributed processing physiological properties of ACCx nociceptive neurons in rats. Brain
of pain and vibration by the human brain. J Neurosci. 1994;14: Res. 1996;735(1):83-92.
4095-4108. 132. Hutchison WD, Davis KD, Lozano AM, et al. Pain-related neurons
107. Mazzola L, Isnard J, Mauguiere F. Somatosensory and pain responses in the human cingulate cortex. Nat Neurosci. 1999;2(5):403-405.
to stimulation of the second somatosensory area (SII) in humans. A 133. Talairach J, Bancaud J, Geier S, et al. The cingulate gyrus and
comparison with SI and insular responses. Cereb Cortex. 2006;16(7): human behaviour. Electroencephalogr Clin Neurophysiol. 1973;34(1):
960-968. 45-52.
108. Russell W. Transient disturbances following gunshot wounds of the 134. Rainville P, Duncan GH, Price DD, et al. Pain affect encoded in
head. Brain. 1945;68:79-97. human anterior cingulate but not somatosensory cortex. Science.
109. Marshall J. Sensory disturbances in cortical wounds with special 1997;277(5328):968-971.
reference to pain. J Neurol Neurosurg Psychiatry. 1951;14(3): 135. Cohen RA, Kaplan RF, Moser DJ, et al. Impairments of attention
187-204. after cingulotomy. Neurology. 1999;53(4):819-824.
110. Bassetti C, Bogousslavsky J, Regli F. Sensory syndromes in parietal 136. Casey KL. Forebrain mechanisms of nociception and pain:
stroke. Neurology. 1993;43(10):1942-1949. analysis through imaging. Proc Natl Acad Sci U S A. 1999;96(14):
111. Ploner M, Freund HJ, Schnitzler A. Pain affect without pain sensa- 7668-7674.
tion in a patient with a postcentral lesion. Pain. 1999;81(1–2): 137. Derbyshire SW. Exploring the pain “neuromatrix.”. Curr Rev Pain.
211-214. 2000;4(6):467-477.
112. Sweet W. Cerebral localization of pain. In: Thompson RaG JR, ed. 138. Derbyshire SW. A systematic review of neuroimaging data during
New Perspectives in Cerebral Localization. New York: Raven Press; visceral stimulation. Am J Gastroenterol. 2003;98(1):12-20.
1982:205-242. 139. Casey KL, Morrow TJ, Lorenz J, et al. Temporal and spatial dynam-
113. Krubitzer L, Clarey J, Tweedale R, et al. A redefinition of somato- ics of human forebrain activity during heat pain: analysis by positron
sensory areas in the lateral sulcus of macaque monkeys. J Neurosci. emission tomography. J Neurophysiol. 2001;85(2):951-959.
1995;15(5 Pt 2):3821-3839. 140. Wager TD, Rilling JK, Smith EE, et al. Placebo-induced changes
114. Disbrow E, Roberts T, Krubitzer L. Somatotopic organization of in FMRI in the anticipation and experience of pain. Science.
cortical fields in the lateral sulcus of Homo sapiens: evidence for 2004;303(5661):1162-1167.
SII and PV. J Comp Neurol. 2000;418(1):1-21. 141. McMahon SB, Lewin GR, Wall PD. Central hyperexcitabil-
115. Frot M, Magnin M, Mauguiere F, et al. Human SII and posterior ity triggered by noxious inputs. Curr Opin Neurobiol. 1993;3:
insula differently encode thermal laser stimuli. Cereb Cortex. 2007; 602-610.
17(3):610-620. 142. Herrero JF, Laird JM, Lopez-Garcia JA. Wind-up of spinal cord
116. Frot M, Garcia-Larrea L, Guenot M, et al. Responses of the neurones and pain sensation: much ado about something? Prog
supra-sylvian (SII) cortex in humans to painful and innocuous Neurobiol. 2000;61(2):169-203.
stimuli. A study using intra-cerebral recordings. Pain. 2001;94(1): 143. Coderre TJ, Katz J, Vaccarino AL, et al. Contribution of central
65-73. neuroplasticity to pathological pain: review of clinical and experi-
117. Ploner M, Schmitz F, Freund HJ, et al. Parallel activation of primary mental evidence. Pain. 1993;52:259-285.
and secondary somatosensory cortices in human pain processing. 144. DeLeo JA, Sorkin LS, Watkins LR, eds. Immune and Glial Regulation
J Neurophysiol. 1999;81(6):3100-3104. of Pain. Seattle: IASP Press; 2007.
118. Ostrowsky K, Isnard J, Ryvlin P, et al. Functional mapping of the 145. Ren K, Dubner R. Central nervous system plasticity and persistent
insular cortex: clinical implication in temporal lobe epilepsy. Epilep- pain. J Orofac Pain. 1999;13(3):155-163.
sia. 2000;41(6):681-686. 146. Willis WD, ed. Hyperalgesia and Allodynia. New York: Raven Press;
119. Ostrowsky K, Magnin M, Ryvlin P, et al. Representation of pain and 1992.
somatic sensation in the human insula: a study of responses to direct 147. Mansour AR, Baliki MN, Huang L, et al. Brain white matter struc-
electrical cortical stimulation. Cereb Cortex. 2002;12(4):376-385. tural properties predict transition to chronic pain. Pain. 2013;
120. Horiuchi T, Unoki T, Yokoh A, et al. Pure sensory stroke caused by 154(10):2160-2168.
cortical infarction associated with the secondary somatosensory 148. Mansour AR, Farmer MA, Baliki MN, et al. Chronic pain: the role
area. J Neurol Neurosurg Psychiatry. 1996;60(5):588-589. of learning and brain plasticity. Restor Neurol Neurosci. 2014;32(1):
121. Schmahmann JD, Leifer D. Parietal pseudothalamic pain syndrome. 129-139.
Clinical features and anatomic correlates. Arch Neurol. 1992;49(10): 149. Beecher HK. Relationship of significance of wound to pain experi-
1032-1037. enced. J Am Med Assoc. 1959;161:1609-1613.
e604 SECTION 6  Pain

150. Heinricher MM, Ingram SL. The brainstem and nociceptive modu- 174. Helmstetter FJ, Tershner SA, Poore LH, et al. Antinociception fol-
lation. In: Bushnell MC, Basbaum AI, eds. The Science of Pain. San lowing opioid stimulation of the basolateral amygdala is expressed
Diego, CA: Academic Press; 2008:593-626. through the periaqueductal gray and rostral ventromedial medulla.
151. Heinricher MM, Fields HL. Central nervous system mechanisms Brain Res. 1998;779(1–2):104-118.
of pain modulation. In: McMahon S, Koltzenburg M, Tracey I, 175. McGaraughty S, Heinricher MM. Microinjection of morphine
et al., eds. Wall and Melzack’s Textbook of Pain. 6th ed. London: into various amygdaloid nuclei differentially affects nociceptive
Elsevier; 2013:129-142. responsiveness and RVM neuronal activity. Pain. 2002;96(1–2):
152. Sherrington CS. The Integrative Action of the Nervous System. New 153-162.
Haven, CT: Yale University Press; 1906. 176. al Absi M, Rokke PD. Can anxiety help us tolerate pain? Pain.
153. Reynolds DV. Surgery in the rat during electrical analgesia induced 1991;46(1):43-51.
by focal brain stimulation. Science. 1969;154:444-445. 177. Rhudy JL, Meagher MW. Fear and anxiety: divergent effects on
154. Watkins LR, Mayer DJ. Organization of endogenous opiate and human pain thresholds. Pain. 2000;84(1):65-75.
nonopiate pain control systems. Science. 1982;216:1185-1192. 178. Benedetti F, Amanzio M, Vighetti S, et al. The biochemical and
155. Barbaro NM. Studies of PAG/PVG stimulation for pain relief in neuroendocrine bases of the hyperalgesic nocebo effect. J Neurosci.
humans. Prog Brain Res. 1988;77:165-173. 2006;26(46):12014-12022.
156. Yaksh TL, Rudy TA. Narcotic analgestics: CNS sites and mecha- 179. Rhudy JL, Dubbert PM, Parker JD, et al. Affective modulation
nisms of action as revealed by intracerebral injection techniques. of pain in substance-dependent veterans. Pain Med. 2006;7(6):
Pain. 1978;4:299-359. 483-500.
157. Yaksh TL, Al-Rodhan NRF, Jensen TS. Sites of action of opiates in 180. Thompson T, Keogh E, French CC, et al. Anxiety sensitivity and
production of analgesia. Prog Brain Res. 1988;77:371-394. pain: generalisability across noxious stimuli. Pain. 2008;134(1–2):
158. Duggan AW, North RA. Electrophysiology of opioids. Pharmacol 187-196.
Rev. 1983;35:219-281. 181. Vierck CJ Jr. Mechanisms underlying development of spatially dis-
159. Lau BK, Vaughan CW. Descending modulation of pain: the GABA tributed chronic pain (fibromyalgia). Pain. 2006;124(3):242-263.
disinhibition hypothesis of analgesia. Curr Opin Neurobiol. 2014; 182. Leistad RB, Nilsen KB, Stovner LJ, et al. Similarities in stress physi-
29C:159-164. ology among patients with chronic pain and headache disorders:
160. Gebhart GF. Descending modulation of pain. Neurosci Biobehavioral evidence for a common pathophysiological mechanism? J Headache
Rev. 2004;27(8):729-737. Pain. 2008;9(3):165-175.
161. Porreca F, Ossipov MH, Gebhart GF. Chronic pain and medullary 183. Chapman CR, Tuckett RP, Song CW. Pain and stress in a systems
descending facilitation. Trends Neurosci. 2002;25(6):319-325. perspective: reciprocal neural, endocrine, and immune interactions.
162. Ossipov MH, Lai J, King T, et al. Antinociceptive and nociceptive J Pain. 2008;9(2):122-145.
actions of opioids. J Neurobiol. 2004;61(1):126-148. 184. Urban MO, Gebhart GF. Supraspinal contributions to hyperalgesia.
163. Fields HL, Heinricher MM. Anatomy and physiology of a nocicep- Proc Natl Acad Sci U S A. 1999;96(14):7687-7692.
tive modulatory system. Philos Trans R Soc Lond B Biol Sci. 1985; 185. Suzuki R, Dickenson A. Spinal and supraspinal contributions to
308:361-374. central sensitization in peripheral neuropathy. Neurosignals. 2005;
164. Martenson ME, Cetas JS, Heinricher MM. A possible neural basis 14(4):175-181.
for stress-induced hyperalgesia. Pain. 2009;142(3):236-244. 186. Ren K, Dubner R. Descending modulation in persistent pain: an
165. Wagner KM, Roeder Z, Desrochers K, et al. The dorsomedial update. Pain. 2002;100(1–2):1-6.
hypothalamus mediates stress-induced hyperalgesia and is the 187. Heinricher MM, Tavares I, Leith JL, et al. Descending control of
source of the pronociceptive peptide cholecystokinin in the rostral nociception: specificity, recruitment and plasticity. Brain Res Rev.
ventromedial medulla. Neuroscience. 2013;238:29-38. 2009;60:214-225.
166. Le Bars D. The whole body receptive field of dorsal horn multire- 188. Carlson JD, Maire JJ, Martenson ME, et al. Sensitization of pain-
ceptive neurones. Brain Res Rev. 2002;40(1–3):29-44. modulating neurons in the rostral ventromedial medulla after
167. Villanueva L, Bouhassira D, Le Bars D. The medullary subnucleus peripheral nerve injury. J Neurosci. 2007;27(48):13222-13231.
reticularis dorsalis (SRD) as a key link in both the transmission and 189. Bingel U, Tracey I. Imaging CNS modulation of pain in humans.
modulation of pain signals. Pain. 1996;67:231-240. Physiology. 2008;23:371-380.
168. Ramirez F, Vanegas H. Tooth pulp stimulation advances both med- 190. Bushnell MC, Ceko M, Low LA. Cognitive and emotional control
ullary off-cell pause and tail flick. Neurosci Lett. 1989;100:153-156. of pain and its disruption in chronic pain. Nat Rev Neurosci. 2013;
169. Kincaid W, Neubert MJ, Xu M, et al. Role for medullary pain 14(7):502-511.
facilitating neurons in secondary thermal hyperalgesia. J Neuro- 191. Petrovic P, Kalso E, Petersson KM, et al. Placebo and opioid
physiol. 2006;95:33-41. analgesia—imaging a shared neuronal network. Science. 2002;
170. Fanselow MS. The midbrain periaqueductal gray as a coordinator 295(5560):1737-1740.
of action in response to fear and anxiety. In: DePaulis A, Bandler R, 192. Bingel U, Lorenz J, Schoell E, et al. Mechanisms of placebo anal-
eds. The Midbrain Periaqueductal Gray Matter. New York: Plenum gesia: rACC recruitment of a subcortical antinociceptive network.
Press; 1991:151-173. Pain. 2006;120(1–2):8.
171. Bandler R, DePaulis A. Midbrain periaqueductal gray control of 193. Mainero C, Zhang WT, Kumar A, et al. Mapping the spinal and
defensive behavior in cat and rat. In: DePaulis A, Bandler R, eds. supraspinal pathways of dynamic mechanical allodynia in the human
Midbrain Periaqueductal Gray Matter. New York: Plenum Press; trigeminal system using cardiac-gated fMRI. Neuroimage. 2007;
1991:175-198. 35(3):1201-1210.
172. Rodgers RJ, Randall JI. Environmentally induced analgesia: situa- 194. Zambreanu L, Wise RG, Brooks JC, et al. A role for the brainstem
tional factors, mechanisms and significance. In: Rodgers RJ, Cooper in central sensitisation in humans. Evidence from functional mag-
SJ, eds. Endorphins, Opiates and Behavioral Processes. New York: Wiley; netic resonance imaging. Pain. 2005;114(3):397-407.
1988:107-144. 195. Tracey I, Ploghaus A, Gati JS, et al. Imaging attentional modulation
173. Lewis JW, Cannon JT, Liebeskind JC. Opioid and nonopioid mech- of pain in the periaqueductal gray in humans. J Neurosci. 2002;
anisms of stress analgesia. Science. 1980;208:623-625. 22(7):2748-2752.
167 Molecular Basis of Nociception
167

Susan L. Ingram

This chapter is a review of key molecular mechanisms of pain tissue damage and the inflammatory process. These substances
transduction and provides several examples of plasticity involved modulate pain at all steps in the process, including gating of
in peripheral and central sensitization of pain. channels involved in transduction, propagation of action poten-
Pain (or nociception) is an evolutionally conserved protective tials, and the release of neurotransmitters from central terminals.
mechanism for detection of a dangerous environment and tissue
damage. The knowledge about cellular mechanisms underlying
the perception of pain and plasticity of pain circuits has increased
PAIN SENSITIZATION
exponentially since the 1990s. Acute inflammatory pain after Persistent pain is often caused by direct and long-lasting damage
tissue damage generally plays a protective role for organisms. to peripheral nerve fibers, through either injury or disease states.
Chronic inflammation can induce pain that persists long after the Peripheral sensitization is defined as either allodynia, pain initi-
initial insult. Direct insult to the nerve can induce neuropathic ated by normally innocuous stimuli, or hyperalgesia, an enhanced
pain that also may persist after the time of healing. In both response to noxious stimuli. In the periphery, the inflammatory
chronic inflammatory and neuropathic conditions, this patho- milieu containing neurotransmitters and peptides (substance P,
logic pain does not appear to be protective; instead, it reflects calcitonin gene-related peptide [CGRP], bradykinin), prostaglan-
activation and plasticity in the central nervous system (CNS). Of dins, leukotrienes, neurotrophins, cytokines, chemokines, extra-
importance is the fact that the central processing of nociceptive cellular proteases, and protons can potentiate normal nociceptive
inputs in the spinal cord are subject to modulation by descending processing. The most common drug targets for peripheral sensi-
projections from several brain circuits. Thus the “perception” of tization are cyclooxygenase-1 and cyclooxygenase-2, inhibition of
tissue damage is rarely a direct reflection of the activated primary which reduces synthesis of prostaglandins.
afferents; instead it is a complex output that includes cognitive Intense stimulation or persistent injury can cause plasticity of
and emotional processing by the brain. This critical distinction the central nociceptive circuits, known as central sensitization.
demonstrates that the brain plays an important role in the per- Several molecular mechanisms mediate plasticity of the CNS and
ception of pain and is particularly relevant for considering the contribute to central sensitization. Plasticity of presynaptic neu-
causes of chronic pain. rotransmitters released from primary afferent terminals and post-
Sensory neurons that innervate the skin, viscera, deep muscles, synaptic responses of dorsal horn neurons has been described, as
and joints are primary afferents. The soma of each of these has plasticity in higher brain centers, all of which contribute to
neurons are in the dorsal root or trigeminal ganglia, and their persistent changes in pain perception. The most well-studied
axons extend bidirectionally into their peripheral targets and cen- mechanism is plasticity of glutamatergic signaling on second-
trally into the spinal cord. Primary afferents include neurons that order nociceptive neurons in the dorsal horn of the spinal cord.
respond to low-intensity touch stimulation, such as the myelin- N-methyl-d-aspartic acid (NMDA) receptor antagonists have
ated A beta fibers (which innervate Pacinian corpuscles) and been developed as potential analgesics, but results from clinical
Merkel cells (which detect vibration and light pressure). Nocicep- trials have not been promising because of serious CNS side
tive primary afferents respond to high-intensity nociceptive effects.
stimulation and express many chemoreceptors that detect tissue Inhibition of nociceptive circuitry in the dorsal horn plays a
damage. Nociceptive afferents are classified as two main subtypes: critical role in pain perception. Descending inhibitory serotoner-
A delta–fiber and C-fiber afferents. A delta fibers are thin and gic, noradrenergic, and dopaminergic pathways from the periaq-
myelinated and have faster conduction velocities than C fibers, ueductal gray matter (PAG), rostral ventral medulla (RVM), locus
which are unmyelinated, and they evoke sharp localized pain coeruleus, and raphe nuclei, as well as local γ-aminobutyric
responses. C fiber nociceptors elicit dull, diffuse, burning pain. acid (GABA)– and glycine-containing interneurons, regulate the
The intensity of a painful stimulus is encoded by action potential excitability of dorsal horn projection neurons. It is clear that
frequency. Nociceptive fibers terminate as free nerve endings in the balance of inhibitory and facilitative control of pain by the
the skin and detect multiple stimuli, including heat, cold, mechan- descending pathways is shifted in states of neuropathic or chronic
ical stimuli, and the myriad of chemicals released with tissue pain. Descending control of nociception is a primary area of
damage. research because in many clinical trials of drugs that specifically
Transduction of pain information occurs through direct stim- target peripheral molecules involved in pain transduction, the
ulation of ligand-gated ion channels or indirectly: for example, results have fallen short of expectations for multiple reasons,
through activation of G protein–coupled receptors that are including lack of efficacy and safety, selectivity, and possible issues
coupled to ion channels through intracellular signaling mol- with animal model translation. Thus attention is beginning to
ecules. Activation of these channels leads to the depolarization turn from a focus on specific molecules to a wider view of pain
of afferent nerve endings. Summation of multiple events leads circuits and their interactions. Further research with optogenetic
to activation of voltage-gated sodium and potassium channels and transgenic techniques to study pain circuits will certainly
involved in action potential generation. Action potentials travel advance the understanding of how this balance is modulated at
to the spinal cord and stimulate voltage-gated Ca2+ channels each level of the pain axis.
in the central terminals. Influx of calcium initiates release of
neurotransmitters from vesicles. Transduction is modulated by
pronociceptive substances and by immune factors released during Full text of this chapter is available online at ExpertConsult.com

1379
167 Molecular Basis of Nociception
167

Susan L. Ingram
Pain (or nociception) is an evolutionally conserved protective nerve endings of nociceptive A delta and C fibers in the skin. This
mechanism for detection of a dangerous environment and tissue damage sets off a cascade of events that constitute neuroinflam-
damage.1 The knowledge of cellular mechanisms underlying the mation.2 Activation of nerve endings stimulates the release of
perception of pain and plasticity of pain circuits has increased pro-nociceptive substances, including the neuropeptides sub-
exponentially since the 1990s. Tissue damage causes the release stance P, CGRP, and neuropeptide Y that trigger inflammatory
of myriad substances that activate nociceptive afferents and responses. Mast cells are recruited to the site and release a host
recruit immune cells to the area to induce inflammation. Acute of inflammatory mediators including: prostaglandins, bradykinin,
inflammatory pain after tissue damage generally plays a protec- cytokines, serotonin and histamine. Vasodilation and plasma
tive role for organisms. Chronic inflammation can induce pain extravasation also contribute to amplification of the inflammatory
that persists long after the initial insult. Direct insult to the nerve response (Fig. 167-1).
can induce neuropathic pain that also may persist after the time
of healing. In both chronic inflammatory and neuropathic condi-
tions, this pathologic pain does not appear to be protective;
Detection and Transduction of Tissue Injury
instead, it reflects activation and plasticity in the CNS. Of impor- The detection of the nociceptive environment and inflammatory
tance is that the central processing of nociceptive inputs in the mediators is accomplished through many specialized receptor
spinal cord are subject to modulation by descending projections proteins that are differentially expressed in primary afferents.
from several brain circuits. Thus the “perception” of tissue The specificity theory of pain, originating back to the writings
damage is rarely a direct reflection of the activated primary affer- of Descartes, describes specific pain modalities having direct but
ents; instead, it is a complex output that includes cognitive and separate pathways from the periphery to the brain.3 Considerable
emotional processing by the brain. This critical distinction dem- effort has been made to identify different populations of nocicep-
onstrates that the brain plays an important role in the perception tive afferents on the basis of their sensitivity to painful stimuli and
of pain and is particularly relevant for considering the causes of responses to pain-promoting molecules. C-fiber nociceptors are
chronic pain. This chapter summarizes key molecular mecha- loosely categorized as peptidergic and nonpeptidergic subpopula-
nisms of pain transduction and several examples of plasticity tions. Peptidergic C-fiber nociceptors contain and release neuro-
involved in peripheral and central sensitization of pain. peptides such as substance P and CGRP. These neurons express
the tyrosine kinase A receptor for nerve growth factor (NGF).
Nonpeptidergic nociceptors do not synthesize these peptides but
ACTIVATION OF PRIMARY AFFERENTS can be identified by their ability to bind the isolectin GS-IB4
Sensory neurons that innervate the skin, viscera, deep muscle, and and by their sensitivity to glia-derived neurotrophic factor.
joints are primary afferents. These neurons have their soma in However, neurons within each of these subpopulations respond
the dorsal root or trigeminal ganglia and extend axons bidirec- to various inflammatory mediators and are generally polymo-
tionally, into their peripheral targets and centrally into the spinal dal, which dramatically increases the complexity of primary
cord. Transduction occurs through direct stimulation of ligand- afferents (Fig. 167-2).
gated ion channels or indirectly, for example via activation of Subtypes of C-fiber and A delta–fiber nociceptors express
G-protein-coupled receptors that are coupled to ion channels many specific receptors for endogenous inflammatory mediators,
through intracellular signaling molecules. Activation of these including bradykinin (both B2 and B1 receptors), serotonin
channels leads to a generator potential or depolarization of (5-HT3 receptors), prostaglandin E2 (EP receptors), histamine,
afferent nerve endings and summation of multiple events leads and protons (the acid-sensitive ion channels 1, 2, and 3). ATP
to activation of voltage-gated sodium and potassium channels receptors include metabotropic P2Y receptors and P2X2 and
involved in action potential generation. ionotropic P2X3 receptors. Many of the receptor proteins are
Primary afferents include neurons that respond to low inten- activated or modulated, or both, by multiple inflammatory sub-
sity touch stimulation such as the myelinated A beta fibers that stances. A prominent example is the family of transient receptor
innervate Pacinian corpuscles and Merkel cells to detect vibration potential (TRP) channels. TRP channels include several proteins
and light pressure. Nociceptive primary afferents respond to that are nonselective cation channels that play a role in nocicep-
high-intensity nociceptive stimulation and express many chemo- tive transduction. The first member of this family to be cloned
receptors that detect tissue damage. Nociceptive afferents are was TRPV1. TRPV1 is gated by multiple chemical stimuli,
split into two main subtypes: A delta–fiber and C-fiber afferents. including capsaicin, the active ingredient in hot peppers and
Thinly myelinated A delta fibers have faster conduction velocities those released during inflammation, as well as by changes in
than unmyelinated C fibers and evoke sharp localized pain temperature in the noxious range (>43°C for humans).4-6 Inflam-
responses as opposed to the dull, diffuse, burning pain elicited by matory mediators that sensitize or activate nociceptors can also
C-fiber nociceptors. The intensity of a painful stimulus is encoded modulate or sensitize TRP channels (Fig. 167-3). Metabolites of
by proportional scaling of action potential frequency. Nocicep- 12- and 15-lipoxygenase (leukotriene B4 and 15[S]-hydroxyeico-
tive fibers terminate as free nerve endings in the skin and detect satrienoic acid), N-arachidonyl dopamine, and protons activate
multiple stimuli, including heat, cold, mechanical stimuli, or the TRPV1. G protein–coupled receptors (e.g., protease-activated
myriad of chemicals released with tissue damage. receptors, bradykinin receptors, P2Y purinergic receptors, pros-
taglandin receptors, metabotropic glutamate receptors) that
Pain-Promoting Substances Released with couple to phosphatidylinositol 4,5-bisphosphate (PIP2) and phos-
pholipase C strongly sensitize TRPV1-mediated currents. Phos-
Tissue Injury phorylation by multiple kinase pathways, including PKC and
Upon tissue damage, injured cells release large quantities of PKA, facilitate activation of TRPV1 by different mechanisms.
adenosine triphosphate (ATP) and glutamate that activate free Thus TRPV1 integrates signals from multiple aspects of the
e605
e606 SECTION 6  Pain

Figure 167-1. Tissue damage sets up an inflammatory cycle. Tissue damage causes release of multiple
substances that excite nociceptive afferents to release neurotransmitters (substance P and calcitonin
gene–related peptide) that in turn recruit immune cells and activate the vasculature to release additional
inflammatory mediators. ATP, adenosine triphosphate; AP, action potential; DRG, dorsal root ganglia;
H+, hydrogen ion; 5-HT, serotonin; NGF, nerve growth factor.

inflammatory cascade and has been shown to be a critical protein


in the development of hyperalgesia.7-9 However, in TRPV1-
deficient mice, heat sensitivity remains intact,10 suggesting that
TRPV1 is not the only molecule involved in heat transduction.
There are several additional subtypes of the TRPV receptors
(TRPV2 to TRPV4). These receptors have slightly different heat
thresholds for activation and are expressed in subpopulations of
primary afferent neurons. TRPV2 has a higher threshold (>52°C)
than TRPV1 and is expressed primarily in A delta–fiber nocicep-
tors. TRPV3 and TRPV4 are expressed in epithelia and have
a narrow activation range, between 25°C and 35°C. These
additional members of the TRP family also respond to mul-
tiple signaling molecules. For example, TRPV4 is activated by
several endogenous chemicals: anandamide, arachidonic acid, and
5′,6′-epoxyeicosatrienoic acid.
Transduction of cold sensation is not as well understood but
probably involves multiple signaling proteins.11 The TRPM8
channel is expressed in a subpopulation of primary afferent A
delta–fiber nociceptors that responds to cold (< 25°C) and is
permeable to calcium ions.12-15 In knockout models of TRPM8,
both normal cold sensation and cold hypersensitivity are defi-
cient,16-18 which indicates that these channels play a primary role
in cold sensation. The TRPM8 channel is sensitive to cooling
agents such as menthol, icilin, and eucalyptol. TRPA1 channels
Figure 167-2. Primary afferent nociceptors are heterogeneous play an important role in injury-evoked cold hypersensitivity.19-21
with regard to transduction molecules. Each nociceptor expresses TRPA1 channels are also activated by a pungent chemical found
a unique complement of receptor and ion channels that are sensitive in mustard, wasabi, and allium plants.21-25 Continued research
to pronociceptive stimuli and inflammatory mediators. (From Basbaum into these receptor subtypes will further define their roles in pain.
AI, Bautista DM, Scherrer G, et al. Cellular and molecular mechanisms The KCNK family of potassium channels expressed in C-type
of pain. Cell. 2009;139:267-284.) nociceptors also plays a role in cold sensation. The TREK-1 (a
“TWIK-related potassium channel,” in which TWIK stands for
“tandem P domain in a weak inward rectifier K+ channel”) and
CHAPTER 167  Molecular Basis of Nociception e607

167

Figure 167-3. An emerging structure-function map for the capsaicin receptor, TRPV1. Pharmacologic
and mutagenesis studies have identified TRPV1 domains that confer sensitivity to various stimuli or contribute
to physiologic modulation of the channel downstream of metabotropic receptors. These include sites of
action for capsaicin (depicted by the chili pepper) and related vanilloid ligands, extracellular protons (depicted
by the lemon), or peptide toxins from the tarantula (depicted by the spider). Regions implicated in channel
modulation by cellular proteins and cytoplasmic second messengers are also indicated. A, ankyrin
repeats; ATP, adenosine triphosphate; C, carboxy terminus; N, amino terminus; PIP2, phosphatidylinositol
4,5-bisphosphate; PKA, protein kinase A; PKC, protein kinase C; PLC, phospholipase C. (From Julius D. TRP
channels and pain. Annu Rev Cell Dev Biol. 2013;29:355-384.)

TRAAK (TWIK-related arachidonic acid–stimulated K+ channel) Inflammation of the target tissues results in hypersensitivity
channels are highly mechanosensitive and thermosensitive chan- of these afferents in association with pain disorders such as
nels.26 Of interest is that another member of the family, KCNK18, irritable bowel syndrome and other functional gastrointestinal
is activated by hydroxy-α-sanshool, the active ingredient of the disorders.35
Szechuan pepper.27 Muscle pain occurs during sustained muscular contraction and
Receptors for mechanical stimuli have long been thought to ischemia, after trauma, or after eccentric exercise.36 The majority
be sensitive to stretching or depression of the skin, or both. of muscle fiber afferents are low-threshold afferents that transmit
Members of the degenerin/epithelial sodium channel family, deep pressure. Less than half of the group III and IV fibers in
including mec-4 and mec-10 of Caenorhabditis elegans, and acid- muscle are thought to be nociceptors. Sensitization of these affer-
sensing ion channels have been proposed as likely candidates for ents may play a role in muscle disorders with generalized pain,
mechanosensation. However, studies of acid-sensing ion channel such as fibromyalgia.37
knock-out mice have demonstrated few deficits in mechanosensa- Skeletal pain results from activation of terminals ending in
tion, which indicates that additional molecules will be implicated various targets, including joints, the periosteum, and the intra-
in mechanosensation.28 TRP channels have also been implicated medullary space. Nociceptive afferents localized throughout
as mechanosensors.29 TRPV2 has been shown to respond to tissues surrounding joints can be sensitized in osteoarthritis.38,39
osmotic stretch,30 and TRPA1 may also detect mechanical stimuli, Degeneration of cartilage and inflammation within the joint
inasmuch as selective inhibition of TRPA1 channels reduces contribute to the development of chronic pain but the cellular
mechanical hypersensitivity associated with inflammation.31,32 mechanisms, both peripheral and central, are still being eluci-
Antisense knockdown of TRPV4 channels reduced mechanical dated. Pain caused by tumor metastasis to bone, or other cancer
hyperalgesia in several animal models of neuropathy.33 However, pain, involves both inflammatory and neuropathic mechanisms.40
few studies have shown that any of these channels are sufficient Approximately 75% of patients with advanced cancer experience
for mechanical transduction in isolated transfection systems; thus bone pain that is characterized by dull and continuous background
additional proteins or lipid environments may be necessary for pain interspersed with spontaneous breakthrough pain.41,42 This
function. mixture of pain symptoms makes cancer-induced bone pain dif-
ficult to treat successfully, and so considerable effort is being
expended to understand the pathophysiologic mechanisms that
Nociception in Other Tissues appear to be unique to cancer pain.
Visceral pain is a prevalent, major clinical problem. The periph-
eral basis of visceral pain differs from that in other body regions.34
Visceral sensory afferents are predominately A delta and C fibers
Peripheral Sensitization
with diffuse receptive fields, so that pain is less localized. These Persistent pain is often caused by direct and long-lasting
visceral afferents travel with sympathetic nerves to the spinal cord damage to peripheral nerve fibers, either through injury or
and express transduction channels and receptors similar to those through disease states. Peripheral sensitization is defined as
expressed by somatic primary afferents (see earlier discussion). either allodynia (pain initiated by normally innocuous stimuli)
e608 SECTION 6  Pain

or hyperalgesia (an enhanced response to noxious stimuli). In the interneurons and projection neurons in different laminae of the
periphery, stimulation of the inflammatory cascade and release of dorsal horn. Centrally, primary afferent axons can extend col-
myriad inflammatory substances (the “inflammatory soup”) after lateral axons into multiple levels of the spinal cord. Communica-
tissue damage initiate gating of ion channels (see Fig. 167-1) tion between the primary afferent central termini and neurons in
through direct or allosteric binding to the receptor proteins or the spinal dorsal horn is predominately glutamatergic but can
through intracellular second messenger signaling. This “inflam- include release of various neuropeptides. The dorsal horn con-
matory soup” contains neurotransmitters and peptides (substance sists of secondary projection neurons to the brain and interneu-
P, CGRP, bradykinin), prostaglandins, leukotrienes, neurotroph- rons that can contain GABA, glycine, or glutamate. The dorsal
ins, cytokines, chemokines, extracellular proteases, and protons. horn has a complex circuitry and a large capacity to modulate the
The most common drug targets for peripheral sensitization are nociceptive inputs from primary afferent central terminals.
cyclooxygenase-1 and cyclooxygenase-2, inhibition of which According to in vivo recordings from neurons in the lamina I
reduces synthesis of prostaglandins. NGF is a component of the dorsal horn, this population of neurons respond specifically to
“inflammatory soup”43 and contributes to peripheral sensitization nociceptive stimuli and are thus termed nociceptive-specific neurons.
by activation of tyrosine kinase A receptors selectively expressed These neurons have small receptive fields and provide informa-
by C-fiber nociceptors.44 Tyrosine kinase A receptors modulate tion as to the localization of the stimuli. In contrast, other neurons
TRPV1 currents45 and induce expression of several pronocicep- in lamina I respond to multiple inputs and are polymodal noci-
tive proteins, including substance P, TRPV1, and Nav1.8 chan- ceptive neurons. The inner lamina II receives thinly myelinated
nels.44,46 Interesting new therapies for peripheral sensitization A delta and unmyelinated C fibers that sense innocuous touch.
include anti-NGF antibodies and antagonists of TRPV1 and Neurons in lamina V respond to nociceptive stimuli, as well as
TRPA1 receptors.21 innocuous stimuli, and have large, complex receptive fields. These
neurons are known as wide dynamic range neurons. Nociceptive
spinal neurons send axons across the midline and ascend to the
MODULATION OF PRIMARY AFFERENT FIRING brain on the contralateral side of the spinal cord.
The frequency of primary afferent firing and the population of Neurotransmitters are released from primary afferent termi-
stimulated afferent fibers underlie the encoding of nociception. nals as the action potentials depolarize terminals and activate
Generator potentials summate to depolarize peripheral terminals voltage-gated calcium channels. As mentioned earlier, inflamma-
and activate voltage-gated sodium, potassium, and calcium chan- tory mediators modulate these channels, and selective inhibition
nels to initiate action potential firing. Many subtypes of voltage- of calcium channels in nociceptive afferents has been an effective
gated sodium channels have been cloned, and these channels strategy for relieving inflammatory pain.54 Peripheral inflamma-
either are blocked by tetrodotoxin (TTX), and are thus termed tion stimulates a similar inflammatory process centrally, so that
TTX-sensitive (Nav1.1, Nav1.6 and Nav1.7), or are TTX-insensitive many of the same molecules modulate the flow of information
channels (Nav1.8 and Nav1.9). Nav1.8 channels are activated by from central presynaptic terminals of primary afferents to post-
membrane repolarization and are also called resurgent sodium synaptic second-order spinal cord neurons (Fig. 167-4). Thus the
channels. Many of these voltage-gated channels involved in action enhanced release of neurotransmitters and peptides from acti-
potential firing are modulated by substances in the “inflammatory vated nociceptive primary afferents stimulates vascular responses
soup”47 and contribute to hyperalgesia and neuropathic pain and recruitment of immune cells, including microglia, astrocytes,
states. Expression of the voltage-gated sodium channel Nav1.748,49 and T cells, that further contribute to the inflammatory response
is upregulated in inflammatory pain and may exacerbate inflam- by releasing inflammatory mediators. The central neuroinflam-
matory pain, but expression of this channel is not changed after matory response and neurotransmission is also under control of
nerve injury.50 descending brain circuits that have been shown to mediate both
Voltage-gated calcium channels involved in release of neu- inhibition and facilitation of pain transmission.55
rotransmitter from primary afferents are also modulated during
inflammatory or neuropathic pain states. Gabapentin, an anticon-
vulsant drug that targets the auxiliary α2δ subunit of calcium
Central Mechanisms of Sensitization
channels has been used to treat neuropathic pain.51 Finally, Intense stimulation or persistent injury can cause plasticity of the
KCNQ channels, which are low-threshold, voltage-activated central nociceptive circuits, which is known as central sensitization.
potassium channels, do not become inactivated and play a role in Plasticity of presynaptic neurotransmitter release from primary
stabilizing membrane potential and decreasing excitability. They afferent terminals and postsynaptic responses of dorsal horn
are negatively regulated by G protein–coupled receptors, such as neurons has been described, as has plasticity in higher brain
the bradykinin receptor, so that activation of the bradykinin centers, all of which contribute to persistent changes in pain
receptor increases excitability of primary afferent nociceptors.52 perception. In some cases, this plasticity leads to abnormal pro-
A family of analgesic agents (flupirtine and retigabine) increase cessing of information from myelinated A beta touch fibers,
activation of KCNQ channels to reduce excitability of primary which leads to paraesthesias and dyasthesias, and increased dis-
afferents during inflammation.53 charge in A delta and C fibers leads to stabbing, burning pain.56
Several molecular mechanisms mediate plasticity of the CNS and
TRANSMISSION OF PAIN SIGNALS TO THE CENTRAL contribute to central sensitization.57,58 Little is known about the
control of vesicle release from primary afferent terminals, but
NERVOUS SYSTEM increased protein kinase G-I and Ca2+ signaling59 has been found
Action potentials in primary afferents travel from the periphery to contribute to an increase in the release probability of C-fiber
to the central terminals in the dorsal horn of the spinal cord. The afferents impinging on spino-periaqueductal gray projection
majority of lightly myelinated A delta– and unmyelinated C-fiber neurons. Postsynaptic synaptic plasticity has been studied
axons form a central branch that projects through the dorsal root more extensively.60 Synaptic plasticity of glutamatergic synapses
to the spinal cord. A subpopulation of these fibers enters via the involves glutamate activation of metabotropic glutamate recep-
ventral roots, but it is not clear whether these fibers carry noci- tors and previously silent NMDA receptors similar to long-term
ceptive information. As the spinal nerve root approaches the potentiation processes in the brain (Fig. 167-5).61-63 NMDA
spinal cord, it splits into smaller rootlets, bifurcates into short receptor antagonists have been developed as potential analgesics,
ascending and descending branches that form Lissauer’s tract on but results from clinical trials have not been promising because
the surface of the dorsal horn, and ultimately synapses with local of serious CNS side effects.64
CHAPTER 167  Molecular Basis of Nociception e609

167

Figure 167-4. Neuronal activity triggers neurogenic neuroinflammation in the central nervous system
(CNS). Neurogenic neuroinflammation at spinal or trigeminal terminals is illustrated. In the CNS, enhanced
neuronal activity from peripheral sources results in neurogenic neuroinflammation owing to vesicular and
nonvesicular release of neurotransmitters and neuropeptides from the primary afferent fiber (listed in the blue
boxes). This induces concerted and interacting immune responses, vascular responses, and higher order
neuronal network responses in the multipartite synapse. As in the periphery, both proinflammatory and
anti-inflammatory mediators, as well as signaling molecules and forces, can be released from all cell types
(key substances listed in pink boxes for astrocytes, the green box for microglial cells, the lavender box for
mast cells, and the tan box for pericytes) in the multipartite synaptic region to further affect receptors or
channels present on all cell types shown (key substances acting on cells are listed in light yellow boxes).
Signaling from higher order CNS centers (descending neurons) can serve to dampen or aggravate neurogenic
neuroinflammation. ADO, adenosine; ATP, adenosine triphosphate; BDNF, brain-derived neurotrophic factor;
CB, cannabinoid; CGRP, calcitonin gene-related peptide; DA, dopamine; DAMP, danger-associated molecular
patterns; D-Ser, D-serine; END, endothelin; GABA, γ-aminobutyric acid; Gly, glycine; His, histamine; IgE,
immunoglobulin E; NA, noradrenaline; NGF, nerve growth factor; NO, nitric oxide; NPY, neuropeptide Y; PAF,
platelet activating factor; PG, prostaglandin; SOM, somatostatin; SP, substance P; TRP, tryptase. (From
Xanthos DN, Sandkühler J. Neurogenic neuroinflammation: inflammatory CNS reactions in response to
neuronal activity. Nature Rev. 2014;15:43-53.)
e610 SECTION 6  Pain

Pre-
synapse

Ca2+

IL-1/6

TNF- α
P

mGluR NMDAR AMPAR


BDNF NK1R

NO

ATP
Ca2+
IP3R
2+
Ca
Key: PKA
PKC
Glutamate
CaMKII
SP NOS
Post- MEK/ERK
synapse Nuclear Ca2+,
CREB and DREAM-
dependent gene
transcription

Figure 167-5. Postsynaptic mechanisms of synaptic plasticity. Synaptic potentiation at some synapses
involves Ca2+ influx via N-methyl-D-aspartate (NMDA) receptors (NMDARs) and increased membrane insertion
of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors (AMPARs). Ca2+ influx via ion
channels, as well as Ca2+ release from intracellular stores, brings about sustained plasticity through synapse-
to-nucleus messengers and subsequent gene transcription. Mediators released by microglia further augment
synaptic potentiation. Abbreviations: ATP, adenosine triphosphate; BDNF, brain-derived neurotrophic factor;
CaMKII, calcium/calmodulin-dependent protein kinase II; CREB, cyclic adenosine monophosphate (cAMP)
response element-binding protein; DREAM, downstream regulatory element antagonist modulator; IL,
interleukin; IP3R, inositol-1,4,5- triphosphate receptor; MEK/ERK, mitogen-activated protein kinase kinase/
extracellular signal-regulated kinase; mGluR, metabotropic glutamate receptor; NK1R, neurokinin 1 receptor;
NO, nitric oxide; NOS, nitric oxide synthase; P, phosphorylation site; PKA, protein kinase A; PKC, protein
kinase C; SP, substance P; TNF-α, tumor necrosis factor α. (From Luo C, Kuner T, Kuner R. Synaptic
plasticity in pathological pain. Trends Neurosci. 2014;37:343-355.)

Inhibition of nociceptive circuitry in the dorsal horn plays a factors, such as the fractalkine receptor and Toll-like receptors,
critical role in pain perception. Descending inhibitory serotoner- have also been implicated in nerve injury.7
gic, noradrenergic, and dopaminergic pathways from the periaq- Despite the advances in understanding molecular mechanisms
ueductal gray matter (PAG), rostral ventral medulla (RVM), locus of pain, many clinical trials of drugs that specifically target these
caeruleus, and raphe nuclei, as well as by local GABA and glycine- molecules have fallen short of expectations for multiple reasons,
containing interneurons, regulate the excitability of dorsal horn including lack of efficacy and safety, selectivity, and possible issues
projection neurons.65 The inhibitory interneurons also release with animal model translation.69 Thus attention is beginning to
endogenous opioid peptides that act at presynaptic terminals to turn from a focus on specific molecules to a wider view of pain
decrease release probability of pronociceptive neurotransmitters circuits and their interactions. It is clear that the balance of
(substance P and CGRP) and postsynaptically to inhibit lamina inhibitory and facilitative control of pain by the descending path-
I and V neurons.66 The ability of these interneurons to inhibit ways is shifted in states of neuropathic or chronic pain.70,71
pain transmission is decreased in neuropathic pain,67 but the Further research with optogenetic and transgenic techniques to
mechanism is still under investigation. One potential mechanism study pain circuits will certainly advance the understanding of
involves activated microglia and astrocytes and release of various how this balance is modulated at each level of the pain axis. In
factors from these glial cells. Release of brain-derived neuro- addition, the current intense focus on interactions between
trophic factor disinhibits lamina I projection neurons leading to neurons and glia may elucidate novel molecular targets for pain
increased responses to pain.68 Other receptors of glia-derived therapies that are useful in various chronic pain states.
CHAPTER 167  Molecular Basis of Nociception e611

Acknowledgments 10. Woodbury CJ, Zwick M, Wang S, et al. Nociceptors lacking TRPV1
The author would like to thank Andy Rekito for assistance with figure and TRPV2 have normal heat responses. J Neurosci. 2004;24(28): 167
preparation and Dr. Tally Largent-Milnes for providing expert feedback 6410-6415.
on the manuscript. 11. McKemy DD. The molecular and cellular basis of cold sensation.
ACS Chem Neurosci. 2013;4(2):238-247.
12. McCoy DD, Knowlton WM, McKemy DD. Scraping through the
SUGGESTED READINGS ice: uncovering the role of TRPM8 in cold transduction. Am J Physiol
Andersson DA, Gentry C, Moss S, et al. Transient receptor potential A1 Regul Integr Comp Physiol. 2011;300(6):R1278-R1287.
is a sensory receptor for multiple products of oxidative stress. J Neu- 13. McKemy DD, Neuhausser WM, Julius D. Identification of a cold
rosci. 2008;28:2485-2494. receptor reveals a general role for TRP channels in thermosensation.
Basbaum AI, Bautista DM, Scherrer G, et al. Cellular and molecular Nature. 2002;416(6876):52-58.
mechanisms of pain. Cell. 2009;139(2):267-284. 14. Peier AM, Moqrich A, Hergarden AC, et al. A TRP channel that
Belmonte C, Viana F. Molecular and cellular limits to somatosensory senses cold stimuli and menthol. Cell. 2002;108(5):705-715.
specificity. Mol Pain. 2008;4:14. 15. Yudin Y, Rohacs T. Regulation of TRPM8 channel activity. Mol Cell
Burgess PR, Perl ER. Myelinated afferent fibres responding specifically Endocrinol. 2012;353(1–2):68-74.
to noxious stimulation of the skin. J Physiol. 1967;190:541-562. 16. Bautista DM, Siemens J, Glazer JM, et al. The menthol receptor
Caterina MJ, Julius D. The vanilloid receptor: a molecular gateway to the TRPM8 is the principal detector of environmental cold. Nature.
pain pathway. Annu Rev Neurosci. 2001;24:487-517. 2007;448(7150):204-208.
Caterina MJ, Schumacher MA, Tominaga M, et al. The capsaicin recep- 17. Colburn RW, Lubin ML, Stone DJ Jr, et al. Attenuated cold sensitiv-
tor: a heat-activated ion channel in the pain pathway. Nature. 1997; ity in TRPM8 null mice. Neuron. 2007;54(3):379-386.
389:816-824. 18. Dhaka A, Murray AN, Mathur J, et al. TRPM8 is required for cold
Davis JB, Gray J, Gunthorpe MJ, et al. Vanilloid receptor-1 is essential sensation in mice. Neuron. 2007;54(3):371-378.
for inflammatory thermal hyperalgesia. Nature. 2000;405(6783): 19. del Camino D, Murphy S, Heiry M, et al. TRPA1 contributes to cold
183-187. hypersensitivity. J Neurosci. 2010;30(45):15165-15174.
Drenth JPH, Waxman SG. Mutations in sodium-channel gene SCN9A 20. Knowlton WM, Bifolck-Fisher A, Bautista DM, et al. TRPM8, but
cause a spectrum of human genetic pain disorders. J Clin Invest. not TRPA1, is required for neural and behavioral responses to acute
2007;117:3603-3609. noxious cold temperatures and cold-mimetics in vivo. Pain. 2010;
Gangadharan V, Kuner R. Pain hypersensitivity mechanisms at a glance. 150(2):340-350.
Dis Model Mech. 2013;6(4):889-895. 21. Moran MM, McAlexander MA, Biro T, et al. Transient receptor
LaMotte RH, Campbell JN. Comparison of responses of warm and noci- potential channels as therapeutic targets. Nat Rev Drug Discov.
ceptive C-fiber afferents in monkey with human judgements of thermal 2011;10(8):601-620.
pain. J Neurophysiol. 1978;41:509-528. 22. Bandell M, Story GM, Hwang SW, et al. Noxious cold ion channel
Leffler A, Fischer MJ, Rehner D, et al. The vanilloid receptor TRPV1 is TRPA1 is activated by pungent compounds and bradykinin. Neuron.
activated and sensitized by local anesthetics in rodent sensory neurons. 2004;41(6):849-857.
J Clin Invest. 2008;118:763-776. 23. Bautista DM, Movahed P, Hinman A, et al. Pungent products from
Luo C, Kuner T, Kuner R. Synaptic plasticity in pathological pain. Trends garlic activate the sensory ion channel TRPA1. Proc Natl Acad Sci
Neurosci. 2014;37(6):343-355. U S A. 2005;102(34):12248-12252.
Moayedi M, Davis KD. Theories of pain: from specificity to gate control. 24. Jordt SE, Bautista DM, Chuang HH, et al. Mustard oils and can-
J Neurophysiol. 2013;109(1):5-12. nabinoids excite sensory nerve fibres through the TRP channel
Tominaga M, Caterina MJ, Malmberg AB, et al. The cloned capsaicin ANKTM1. Nature. 2004;427(6971):260-265.
receptor integrates multiple pain-producing stimuli. Neuron. 1998;21: 25. Macpherson LJ, Geierstanger BH, Viswanath V, et al. The pungency
531-543. of garlic: activation of TRPA1 and TRPV1 in response to allicin.
Torebjörk HE, Ochoa JL. Specific sensations evoked by activity in single Curr Biol. 2005;15(10):929-934.
identified sensory units in man. Acta Physiol Scand. 1980;110:445-447. 26. Noel J, Zimmermann K, Busserolles J, et al. The mechano-activated
Vallbo AB, Hagbarth K-E, Torebjörk HE, et al. Somatosensory, proprio- K+ channels TRAAK and TREK-1 control both warm and cold
ceptive, and sympathetic activity in human peripheral nerves. Physiol perception. EMBO J. 2009;28(9):1308-1318.
Rev. 1979;59:919-957. 27. Bautista DM, Sigal YM, Milstein AD, et al. Pungent agents from
Xanthos DN, Sandkuhler J. Neurogenic neuroinflammation: inflamma- Szechuan peppers excite sensory neurons by inhibiting two-pore
tory CNS reactions in response to neuronal activity. Nat Rev Neurosci. potassium channels. Nat Neurosci. 2008;11(7):772-779.
2014;15(1):43-53. 28. Lumpkin EA, Caterina MJ. Mechanisms of sensory transduction in
the skin. Nature. 2007;445(7130):858-865.
29. O’Neil RG, Heller S. The mechanosensitive nature of TRPV chan-
REFERENCES nels. Pflugers Arch. 2005;451(1):193-203.
1. Woolf CJ. What is this thing called pain? J Clin Invest. 2010; 30. Soya M, Sato M, Sobhan U, et al. Plasma membrane stretch activates
120(11):3742-3744. transient receptor potential vanilloid and ankyrin channels in
2. Xanthos DN, Sandkuhler J. Neurogenic neuroinflammation: inflam- Merkel cells from hamster buccal mucosa. Cell Calcium. 2014;55(4):
matory CNS reactions in response to neuronal activity. Nat Rev 208-218.
Neurosci. 2014;15(1):43-53. 31. Brierley SM, Castro J, Harrington AM, et al. TRPA1 contributes to
3. Moayedi M, Davis KD. Theories of pain: from specificity to gate specific mechanically activated currents and sensory neuron mechan-
control. J Neurophysiol. 2013;109(1):5-12. ical hypersensitivity. J Physiol. 2011;589(Pt 14):3575-3593.
4. Caterina MJ, Julius D. The vanilloid receptor: a molecular gateway 32. Brierley SM, Hughes PA, Page AJ, et al. The ion channel TRPA1 is
to the pain pathway. Annu Rev Neurosci. 2001;24:487-517. required for normal mechanosensation and is modulated by algesic
5. Huang J, Zhang X, McNaughton PA. Inflammatory pain: the cellular stimuli. Gastroenterology. 2009;137(6):2084-2095, e3.
basis of heat hyperalgesia. Curr Neuropharmacol. 2006;4(3):197-206. 33. Alessandri-Haber N, Dina OA, Yeh JJ, et al. Transient receptor
6. Tominaga M, Caterina MJ, Malmberg AB, et al. The cloned capsaicin potential vanilloid 4 is essential in chemotherapy-induced neuro-
receptor integrates multiple pain-producing stimuli. Neuron. 1998; pathic pain in the rat. J Neurosci. 2004;24(18):4444-4452.
21(3):531-543. 34. Cervero F. Visceral nociception: peripheral and central aspects
7. Basbaum AI, Bautista DM, Scherrer G, et al. Cellular and molecular of visceral nociceptive systems. Philos Trans R Soc Lond B Biol Sci.
mechanisms of pain. Cell. 2009;139(2):267-284. 1985;308(1136):325-337.
8. Davis JB, Gray J, Gunthorpe MJ, et al. Vanilloid receptor-1 is essen- 35. Sikandar S, Dickenson AH. Visceral pain: the ins and outs, the ups
tial for inflammatory thermal hyperalgesia. Nature. 2000;405(6783): and downs. Curr Opin Support Palliat Care. 2012;6(1):17-26.
183-187. 36. Mense S. Nociception from skeletal muscle in relation to clinical
9. Caterina MJ, Leffler A, Malmberg AB, et al. Impaired nociception muscle pain. Pain. 1993;54(3):241-289.
and pain sensation in mice lacking the capsaicin receptor. Science. 37. Staud R. Peripheral pain mechanisms in chronic widespread pain.
2000;288(5464):306-313. Best Pract Res Clin Rheumatol. 2011;25(2):155-164.
e612 SECTION 6  Pain

38. Lee AS, Ellman MB, Yan D, et al. A current review of molecular 55. Heinricher MM, Ingram SL. The brainstem and nociceptive modu-
mechanisms regarding osteoarthritis and pain. Gene. 2013;527(2): lation. In: Bushnell MC, Basbaum AI, eds. The Senses: A Comprehensive
440-447. Reference. Vol 5: Pain. San Diego, CA: Academic Press; 2008:
39. Schaible HG. Mechanisms of chronic pain in osteoarthritis. Curr 593-626.
Rheumatol Rep. 2012;14(6):549-556. 56. Nickel FT, Seifert F, Lanz S, et al. Mechanisms of neuropathic pain.
40. Falk S, Dickenson AH. Pain and nociception: mechanisms of cancer- Eur Neuropsychopharmacol. 2012;22(2):81-91.
induced bone pain. J Clin Oncol. 2014;32(16):1647-1654. 57. Kuner R. Central mechanisms of pathological pain. Nat Med. 2010;
41. Portenoy RK, Lesage P. Management of cancer pain. Lancet. 1999; 16(11):1258-1266.
353(9165):1695-1700. 58. Gangadharan V, Kuner R. Pain hypersensitivity mechanisms at a
42. Foley KM. Treatment of cancer-related pain. J Natl Cancer Inst glance. Dis Model Mech. 2013;6(4):889-895.
Monogr. 2004;32:103-104. 59. Luo C, Gangadharan V, Bali KK, et al. Presynaptically localized
43. Ritner HL, Machelska H, Stein C. Immune system pain and analge- cyclic GMP-dependent protein kinase 1 is a key determinant of
sia. In: Basbaum AI, Bushnell M, eds. Science of Pain. Oxford, UK: spinal synaptic potentiation and pain hypersensitivity. PLoS Biol.
Academic Press; 2009:407-427. 2012;10(3):e1001283.
44. Chao MV. Neurotrophins and their receptors: a convergence point 60. Luo C, Kuner T, Kuner R. Synaptic plasticity in pathological pain.
for many signaling pathways. Nat Rev Neurosci. 2003;4:299-309. Trends Neurosci. 2014;37(6):343-355.
45. Chuang HH, Prescott ED, Kong H, et al. Bradykinin and nerve 61. Drdla R, Sandkuhler J. Long-term potentiation at C-fibre synapses
growth factor release the capsaicin receptor from PtdIns(4,5) by low-level presynaptic activity in vivo. Mol Pain. 2008;4:18.
P2-mediated inhibition. Nature. 2001;411(6840):957-962. 62. Randic M, Jiang MC, Cerne R. Long-term potentiation and long-
46. Ji RR, Samad TA, Jin SX, et al. p38 MAPK activation by NGF in term depression of primary afferent neurotransmission in the rat
primary sensory neurons after inflammation increases TRPV1 levels spinal cord. J Neurosci. 1993;13(12):5228-5241.
and maintains heat hyperalgesia. Neuron. 2002;36(1):57-68. 63. Liu XG, Sandkuhler J. Long-term potentiation of C-fiber–evoked
47. Tan ZY, Piekarz AD, Priest BT, et al. Tetrodotoxin-resistant sodium potentials in the rat spinal dorsal horn is prevented by spinal
channels in sensory neurons generate slow resurgent currents that N-methyl-d-aspartic acid receptor blockage. Neurosci Lett. 1995;
are enhanced by inflammatory mediators. J Neurosci. 2014;34(21): 191(1–2):43-46.
7190-7197. 64. Zhou H-Y, Chen S-R, Pan H-L. Targeting N-methyl-d-aspartate
48. Cox JJ, Reimann F, Nicholas AK, et al. An SCN9A channelopa- receptors for treatment of neuropathic pain. Expert Rev Clin Phar-
thy causes congenital inability to experience pain. Nature. 2006; macol. 2011;4(3):379-388.
444(7121):894-898. 65. Millan MJ. Descending control of pain. Prog Neurobiol. 2002;66(6):
49. Dib-Hajj SD, Yang Y, Waxman SG. Genetics and molecular patho- 355-474.
physiology of Na(v)1.7-related pain syndromes. Adv Genet. 2008;63: 66. Yaksh TL. Pharmacology and mechanisms of opioid analgesic activ-
85-110. ity. Acta Anaesthesiol Scand. 1997;41(1 Pt 2):94-111.
50. Nassar MA, Stirling LC, Forlani G, et al. Nociceptor-specific gene 67. Zeilhofer HU. Loss of glycinergic and GABAergic inhibition in
deletion reveals a major role for Nav1.7 (PN1) in acute and inflam- chronic pain—contributions of inflammation and microglia. Int
matory pain. Proc Natl Acad Sci U S A. 2004;101(34):12706-12711. Immunopharmacol. 2008;8(2):182-187.
51. Davies A, Hendrich J, Van Minh AT, et al. Functional biology of 68. Coull JA, Beggs S, Boudreau D, et al. BDNF from microglia causes
the alpha(2)delta subunits of voltage-gated calcium channels. Trends the shift in neuronal anion gradient underlying neuropathic pain.
Pharmacol Sci. 2007;28(5):220-228. Nature. 2005;438(7070):1017-1021.
52. Liu B, Linley JE, Du X, et al. The acute nociceptive signals induced 69. Hayes AG, Arendt-Nielsen L, Tate S. Multiple mechanisms have
by bradykinin in rat sensory neurons are mediated by inhibition of been tested in pain—how can we improve the chances of success?
M-type K+ channels and activation of Ca2+-activated Cl− channels. Curr Opin Pharmacol. 2014;14:11-17.
J Clin Invest. 2010;120(4):1240-1252. 70. Carlson JD, Maire JJ, Martenson ME, et al. Sensitization of pain-
53. Brown DA, Passmore GM. Some new insights into the molecular modulating neurons in the rostral ventromedial medulla after
mechanisms of pain perception. J Clin Invest. 2010;120(5):1380-1383. peripheral nerve injury. J Neurosci. 2007;27(48):13222-13231.
54. Vink S, Alewood PF. Targeting voltage-gated calcium channels: 71. Cleary DR, Heinricher MM. Adaptations in responsiveness of brain-
developments in peptide and small-molecule inhibitors for the treat- stem pain-modulating neurons in acute compared with chronic
ment of neuropathic pain. Br J Pharmacol. 2012;167(5):970-989. inflammation. Pain. 2013;154(6):845-855.
PART 2 Nonsurgical Therapy

168 Approach to the Patient with Chronic Pain


Joshua M. Rosenow

Evaluating patients with chronic pain can be a challenging reassure the patient that obtaining this information is a key part
exercise for even the experienced clinician. Many of these of determining both why the prior treatments have not succeeded
patients have been through numerous previous evaluations and and whether there are any further surgical treatments that
treatments and have experienced many disappointing outcomes. may provide the desired outcome. However, the neurosurgeon
Despite this, they may continue to have expectations that are dif- should also be focused in eliciting these details so as to avoid
ficult to meet. This chapter contains an overview of some of the wasting time on irrelevant details. Also, the patient should under-
considerations that need to be taken into account in evaluating stand that more than a single visit may be necessary for a full
and treating these patients. This process may be time consuming, evaluation and to determine a therapeutic course of action.
but it is time well spent, insofar as a hasty encounter will leave Further diagnostic testing or other consultative evaluations, such
many issues unresolved on both sides of the physician-patient as psychological or neurophysiologic testing or further imaging,
relationship. may also be necessary.
The failure of previous medical or surgical attempts to relieve
pain may be attributable to the failure of the treatment itself, but
BEFORE THE PATIENT’S ARRIVAL it might also be related to a variety of underlying psychosocial
Although not an absolute necessity, a letter from the patient’s issues. Pain relief that is judged adequate by a previous physician
referring physician often provides valuable insight into the may not be so according to the patient. Possible reasons for prior
patient’s state of mind, as well as the pain complaint. In addition, treatment failure are listed in Box 168-1.
this also helps focus the encounter, insofar as the patient may
have numerous somatic complaints. Of importance is that it also
establishes a line of communication with the referring physician
Obtaining a History
and aids in the coordination of care.
The physician should obtain as complete a record of prior
General Aspects
treatments and outcomes as possible, including operative reports, A detailed history may be the most helpful factor in establishing
psychological reports, results of diagnostic testing (e.g., electro- a diagnosis and forming a treatment plan. The history provides
myography, discography), imaging studies and reports (before important information not only about the possible mechanisms
and after each surgical procedure), specialist consultations, physi- and pathophysiologic aspects of the patient’s pain but also about
cal therapy summaries, and relevant office notes. It is also helpful the emotional and psychological status of the patient. As previ-
to know whether the patient has already applied for long-term ously stated, the physician must guide the interview while being
worker’s compensation or social security disability, as well as flexible enough to allow the patient to volunteer valuable infor-
whether litigation is involved. mation. Keeping sometimes complex patient histories chrono-
Of note, while obtaining comprehensive prior records is logically organized is helpful.
useful, this may result in an overwhelming amount of data. The Prior failures may also have made the patient distrustful of
neurosurgeon needs to become facile at extracting the key docu- physicians and suspicious of their motives for suggesting inter-
ments that will provide the highest yield information and retain- ventional treatments. Such distrust and suspicion need to be
ing the rest as reference material. overcome for the physician-patient relationship to be truly thera-
Sending the patient a document explaining the policies and peutic. Although the patient needs to understand that the neuro-
procedures of the clinic before the visit establishes ground rules surgeon must believe the patient, the neurosurgeon should also
on such items as missed appointments, medication prescribing listen objectively.
and refills, general time frame for routine return phone calls, and
paperwork completion.
Pain History and Onset
Once an understanding of the current pain situation is
THE INITIAL VISIT established—including standard factors such as location, quality,
Understanding the patient’s goals is crucial not just for the initial radiation, duration, and exacerbating and alleviating factors—the
encounter but also for the physician-patient relationship. Con- pain may be analyzed in relation to the history.
versely, the patient must understand the capabilities and limits of Determining the evolution of the original pain syndrome into
the relationship (which parts of care the evaluating physician may the current one allows the physician to determine whether this
or may not be willing to assume) to avoid future misunderstand- pain represents new pain or a continuation/progression of an
ings that can erode the patient’s trust in the physician. For original pain complaint. What prior treatments were used? What
example, a patient may expect the evaluating physician to assume effect on the pain did they have? Were prior therapies inappropri-
prescribing responsibility for chronic opioids or to aid in the ate (possibly guided by the wrong diagnosis), or were they just
patient’s quest for disability benefits. not used to their fullest extent?
Patients in pain have already seen numerous physicians, to If the current pain is substantially different from the original
whom they have already explained their problems repetitively, pain, this may imply that a different process may be responsible
and may become disenchanted or angry at the thought of having for the current pain or that the current pain is the result of a treat-
to go through this again. To avoid this, the neurosurgeon should ment side effect. Patients often undergo misguided treatments for
1380
CHAPTER 168  Approach to the Patient with Chronic Pain 1381

whose pain either never varies from 10 or is rated as “11” on a


BOX 168-1  Possible Reasons for Prior Treatment Failure scale of 0 to 10. 168
Pain may be classified as localized, radiating, or referred.
• Incorrect diagnosis
Localized pain continues at the same location as its origination.
• Incorrect choice of therapy (medical or surgical)
This pain may be associated with other anatomic and sensory
• Incorrect application of therapy (wrong level, wrong site)
changes at the site of pain, such as hyperalgesia, wind-up hyper-
• Failure to apply the therapy to the full extent necessary for
pathia, color change, and trophic changes of the skin. Pain may
symptomatic relief (i.e., declaring a medication to have failed
also be described as radiating along the distribution of a nerve
before increasing the dose to a sufficient extent; not
root’s dermatome or the innervation of a peripheral nerve. This
sufficiently decompressing a stenotic spine)
is distinct from pain that is referred from a deep visceral structure
• Side effects limiting full application of a treatment
to a separate distinct anatomic location. Classic examples of
• Treatment-related side effect (e.g., adjacent segment
referred pain are back pain or superficial abdominal pain from
degeneration)
chronic pancreatitis and scapular pain from cholecystitis.
• Patient factors (health, anatomy, allergies) that prevent full
application of a treatment
• Intraprocedural complication Pain Treatment History
• Technical failure of procedure (e.g., nonunion)
A recounting of past medical and interventional treatments and
• Incorrect expectations of outcome (either on the part of the
the response to each is an invaluable part of the evaluation. A
patient or poor expectation management on the part of the
thorough analysis of this part of the history prevents the clinician
physician)
from repeating past failures. Which classes of medications have
• Medicolegal factors
been tried? Were the medication trials adequate? Were interven-
• Occupational factors
tional techniques used appropriately? Did each subsequent treat-
• Psychological and social factors
ment follow logically on the basis of past information? Has every
treatment failed to provide any relief? Moreover, this history
helps to ascertain patient compliance and assess for causes of past
treatment failures. The clinician needs to take a very critical view
of this section of the history, always asking why treatments failed
pain syndromes. These treatments can themselves cause further and whether the neurosurgeon can offer anything else to the
pain. As previously stated, procedural complications can also patient.
result in pain that is unlike the presenting pain (as with deaf- Patients may not recall each treatment and outcome. For
ferentation pain). The physician should be cautious in treating instance, the neurosurgeon should not waste valuable time forcing
patients whose pain changes significantly in location and charac- a patient to remember the outcome from each of their multitude
ter after each treatment. of injections if it is clear that all the other similar injections pro-
Was an obvious inciting incident associated with the pain? vided only short-term relief at best.
Was this mechanism consistent with the patient’s original com-
plaints? The location and distribution, quality, intensity or sever-
ity, and duration of the first pain should be ascertained. Were
Medical and Surgical History
there any other associated neurological symptoms at the time, Concomitant medical conditions can adversely affect both
and did any of these develop in a subacute manner? Was any medical and surgical treatments. Obesity, diabetes, hypertension,
treatment applied immediately? hypothyroidism, chronic obstructive pulmonary disease, and
Pain that is the result of a workplace-related activity presents inflammatory arthritis can complicate therapy. Patients fre-
a special challenge because of the complexities unique to worker’s quently do not mention conditions they do not consider impor-
compensation claims. In such cases, the history needs to be docu- tant or omit conditions they consider treated (e.g., not mentioning
mented even more fastidiously, including the exact time and date hypertension because they are taking medication for it and no
of the injury. Other information that should be obtained includes longer consider it an active medical problem). For completeness,
whether the patient continued to work after the injury, when and the inquiry should proceed through a comprehensive list of organ
to whom the injury was reported, and the response of the patient’s systems.
employer to the injury with regard to the patient’s work status. If the patient has undergone multiple surgical procedures that
The physician should also ascertain the effect of the pain on failed to ameliorate their pain syndrome, did they also experience
the patient’s daily life. To accomplish this, the patient should surgical failures for other conditions as well? Intrinsic patient
provide a timeline of an average day. This provides important medical factors, poor surgical techniques, unrealistic expecta-
insight into the psychological effects of the pain on the patient’s tions, insistence on unnecessary surgical treatments, and bad
life and the level of disability experienced. fortune may all reduce the likelihood of a good outcome. It is
important to ascertain whether the patient appears compliant
with their physician instructions for their other ailments. Poor
Pain Characteristics compliance may also contribute to consistently disappointing
The patient should be asked to describe in detail the character- results from health interventions.
istics of the pain at the time of evaluation and to indicate whether
these have changed since the onset of the pain syndrome or after
any prior procedures. Factors that have no effect on the pain are
Family History
important as well. The neurosurgeon should inquire about such Patterns may be recognized from this effort. Have multiple
factors as stress and other emotional disturbances, movement, family members undergone spinal surgery? Are they all employed
pressure, heat or cold, coughing, sneezing, straining, and deep in high-impact professions? Is there is family tendency to develop
breathing. The 0-to-10 visual analogue scale (VAS) is the most spinal degeneration or stenosis? Are multiple family members
commonly used pain rating scale, even if it is not the most robust receiving disability insurance? Chronic pain behavior may be a
measure. It is often more useful to collect the visual analogue learned trait within families.1 Cohort studies have shown that
scale by asking the patient to mark the level of pain on an unla- a history of child abuse may increase pain and pain-related
beled 10-cm line. Physicians should be wary of those patients behaviors.1-4
1382 SECTION 6  Pain

of neurosurgical evaluation. The neurosurgeon must still be vigi-


Social and Psychological History lant for signs such as weakness and pathologic reflexes that may
A complete psychological and psychosocial assessment is crucial indicate conditions such as nerve root or spinal cord compression
for the overall management of the patient with chronic pain.5-9 that could warrant urgent further evaluation or treatment. These
The psychological and psychosocial part of the history helps findings should not be discounted or neglected just because the
determine the contribution of affective or environmental factors patient may have already seen other physicians or may have had
to the patient’s pain syndrome. Referral to a psychologist with other surgical procedures.
special expertise in the evaluation and treatment of patients with The central sensitization that occurs in many patients with
chronic pain is an essential part of the total care of these patients. chronic pain can result in certain characteristic exam findings.
At the minimum, this part of the history should include a listing Central sensitization affects the wide dynamic range (WDR)
of current and prior psychological and psychiatric illnesses. neurons in the dorsal horn of the spinal cord that receive input
Special attention should be paid to depression, a common condi- from both A beta and C fibers. In neuropathic pain states, A beta
tion in patients with chronic pain. The astute neurosurgeon will fiber depolarization also results in stimulation of these other
delve deeper into this issue than simply inquire about the patient’s fibers, resulting in allodynia, the perception of pain from light
mood. The patient’s daily schedule provides important data about touch. Patients with chronic pain may exhibit generalized hyper-
depression. Other clues are irritability, insomnia, abulia, weight pathia (exaggerated and prolonged reactions to painful stimuli).
gain or loss, and suicidal ideation. The patient’s family may An example of hyperpathia is the perception that a pinprick is
provide important perspective as well. intensely painful over the entire body. Repetitive stimulation of
The neurosurgeon should also explore the patient’s vocational C fibers may result in an augmented response to each subsequent
status and vocational stressors, including compensation-litigation stimulus, a process known as wind-up. Repetitive light stroking of
issues. These can affect the patient’s motivation and outcome of the painful area is interpreted by the patient as increasingly
treatment.10-13 painful with each iteration.
Although the history should include current prescription Red flags on examination should engender skepticism in the
drugs, an appropriately thorough history should also include examiner. These may include Waddell’s signs15-17 for nonphysio-
over-the-counter and alternative (e.g., herbal) medicines. Infor- logic back pain and other inconsistencies on physical and neuro-
mation about illicit pharmaceutical use (both current and past) logical examination. For instance, does a patient with apparent
should be elicited by direct questioning, even with specific ques- ankle dorsiflexor weakness when tested seated have the ability to
tions about the most commonly used abused pharmaceuticals. It heel walk without much difficulty? Do the findings fail to conform
is important to determine not only what the patient is using but to a peripheral or spinal nerve distribution? Fishbain and col-
also whether he or she is using it appropriately. For instance, is leagues18 published a structured review of the medical literature
the patient using another person’s prescription medication for his on the validity of Waddell’s signs and noted that Waddell’s signs
or her own use? Is the patient using medication up earlier than are not reliable as a discriminator of physiologic from nonphysi-
prescribed? Is he or she obtaining substitutes illicitly? A history ologic pain.
of tobacco and alcohol use, including type and frequency of use
of each, should be included as well. Physical and psychological
dependence on drugs and alcohol is a common impediment to
Formulating a Treatment Plan
treatment. The patient with chronic pain requires an individualized treat-
A useful exercise is to conceptualize the patient’s pain syn- ment plan. First, the treatment team should be able to work in a
drome into separate components of pain and suffering. Pain is cohesive manner and present a united front to the patient. Rep-
the purely physical and physiologic component of the syndrome, resentative members of a pain team are listed in Box 168-2. Lines
whereas suffering includes the individual’s reaction to the pain, of communication among the clinicians involved should be clear
as well the pain’s effect on the rest of the psychological and emo- and open. The best method to ensure this is a regularly scheduled
tional environment. Suffering includes depression, anxiety, loss of patient management meeting attended by the pain team. A team
self-esteem, failure of relationships, past emotional and physical consensus may be reached and be discussed with the patient and
abuses that shape the pain response, poor coping strategies, and family.
withdrawal from friends and family. Surgical interventions may Next, in defining a plan, it is important to outline the plan in
be an excellent method for dealing with the pain, but they are as much specificity as possible and then stick to it. The plan
inadequate at handling an impressive suffering component. should include steps for dealing with medication-related side
Again, a psychologist skilled at evaluating patients in pain is adept effects and procedural failures. This gives the patient a clear
at determining the balance between the pain and suffering com-
ponents, along with identifying maladaptive coping strategies
and other pitfalls of which the surgeon should be aware before
embarking on a therapeutic relationship with the patient.
Although a small population of individuals have no physical BOX 168-2  Members of a Pain Team
basis for their pain complaints,14 the majority of patients with
chronic pain have a complex interplay between psychological and Surgical specialist(s)
physical components of their pain syndrome in varying propor- Anesthesia
tions. Among the neurosurgeon’s challenges in evaluating and Neurology
managing patients with chronic pain are first determining the Medical specialists (e.g., internal medicine, oncology)
relative balance between these factors and then using that estima- Physiatry
tion to drive the selection of an individualized combination of Pain psychology
medical, interventional, surgical, and psychological treatments Psychiatry
for each patient. Nursing
Physical therapy
Occupational/vocational therapy
Physical and Neurological Examinations Social work
The examination of the patient with chronic pain is essentially Addiction medicine
no different from that of a patient presenting for any other type
CHAPTER 168  Approach to the Patient with Chronic Pain 1383

understanding of what steps will be taken and in what order. for a patient to return to the same line of work after treatment,
Moreover, the patient will understand what the expected out- but it is critical to understand whether the patient believes that 168
comes are for each step and what will be done if the actual he or she will do so. The clinician’s and patient’s ideas of a “good”
outcome does not meet expectations at each step. As part of outcome may be divergent, and this disparity needs to be under-
outlining this plan, a treatment contract may be signed by the stood and managed.
patient and clinician.19-21 If a contract is signed, the patient should Goal and expectation setting should include lifestyle modifica-
have a copy to keep. Contracts should denote the obligations of tion as needed. Smoking and obesity are negative determinants
both parties, as well as the consequences for violations. of outcome,22-24 and the patient’s willingness to resolve these
In formulating a plan with the patient, the neurosurgeon may problems is a good determinant of the patient’s level of motiva-
be requested to prescribe medications (either opioid or other- tion. Return visits may need to be scheduled to evaluate progress
wise). This may introduce a complication into an otherwise on certain goals or to discuss consultation or imaging results
straightforward evaluation. As a consultant, the neurosurgeon before the physician agrees to embark on a therapeutic relation-
may not see his or her role as one of assuming chronic care ship. Return visits also allow the physician to assess the amount
of the patient. Moreover, many neurosurgeons are not skilled in of material internalized by the patient from previous visits.
the long-term management of patients on what are often regi- Written materials given to the patient to read at home may
mens consisting of multiple classes of medications (e.g., opioids, aid in increasing retention of material discussed during the
selective serotonin reuptake inhibitors [SSRIs] or serotonin- office visit.
norepinephrine reuptake inhibitors [SNRIs], anticonvulsants, Some patients may not be accepted for care. A true pain team
nonsteroidal anti-inflammatory drugs [NSAIDs], muscle relax- is not just a dumping ground for all patients whom other special-
ants). Even if not requesting that the surgeon manage the entire ists do not want to manage. Patients with a significant ongoing
pain medication system, the patient may ask the surgeon to pre- pattern of pharmaceutical misuse, whether legal or illegal, must
scribe something in addition to the current regimen. In general, first resolve that issue. Moreover, severe overwhelming emotional
neurosurgeons should be cautious in these situations. It is always and psychological problems also need to be brought under
best that the patient’s medication treatment be handled by one control, especially before nonurgent surgical procedures.
provider so as to avoid inadvertent overdoses, medication interac-
tions, and generalized confusion, as well as to reduce the likeli-
SUGGESTED READINGS
hood of unscrupulous behavior by patients. The surgeon may Burns JW, Sherman ML, Devine J, et al. Association between workers’
state that he or she will communicate and coordinate any medica- compensation and outcome following multidisciplinary treatment for
tion changes with the prescribing provider while emphasizing the chronic pain: roles of mediators and moderators. Clin J Pain. 1995;
importance of the single person to contact for medications. Most 11(2):94-102.
states now have prescription monitoring databases that allow a Doleys DM. Psychologic evaluation for patients undergoing neuroaug-
clinician to see where, when, and from which physician patients mentative procedures. Neurosurg Clin N Am. 2003;14(3):409-417.
receive their medications. Neurosurgeons should be familiar with Doleys DM. Psychological factors in spinal cord stimulation therapy:
their own state’s system and how to obtain access to these data. brief review and discussion. Neurosurg Focus. 2006;21(6):E1.
If there is any concern about the patient’s use of medications, this Fishbain DA, Cole B, Cutler RB, et al. A structured evidence-based
review on the meaning of nonorganic physical signs: Waddell signs.
database should be consulted and the other providers brought in Pain Med. 2003;4(2):141-181.
to coordinate a response. Fishman SM, Mahajan G, Jung SW, et al. The trilateral opioid contract.
Setting expectations of treatment is a crucial part of the overall Bridging the pain clinic and the primary care physician through the
plan. In many cases, the patient’s expectations were not met by opioid contract. J Pain Symptom Manage. 2002;24(3):335-344.
past medical and surgical treatments, either because the treat- Turner JA, Romano JM. Psychological and psychosocial evaluation. In:
ments did not deliver expected results or because the patient’s Loeser JD, ed. Bonica’s Management of Pain. Philadelphia: Lippincott
expectations were not realistic. Substantial time should be spent Williams & Wilkins; 2001.
discussing realistic outcomes for each care step, as well as for the
overall plan of care. For instance, it may or may not be realistic See a full reference list on ExpertConsult.com
CHAPTER 168  Approach to the Patient with Chronic Pain 1383.e1

REFERENCES 13. Burns JW, Sherman ML, Devine J, et al. Association between
1. Craig KD. Social modeling influences: pain in context. In: Sternbach workers’ compensation and outcome following multidisciplinary 168
RA, ed. Psychology of Pain. 2nd ed. New York: Raven Press; 1986: treatment for chronic pain: roles of mediators and moderators. Clin
67-95. J Pain. 1995;11(2):94-102.
2. Koss MP, Heslet L. Somatic consequences of violence against 14. Sullivan MD, Turk DC. Psychiatric illness, depression, and
women. Arch Fam Med. 1992;1(1):53-59. psychogenic pain. In: Loeser JD, ed. Bonica’s Management of Pain.
3. Lampe A, Solder E, Ennemoser A, et al. Chronic pelvic pain and Philadelphia: Lippincott Williams & Wilkins; 2001.
previous sexual abuse. Obstet Gynecol. 2000;96(6):929-933. 15. Waddell G, Bircher M, Finlayson D, et al. Symptoms and signs:
4. Walker EA, Stenchever MA. Sexual victimization and chronic pelvic physical disease or illness behaviour? Br Med J (Clin Res Ed).
pain. Obstet Gynecol Clin North Am. 1993;20(4):795-807. 1984;289(6447):739-741.
5. Turner JA, Romano JM. Psychological and psychosocial evaluation. 16. Waddell G, McCulloch JA, Kummel E, et al. Nonorganic physical
In: Loeser JD, ed. Bonica’s Management of Pain. Philadelphia: signs in low-back pain. Spine. 1980;5(2):117-125.
Lippincott Williams & Wilkins; 2001. 17. Waddell G, Pilowsky I, Bond MR. Clinical assessment and interpre-
6. Doleys DM. Psychologic evaluation for patients undergoing neu- tation of abnormal illness behaviour in low back pain. Pain. 1989;
roaugmentative procedures. Neurosurg Clin N Am. 2003;14(3): 39(1):41-53.
409-417. 18. Fishbain DA, Cole B, Cutler RB, et al. A structured evidence-based
7. Doleys DM. Psychological factors in spinal cord stimulation therapy: review on the meaning of nonorganic physical signs: Waddell signs.
brief review and discussion. Neurosurg Focus. 2006;21(6):E1. Pain Med. 2003;4(2):141-181.
8. Doleys DM, Dinoff BL. Psychological aspects of interventional 19. Doleys DM, Rickman L. Other benefits of an opioid “agreement.”
therapy. Anesthesiol Clin North America. 2003;21(4):767-783. J Pain Symptom Manage. 2003;25(5):402-403.
9. Seres G, Kovacs Z, Kovacs A, et al. Different associations of health 20. Fishman SM, Kreis PG. The opioid contract. Clin J Pain. 2002;
related quality of life with pain, psychological distress and coping 18(4 suppl):S70-S75.
strategies in patients with irritable bowel syndrome and inflam- 21. Fishman SM, Mahajan G, Jung SW, et al. The trilateral opioid con-
matory bowel disorder. J Clin Psychol Med Settings. 2008;15(4): tract. Bridging the pain clinic and the primary care physician through
287-295. the opioid contract. J Pain Symptom Manage. 2002;24(3):335-344.
10. Klekamp J, McCarty E, Spengler DM. Results of elective lumbar 22. Jamison RN, Stetson BA, Parris WC. The relationship between ciga-
discectomy for patients involved in the workers’ compensation rette smoking and chronic low back pain. Addict Behav. 1991;16(3–4):
system. J Spinal Disord. 1998;11(4):277-282. 103-110.
11. LaCaille RA, DeBerard MS, LaCaille LJ, et al. Obesity and litigation 23. Vogt MT, Hanscom B, Lauerman WC, et al. Influence of smoking
predict workers’ compensation costs associated with interbody cage on the health status of spinal patients: the National Spine Network
lumbar fusion. Spine J. 2007;7(3):266-272. database. Spine. 2002;27(3):313-319.
12. Vaccaro AR, Ring D, Scuderi G, et al. Predictors of outcome in 24. Patel N, Bagan B, Vadera S, et al. Obesity and spine surgery: rela-
patients with chronic back pain and low-grade spondylolisthesis. tion to perioperative complications. J Neurosurg Spine. 2007;6(4):
Spine. 1997;22(17):2030-2034. 291-297.
169 Pharmacologic Treatment of Pain
Diaa Bahgat, Heather N. Pinckard-Dover, and Erika A. Petersen

Pain—the unpleasant physical and emotional experience associ- channels, cause conformational changes in transducer proteins,
ated with tissue damage, whether impending or actual—involves and increase calcium influx in second-messenger systems, all
sensations transmitted from the periphery to the central nervous leading to sensitization of already activated nociceptors and the
system for processing and perception. The experience of pain, activation of dormant ones.
however, is different for each individual and is influenced by Nociceptive neurons transmit their signal to the dorsal horn
physical, mental, cultural, and religious factors and by the of the spinal cord, where the signal is modified by inhibitory and
individual’s current situation. Because the experience of pain is excitatory interneurons and by descending inhibitory neurons
diverse, treatment can be difficult. Accurate diagnosis and a thor- from the central nervous system. If this inhibition is lost, it can
ough understanding of the nature of the pain to be addressed lead to peripheral sensitization. The C fibers are temporally sum-
facilitate appropriate treatment recommendations. Many attempts mated in the dorsal horn, and the combined effects of the fiber
have been made to categorize pain. Some systems distinguish signal, interneurons, and descending input result in a new signal
between acute and chronic pain; some are based on the difference that is transmitted from the dorsal horn to the thalamus through
between cancer and noncancer pain causes; some differentiate by the spinothalamic tract.
diagnosis; and some are attempts to identify the processes under- The thalamus is not the only location where pain is processed.
lying the pain. Pain signals are also received by the periaqueductal gray matter
Acute pain is a reaction to focal peripheral nerve or tissue in the midbrain, somatosensory cortex, periventricular gray
injury and usually resolves as tissue heals and inflammation sub- matter, ventral medulla, nucleus raphe magnus, limbic system,
sides. Chronic pain persists beyond the usual course of injury and and insula. The importance of the pain signal is perceived through
healing and does not respond to routine pain control measures.1 a pathway involving the insula, striatum, parietal association
Pain directly related to cancer can be caused by multiple pro- cortex, and lateral prefrontal cortex. The perception of pain can
cesses, including vertebral involvement, and can take the form of be suppressed by supratentorial structures that release endor-
referred pain. Certain pain conditions such as trigeminal neural- phins, enkephalins, dynorphins, norepinepherine, γ-aminobutyric
gia and diabetic neuropathy have been shown to respond better acid (GABA), and serotonin. All of these neurotransmitters
to specific medications; therefore, appropriate diagnosis governs are also the targets for specific medications in the treatment
treatment strategy. of pain.3,4
On the basis of underlying characteristics, pain can be catego-
rized as nociceptive, neuropathic, or inflammatory. Nociceptive
pain is usually acute and comparable to the degree of damage to
PHARMACOLOGIC TREATMENT OF PAIN
tissue. Neuropathic pain is caused by nerve dysfunction related As knowledge about the underlying mechanisms of pain increases,
to injury or inappropriate signaling. Usually chronic and the so has the ability to apply medications to target these mechanisms.
most resistant to treatment, neuropathic pain is described as The main classes of treatment currently include nonsteroidal
burning or lancinating. Inflammatory pain results from release anti-inflammatory drugs (NSAIDs), acetaminophen, antidepres-
of inflammatory mediators and hypersensitization of nerve sants, ion channel blockers, and opioids. These medications can
endings to innocuous stimulus, and its degree can differ between be delivered through multiple routes of administration. Their
patients.2 Regardless of the system used to approach each case, effectiveness, dosing, and tolerability often vary with the route
it is important for the physician to understand the basic patho- of administration.
physiologic process behind pain, the subjective influences on
pain, and the various tools designed to relieve pain and how
they work.
Nonsteroidal Anti-inflammatory Drugs
NSAIDs are a large group of medications that have antipyretic,
analgesic, and anti-inflammatory effects. They are divided into
PATHOPHYSIOLOGY selective cyclooxygenase-2 (COX-2) inhibitors and nonselective
The exact mechanisms involved in pain signaling and perception cyclooxygenase (COX) inhibitors. COX is an enzyme responsible
are not known, but research has led to improved understanding for prostaglandin synthesis from arachidonic acid. Inhibition of
of a few of the underlying processes, which allows drug therapies COX results in downregulation of the synthesis of prostaglan-
to be more targeted. Nociceptive neurons are now known to be din and thromboxane. Common nonselective COX inhibitors
responsible for the conversion of mechanical, chemical, or include aspirin (acetylsalicylic acid), ibuprofen, and naproxen.
thermal stimuli to electrical signals, which is the process of trans- Common selective COX-2 inhibitors include diclofenac and
duction. Nociceptive neurons are usually bipolar and can be ketorolac.
either myelinated or unmyelinated. The myelinated A delta–fiber NSAIDs interfere with the inflammatory response to tissue
nociceptive neurons transmit their signals rapidly, whereas damage and thus are best used for acute nociceptive, neuro-
the unmyelinated C-fiber nociceptive neurons transmit their pathic, or inflammatory pain. They are generally well tolerated
signal more slowly. Both types propagate the signals through but can have some gastrointestinal side effects nonselective COX
voltage-gated sodium channels, which are the target of multiple inhibitors have more pronounced gastrointestinal side effects
medications. When tissue damage ensues, multiple inflammatory than do selective COX-2 inhibitors. Some manufacturers have
mediators are released, including histamine, serotonin, bradyki- started combining NSAIDs with proton pump inhibitor or his-
nin, substance P, leukotrienes, cytokines, neurotropic factors, and tamine blockers in an aim to offset these side effects. Because of
prostaglandins. These mediators upregulate voltage-gated sodium NSAIDs’ effect on thromboxane A (an enzyme that plays a
1384
CHAPTER 169  Pharmacologic Treatment of Pain 1385

key role in the aggregation of platelets), NSAIDs should be ankylosing spondylitis. Patients with headaches showed the
prescribed with special caution in the preoperative period. greatest response to TCAs among classes of antidepressants.17 169
There are reported drug-drug interactions between NSAIDs Despite their usefulness in multiple pain disorders, TCAs have
and angiotensin-converting enzyme inhibitors, corticosteroids, not been found useful in the treatment of neuropathy related to
warfarin, sulfonylurea, and methotrexate. Combining these medi- human immunodeficiency virus (HIV) infection, cisplatin-related
cations causes a risk of bradycardia and gastrointestinal ulceration neuropathy, neuropathic cancer pain, phantom limb pain, or
and increases the risk of bleeding. chronic lumbar root pain.17
TCAs have multiple side effects, and patients taking them
must be monitored closely. Before starting TCAs, patients
Acetaminophen should complete a cardiac workup, including orthostatic blood
Like NSAIDs, acetaminophen (paracetamol) has antipyretic and pressure measurements and electrocardiography for measure-
analgesic effects. It does not, however, have any anti-inflammatory ment of the corrected QT (QTc) interval. TCA therapy should
effects. The mechanism of action of acetaminophen is poorly always be started at a low dosage and titrated upward every 3 to
understood, but it is believed to reduce the oxidized form of COX 7 days. An adequate trial requires 6 weeks of therapy with 1 to
and selectively inhibit prostaglandin H2 synthase. Its metabolites 2 weeks at the maximum tolerated dosage. Patients’ serum
may also block the uptake of anandamide, therefore inhibiting drug concentrations should be monitored, and routine electro-
vanilloid receptor activation in nociceptors. There is also evi- cardiograms to monitor the QTc interval may be obtained.
dence that acetaminophen decreases prostaglandin E2 synthesis Side effects to watch for include sedation, dry mouth, blurred
in the brain by COX-3 inhibition. It is generally well tolerated, vision, weight gain, urinary retention, sinus tachycardia and ven-
the most noted side effects being allergic reactions and effects on tricular ectopy, and QTc interval prolongation. The medications
renal function. Acetaminophen causes minimal inhibition of can also potentiate the actions of cytochrome P-450, and so
platelet aggregation. It does, however, carry risk of hepatotoxicity the patient’s other medications must be checked for interactions
when taken in large quantities, as in overdose; therefore, the daily with TCAs.
dose should be less than 4 g. Like TCAs, venlafaxine (an SNRI) is effective in treating neu-
ropathic pain; however, it is more tolerable and has fewer side
effects. Venlafaxine and other SNRIs have been found to be effec-
Antidepressants tive in the treatment of fibromyalgia. They also improve the poor
Three main classes of antidepressants are used for the sleep and fatigue associated with fibromyalgia.18,19 There are no
treatment of pain: tricyclic antidepressants (TCAs), serotonin- studies to support the use of SNRIs in low back pain. Venlafaxine,
norepinephrine reuptake inhibitors (SNRIs), and selective sero- however, has been show to be just as effective as amitriptyline for
tonin reuptake inhibitors (SSRIs). These drugs primarily increase migraine prophylaxis. The most common side effects experienced
the serotonin and norepinephrine available in the synaptic cleft, with SNRIs including nausea, vomiting, constipation, somno-
thereby reinforcing the descending inhibitory pathways from lence, dry mouth, hyperhidrosis, anorexia, weakness, agitation,
the central nervous system to nociceptive neurons in the dorsal diarrhea, hypertension, and hyponatremia. Venlafaxine has been
horn.5 They may also act by decreasing central prostaglandin associated with increased liver enzyme levels, and therefore liver
E2 and tumor necrosis factor α production, activating µ and δ function must be monitored. Because of the serotonergic effects
opioid receptors, increasing GABA type B receptor function, of SNRIs, there is a slight risk of serotonin syndrome: headache,
increasing adenosine availability and activating α1 receptors, agitation, mental confusion, sweating, hyperthermia, hyperten-
blocking calcium and voltage-gated sodium channels, activat- sion, tachycardia, diarrhea, myoclonus, tremor, hallucinations,
ing potassium channels, and inhibiting N-methyl-d-aspartate and coma.
(NMDA) receptors.5,6 The best analgesic effects are achieved by Although studies have shown that SSRIs are superior to
medications with primarily norepinephrine or mixed receptor placebo in the treatment of neuropathic pain and fibromyalgia,
activity.5,7,8 they are less effective than TCAs.11 They are, however, better
There is a high correlation between pain and depression. In tolerated; side effects include nausea, vomiting, anorexia, diar-
fact, in an epidemiologic study, Tunks and colleagues9 showed rhea, decreased libido, delayed orgasm, hyperhidrosis, somno-
that the presence of pain doubled, and in some cases almost lence, insomnia, drowsiness, fatigue, weight gain, increased risk
tripled, the likelihood of having depression, dysthymia, anxiety of bleeding, and syndrome of inappropriate diuretic hormone
disorder, agoraphobia or panic disorder, and social phobia.5 secretion. There is a small risk of serotonin syndrome. Unlike
Because of their analgesic effects, prescription of antidepres- TCAs and SNRIs, SSRIs are not effective in the treatment of
sants is not limited to individuals with depression or other psy- headache, low back pain, or rheumatoid arthritis.5
chological issues. Instead, the analgesic effect has been found to
be independent of the antidepressant effect and typically occurs
at lower dosages than those required for antidepressant
Antiepileptics
effect.5,10-12 Antiepileptics with varying mechanisms of action are commonly
TCAs such as amitriptyline and nortriptyline have become the used for the treatment of neuropathic pain. Some antiepileptics
“gold standard” of antidepressants for treating neuropathic work by blocking calcium channels on the nerve terminal, thereby
pain.5,13,14 Level 1 evidence supports the use of amitriptyline for preventing release of neurotransmitters into the synaptic cleft.
postherpetic neuralgia and polyneuropathy.5,14 Level 2 evidence Others target sodium channels to suppress ectopic discharges
supports the use of TCAs in the treatment of central neuropathic while preserving normal nerve conduction.
pain caused by stroke, spinal cord injury, or multiple sclerosis.15 Calcium channel blockers include zonisamide, ziconotide,
Other studies have shown that TCAs are helpful in the treatment levetiracetam, gabapentin, and pregabalin. Zonisamide is an anti-
of fibromyalgia, although they showed greater effects on the sleep epileptic used in the treatment of diabetic neuropathy,20,21 post-
disorders and fatigue than on pain ratings in this population. stroke central pain,20,22 and migraine prophylaxis.23 Because it is
TCAs have also been found to be as effective as NSAIDs in the usually a component of multidrug regimens, its individual effi-
treatment of low back pain.16 They have been found to have weak cacy and tolerability are not well known. Zonisamide is believed
analgesic effects in patients with rheumatoid arthritis.17 Amitrip- to act by blocking both voltage-gated and N-type calcium chan-
tyline, but not other TCAs, has been shown effective in the nels, reducing monoamine release. It has also been shown to work
treatment of pain, fatigue, and sleep disorder related to as a free radical scavenger. Ziconotide is a synthetic peptide
1386 SECTION 6  Pain

analogue of ω-conotoxin from marine snails that selectively some clinical scenarios. It has been effective in the treatment of
blocks N-type voltage-sensitive calcium channels. It is given diabetic neuropathy, sleep disorders, and tension, cluster, and
intrathecally and has been approved by the U.S. Food and Drug migraine headaches.20,32,33 Its effectiveness in other neuropathic
Administration for use in only patients with chronic severe pain disorders is unclear.
refractory to other therapies, including intrathecal morphine. It Two other medications work by blocking voltage-gated
has been used to treat complex regional pain syndrome, chronic sodium channels but are not antiepileptics: lidocaine and mexi-
pain from cancer and acquired immunodeficiency syndrome letine. Lidocaine is a local anesthetic and an antiarrhythmic
(AIDS), and trigeminal neuralgia.20,24,25 drug that has been used diagnostically for neuropathic pain.
Levetiracetam’s mechanism of action is not well known, but Petersen and associates34 showed that 78% of pain cases have
it has been shown to target N-type voltage-sensitive calcium good responses to intravenous lidocaine but that this treatment
channels and to modulate the dopaminergic, GABAergic, and did not provide long-term relief. Topical formulations, including
glutamatergic systems. Studies have shown it to be effective in patches and creams, have been used. They have good effect
the treatment of neoplastic plexopathies, peripheral neuropathy, locally but little to no systemic effect. They are beneficial in
and postherpetic neuralgia and to be useful in migraine prophy- the treatment of diabetic neuropathy, postherpetic neuralgia,
laxis.20,26 It is generally well tolerated; most common side effects other peripheral neuropathies, and chronic low back pain.34
are fatigue, dry eyes, and dizziness. The patches can cause local irritation and rash, which is why it
Both gabapentin and pregabalin work through binding the is recommended that patients wear patches for only 12 hours of
α2δ subunit of voltage-gated calcium channels, decreasing the the day and rotate location with each application. Mexiletine is
release of excitatory neurotransmitters such as glutamate, sub- an oral form of lidocaine that allows more prolonged pain relief
stance P, and calcitonin gene–related peptide. Their side effect in patients who have a positive response to the intravenous lido-
profiles and time to effectiveness differ; this is believed to be caine test. It is useful in the treatment of diabetic neuropathy,
attributable to pregabalin’s higher affinity for the α2δ subunit. phantom pain, fibromyalgia, myofascial pain, and spinal cord
Both medications are useful in the treatment of diabetic neuropa- injury.20
thy, peripheral neuropathy after spinal cord injury, and posther-
petic neuralgia.20 Gabapentin has also been shown effective in
the treatment of trigeminal neuralgia, HIV neuropathy, cancer-
Opioids
related neuropathy, multiple sclerosis, complex regional pain syn- Probably the most well-known and controversial class of analge-
drome, Eagle’s syndrome, and glossopharyngeal neuralgia.20 sic medications is that of the opioids. These medications activate
There are also data to support its use in treating postoperative µ receptors on the primary afferent nerves, dorsal horn neurons,
pain. Gabapentin is usually well tolerated, but patients may expe- and supraspinal pain centers. µ Receptors are located on both
rience dizziness and diarrhea, which typically subsides in 7 to sides of a synapse. At presynaptic µ receptors, opioids act to
10 days and can usually be prevented with slow tapering up of decrease neurotransmitter release; at postsynaptic µ receptors,
medication. Pregabalin is also well tolerated; common side effects they act to hyperpolarize the neuron by increasing potassium
are dizziness, somnolence, and mild to moderate peripheral conductance. Studies have shown than in patients with chronic
edema. Pregabalin tends to have a quicker onset of action, and central pain, pain intensity scores were significantly lower after
sometimes a patient may experience pain relief from the first opioid administration than with placebo. There is some evidence
doses. It has been shown to be effective in treatment of fibromy- of efficacy in the use of opioids for neuropathic pain, but the
algia, facial neuropathic pain, and neuropathic pain refractory to evidence is inconclusive because of small study size, bias, and
gabapentin.20 duration.35 Although opioids are best suited for the treatment of
Antiepileptics that treat pain by sodium channel blockade acute nociceptive and inflammatory pain for short durations,
include lamotrigine, carbamazepine, oxcarbazepine, and topira- their long-term use has not been studied adequately, but it has
mate. Lamotrigine works by blocking voltage-gated sodium become common practice.1,36 The use of opioids is cultural: in
channels, as well as suppressing calcium influx to the nerve some nations, such as Japan, there are very few prescriptions for
terminal and therefore decreasing excitatory neurotransmitter opioids37; although the United States has 4.6% of the world’s
release. It is used in the treatment of trigeminal neuralgia,27 population, more than 80% of the world’s oxycodone and hydro-
peripheral nerve or spinal cord injury,28,29 and HIV neuropa- codone was consumed in the United States in 2007.1 This trend
thy.20,30 Lamotrigine can have severe side effects, such as rash has continued: According to the International Narcotics Control
and Stevens-Johnson syndrome. It can also cause dizziness, som- Board (INCB), the United States consumed over six times the
nolence, nausea, and constipation. It is very important to start global mean of oxycodone in 2013.37a The most common opioids
with a low dosage, titrate up over 2 to 3 weeks, and monitor for are morphine, codeine, oxycodone, hydrocodone, fentanyl,
side effects. Carbamazepine also has rare severe side effects, meperidine, methadone, hydromorphone, propoxyphene, and
including aplastic anemia and agranulocytosis, and can have alfentanil. Opioids and their morphine equivalents are listed in
significant drug-drug interactions and central nervous system Table 169-1.
effects. Opioids have a wide range of frequent side effects, including
With the development of the better tolerated carbamazepine nausea, vomiting, constipation, and sedation. Other effects
analogue oxcarbazepine, carbamazepine is no longer the first-line include perceptual distortion, urinary retention, anorexia, dry
medication for trigeminal neuralgia. Oxcarbazepine, a sodium mouth, and respiratory depression. Addiction, habituation, and
channel blocker and neuronal membrane stabilizer, has the same hyperalgesia may occur with long-term use of opioids. Both the
efficacy as carbamazepine for the treatment of trigeminal neural- magnitude of the side effects and how well they are tolerated vary
gia but with fewer serious side effects.20,31 It has also been shown among patients; they also vary among different opioids and
effective in the treatment of diabetic neuropathy, postherpetic depending on the mode of delivery of the medication. Transder-
neuralgia, and complex regional pain syndrome. Although more mal fentanyl, for example, has been shown to produce less
tolerable than carbamazepine, oxcarbazepine can nonetheless constipation than do other opioids, including oxycodone and
cause dizziness, drowsiness, hypotension, nausea, and hyponatre- morphine.38-40 Long-term intrathecal morphine has been shown
mia; therefore, it also must be started at a low dosage and titrated to cause hypogonadotropic hypogonadism and other hormonal
up over a couple of weeks, and sodium levels must be monitored. disturbances.41
Topiramate has side effects similar to those of oxcarbazepine; Managing the side effects of opioids is vital for adequate pain
however, it can also cause weight loss, which can be beneficial in control; this can be done by symptomatic treatment, such as use
CHAPTER 169  Pharmacologic Treatment of Pain 1387

TABLE 169-1  Parenteral-Oral Opioid Equianalgesia Conversion


8 169
Duration Equianalgesic
Medication of Action Half-Life Dosage
7
Morphine 3-7 hr 1.5-2 hr 10 mg IM or IV
30 mg PO
Morphine, sustained 8-12 hr NA 30 mg PO 6
release
Oxycodone 4-6 hr NA 20 mg PO 5
Oxycodone, 8-12 hr NA 20 mg PO
controlled release

Rate
Fentanyl 1-2 hr 1.5-6 hr 100 µg IV or IM 4
15 µg transdermal
Hydromorphone 4-5 hr 2-3 hr 1.5 mg IV or IM 3
7.5 mg PO
Methadone 4-6 hr 2-3 hr 5 mg IM or IV
2
10-20 mg PO
Codeine 4-6 hr 3 hr 120 mg IM or IV
200 mg PO 1
Hydrocodone 4-8 hr 3.3-4.5 hr 30 mg PO
Meperidine 2-4 hr 3-4 hr 75 mg IM
0
300 mg PO
1999 2001 2003 2005 2007 2009
Propoxyphene 4-6 hr 6-12 hr 130-200 mg PO
The following dosages are equianalgesic but not standard dosing. Year
Tramadol 4-6 hr NA 300 mg PO OPR deaths/100,000 Treatment
Ketorolac 6 hr NA 10-30 mg IV or IM admissions/10,000
10 mg PO OPR sales kg/10,000
Ibuprofen 6-8 hr NA 1300 mg PO
Acetaminophen 4 hr NA 3900 mg PO
Aspirin 4 hr NA 3900 mg PO Figure 169-1. Rates of opioid pain reliever (OPR)–related
overdose deaths and treatment admissions and kilograms of
IM, intramuscularly; IV, intravenously; NA, not applicable; PO, orally.
OPR sold: United States, 1999-2010. Rates are age adjusted per
100,000 population for OPR-related deaths, per 10,000 population for
admissions for treatment of OPR abuse, and per 10,000 population
for kilograms of OPR sold. (From Paulozzi L, Jones C, Mack K, et al;
Centers for Disease Control and Prevention. Vital signs: overdoses of
of laxative for constipation, antihistamines for itching, and anti- prescription opioid analgesics—United States, 1999-2008. MMWR
emetics for nausea and vomiting. Another approach is titration Morb Mortal Wkly Rep. 2011;60[43]:1487-1492.)
of dosage and opioid rotation, which involves the switch from
one opioid to another in an effort to improve pain control and
reduce side effects.42 When opioids are rotated, it is important to TABLE 169-2  Mortality and ED Visits Related to Use of Prescription
use equivalent dosages, as well as provide breakthrough doses, Medication in 2010
which are generally 5% to 15% of the total daily dose.43 The most
effective way to minimize side effects is to treat with the lowest Prescription No. of Medication-
Medication Related Deaths No. of ED Visits†
possible effective dosage.
Opioid 16,651 420,000
Current Practices with Opioids and Their Consequences.  Benzodiazepine 6,497 425,000
The use of opioids has increased five times since the 1990s (Fig. Antidepressants* 3,889 NA
169-1),44 and these increases are reflected across all age groups Data from Behavioral Health Coordinating Committee, Prescription
and both genders (Fig. 169-2).45 Between 2007 and 2010, opioids Drug Abuse Subcommittee, U.S. Department of Health and Human
were prescribed in 2.6% of clinic visits.46 There has been an Services. Addressing Prescription Drug Abuse in the United States:
increase in mortality and morbidity with this rise in consumption; Current Activities and Future Opportunities. Atlanta: Centers for
according to statistics from 2011, more than 1.4 million deaths Disease Control and Prevention; 2013.
and emergency room visits were related to pharmaceutical misuse ED, emergency department; NA, not available.
*Usually associated with other medication.
or abuse (Table 169-2).47,48 The rapid increases in opioid prescrib- †
Alcohol was included in 18% of cases.
ing and associated opioid-related mortality have led to increased
awareness of tolerance, dependence, and addiction. It is of para-
mount importance to be able to differentiate between these
problems. attributed to activation of NMDA receptors and increased spinal
In clinical tolerance, increased dosages are needed to achieve dynorphin levels.52,53
adequate pain relief49; however, clinicians should bear in mind Dependence, on the other hand, presents with symptoms
that other factors, such as disease progression and depression, and sign of withdrawal: fatigue, diaphoresis, pupillary dilation,
may also lead to increased dosage requirements. Understanding anxiety, diarrhea, tachycardia, abdominal cramps, lacrimation,
the underlying cause of poor pain control will help guide therapy. and increased pain. These symptoms may be subtle, and their
Pain caused by disease progression may be better controlled with peak varies according to the type of opioid used. With long-
disease-modifying agents, and psychological problems may be acting opioids, withdrawal symptoms occur after 48 to 72 hours,
better managed with antidepressants or anxiolytics. Also, it is whereas with short-acting opioids, withdrawal symptoms peak at
important to distinguish tolerance from hyperalgesia, which is 6 to 12 hours.49 The patient must restart the prior medication
usually a more diffuse pain, less defined in quality. In hyperalge- dosages, and symptomatic control is needed once symptoms are
sia, the pain threshold decreases, and an increased dose of opioids noticed. Clonidine, an α2 agonist, can be used to help control
will cause an increase in the pain.50,51 Hyperalgesia has been symptoms (Table 169-3).
1388 SECTION 6  Pain

Nonmedical Use of Opioids in the Nonmedical Use of Opioids in the United States by Age
United States 8

6
7

5 6

Percentage of population
Percentage of population

4 5

4
3

3
2
2

1
1

0 0
All ages Male Female 12-17 18-25 26-34 34-49 50 and above
Age group
2002-2003 2009-2010 2002-2003 2009-2010

Figure 169-2. Percentage of population using prescription opioids for chronic nonmedical reasons.47
Chronic is defined here as more than 200 days.

TABLE 169-3  Management of Tolerance, Hyperalgesia, and Dependence


Tolerance Hyperalgesia Withdrawal
• Increase the opioid dosage by 15%-20%. • Taper dosage slowly until pain control • Restart opioids at prior dosage.
• Rotate opioids. is achieved. • Taper dosage slowly and gradually.
• Add NMDA receptor antagonist to regimen. • Rotate opioids. • Prescribe symptomatic treatment, including
• Prescribe adjuvant medications to target • Prescribe adjuvant pain medications. clonidine.
analgesia at different pain mechanisms. • Prescribe NMDA receptor antagonists. • Instruct patient to avoid sudden discontinuation
of opioids and to avoid simultaneous use of
agonists and antagonists.
NMDA, N-methyl-D-aspartate.

Understanding how long-term opioid use leads to tolerance use of opioids in the United States. Of patients with chronic pain
and dependence is crucial in the perioperative management of who are receiving opioids, one third are not taking them as pre-
patients receiving long-term opioid therapy. Postoperative pain scribed or are abusing them. Further ASIPP investigation into
often becomes a major problem and a source of frustration the practice patterns of physicians who prescribe opioid medica-
for the patient and the surgeon. The use of short-acting analge- tion revealed that most patients taking opioids receive both long-
sics, such as remifentanil, has been shown to increase hyperalge- acting and short-acting opioids for more than 3 months and
sia.54 Longer acting opioids, such as fentanyl or sufentanil, prescribed by a physician who is not a pain specialist. Patients
have been shown to be more effective.55 Other options include receiving combined long- and short-acting opioids account for a
ketamine56 and COX inhibitors, which act on NMDA receptors significant proportion of the fatalities associated with opioid use.
to reduce hyperalgesia.57 Scheduled medication provides better Of opioid-related fatalities, 60% occur in patients taking medica-
pain control than does as-needed dosing. Knowledge of prior tion prescribed within the current guidelines. In view of these
home medication dosage and calculating equivalent dosage findings and the dearth of studies on the effectiveness of long-
also help in avoiding withdrawal symptoms and suboptimal pain term opioid use, it is important for physicians to weigh all benefits
control.55 and risks when prescribing these medications for long-term use.
Addiction remains a major problem with opioid prescription: There is evidence to support the use of urine drug tests to identify
the number of people addicted to prescription medication is patients who are noncompliant or abusing their medications.
higher than the numbers heroin and cocaine addicts combined. Furthermore, prescription drug monitoring programs may
In a review on opioid use, the American Society of Interventional reduce abuse and “doctor shopping.” An ASIPP-developed set
Pain Physicians (ASIPP) found a pattern of extensive nonmedical of guidelines related to opioid prescribing intended to guide
CHAPTER 169  Pharmacologic Treatment of Pain 1389

TABLE 169-4  Classification of Pain and Characteristics


169
Pain Type Description Onset and Course Distribution Intensity
Nociceptive Sharp; may be dull if Correlated with injury Related to area of injury or trauma Varies widely between individuals
not severe
Inflammatory Aching, dull, stabbing Progressive; affected by activity Localized to the body part that is Is variable and can affect functioning
inflamed or infected
Neuropathic Burning, shooting, Insidious, usually progressive, Radiating both proximally and Varies widely between individuals
squeezing, numb and possibly lasting for years distally from the site of nerve Is usually intense and profound
damage

physicians in patient management can be used as a reference for the Study of Pain (IASP) as “pain initiated or caused by a
point, but clinical management should be tailored to each indi- primary lesion, or dysfunction of the nervous system.”59 Neuro-
vidual case.24,33 pathic pain often exceeds the severity of the injury leading to the
pain. It can be described as burning, lancinating, cramping, or
electric, and it tends to be more diffuse and less focal. It can also
Mechanistic Approach to Pain be more intense distally, increase at night, and be aggravated by
With different pharmacologic agents available for pain treat- factors such as cold, touch, and movement.60,61
ment, it is important to match the patient with the appropriate It remains important to identify the cause of the pain as much
medication. Clinicians must understand that certain classes of as possible because this can direct the therapy. Causes of neuro-
medication are most effective for certain broad classes of pain, pathic pain can be from trauma, nerve avulsion injury, polyneu-
depending on the pathophysiologic mechanisms of the pain ropathy (as in diabetes), postherpetic pain, phantom pain, or
process. However, depending solely on this approach is not complex regional pain syndrome.
always effective, and other factors such as pain intensity, toler- Current therapies for neuropathic pain aim at regulating the
ability, safety, use of disease-modifying agents, compliance, and action of neurotransmitters, receptors, and ion channels. Antide-
cost have to be considered. pressants and antiepileptics remain the first choices for pain
Because of the extensive controversy over the categorization control and can be used in combination. Although opioids are
of pain, a broad classification may be more applicable for clinical more effective in treating nociceptive pain, opioid use has been
purposes. Pain can be categorized as nociceptive, which is a shown to have synergistic value in select refractory cases of neu-
response to injury; inflammatory, which results from upregulation ropathic pain. Methadone, for example, acts as an NMDA recep-
of inflammatory mediators; and neuropathic, which is caused by tor antagonist and inhibits serotonin and norepinephrine uptake,
nerve injury. Pain should also be characterized as either acute or which helps with neuropathic pain control.62 Other alternatives
chronic. With either system, clinicians can classify pain by obtain- include local anesthetics, capsaicin, clonidine, and baclofen
ing a careful history and performing a thorough examination on (Tables 169-5 and 169-6).
the patient in which the qualities and characteristics of the pain
are assessed. Some diagnoses are associated with specific types of
pain; others can manifest with multiple types of pain that syner-
Acute Pain
gistically affect the patient (Table 169-4). Certain diagnoses tran- Acute pain results from direct injury and trauma. The most
scend this classification system, lending themselves to specific common occurrence of acute pain in neurosurgical practice is
treatment; for example, trigeminal neuralgia is best treated with postoperative pain. The main processes involved in acute pain are
oxcarbazepine. inflammatory and nociceptive. NSAIDs can be used to disrupt
the inflammatory process and opioids to further diminish the
pain. Other options aim to interrupt signal conduction and
Medication Selection transmission from the peripheral nervous system to the central
Once the type of pain is established, the class of pharmacologic nervous system. These options include local anesthetics that
agents can be established. The challenge lies in appropriately block voltage-gated ion channels, as well as epidural and intrathe-
adjusting medications according to the pain intensity, efficacy of cal opioids and anesthetics.
the drug, and side effects. At times, multiple lines of therapy are Patient-controlled analgesia has been shown to be effective
needed for synergistic effects, to reduce side effects, and to against postoperative pain. The use of multimodal therapy and a
promote safety. This is possible when more than one pain modal- stepladder approach can help with better control and reduce side
ity is involved, as in the case of prolapsed lumbar disk, whereby effects. The control of postoperative pain starts in the preopera-
the disk releases inflammatory mediators that cause inflammatory tive period with patient education and shaping expectations.
pain and may also physically compress the nerve and thus causes This can be applied to both the adult and pediatric populations
neuropathic pain. In this situation, it is important to remember (Table 169-7).
that some medications can be used for multiple types of pain; for
example, NSAIDs can be used for both nociceptive and inflam- Acute Pain in Children.  Identifying pain in children can be
matory pain. Drug-drug interactions should always be assessed challenging because children often cannot express their pain and
when a polypharmacy approach is considered. cannot describe its severity or character. Therefore, it remains
the charge of the physician to identify subtle signs such as irrita-
bility, crying, guarding, and apathy as signs of pain. It is also useful
Neuropathic Pain to listen to the parents and addressing their concern because they
In general, neuropathic pain results from neural dysfunction or are likely to know how their child responds when in pain or
damage to the somatosensory pathway; unlike nociceptive pain, discomfort.
neuropathic pain has no functional or protective benefit.58 Neu- Pain control can start before a procedure with the use of
ropathic pain has been defined by the International Association long-lasting local anesthetics, lidocaine and prilocaine (EMLA)
1390 SECTION 6  Pain

TABLE 169-5  First-Line Medications for Neuropathic Pain


Duration of
Medication Beginning Dosage Titration Maximum Dosage Adequate Trial
CALCIUM CHANNEL α2δ LIGANDS
Gabapentin 100-300 mg every Increase by 100-300 mg for 3600 mg/day (1200 mg three 3-8 wk for titration plus
night or 100-300 mg three times daily dosing times daily); reduce if 1-2 wk at maximum
three times daily every 1-7 days as tolerated creatinine clearance is low tolerated dosage
Pregabalin 50 mg three times daily Increase to 300 mg daily after 600 mg daily (200 mg three 4 wk
or 75 mg twice daily 3-7 days, then by 150 mg/ times daily or 300 mg twice
day every 3-7 days as daily); reduce dosage if renal
tolerated function is impaired
Topical 5% lidocaine Maximum of 3 patches None needed Maximum of three patches daily 3 wk
patch daily for a maximum for a maximum of 12 hr
of 12 hr
OPIOIDS
Morphine 5-15 mg every 4 hr as After 1-2 wk, convert total daily No maximum with careful 4-6 wk
needed dosage to long-acting opioid titration; consider evaluation
analgesic and continue by pain specialist at dosages
short-acting medication as exceeding 120-180 mg/day
needed
Tramadol hydrochloride 50 mg once or twice Increase by 50-100 mg/day in 400 mg/day (100 mg four times 4 wk
daily divided doses every 3-7 days daily); in patients older than
as tolerated 75 years, 300 mg/day in
divided doses
TRICYCLIC ANTIDEPRESSANTS
Nortriptyline hydrochloride 10-25 mg every night Increase by 10-25 mg/day 75-150 mg/day; if blood level of 6-8 wk, with at least
or desipramine every 3-7 days as tolerated active drug and its metabolite 1-2 wk at maximum
hydrochloride is <100 ng/mL, continue tolerated
titration with caution
SELECTIVE SEROTONIN REUPTAKE INHIBITORS (SSNRIS)
Duloxetine 30 mg once daily Increase to 60 mg once daily 60 mg twice daily 4 wk
after 1 wk
Venlafaxine 37.5 mg once or twice Increase by 75 mg each wk 225 mg daily 4-6 wk
daily
Modified from Dworkin RH, O’Connor AB, Backonja M, et al. Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain.
2007;132:237-251.

TABLE 169-6  Treatment Effectiveness in Neuropathic Pain anxiolytic medication. In children, dosing is based primarily on
weight, and thus it is very important to verify dosages carefully
Neuropathic Pain Medication Class Number Needed to Treat*
(Table 169-8).
Tricyclic antidepressants 2.4 Different nonmedical interventions have been shown to
Carbamazepine 2.5 reduce pain and anxiety and shorten hospital stays. These tech-
Tramadol 3.5 niques aim to distract the patient from pain; examples include
Gabapentin 3.7 playing games, muscular relaxation, guided imagination, hypno-
Selective serotonin reuptake inhibitor 6.7
sis, and massage. Other alternatives include acupuncture, bio-
Dextromethorphan 1.9
Levodopa 3.4 feedback, art therapy, music therapy, herbal medicine, and
Capsaicin 5.9 chiropractic care. Reductions in the duration of the procedure,
Mexiletine 38 preoperative delay, waiting time, and noise level can reduce the
child’s anxiety level and enable better pain control.
*Number needed to treat is an epidemiologic calculation of treatment
effectiveness, defined as the number of patients who need to be
treated in order to achieve benefit from the treatment in one patient. CONCLUSION
Pharmacologic treatment of pain has become increasingly
complex with the growing understanding of pain mechanisms
cream, and topical agents. In general, the use of a scheduled and the development of medications targeting these mechanisms.
pain regimen is preferred over as-needed dosing. Oral and intra- Multiple drug classes for the management of pain can be used
venous acetaminophen and NSAIDs remain the cornerstone of individually or in combination to provide synergistic effects.
pharmacologic agents for control of mild and moderate pain Management techniques vary: some therapies are chosen accord-
because of their analgesic and anti-inflammatory properties. If ing to type of pain and underlying mechanism of action, whereas
pain is not controlled, opioids can be added to the regimen, with others are selected on the basis of the diagnosis. Either way, it is
caution because of the potential side effects of sedation, respira- important to consider the mechanism of action, side effects,
tory depression, nausea, and vomiting. The use of opioids dosing, drug-drug interactions, and the individual patient in
in mechanically ventilated patients can have good results, but determining the appropriate therapies. When prescribing opioids,
with prolonged treatment, patients may develop dependence clinicians must also be vigilant for abuse and counsel patients on
and will need to be weaned off them. Other options include the appropriate use of the medication.
1392 SECTION 6  Pain

SUGGESTED READINGS Knotkova H, Fine PG, Portenoy RK. Opioid rotation: the science and
Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharmaco- the limitations of the equianalgesic dose table. J Pain Symptom Manage.
logical treatment of neuropathic pain: 2010 revision. Eur J Neurol. 2009;38(3):426-439.
2010;17(9):1113-1123, e67-e88. Kuner R. Central mechanisms of pathological pain. Nat Med. 2010;
Behavioral Health Coordinating Committee, Prescription Drug Abuse 16(11):1258-1266.
Subcommittee, U.S. Department of Health and Human Services. Manchikanti L, Abdi S, Atluri S, et al. American Society of Interventional
Addressing Prescription Drug Abuse in the United States: Current Activities Pain Physicians (ASIPP) guidelines for responsible opioid prescribing
and Future Opportunities. Atlanta, GA: Centers for Disease Control and in chronic non-cancer pain: Part I—evidence assessment. Pain Physi-
Prevention; 2013:8-10. cian. 2012;15(3 suppl):S1-S65.
Bryson HM, Wilde MI. Amitriptyline. A review of its pharmacological McNicol ED, Midbari A, Eisenberg E. Opioids for neuropathic pain.
properties and therapeutic use in chronic pain states. Drugs Aging. Cochrane Database Syst Rev. 2013;(8):CD006146.
1996;8(6):459-476. Paulozzi L, Jones C, Mack K, et al; Centers for Disease Control and
Chen L, Mao J. Update on neuropathic pain treatment: ion channel Prevention (CDC). Vital signs: overdoses of prescription opioid
blockers and gabapentinoids. Curr Pain Headache Rep. 2013;17(9): analgesics—United States, 1999-2008. MMWR Morb Mortal Wkly Rep.
359. 2011;60(43):1487-1492.
Dharmshaktu P, Tayal V, Kalra BS. Efficacy of antidepressants as analge- Stromer W, Michaeli K, Sandner-Kiesling A. Perioperative pain therapy
sics: a review. J Clin Pharmacol. 2012;52(1):6-17. in opioid abuse. Eur J Anaesthesiol. 2013;30(2):55-64.
Jones CM, Mack KA, Paulozzi LJ. Pharmaceutical overdose deaths,
United States, 2010. JAMA. 2013;309(7):657-659. See a full reference list on ExpertConsult.com
CHAPTER 169  Pharmacologic Treatment of Pain 1392.e1

REFERENCES 29. Finnerup NB, Sindrup SH, Bach FW, et al. Lamotrigine in spinal
1. Manchikanti L, Abdi S, Atluri S, et al. American Society of Interven- cord injury pain: a randomized controlled trial. Pain. 2002;96(3): 169
tional Pain Physicians (ASIPP) guidelines for responsible opioid 375-383.
prescribing in chronic non-cancer pain: Part I—evidence assessment. 30. Simpson DM, McArthur JC, Olney R, et al. Lamotrigine for HIV-
Pain Physician. 2012;15(3 suppl):S1-S65. associated painful sensory neuropathies: a placebo-controlled trial.
2. Murinson B. A mechanism-based approach to pain pharmacother- Neurology. 2003;60(9):1508-1514.
apy: targeting pain modalities for optimal treatment efficacy. In: 31. Gomez-Arguelles JM, Dorado R, Sepulveda JM, et al. Oxcarbazepine
Smith H, ed. Current Therapy in Pain. 1st ed. Philadelphia: Saunders; monotherapy in carbamazepine-unresponsive trigeminal neuralgia.
2009. J Clin Neurosci. 2008;15(5):516-519.
3. Basbaum AI, Bautista DM, Scherrer G, et al. Cellular and molecular 32. Hershey LA, Bednarczyk EM. Treatment of headache in the elderly.
mechanisms of pain. Cell. 2009;139(2):267-284. Curr Treat Options Neurol. 2013;15(1):56-62.
4. Kuner R. Central mechanisms of pathological pain. Nat Med. 2010; 33. Donofrio PD, Raskin P, Rosenthal NR, et al. Safety and effectiveness
16(11):1258-1266. of topiramate for the management of painful diabetic peripheral
5. Dharmshaktu P, Tayal V, Kalra BS. Efficacy of antidepressants as neuropathy in an open-label extension study. Clin Ther. 2005;27(9):
analgesics: a review. J Clin Pharmacol. 2012;52(1):6-17. 1420-1431.
6. Verdu B, Decosterd I, Buclin T, et al. Antidepressants for the treat- 34. Petersen P, Kastrup J, Zeeberg I, et al. Chronic pain treatment with
ment of chronic pain. Drugs. 2008;68(18):2611-2632. intravenous lidocaine. Neurol Res. 1986;8(3):189-190.
7. Salerno SM, Browning R, Jackson JL. The effect of antidepressant 35. McNicol ED, Midbari A, Eisenberg E. Opioids for neuropathic pain.
treatment on chronic back pain: a meta-analysis. Arch Intern Med. Cochrane Database Syst Rev. 2013;(8):CD006146.
2002;162(1):19-24. 36. Manchikanti L, Atluri S, Hansen H, et al. Opioids in chronic non-
8. Hirt H, Gartner A, Heberle-Bors E. An alfalfa cDNA encodes a cancer pain: have we reached a boiling point yet? Pain Physician.
protein with similarity to human snRNP-E. Nucleic Acids Res. 1992; 2014;17(1):E1-E10.
20(3):613. 37. Kitahara M, Kojima KK, Ohmura A. Efficacy of interdisciplinary
9. Tunks ER, Crook J, Weir R. Epidemiology of chronic pain with treatment for chronic nonmalignant pain patients in Japan. Clin J
psychological comorbidity: prevalence, risk, course, and prognosis. Pain. 2006;22(7):647-655.
Can J Psychiatry. 2008;53(4):224-234. 37a. University of Wisconsin-Madison, Pain and Policy Studies Group, WHO
10. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain: a Cochrane Regional Office for the Americas (AMRO). AMRO opioid consumption,
review. J Neurol Neurosurg Psychiatry. 2010;81(12):1372-1373. 2013. <http://www.painpolicy.wisc.edu/amro-0>; 2013 Accessed
11. Max MB, Kishore-Kumar R, Schafer SC, et al. Efficacy of desipra- 19.01.16.
mine in painful diabetic neuropathy: a placebo-controlled trial. Pain. 38. van Seventer R, Smit JM, Schipper RM, et al. Comparison of TTS-
1991;45(1):3-9. fentanyl with sustained-release oral morphine in the treatment of
12. Max MB, Culnane M, Schafer SC, et al. Amitriptyline relieves dia- patients not using opioids for mild-to-moderate pain. Curr Med Res
betic neuropathy pain in patients with normal or depressed mood. Opin. 2003;19(6):457-469.
Neurology. 1987;37(4):589-596. 39. Staats PS, Markowitz J, Schein J. Incidence of constipation associated
13. Galer BS. Neuropathic pain of peripheral origin: advances in phar- with long-acting opioid therapy: a comparative study. South Med J.
macologic treatment. Neurology. 1995;45(12, suppl 9):S17-S25. 2004;97(2):129-134.
14. Bryson HM, Wilde MI. Amitriptyline. A review of its pharmacologi- 40. Allan L, Richarz U, Simpson K, et al. Transdermal fentanyl versus
cal properties and therapeutic use in chronic pain states. Drugs Aging. sustained release oral morphine in strong-opioid naive patients
1996;8(6):459-476. with chronic low back pain. Spine (Phila Pa 1976). 2005;30(22):
15. Attal N, Cruccu G, Baron R, et al. EFNS guidelines on the pharma- 2484-2490.
cological treatment of neuropathic pain: 2010 revision. Eur J Neurol. 41. Abs R, Verhelst J, Maeyaert J, et al. Endocrine consequences of long-
2010;17(9):1113-1123, e67-e88. term intrathecal administration of opioids. J Clin Endocrinol Metab.
16. Schnitzer TJ, Ferraro A, Hunsche E, et al. A comprehensive review 2000;85(6):2215-2222.
of clinical trials on the efficacy and safety of drugs for the treatment 42. Knotkova H, Fine PG, Portenoy RK. Opioid rotation: the science
of low back pain. J Pain Symptom Manage. 2004;28(1):72-95. and the limitations of the equianalgesic dose table. J Pain Symptom
17. Jenkins DG, Ebbutt AF, Evans CD. Tofranil in the treatment of low Manage. 2009;38(3):426-439.
back pain. J Int Med Res. 1976;4(suppl 2):28-40. 43. Smith HS, McCleane G. Opioids issues. In: Smith HS, ed. Current
18. Sayar K, Aksu G, Ak I, et al. Venlafaxine treatment of fibromyalgia. Therapy in Pain. 1st ed. Philadelphia: Elsevier; 2009.
Ann Pharmacother. 2003;37(11):1561-1565. 44. Paulozzi LJ, Mack KA, Hockenberry JM. Vital signs: variation among
19. Dwight MM, Arnold LM, O’Brien H, et al. An open clinical trial states in prescribing of opioid pain relievers and benzodiazepines—
of venlafaxine treatment of fibromyalgia. Psychosomatics. 1998;39(1): United States, 2012. MMWR Morb Mortal Wkly Rep. 2011;63(26):
14-17. 563-568.
20. Chen L, Mao J. Update on neuropathic pain treatment: ion channel 45. Substance Abuse and Mental Health Services Administration, Center for
blockers and gabapentinoids. Curr Pain Headache Rep. 2013;17(9): Behavioral Health Statistics and Quality. The TEDS report: gender
359. differences in primary substance of abuse across age groups. <http://
21. Atli A, Dogra S. Zonisamide in the treatment of painful diabetic www.samhsa.gov/data/sites/default/files/sr077-gender-differences-
neuropathy: a randomized, double-blind, placebo-controlled pilot 2014.pdf>; 2014 Accessed 19.01.16.
study. Pain Med. 2005;6(3):225-234. 46. Olfson M, Wang S, Iza M, et al. National trends in the office-based
22. Takahashi Y, Hashimoto K, Tsuji S. Successful use of zonisamide for prescription of schedule II opioids. J Clin Psychiatry. 2013;74(9):
central poststroke pain. J Pain. 2004;5(3):192-194. 932-939.
23. Drake ME Jr, Greathouse NI, Renner JB, et al. Open-label 47. Behavioral Health Coordinating Committee, Prescription Drug
zonisamide for refractory migraine. Clin Neuropharmacol. 2004;27(6): Abuse Subcommittee, U.S. Department of Health and Human Ser-
278-280. vices. Addressing Prescription Drug Abuse in the United States: Current
24. Michiels WB, McGlthlen GL, Platt BJ, et al. Trigeminal neuralgia Activities and Future Opportunities. Atlanta: Centers for Disease
relief with intrathecal ziconotide. Clin J Pain. 2011;27(4):352-354. Control and Prevention; 2013:8-10.
25. Backonja MM. Neuropathic pain therapy: from bench to bedside. 48. Jones CM, Mack KA, Paulozzi LJ. Pharmaceutical overdose deaths,
Semin Neurol. 2012;32(3):264-268. United States, 2010. JAMA. 2013;309(7):657-659.
26. Crepeau AZ, Treiman DM. Levetiracetam: a comprehensive review. 49. Chang G, Chen L, Mao J. Opioid tolerance and hyperalgesia. Med
Expert Rev Neurother. 2010;10(2):159-171. Clin North Am. 2007;91(2):199-211.
27. Carim L, Sumoy L, Andreu N, et al. Cloning, mapping and expres- 50. Brush DE. Complications of long-term opioid therapy for manage-
sion analysis of VPS33B, the human orthologue of rat Vps33b. Cyto- ment of chronic pain: the paradox of opioid-induced hyperalgesia.
genet Cell Genet. 2000;89(1–2):92-95. J Med Toxicol. 2012;8(4):387-392.
28. Sandner-Kiesling A, Rumpold Seitlinger G, Dorn C, et al. Lamotri­ 51. Mao J, Price DD, Mayer DJ. Mechanisms of hyperalgesia and mor-
gine monotherapy for control of neuralgia after nerve section. Acta phine tolerance: a current view of their possible interactions. Pain.
Anaesthesiol Scand. 2002;46(10):1261-1264. 1995;62(3):259-274.
1392.e2 SECTION 6  Pain

52. Mao J. Opioid-induced abnormal pain sensitivity: implications in 58. Woolf CJ. Pain: moving from symptom control toward mechanism-
clinical opioid therapy. Pain. 2002;100(3):213-217. specific pharmacologic management. Ann Intern Med. 2004;140(6):
53. Mao J. Opioid-induced abnormal pain sensitivity. Curr Pain Headache 441-451.
Rep. 2006;10(1):67-70. 59. Merskey H, Bogduk N. Task Force on Taxonomy of the International
54. Konopka KH, van Wijhe M. Opioid-induced hyperalgesia: pain Association for the Study of Pain. Seattle: IASP Press; 1994.
hurts? Br J Anaesth. 2010;105(5):555-557. 60. Backonja MM. Defining neuropathic pain. Anesth Analg. 2003;97(3):
55. Stromer W, Michaeli K, Sandner-Kiesling A. Perioperative pain 785-790.
therapy in opioid abuse. Eur J Anaesthesiol. 2013;30(2):55-64. 61. Otto M, Bak S, Bach FW, et al. Pain phenomena and possible mecha-
56. Laulin JP, Maurette P, Corcuff JB, et al. The role of ketamine in nisms in patients with painful polyneuropathy. Pain. 2003;101(1–2):
preventing fentanyl-induced hyperalgesia and subsequent acute 187-192.
morphine tolerance. Anesth Analg. 2002;94(5):1263-1269. table of 62. Hays L, Reid C, Doran M, et al. Use of methadone for the treatment
contents. of diabetic neuropathy. Diabetes Care. 2005;28(2):485-487.
57. Koppert W, Wehrfritz A, Korber N, et al. The cyclooxygenase
isozyme inhibitors parecoxib and paracetamol reduce central hyper-
algesia in humans. Pain. 2004;108(1–2):148-153.
PART 3 Treatment of Trigeminal Neuralgia 170

170 ofEvidence-Based
Facial Pain
Approach to the Treatment

Turo J. Nurmikko and Francis O’Neill

The evolution of the surgical management of pain has been tomatic people9; (2) in up to 28% of patients with classic trigemi-
shaped by shared clinical experience and observation, with limited nal neuralgia, no compression can be found10; and (3) trigeminal
effect from well-designed clinical trials. The common successful neuralgia does recur over the years in 25% of affected patients
scenario follows a typical path: an innovative novel procedure, without evidence of a new compression.11-13 A more plausible
supported by a single or very few successful case series, is pro- explanation is that compression is only one (albeit perhaps the
posed; it is followed by further case reports by interested sur- most common) cause of alterations in anatomy and physiology of
geons; interest becomes more widespread; the procedure is the nerve and its central connections.14-16 Although decompres-
gradually adopted into general surgical practice; and finally it sion undoubtedly improves nerve function significantly,17 it has
becomes the “gold standard.” In many cases, however, the early not been shown that such change is needed to render the condi-
success is not repeated in other centers, and interest in the inno- tion asymptomatic.
vation wanes. Even procedures that become established may face Second, MVD is less effective in patients with atypical tri-
rejection if in popular use they are shown to be associated with geminal neuralgia who have concomitant multiple sclerosis
excessive risk or poor tolerability or are simply perceived as less and when the compression is caused by a vein13,18-21 (but see
effective than an alternative intervention. In the process, many Sindou and associates22). The reason may be that the pathophysi-
patients will have been subjected to surgical interventions that ologic process affecting the trigeminal system remains unaffected
have limited value. Although multiple mechanisms are necessary by MVD.
to dictate the fate of a particular intervention, an important role Third, repeated MVD usually provides no better outcome
is played by key opinion leaders whose views remain unchal- than less invasive percutaneous procedures.23 As a result, most
lenged because of the lack of objective evidence to the contrary. neurosurgeons favor the latter in cases of recurrent pain after
This scenario was played out in the 20th century in the surgi- initial MVD.8
cal management of trigeminal neuralgia. The notable limitations Fourth, the risks of MVD, including hearing loss, brainstem
of pharmacologic treatment paved the way to the evolution of infarction, and mortality, are marginally higher than with other
surgical methods ranging from division of the trigeminal nerve procedures.1
to alcohol injections, neurectomy, radiofrequency rhizotomy, Because the primary aim in the management of trigeminal
glycerol rhizotomy, and balloon microcompression. By the 1970s neuralgia is pain relief, all procedures that sufficiently alter the
the ganglion-level minimally invasive neuroablative procedures pathophysiologic process may be clinically useful. As described
(NAPs) had become the surgical “gold standard.” Coincidentally elsewhere in this book (Chapter 172), minimally invasive NAPs—
with the development of the operative microscope, microvascular currently in widespread use—have a respectable record for com-
decompression (MVD) became a therapeutic option.1 After Peter plete or near-complete control of the pain, with an excellent
Janetta reintroduced Walter Dandy’s original proposal of MVD safety profile. When the surgeon discusses the advantages and
from 1932 and converted it into a procedure in 1967, it was slowly disadvantages of various procedures with the patient, results from
adopted into the neurosurgeon’s armamentarium over the follow- comparative trials showing superiority of a given procedure over
ing two decades.2-4 Both enthusiasm and criticism accompanied the others would be helpful; however, there are none.1,8 There-
its early adoption; opponents and proponents substantiated their fore, the surgeon must extrapolate from published outcomes of a
views on the basis of their own experience rather than well- large number of observational studies and adapt the conclusions
designed studies. MVD is now the commonest operation for to the individual needs of the patient.
trigeminal neuralgia in the United States.5,6 Observational studies, however, have been conducted with
MVD has reached this status by common surgical consensus. clear patient selection, careful data collection, independent asses-
It is easy to understand why: it is an elegant way of removing a sors, and actuarial outcome presentation.7,8,22,24 There are ample
cause that can be visualized on magnetic resonance imaging published clinical data that allow the surgeon to describe what a
(MRI); it avoids injury to the nerve and, in contrast, restores its given patient may expect from a particular procedure. However,
function; patients find the concept easy to understand; the pro- it does not answer the question of whether in a particular case
cedure is among the safest of intracranial operations; it is well the chosen method is optimal.
tolerated by the majority of patients who undergo it; and the The currently available procedures are, without doubt,
early and long-term results are excellent. In contrast, NAPs— extraordinarily effective in treating trigeminal neuralgia. In a
that is, radiofrequency rhizotomy, balloon microcompression patient with refractory trigeminal neuralgia, the remarkable
of the gasserian ganglion, glycerol rhizotomy, and stereotactic outcome of postoperative freedom of pain for several years is
radiosurgery—do not treat the cause, are associated with inten- evidence enough.8,24-26 It is obvious that subjecting both MVD
tional injury to the nerve, yield poorer long-term results, and are and ganglion-level NAPs to efficacy trials against sham treatment
associated with side effects that include annoying paresthesias is meaningless.27 In fact, considering the very large effect size and
and frank anesthesia dolorosa.7,8 a substantial number of patients in prospective case series with
Many of the reasons to perform MVD are less compelling if independent assessors, the level of evidence from them, if assessed
subjected to scrutiny. First, vascular compression or contact according to Grades of Recommendation, Assessment, Develop-
cannot be the primary cause because (1) it is common in asymp- ment, and Evaluation (GRADE) criteria, would be considered
1393
1394 SECTION 6  Pain

high.28 The neurosurgical literature contains sparse preoperative steadily growing in the United States.6 Not only that, NAPs are
pain data; when such data are available, however, the effect sizes being performed less frequently than before, which suggests that
calculated from them are extremely high, between 2 and 3, after the overall demand for operative treatment of trigeminal neural-
radiofrequency rhizotomy at 3 years and MVD at 2 years, respec- gia is mostly satisfied. The very significant difference in cost has
tively.29,30 Because of these large effect sizes that characterize both not stifled this trend.5
MVD and all NAPs, comparative trials are necessary that focus Trigeminal neuralgia is by no means the only example of facial
on the balance between benefit on the one hand and safety and pain in which evidence of efficacy of surgical interventions is
tolerability on the other. conspicuously lacking. The surgical options available for chronic
Unfortunately, only low-quality comparative studies to that cluster headache are limited. We carried out a systematic review
effect have been published. They are mostly single-center retro- of publications from 1980 to 2013 and found 14 procedures that
spective assessments of outcomes from two or more procedures were advocated by various authors. Practically all material was
with inconstant follow-up (for reviews, see Pollock1; Tatli and based on uncontrolled case series (eTable 170-1). Of interest was
associates11; and Zakrzewska and colleagues31). The picture that no intervention was superior to the rest in efficacy or safety.
emerging is confusing. As the first operation, MVD in most None yielded such poor results that it would be rejected outright.
comparisons emerges superior in long-term effectiveness over The relative similarity of benefit across all reported methods
NAPs.2,3,32-34 However, some authors either claim better effective- reveals the same problem as in the surgical management of tri-
ness with NAPs35 or point to the associated risks of mortality and geminal neuralgia: for the surgeon, the choice of the best method
morbidity and the marginal difference between the procedures’ for the patient remains arbitrary.
effectiveness and therefore favor radiofrequency rhizotomy.26 In
a valiant attempt to resolve the question, Tatli and associates11
evaluated data from approximately 5000 patients in studies pub-
THE BENEFIT OF EVIDENCE-BASED MEDICINE
lished between 1970 and 2005 and concluded that although the In the 1970s, physicians realized that treatment decisions must
quality of many of the studies was not high, the overall picture not be based on evidence that is derived from clinical practice
emerging was that of considerable success in controlling the pain alone and that any treatment must undergo rigorous scientific
in nearly 40%, more or less permanently, with any intervention. evaluation. This led to the development of evidence-based medi-
Of importance is that after 20 years, there was little difference cine (EBM), which is now an integral part of pharmacologic
between those undergoing MVD and those undergoing NAPs. medicine and should be essential for all new surgical procedures
Practically all authors acknowledge greater effectiveness of and some in common use now. EBM aims to ensure that a given
MVD over stereotactic radiosurgery, but the benefits of the latter treatment is as accurate, effective, and unbiased as possible. By
are also promoted.36,37 Comparisons between various NAPs yield definition, EBM is the conscientious, explicit, and judicious use
variable results; no single procedure is clearly superior when the of current best evidence in making decisions about the care of
outcome is based on recurrence rate, morbidity (especially individual patients.41 In the previously described examples of tri-
sensory loss), and patient satisfaction.8,38 In systematic reviews of geminal neuralgia and chronic cluster headache, clinical trials
better quality prospective case series with actuarial outcome with rigorous scientific methods stand the best chance of identi-
reporting, authors could not find a real difference between fying the preferred treatment for a given patient.
these procedures.7 In a situation in which preoperative three- It is widely accepted that the strongest evidence comes from
dimensional MRI reconstruction fails to show vascular compres- randomized controlled trials (RCTs) that are adequately powered
sion, the choice of the operation probably represents the personal and whose design limits bias and ensures that the outcome will
preference of the surgeon. be measured objectively.28,42,43 The weakest evidence comes from
The uncertainty over the best method becomes even greater uncontrolled case series, case reports, and expert opinion.43 Criti-
if the clinician wishes to establish which approach is best for cal EBM appraisal is increasingly applied to novel diagnostic
subgoups of patients with trigeminal neuralgia associated with techniques, drugs, and noninterventional treatments, and there
multiple sclerosis, trigeminal neuralgia with atypical features, and are ever stronger calls for its application to surgery. However,
trigeminal neuralgia that recurs after an earlier operation. Advo- implementation of EBM in surgery is not as straightforward as
cates and opponents of MVD report completely different results it is in other areas of medicine, and it is not surprising that the
in patients with multiple sclerosis–associated trigeminal neural- surgical community approaches the idea with circumspection and
gia.18,39 In most case series, patients with atypical trigeminal neu- reservation.44
ralgia have a poorer response to any procedure than do patients We posit that there are particular reasons for application of
with typical trigeminal neuralgia4,12,19,40; however, whether MVD EBM in the surgical management of chronic pain. First, chronic
or any other intervention is superior in this subgroup of patients pain is subjective, and its intensity notoriously fluctuates over
remains unclear. time; some conditions are even known to go into long spontane-
The failure of any of the comparative studies published so far ous remissions. Second, for chronic pain, there are many treat-
to identify a superior treatment results from their inherent biases. ments easily accessed by patients, seemingly similar in outcome,
The major bias concerns the difference between compared which enhances the risk of bias from patients. Third, the placebo
patient groups because of lack of randomization. The groups tend reaction is greater in chronic pain than in any other condition,
to differ in previous treatment received, duration of trigeminal except depression, and accounts for up to 44% of the treatment
neuralgia, associated disability, clinical features (especially those response.45 The resulting risk of bias may be high enough to
of atypical trigeminal neuralgia), and previous responses to question the very foundation of a standard treatment. Indeed,
therapy. There is an inherent bias in the operator’s choice of the when RCTs were conducted to evaluate the efficacy of arthroscopic
procedure and patient’s preference, which is based on informa- débridement or partial meniscectomy for symptom relief in
tion that he or she has received, which itself may be inaccurate. degenerative knee pain or adhesiolysis in abdominal pain, no
In addition, because of the high attrition rate in long-term studies difference was found between the outcomes of the active and
and the general tendency not to use an independent party for sham treatments.46-48
outcome assessment, the risk of overall bias is too high for The two critical aspects of studies that determine the strength
conclusions. of evidence are randomization and control. Random assignment
It is intriguing that despite the obvious lack of evidence for of patients to two or more treatment conditions provides the best
the optimal intervention in trigeminal neuralgia, the popularity protection against a wide array of confounding factors that are
of MVD over radiofrequency rhizotomy and other NAPs is either unknown or unavoidable and will inevitably influence the
CHAPTER 170  Evidence-Based Approach to the Treatment of Facial Pain 1394.e1

eTABLE 170-1  Reported Interventions with Outcomes for the Management of Chronic Cluster Headache, 1980 to 2013
170
Total No. Duration of Good-to-Excellent
Intervention No. Trials Publication Years Patients Follow-up Long-Term Response
Sphenopalatine ganglion blocks 2 2006-2010 36 8-28 mo 5/36
Sphenopalatine ganglion RFL 4 1987-2012 35 6-34 mo 15/35
Sphenopalatine ganglion pulsed RFL 2 2005-2011 10 4-52 mo 2/3 (other data N/A)
Sphenopalatine ganglion stimulation 1 2013 28 3-8 wk N/A
Vagus nerve stimulation 3 2005-2013 11 N/A 8/11
Occipital nerve stimulation 6 2007-2011 56 3-64 mo 29/56
Supraorbital nerve stimulation 2 2007-2012 5 16-36 mo 4/5
Cervical cord stimulation 1 2011 7 3-78 mo 4/7
Trigeminal glycerol rhizotomy 3 1985-2000 29 9-78 mo 11/29
Trigeminal RFR 4 1982-1995 55 7 mo–20 yr 35/55
Stereotactic radiosurgery 4 1998-2011 29 5-88 mo 7/29
Trigeminal open rhizotomy 2 1983-2003 25 1 mo–19 yr 17/25
Microvascular decompression 1 1998 28 12-144 mo 9/28
Deep brain stimulation 7 2005-2013 55 9-144 mo 25/55
N/A, not available; RFL, radiofrequency lesioning; RFR, radiofrequency rhizotomy.
CHAPTER 170  Evidence-Based Approach to the Treatment of Facial Pain 1395

outcome. Comparison of a novel treatment with sham treatment therapeutic evolution. Rather, a further step would be a compara-
will show whether the novel treatment has some efficacy in a tive trial between balloon microcompression of the gasserian 170
given condition and, in most cases, the magnitude of the effect. ganglion or radiofrequency lesioning of the sphenopalatine/
Comparison of a novel treatment with an established treatment trigeminal ganglion for both efficacy and cost effectiveness.
(e.g., the “gold standard”) will allow clinicians to judge whether As discussed previously, in trigeminal neuralgia little benefit
the efficacy of the novel treatment is similar to that of the estab- is expected from any sham-controlled efficacy studies, now that
lished treatment and how the two compare in terms of patient case series in large patient populations have shown a substantial
preference, safety, and cost. In contrast, studies that lack random- effect size. However, comparative studies involving subgroups
ization and control yield results applicable only to that particular of patients with trigeminal neuralgia are clearly needed and
study population, and generalizability is limited. With few excep- have significant potential in providing clinically meaningful
tions, such studies fail to address the very essence of the matter: information.54 We suggest that in patients with either multiple
whether a particular treatment is superior to an alternative treat- sclerosis or atypical trigeminal neuralgia shown to have vascular
ment or sham treatment. compression on preoperative MRI, noninferiority trials could be
carried out between a NAP (e.g., balloon compression) and
IMPLEMENTATION OF EVIDENCE-BASED MEDICINE MVD. Patients with recurrent trigeminal neuralgia in which the
shooting pain typical of classic trigeminal neuralgia remains pre-
IN SURGERY FOR FACIAL PAIN dominant, posterior fossa exploration could be compared with a
There are increasing calls from study reviewers and authors NAP. This idea is based on reports of relative success both
themselves for controlled studies. In cluster headache, both indi- from posterior fossa exploration and balloon compression and
vidual experts and the Task Force of the European Headache radiofrequency rhizotomy.23,55,56 Table 170-1 contains more
Society emphasize the need for adequately powered controlled recommendations.
trials before treatment recommendations can be formulated.49,50 One should not be reluctant to conduct such studies because
In accordance with this, a protocol for a multicenter study to start of the difficulties that they entail, but without them (or similar
in Europe has been published in which researchers will compare controlled trials), a large percentage of patients with trigeminal
low- and high-amplitude stimulation of the occipital nerves in neuralgia remain in danger of receiving inferior care. For novel
144 patients.51 Low stimulation for the control was chosen over interventions, which are likely to be less invasive than MVD, the
nonstimulation to maintain blinded conditions. For treatment of appropriate approach is to first conduct a sham-controlled effi-
attacks, another multicenter study focused on the efficacy of cacy study and then conduct a controlled comparison with a
sphenopalatine ganglion stimulation versus control for 15 minutes NAP; patients should be chosen because of lack of vascular
before use of acute medication. The latter trial builds on the first contact on MRI or because of their inability or unwillingness to
RCT in which such stimulation was found to be effective.52 Of undergo MVD.
interest, a sham-controlled study design was implemented in a
small study of patients treated with deep brain stimulation.53
Because of the nature of deep brain stimulation, a genuine sham
OTHER TRIAL OPTIONS
control condition was possible during the trial period. Although Not all surgery is suited to clinical trial methods. Clearly,
the results were negative, they reflected the practicalities of the blind conditions may be impossible or must be compromised
study rather than its design. to an unsatisfactory degree. Many surgeons are uncomfortable
These trials are examples that should allay the concern some with the concept of equipoise. Research by its very nature is
surgeons have expressed for EBM.44 The results will not be time consuming and expensive. These issues, however, are not
known for years, but they are likely to end the 30 or more years insurmountable, and alternative trial designs can be used to
of uncertainty about interventional treatment of cluster head- address them.
ache. One caveat has to be pointed out in this development: The Idea, Development, Exploration, Assessment, Long-term
if the new neuromodulation studies based on RCT methods show follow-up (IDEAL) collaboration (http://www.ideal-collaboration
efficacy in attack prophylaxis, it may lead to their adoption into .net) is an international network of researchers, surgeons, and
clinical practice without comparison of neuroablative methods methodologists formed to address issues of experimental design
for which a similar level of evidence is not available. Unless the in surgery and other areas of complex interventions. It has devel-
effect sizes are substantial, this would not be in the interest of oped a framework for describing the stages of development in

TABLE 170-1  Trigeminal Neuralgia: Some Unanswered Questions Benefiting from Further Research
Research Question Justification for Trial Possible Trial Designs
If trigeminal neuralgia pain recurs after a No controlled trials; inconsistent results from 1. Following MVD: RCT of PFE vs. BC/RFR/GKS
remission period, what is the best case series with preferred option status 2. Following NAP: same or alternative NAP
intervention? claimed for PFE, RFR,BC,GSK
What is the preferred first-line intervention All procedures less effective than in classic 1. If vascular contact on MRI: RCT of MVD vs. BC/GKS
for atypical trigeminal neuralgia (TN2)? trigeminal neuralgia (TN1) 2. If no vascular contact: RCT of BC vs. GSK
What is the preferred first-line intervention Dual mechanism of trigeminal neuralgia16; 1. RCT of MVD vs. NAP (noninferiority trial)
for MS-related trigeminal neuralgia conflicting outcomes from case series18,39 2. RCT of MVD vs. NAP (superiority trial)
with MRI confirmed vascular
compression of V nerve?
In patients >80 yr, does MVD offer any All interventions tolerated by the elderly. Given 1. Noninferiority RCT of MVD vs. NAP
advantage over NAP? length of life expectancy, NAPs (repeated if 2. Cost effectiveness of MVD vs. NAP
necessary) may provide same level of pain (Consider lowering the age limit to 75 yr)
relief/satisfaction than MVD.
BC, balloon compression of the gasserian ganglion; GR, glycerol rhizotomy; GSK, Gamma Knife surgery; MS, multiple sclerosis; MVD, microvascular
decompression; NAP, neuroablative procedure; PFE, posterior fossa exploration; RFR, radiofrequency rhizotomy.
1396 SECTION 6  Pain

surgery and innovation and for issuing recommendations for how the case of trigeminal neuralgia, substantial effect sizes in large
research at each of these different stages can be optimized. patient populations undergoing MVD or NAPs are sufficient to
Among these recommendations are suggestions for modifica- provide solid evidence of their efficacy. Trigeminal neuralgia sub-
tions and additions to the standard RCT design, as well as alter- types, in contrast, have not been rigorously assessed in this regard,
natives to RCT design that are appropriate for some surgical and there are no reliable data to be used for treatment recom-
questions (i.e., case-matching studies, controlled interrupted time mendations. In chronic cluster headache, the emergence of novel
series, and step-wedge study designs). When a stringent RCT neurostimulation methods is welcome, but their value remains
design is not possible, then full description of the design with uncertain as long as adequately powered RCTs have not been
minimalization and explanation of confounders will allow more conducted.
realistic interpretation of results.57
Although modern health care is increasingly based on evi-
SUGGESTED READINGS
dence that confirms the efficacy and cost effectiveness of surgical Asher AL, McCormick PC, Selden NR, et al. The National Neurosur-
procedures, observational studies still have their role. They are gery Quality and Outcomes Database and NeuroPoint Alliance: ratio-
appropriate when used to test early innovative ideas and when nale, development, and implementation. Neurosurg Focus. 2013;34:E2.
researchers gather data to allow sample size estimates for pivotal Balshem H, Helfand M, Schunemann HJ, et al. GRADE guidelines: 3.
controlled trials. The mistake in the past was to assume that Rating the quality of evidence. J Clin Epidemiol. 2011;64:401-406.
observational data per se suffice to prove a hypothesis, which is Glasziou P, Chalmers I, Rawlins M, et al. When are randomised con-
not the case. Scientifically robust methods to assess the value of trolled trials unnecessary? Picking signals from noise. BMJ. 2007;334:
a new intervention should be rigorously employed. 349-351.
Trials such as the ones just proposed would undoubtedly add Haines SJ. Evidence-based neurosurgery. Neurosurgery. 2003;52:36-47.
Jurgens TP, May A. Role of sphenopalatine ganglion stimulation in
to clinical practice but are costly and time consuming. They cluster headache. Curr Pain Headache Rep. 2014;18:433.
require multicenter and even international collaborations to be McCulloch P, Altman DG, Campbell WB, et al. No surgical innovation
adequately powered, with added attention to standardization of without evaluation: the IDEAL recommendations. Lancet. 2009;374:
surgical procedures and patient selection. In an effort to encour- 1105-1112.
age greater engagement, groups such as the NeuroPoint Alliance Sackett DL, Rosenberg WM, Gray JA, et al. Evidence-based medicine:
(http://www.neuropoint.org/index.html) have been developed by what it is and what it isn’t. BMJ. 1996;312:71-72.
the American Association of Neurological Surgeons to collect, Sivakanthan S, van Gompel JJ, Alikhani P, et al. Surgical management of
analyze, and report nationwide clinical data.58 This alliance also trigeminal neuralgia: use and cost-effectiveness from an analysis of the
provides clinical trial management, study design, and survey Medicare Claims Database. Neurosurgery. 2014;75:220-226.
Truini A, Galeotti F, Cruccu G. New insight into trigeminal neuralgia.
facilitation. J Headache Pain. 2005;6:237-239.
Zakrzewska JM, Akram H. Neurosurgical interventions for the treatment
CONCLUSION of classical trigeminal neuralgia. Cochrane Database Syst Rev. 2011;(9):
CD007312, doi:10.1002/14651858.CD007312.pub2.
Surgical management of facial pain is based almost exclusively on
observational data, and controlled studies are all but lacking. In See a full reference list on ExpertConsult.com
CHAPTER 170  Evidence-Based Approach to the Treatment of Facial Pain 1396.e1

REFERENCES 25. Barker IIFG, Jannetta BJ, Bissonette DJ, et al. The long-term
1. Pollock BE. Surgical management of medically refractory trigeminal outcome of microvascular decompression for trigeminal neuralgia. N 170
neuralgia. Curr Neurol Neurosci Rep. 2012;12:125-131. Engl J Med. 1996;334:1077-1083.
2. Apfelbaum RI. A comparison of percutaneous radiofrequency tri- 26. Taha JM, Tew JM Jr. Comparison of surgical treatments for trigemi-
geminal neurolysis and microvascular decompression of the trigemi- nal neuralgia: re-evaluation of radiofrequency rhizotomy. Neurosur-
nal nerve for the treatment of tic doloureux. Neurosurgery. 1977;1: gery. 1996;38:865-871.
16-21. 27. Glasziou P, Chalmers I, Rawlins M, et al. When are randomised
3. Burchiel KJ, Stege TD, Howe JF, et al. Comparison of percutaneous controlled trials unnecessary? Picking signals from noise. BMJ.
radiofrequency gangliolysis and microvascular decompression for 2007;334:349-351.
the surgical management of tic douloureux. Neurosurgery. 1981;9: 28. Balshem H, Helfand M, Schunemann HJ, et al. GRADE guide-
111-118. lines: 3. Rating the quality of evidence. J Clin Epidemiol. 2011;64:
4. Patil CC, Veeravagu A, Bower RS, et al. CyberKnife radiosurgical 401-406.
rhizotomy for the treatment of atypical trigeminal nerve pain. Neu- 29. Reddy VK, Parker SL, Lockney DT, et al. Percutaneous stereotactic
rosurg Focus. 2007;23:E9. radiofrequency lesioning for trigeminal neuralgia: determination of
5. Sivakanthan S, van Gompel JJ, Alikhani P, et al. Surgical management minimum clinically important difference in pain improvement for
of trigeminal neuralgia: use and cost-effectiveness from an analysis patient-reported outcomes. Neurosurgery. 2014;74:262-266.
of the Medicare Claims Database. Neurosurgery. 2014;75:220-226. 30. Reddy VK, Parker SL, Patrawala SA, et al. Microvascular decom-
6. Rosenbaum BP, Kelly ML, Kshettry VR, et al. Practice patterns of pression for classic trigeminal neuralgia: determination of minimum
in-hospital surgical treatment of trigeminal neuralgia from 1980 to clinically important difference in pain improvement for patient-
2010. Clin Neurol Neurosurg. 2014;120:55-63. reported outcomes. Neurosurgery. 2013;72:749-754.
7. Lopez BC, Hamlyn PJ, Zakrzewska JM. Systematic review of ablative 31. Zakrzewska JM, Akram H. Neurosurgical interventions for the treat-
neurosurgical techniques for the treatment of trigeminal neuralgia. ment of classical trigeminal neuralgia. Cochrane Database Syst Rev.
Neurosurgery. 2004;54:973-983. 2011;(9):CD007312, doi:10.1002/14651858.CD007312.pub2.
8. Cheng JS, Lim DA, Chang EF, et al. A review of percutaneous 32. Meglio M, Cioni B, Moles A, et al. Microvascular decompression
treatments for trigeminal neuralgia. Neurosurgery. 2014;10(suppl 1): versus percutaneous procedures for typical trigeminal neural-
25-33. gia: personal experience. Stereotact Funct Neurosurg. 1990;54-55:
9. Peker S, Dincer A, Pamir N. Vascular compression of the trigeminal 76-79.
nerve is a frequent finding in asymptomatic individuals: 3-T MR 33. Zakrzewska JM, Lopez BC, Kim SE, et al. Patient reports of satis-
imaging of 200 trigeminal nerves using CISS sequences. Acta Neuro- faction after microvascular decompression and partial sensory rhi-
chir (Wien). 2009;151:1081-1088. zotomy for trigeminal neuralgia. Neurosurgery. 2005;56:1304-1311.
10. Lee A, McCartney S, Burbridge C, et al. Trigeminal neuralgia occurs 34. Degn J, Brennum J. Surgical treatment of trigeminal neuralgia.
and recurs in the absence of neurovascular compression. J Neurosurg. Results from the use of glycerol injection, microvascular decompres-
2014;120:1048-1051. sion, and rhizotomia. Acta Neurochir (Wien). 2010;152:2125-2132.
11. Tatli M, Satci O, Kanpolat Y, et al. Various surgical modalities for 35. Chen J-F, Lee S-T. Comparison of percutaneous trigeminal gan-
trigeminal neuralgia: literature study of respective long-term out- glion compression and microvascular decompression for the man-
comes. Acta Neurochir (Wien). 2008;15:243-255. agement of trigeminal neuralgia. Clin Neurol Neurosurg. 2003;105:
12. Sarsam Z, Garcia-Finana M, Nurmikko T, et al. The long-term 203-208.
outcome of microvascular decompression for trigeminal neuralgia. 36. Dhople AA, Adams JR, Maggio WW, et al. Long-term outcomes of
Br J Neurosurg. 2010;24:18-25. Gamma Knife radiosurgery for classic trigeminal neuralgia: implica-
13. Zhang H, Lei D, You C, et al. The long-term outcome predictors of tions of treatment and critical review of the literature. J Neurosurg.
pure microvascular decompression for primary trigeminal neuralgia. 2009;111:351-358.
World Neurosurg. 2013;79:756-762. 37. Regis J, Tuleasca C. Fifteen years of Gamma Knife surgery for tri-
14. Nurmikko TJ, Eldridge PR. Trigeminal neuralgia—aetiology, patho- geminal neuralgia in the Journal of Neurosurgery: history of a revolu-
physiology and current management. Br J Anaesth. 2001;87:117-132. tion in functional neurosurgery. J Neurosurg. 2011;115:205.
15. Devor M, Amir R, Rappaport ZH. Pathophysiology of trigeminal 38. Parmar M, Sharma N, Modgill V, et al. Comparative evaluation of
neuralgia: the ignition hypothesis. Clin J Pain. 2002;18:4-13. surgical procedures for trigeminal neuralgia. J Maxillofac Oral Surg.
16. Truini A, Galeotti F, Cruccu G. New insight into trigeminal neural- 2013;12:400-409.
gia. J Headache Pain. 2005;6:237-239. 39. Eldridge PR, Sinha AK, Javadpour M, et al. Microvascular decom-
17. Leandri M, Eldridge P, Miles J. Recovery of nerve conduction fol- pression for trigeminal neuralgia in patients with multiple sclerosis.
lowing microvascular decompression for trigeminal neuralgia. Neu- Stereotact Funct Neurosurg. 2003;81:57-64.
rology. 1998;51:1641-1646. 40. Pollock BE. Percutaneous retrogasserian glycerol rhizotomy for
18. Broggi G, Ferroli P, Franzini A, et al. Operative findings and out- patients with idiopathic trigeminal neuralgia: a prospective analysis
comes of microvascular decompression for trigeminal neuralgia in of factors related to pain relief. J Neurosurg. 2005;102:223-228.
35 patients affected by multiple sclerosis. Neurosurgery. 2014;55: 41. Sackett DL, Rosenberg WM, Gray JA, et al. Evidence-based medi-
830-839. cine: what it is and what it isn’t. BMJ. 1996;312:71-72.
19. Tyler-Kabara EC, Kassam AB, Horowitz MH, et al. Predictors of 42. Glasziou P, Vandenbroucke J, Chalmers I. Assessing the quality of
outcome in surgically managed patients with typical and atypical research. BMJ. 2004;328:39-41.
trigeminal neuralgia: comparison of results following microvascular 43. Brainin M, Barnes M, Baron J-C, et al. Guidelines for preparation of
decompression. J Neurosurg. 2002;96:527-531. neurological management guidelines by EFNS scientific task forces.
20. Zacest AC, Magill ST, Miller J, et al. Preoperative magnetic reso- Eur J Neurol. 2004;11:577-581.
nance imaging in type 2 trigeminal neuralgia. J Neurosurg. 2010; 44. Haines SJ. Evidence-based neurosurgery. Neurosurgery. 2003;52:
113:511-515. 36-47.
21. Hong W, Cheng X, Wu Z, et al. Clinical features and surgical treat- 45. Quessy SN, Rowbotham MC. Placebo response in neuropathic pain
ment of trigeminal neuralgia caused solely by venous compression. trials. Pain. 2008;138:479-483.
Acta Neurochir (Wien). 2011;153:1037-1042. 46. Moseley JB, O’Malley K, Petersen JD, et al. A controlled trial of
22. Sindou M, Leston J, Howeidy T, et al. Micro-vascular decompression arthroscopic surgery for osteoarthritis of the knee. N Engl J Med.
for primary trigeminal neuralgia (typical or atypical). Long term 2002;347:81-88.
effectiveness on pain: prospective study with survival analysis in a 47. Sihvonen R, Paavola M, Malmivaara A, et al. Arthroscopic partial
consecutive series of 362 patients. Acta Neurochir (Wien). 2006;148: meniscectomy versus sham surgery for a degenerative meniscal tear.
1235-1245. N Engl J Med. 2013;369:2515-2524.
23. Amador N, Pollock BE. Repeat posterior fossa exploration for 48. Swank DJ, Swank-Bordewijk SCG, Hop WCJ, et al. Laparoscopic
patients with persistent or recurrent idiopathic trigeminal neuralgia. adhesiolysis in patients with chronic abdominal pain. A blinded ran-
J Neurosurg. 2008;108:916-920. domised controlled multi-centre trial. Lancet. 2003;361:1247-1251.
24. Zakrzewska JM, Coakham HB. Microvascular decompression for 49. Jurgens TP, May A. Role of sphenopalatine ganglion stimulation in
trigeminal neuralgia: update. Curr Opin Neurol. 2012;25:296-301. cluster headache. Curr Pain Headache Rep. 2014;18:433.
1396.e2 SECTION 6  Pain

50. Martelletti P, Jensen RH, Antal A, et al. Neuromodulation of chronic 55. Huang CF, Chou SY, Wu MF, et al. Gamma Knife surgery for recur-
headaches: position statement from the European Headache Federa- rent or residual trigeminal neuralgia after a failed initial procedure.
tion. J Headache Pain. 2013;14:86. J Neurosurg. 2010;113(suppl):172-177.
51. Willbrink LA, Teernstra OPM, Haan J, et al. Occipital nerve stimula- 56. Bender M, Pradilla G, Batra S, et al. Effectiveness of repeat glycerol
tion in medically intractable, chronic cluster headache. The ICON rhizotomy in treating recurrent trigeminal neuralgia. Neurosurgery.
study: rationale and protocol of a randomised trial. Cephalalgia. 2012;70:1125-1233.
2013;33:1238-1247. 57. McCulloch P, Altman DG, Campbell WB, et al. No surgical innova-
52. Fontaine D, Lazorthes Y, Mertens P, et al. Safety and efficacy in deep tion without evaluation: the IDEAL recommendations. Lancet.
brain stimulation for refractory cluster headache: a randomized, 2009;374:1105-1112.
placebo-controlled, double-blind trial followed by an open extension. 58. Asher AL, McCormick PC, Selden NR, et al. The National Neuro-
J Headache Pain. 2010;11:23-31. surgery Quality and Outcomes Database and NeuroPoint Alliance:
53. Schoenen J, Jensen RH, Lanteri-Minet M, et al. Stimulation of the rationale, development, and implementation. Neurosurg Focus. 2013;
sphenopalatine ganglion (SPG) for cluster headache treatment. 34:E2.
Pathway CH-1: a randomized, sham-controlled study. Cephalalgia.
2013;33:816-830.
54. Dworkin DH, O’Connor AB, Kent J, et al. Interventional manage-
ment of neuropathic pain: NeuPSIG recommendations. Pain. 2013;
154:2249-2261.
171 Nonoperative
171
Trigeminal Neuralgia: Diagnosis and
Management
Mark E. Linskey

According to the Headache Classification Committee of the described “a type of pain which affects the teeth on one side and
International Headache Society (IHS), trigeminal neuralgia (TN) the whole of the jaw on the side which is painful”.16 The 17th
is a disorder characterized by recurrent unilateral brief, electric, century physician John Locke described the symptoms of TN in
shock-like pains, abrupt in onset and termination, limited to the the wife of the English ambassador to France. Faced with limited
distribution of one or more divisions of the trigeminal nerve, treatment options for the patient’s excruciating pain, the physi-
often triggered by innocuous trigeminal tactile stimuli.1 It may cian opted for eight rounds of cleansing of the gastrointestinal
develop without apparent cause (classic or typical TN) or may be tract, which reportedly resulted in remission of symptoms.17
a result of another diagnosed disorder (secondary or symptomatic An important breakthrough in the management of TN
TN).2-4 Other diagnosed disorders that can lead to secondary TN occurred fortuitously in 1942 when Bergouignan administered
include intrinsic brainstem pathology with trigeminal nerve, his patients the new anticonvulsant diphenylhydantoin (phenyt-
nuclei, or tract involvement (e.g., multiple sclerosis [MS] or oin).18 Before this, the condition had been referred to as neuralgia
lacunar infarction), or extrinsic cerebellopontine angle pathology epileptiform because of a prior hypothesis by Trousseau in 1853
(e.g., neoplasms, benign or malignant or nonneoplastic cysts such that TN, being paroxysmal, was related to abnormal impulse
as epidermoid, dermoid, or arachnoid cysts or vascular lesions conduction, analogous to epilepsy.19 When carbamazepine, a new
such as aneurysms or arteriovenous malformations). In addition, medication for epilepsy, was introduced in 1962, it was soon
there may or may not be persistent background facial pain of found to be useful in patients with TN.20,21 It had greater efficacy
moderate intensity, usually comprising less than 50% of the and less toxicity than the hydantoins and remains to this day the
overall syndrome (classic TN with concomitant persistent facial mainstay of medical therapy.4
pain, sometimes called atypical TN).1-5
Considerable progress has occurred in determining the etiol-
ogy of TN. In the majority of patients with classic TN, the pain
EPIDEMIOLOGY
is generated because of compression of the trigeminal nerve at In the United States the annual incidence of TN (age adjusted
the root entry zone (point in the proximal nerve root central to the 1980 age distribution of the United States) is 5.9 per
oligodendrocyte myelin persists and has not yet given way to 100,000 women and 3.4 per 100,000 men.22 The incidence tends
peripheral Schwann cell myelin). The plaques of demyelination to be slightly higher in women at all ages, and increases with age.
that occur lead to hyperexcitability of exposed and potentially Peak incidence is between ages 50 and 60. Point prevalence has
injured afferents, which results in afterdischarges large enough been estimated to be 0.001.2 A retrospective cohort study in
to cause a nonnociceptive signal being perceived as pain.5 Cur- U.K. primary care, with a very loose definition of TN that
rently, the most widely accepted theory to explain TN is the one likely included many patients with other forms of facial pain,
proposed by Devor—the ignition theory.6 It is likely that both reported a higher incidence of 26.8 per 100,000 person-years.23
central nervous system and nerve root changes occur over time, A similar primary care study carried out in Holland reported an
which would account for why not all patients get permanent relief incidence of 12.5 per 100,000 person-years (95% CI, 10.4-14.9)
after relief of vascular compression of the nerve root. when trained neurologists reviewed the data.24 A population-
There may also be genetic and/or myelin biologic predisposi- based study in Germany reported a lifetime prevalence of
tions, given that rare reports of genetic and familial associations 0.3% (95% CI, 0.1% to 0.5%).25 Pediatric TN does exist, but is
do exist.7-15 However, at least one fascinating study suggests that even more rare and is estimated to represent less than 1.5% of
environmental factors may also play a role. Hemminki and col- all TN cases.26-28
leagues constructed a nationwide neurological database from the Bilateral involvement is present in only 5% of cases. In a
Swedish multigenerational register on 0- to 69-year-old siblings retrospective study by Pollack and colleagues, patients with bilat-
that was linked to the hospital discharge register for the 15-year eral TN were more commonly females (74% versus 58%, P < .1),
period 1987 to 2001.12 They found that if one sibling had TN, had a higher rate of “familial” TN (17% versus 4.1%, P < .001),
the standardized risk ratio for another sibling having TN was and had a greater increased incidence of additional cranial nerve
2.36 (95% confidence interval [CI], 0.85-5.99), confirming a dysfunction (17% versus 6.6%, P < .05) and hypertension (34%
familial tendency. However, in an interesting environmental versus 19%, P < .05).13
control analysis, they showed that if a wife had TN, the standard-
ized risk ratio for development of TN in her spouse was 2.21
(95% CI, 1.10-3.97), and if a husband had TN, the standardized
RISK FACTORS
risk ratio for development of TN in his spouse was 1.62 (95% The major risk factor for TN is MS (for unilateral TN—risk ratio
CI, 0.69-3.21)—suggesting that common familial environmental [RR], 20.0; 95% CI, 4.1-59.0).29 Despite this risk factor, less than
exposures could account for a significant portion of this familial 5% of patients with unilateral TN will be found to have MS.
association.12 Conversely, less than 2% of patients with MS will eventually
develop TN, and in approximately 85% of patients the diagnosis
of MS will already have been made based on the basis of other
HISTORICAL PERSPECTIVE symptoms, imaging, or laboratory findings before TN develops.30
One of the earliest known descriptions of paroxysmal facial pain With MS-associated TN, the demyelination plaque is character-
was by the Arab physician Jurjani in the 11th century. He istically located either in the nerve root entry zone itself or in the
1397
1398 SECTION 6  Pain

intrapontine fascicular fibers of the trigeminal nerve, sometimes last years. Between paroxysms, the person is asymptomatic.
also including the trigeminal nucleus sensorius principalis or the Although the attacks last only seconds to minutes (Fig. 171-2),
trigeminal nucleus spinalis.31 In patients with bilateral TN, the they can cluster together in “tetanic bursts” that can last minutes
incidence of MS is much higher (>18%), and bilateral TN, espe- to hours. In some cases the bursts are individual shocks with reso-
cially in a younger patient (<40 years old), should trigger consid- lution between the shocks (Fig. 171-3A). In others, the bursts
eration of MS as an underlying cause.5,30,32 have incomplete resolution between the shocks (Fig. 171-3B). A
Hypertension is a risk factor in women (RR, 2.1; 95% CI, pain that is continuous and unvarying for many minutes to hours
1.2-3.4), but the evidence is less clear for men (RR, 1.53; 95% (Fig. 171-3C and D) should lead to consideration of differential
CI, 0.30-4.50).29 A history of TN in a first-degree relative is also diagnoses.3,4
a minor risk factor.7-15 Although approximately 10% of TN patients do not respond
to antiepileptic drug (AED) treatment, up to 90% of TN patients
do, at least initially.33 Thus a trial of appropriate AED treatment
DIAGNOSIS can be used as both a therapeutic option and as a diagnostic chal-
TN is a characteristic pain in the distribution of one or more lenge, as long as it is not considered to be an absolute criterion
branches of the fifth cranial nerve (Fig. 171-1). In the majority or pathognomonic.
of cases the pain begins in the second or third divisions of the Despite the exclusion of patients with evidence of sensory
trigeminal nerve (V2 and V3, respectively). With time it can deficit from the IHS definition of TN,1 abundant evidence sug-
spread to other divisions including the first division (V1). gests that a subset of TN patients have subtle sensory deficits that
However, TN involves V1 in isolation in only 5% of cases.2-5,29 can be picked up by either careful and thorough neurological
Isolated V1 involvement should lead to careful consideration of examination34-36 or by trigeminal sensory evoked potential
differential diagnoses.3,4 study.37,38 In one TN experience looking mostly at patients who
The diagnosis is made on the history alone, based on charac- had had the syndrome for many years, subtle sensory deficits were
teristic features of the pain.1-5 It occurs in sudden paroxysms, with detectable in the medial aspect of the V2 or less commonly the
each pain lasting a few seconds to several minutes. The pain is V3 division in up to 30% of cases.3 Nevertheless, early, signifi-
characteristically so severe that it is “arresting.” Whatever the cant, and nonsurgical sensory loss, in addition to bilateral involve-
patient is doing is immediately stopped, and the patient usually ment, should trigger a thorough investigation for symptomatic
either is frozen in position or contortion or suddenly reaches (i.e., secondary) TN.
up to grab the face. The pain is usually described as sharp, stab- There is growing evidence to suggest that classic TN with no
bing, electric, or shock-like. It characteristically resolves as sud- pain between episodes evolves over time into a syndrome in
denly as it started. The first time the pain is experienced is usually which a minor component of constant background trigeminal
memorable. Patients may not be able to name a date or time, pain (<50% of the overall facial pain syndrome) can develop while
but they can usually describe the circumstances and exactly the syndrome remains dominated by intermittent sudden sharp,
what they were doing when it first occurred. Typically, the pain stabbing, electric, shock-like episodes.39
can be triggered by light touch on any area innervated by the The condition can impair activities of daily living, impair job
trigeminal nerve, including such things as wind, chewing, talking, performance and/or professional advancement, and lead to social
washing the face, applying or removing makeup, shaving, or withdrawal and/or depression.5 Before the discovery of AEDs,
cleaning the teeth. patients occasionally resorted to suicide to escape the fear of
The frequency of paroxysms is highly variable, ranging from severe and unpredictable attacks.17 It was during this period that
hundreds of attacks a day to long periods of remission that can TN developed the moniker of “the suicide disease.”

Seconds, no afterpain

Shooting, sharp Attacks Seconds, dull afterpain


for minutes
Terrifying, exhausting Character Timing

Unbearable Periods of complete remission


weeks, months

Eating
Clinical features of Light touch
Moderate to severe Severity
trigeminal neuralgia
Provoking Talking

Right up to 60% Trigeminal nerve Spontaneous


Washing
Site
First division rare Sometimes sleep
Unilateral 97%
Relieving
Drugs

Trigger points
Associated
May be sensory change

Figure 171-1. Major clinical features of trigeminal neuralgia. (From Zakrzewska JM, Linskey ME. Trigeminal
neuralgia. In: Zakrzewska JM, ed. Orofacial Pain. New York: Oxford University Press; 2008:119-134.)
CHAPTER 171  Trigeminal Neuralgia: Diagnosis and Nonoperative Management 1399

171

Pain intensity
Pain intensity
Pain intensity

A Time B Time C Time

Figure 171-2. Pictogram describing the typical time onset, resolution, and severity early in the course of the
syndrome (A) and later in the course as the pain frequency and severity increase (B). The pain is
characteristically sudden in onset and resolution without pain between episodes and lasts only seconds to a
few minutes. With chronicity, a constant persisting background dull, aching, or burning pain may develop (C),
but this component is usually minor in comparison to the characteristic episodes from the patient perspective.

Minutes-to-hours Minutes-to-hours Minutes-to-hours Minutes-to-hours


Pain intensity

Pain intensity

Pain intensity

Pain intensity
A Time B Time C Time D Time

Figure 171-3. Pictogram demonstrating how short bursts of episodic pain can be interpreted by trigeminal
neuralgia (TN) patients as lasting many minutes or even a few hours. If the transient spikes cluster in tetanic
bursts, the patient may time the length of the cluster rather than the component episodes, and the clinician
may be fooled into discounting the syndrome as TN. These bursts may cluster with either complete (A) or
partial (B) resolution between spikes. In our experience, patients who describe a sudden onset of pain that
lasts many minutes to hours and remains at the same intensity level until sudden (C) or gradual (D) resolution
are not likely to have TN.

Certainly, before the AED treatment era, patients with TN (SUNA). In a study of V1 division neuralgic pain TN patients, a
were so desperate for relief that they were willing to undergo median pain episode duration of 4 seconds (range, 2-32 seconds)
brain surgery at a time when the risk of brain surgery mortality was found, compared with 40 seconds (range, 5-250 seconds) for
was still relatively high. As a result, TN surgery became strongly SUNCT syndrome patients, 10 minutes (range, 3-46 minutes)
associated with early neurosurgery pioneers and neurosurgical for chronic paroxysmal hemicrania patients, and 38 minutes
technical developments.40-43 (range, 8-238 minutes) for cluster headache patients.44
Association of trigeminal pain with autonomic symptoms
including dermatomal flushing, ptosis, conjunctival injection,
Differential Diagnosis tearing, or rhinitis should trigger additional work-up to rule out
Common conditions that mimic TN as well as their presenting other conditions such as cluster headache syndrome, SUNCT,
features are listed in Table 171-1. Continuous trigeminal pain, if SUNA, or chronic regional pain syndrome (CRPS), formerly
the dominant complaint (>50% of the overall orofacial pain syn- known as reflex sympathetic dystrophy.3-5
drome), is usually a sign of a disorder other than TN and should Less than 5% of TN patients have bilateral involvement. If
trigger an appropriate differential work-up. present, it overwhelmingly manifests as first unilateral involve-
Isolated V1 involvement occurs in less than 5% of TN ment, to be later followed by contralateral involvement. A diag-
patients and should trigger additional work-up to rule out other nosis of bilateral TN, particularly developing in a patient younger
conditions such as temporal arteritis, herpes zoster, ocular than age 40, should lead to a very careful investigation into the
migraine, paroxysmal hemicranias, cluster headache syndrome, possibility of MS. Simultaneous onset of bilateral involvement
short-lasting unilateral neuralgiform headaches with conjuncti- should trigger work-up for a broader orofacial pain differential
val injection and tearing (SUNCT), or short-lasting unilateral diagnosis.3-5 Bilateral TN occurs in 11% to 30% of MS TN
neuralgiform headaches with cranial autonomic symptoms patients versus 3% to 10% of non-MS TN patients.32,45 Patients
1402 SECTION 6  Pain

with MS-associated TN have a younger median age of onset to be effective for TN,18 it has not been tested in a prospective
(45 years) compared with their non-MS–associated TN counter- randomized clinical trial and has been largely bypassed for
parts (54 years).30 chronic therapy owing to its relatively high toxicity profile.
In the absence of prior surgery for TN, demonstrable trigemi- However, it is still useful in emergency room, acute extremis situ-
nal hypesthesia and/or hypalgesia are rare and is usually present ations because it can be rapidly loaded to full therapeutic dose by
only in patients with chronic TN. Presence in a newly diagnosed intravenous bolus.
patient should trigger work-up for diseases other than TN, par- Carbamazepine is considered the most proven, first-line treat-
ticularly for lesions such as cavernous sinus or cerebellopontine ment for TN. In the United Kingdom, this position has now been
angle masses, or perineural spread of head and neck cancer formally codified by the National Institute for Health and Care
(including cutaneous malignancy and adenoid cystic carcinoma). Excellence (NICE), which advocates that carbamazepine should
Magnetic resonance imaging (MRI) with and without contrast is be offered as the initial treatment for patients with TN.47
very useful for ruling out structural, nonvascular, compressive Carbamazepine is associated with a very rare risk of poten-
causes of TN or other orofacial pain syndromes.3-5 In patients tially life-threatening skin reactions including Stevens-Johnson
with trigeminal sensory loss studied with MRI, MS, infarct, and syndrome. However, the risk of this condition is markedly
glioma are the most common abnormalities found in the brain- increased in people with the allele HLA-B*1502. The frequency
stem; acoustic and trigeminal schwannomas, meningiomas, epi- of this allele varies worldwide and is highest in some Asian popu-
dermoid cysts, lipomas, and metastases are the most common lations. Individuals of Han Chinese, Hong Kong Chinese, or
abnormalities identified in the cistern; meningiomas, trigeminal Thai origin should be screened for HLA-B*1502 before starting
schwannomas, epidermoid cysts, metastases, pituitary adenomas, treatment with carbamazepine.48
and aneurysms are the most common abnormalities identified in Carbamazepine is an enzyme inducer and interacts with the
Meckel’s cave and the cavernous sinus; and malignant tumors, renal distal tubule antidiuretic hormone receptor, which can lead
which may demonstrate perineural tumor spread, are the most to hyponatremia. A full blood count; measurements of electro-
common extracranial abnormality identified.46 lytes, liver enzymes, and vitamin D levels; and other tests of bone
metabolism (e.g., serum calcium and alkaline phosphatase) should
be performed before and a few weeks after the start of carbam-
NONOPERATIVE MANAGEMENT azepine therapy and then every 2 to 5 years.49 Clinicians should
The first-line treatment for TN is medical or pharmacologic start or stop treatment by changing the dose in increments over
(Table 171-2). Although phenytoin was the first AED ever shown several days to reduce common adverse effects. After treatment

TABLE 171-2  Common Drugs Used for the Medical Management of Trigeminal Neuralgia (TN)
Drug Level of Evidence Effect Adverse Effects Suggested Dose Comment
Carbamazepine Systematic review NNT for pain Drowsiness, ataxia, 100 mg twice daily; Dose may need to be
(the only of four relief is 1.9; nausea, increase as necessary adjusted after 3 weeks
first-line randomized 72% of constipation by 50-100 mg every because of enzyme
agent) controlled trials patients had (minor); NNT, 3.7 3-4 days; target range induction.
(n = 160) excellent or 400-1000 mg/day
good response
Baclofen One controlled trial 7/10 improved Drowsiness, 10 mg three times daily; May be useful in patients
compared with baclofen; hypotonia; avoid increase as necessary with multiple sclerosis, in
baclofen with 0/10 improved abrupt by 10 mg/day; target whom its antispasticity
placebo (n = 10) with placebo withdrawal dose 50-60 mg daily effects can be harnessed.
(P = .05)
Gabapentin Five uncontrolled Good to excellent Drowsiness, ataxia, 300 mg once daily; Widely used for TN although
studies (n = 123) pain relief in diarrhea (minor); increase as necessary evidence is weak;
40%; any pain NNT, 2.5 by 300 mg every 3 evidence base in other
relief in 53% days in divided doses types of neuropathic
(three times daily); pains much stronger.
target dose
900-2400 mg daily
Lamotrigine One randomized 10/13 improved Drowsiness, 25 mg twice daily; Probably better tolerated
controlled trial on lamotrigine; dizziness, increase by 50 mg than carbamazepine but
with lamotrigine 8/14 improved constipation, weekly; target dose needs slow titration; may
as add-on to on placebo; ns nausea; no 200-600 mg daily therefore have a role in
carbamazepine different from the elderly or patients with
or phenytoin placebo multiple sclerosis who
(n = 14) have less severe disease.
Oxcarbazepine Two uncontrolled Pain relief in all Dizziness, fatigue, 300 mg twice daily; Evidence weak; structurally
studies (n = 21) 21 patients rash, and increase by 600 mg similar to carbamazepine,
hyponatremia weekly; target dose although probably better
600-2400 mg daily tolerated.
Phenytoin Three uncontrolled 77% of patients Drowsiness, ataxia, 300 mg/day; dose First drug used in the
studies (n = 30) reported some dizziness, gum altered to achieve successful management
pain relief hypertrophy therapeutic plasma of TN; little evidence but
concentration rapid dose titration and
once-daily administration
are advantages.
Modified from Zakrzewska JM, Linskey M. Trigeminal neuralgia. In: Zakrzewska JM, ed. Orofacial Pain. London: Oxford University Press; 2009:119-134.
NNT, number needed to treat; ns, not statistically significant.
CHAPTER 171  Trigeminal Neuralgia: Diagnosis and Nonoperative Management 1403

is started, a dose adjustment is often necessary at about 3 weeks medication dose to achieve the same degree of pain relief, until
owing to induction of liver enzymes. a point is reached at which either the medication no longer con- 171
On the basis of observational studies, including a 15-year trols the syndrome or the patient can no longer tolerate the
prospective cohort study,50 oxcarbazepine has also been shown to medication side effects.
be effective for treating patients with TN. It is the first-line treat- Although adding medications in combinations can be consid-
ment for TN in Scandinavian countries and second-line treat- ered, there is no good evidence that patients will achieve signifi-
ment after carbamazepine in North America.5 One nonsystematic cantly durable pain control once an adequate trial of a single
review (three randomized clinical trials totaling 130 people)51 first-line agent (such as carbamazepine or oxcarbazepine) has
found that oxcarbazepine and carbamazepine were associated failed at the maximum tolerable dose. Consideration should be
with similar reductions in attacks (pain, global symptoms) of TN. given to surgical consultation at that point. For carbamazepine,
Although oxcarbazepine has not demonstrated superiority over maximum tolerable doses are usually 800 to 1200 mg/day, but
carbamazepine for pain control, and is more expensive in coun- elderly patients may reach tolerance limits at even lower doses.
tries where it is still under patent, many clinicians prefer it for its
lower toxicity profile.5 TIMING OF SURGICAL CONSULTATION
Although gabapentin has been shown to be effective in treat-
ing some neuropathic pain conditions,52 particularly MS,53 evi- AND INTERVENTION
dence for its use in TN is weak. One randomized controlled trial Unfortunately, many practitioners currently wait too long to
showed that gabapentin plus ropivacaine compared with gaba­ refer patients for surgery. In one postoperative survey of patients
pentin alone can improve pain and functional health status in TN who underwent surgery for their TN, approximately 70% wished
with few or no side effects.54 Pregabalin is another anticonvulsant that they had had their surgery earlier.59 In our practice, although
in the same family as gabapentin that is used for patients with there are certainly exceptions wherein patients reach tolerance
TN with little published evidence to date to support it. Anecdot- sooner or tolerate the medications longer, the large majority of
ally, both gabapentin and pregabalin are often used for patients patients referred for surgical intervention have reached medica-
with MS, patients with a “burning” component to their pain, or tion intolerance or ineffectiveness somewhere between 5 and 10
as an add-on agent for patients already taking carbamazepine or years after symptom onset.
oxcarbazepine. Patients with TN are best managed by a coordinated multi-
Baclofen is also used for patients with TN, despite very weak disciplinary approach. This approach includes early referral to a
evidence in the literature supporting its use.5 Consensus suggests surgeon experienced and skilled in TN surgery for initial consul-
that it may be useful in people with MS who develop TN. These tation and counseling. This should ideally be done well before
patients are often taking baclofen already, and may achieve the nonsurgical members of the team feel that a patient actually
control of symptoms without having to add carbamazepine. needs surgery. It is neither fair to the patient nor likely best
Lamotrigine is also used for patients with TN, despite very practice to have the patient be seen by the surgeon only when
weak evidence in the literature supporting its use. Consensus medical therapy has failed. Under these circumstances, the patient
suggests that it may be useful in people who cannot tolerate or is usually in very severe pain to the point of desperation and will
are allergic to carbamazepine, or as an add-on agent augmenting likely agree to anything offered to help. He or she is also likely
either carbamazepine or oxcarbazepine as their effectiveness to be taking high doses of AEDs, which can impair focus, con-
wains.5 centration, memory, and ability to learn and to make decisions.
CNV1014802 is a novel new sodium channel blocker that has The time to learn about surgical options, their relative pros
selectivity for the NaV 1.7 sodium channel and is being assessed and cons, and the providers available to deliver these options is
in the treatment of TN.55 Initial reports presented June 8, 2015 in the “cool of the moment” when patients are neither desperate
at the IHS International Headache Congress suggested promis- from pain nor relatively incapacitated by high doses of medica-
ing initial results with the 31 patients randomized, with tions. Patients may also have their own ideas about optimal
CNV1014802 therapy failing in only 33% of patients (defined as timing for their lives for moving on to a surgical treatment once
≥50% of the original pain frequency and severity returning) they understand the prognosis, the procedures, and their relative
versus a 64% failure rate in patients taking placebo over the success rates. Contingency plans for surgery are best made well
course of the study.56 While still early in development and assess- in advance, just as one would do for a trust fund or a will. These
ment, CNV1014802 is significant as the first pharmacologic plans can then be implemented with confidence once the appro-
agent being specifically developed to treat TN. priate agreed-on trigger point is reached.
Side effects of treatment with AEDs are quite common, and The paradigm of pursuing surgical intervention for patients
their frequency and impact are usually underestimated by clini- with TN only after medical therapy has failed is in most need
cians treating TN patients.57 Drowsiness and cognitive impair- of rethinking with younger adult patients and especially
ment are the side effects most disliked by patients.57 These side pediatric patients.60,61 For patients in the United States, a general
effects are often dose related and therefore worsen as the syn- idea of median life expectancy for any given patient can be deter-
drome progresses and AED doses increase. mined by going to the Social Security Administration life expec-
tancy calculator website (https://www.ssa.gov/oact/population/
longevity.html) and entering in the patient’s sex and date of birth.
PROGNOSIS This will give the clinician some idea of the timeline for necessary
Consensus experience suggests that TN is a progressive syn- pain control in any given TN patient. If the median life expec-
drome with progression rates that vary widely, patient-to-patient. tancy for a given patient is longer than 20 years (currently age
Life table analytic methods demonstrate that 65% of newly diag- <63.5 for men and <66 for women), then it is not very likely that
nosed TN patients will have a second episode within 5 years, and a strategy consisting solely of medical therapy is likely to be
77% within 10 years.29 Fifty percent of TN patients will experi- effective in the long run. Surgical intervention should probably
ence remissions of at least 6 months’ duration.58 be considered before conditions develop with age that might
The main problem with treating TN patients with AEDs, make general anesthesia or surgical intervention excessively risky.
even if they are tolerated initially, is that the treatment does not Unfortunately, AEDs are not selective. They have general
address the vascular compression cause present in 95% of patients, electrical suppressive effects throughout the brain. In children
nor the fact that TN is a progressive syndrome. Therefore the the brain is still developing, and the effects of these drugs at these
usual result is that, over time, patients require a higher and higher doses are potentially much more severe than in adults. These
1404 SECTION 6  Pain

drugs at TN doses interfere with learning and memory during SUGGESTED READINGS
the key learning period in life. This interferes with the child’s Headache Classification Committee of the International Headache
grades and impairs competitiveness for higher educational and Society (IHS). The International Classification of Headache Disor-
career opportunities. AEDs are general suppressants, and they ders, 3rd ed (beta version). Cephalalgia. 2013;33:629-808.
interfere with successful peer socialization and interaction at a Linskey ME. Trigeminal neuralgia. American College of Physicians (ACP)
Smart Medicine. <http://pier.acponline.org/physicians/diseases/d625/
time when adolescents are developing their social self-confidence d625-pdf.htnl> <http://smartmedicine.acponline.org/content.aspx?gb
and a feel for their place in society. If the child is young enough, osid=299>.
he or she may even be erroneously diagnosed with attention- Linskey ME, Zakrzewska JM Trigeminal neuralgia. BMJ Talk Medicine.
deficit/hyperactivity disorder (ADHD).60,61 April 2015 <https://soundcloud.com/bmjpodcasts> or via <http://
Even the syndrome of TN by itself is potentially devastating journals.bmj.com/site/podcasts/>.
for adolescents from a socialization perspective. Imagine being Zakrzewska JM, Linskey M. Trigeminal neuralgia. In: Zakrzewska JM, ed.
afraid to have someone touch your face or kiss you if you are an Orofacial Pain. London: Oxford University Press; 2009:119-134.
adolescent (especially girls) and the effect that would have on Zakrzewska JM, Linskey ME. Trigeminal neuralgia. BMJ. 2015;350:h1238.
you socially at this vulnerable time. Many even withdraw and doi:10.1136/bmj.h1238; PMID:2576710.
become almost reclusive. Getting them off these medications and
pain free, allowing them to learn, compete, develop profession-
ally, socialize, and gain personal self-confidence is crucial. In
these patients, early surgical intervention should probably be
considered.60,61 See a full reference list on ExpertConsult.com
CHAPTER 171  Trigeminal Neuralgia: Diagnosis and Nonoperative Management 1404.e1

REFERENCES 29. Katusic S, Beard CM, Bergstralh E, et al. Incidence and clinical
1. Headache Classification Committee of the International Headache features of trigeminal neuralgia, Rochester, Minnesota, 1945-1984. 171
Society (IHS). The International Classification of Headache Disor- Ann Neurol. 1990;27:89-95.
ders, 3rd edition (beta version). Cephalalgia. 2013;33:629-808. 30. Brisman R. Trigeminal neuralgia and multiple sclerosis. Arch Neurol.
2. Zakrzewska JM, Linskey M. Trigeminal neuralgia. In: Zakrzewska 1987;44:379-381.
JM, ed. Orofacial Pain. London: Oxford University Press; 2009: 31. Gass A, Kitchen N, MacManus DG, et al. Trigeminal neuralgia in
119-134. patients with multiple sclerosis: lesion localization with magnetic
3. Linskey ME, Jannetta PJ. Differential diagnosis of trigeminal neu- resonance imaging. Neurology. 1997;49:1142-1144.
ralgia: look-a-like diseases and atypical trigeminal neuralgia. In: Jan- 32. Brisman R. Bilateral trigeminal neuralgia. J Neurosurg. 1987;67:
netta PJ, ed. Trigeminal Neuralgia. London: Oxford University Press; 44-48.
2010:74-86. 33. Sato J, Saitoh T, Notani K, et al. Diagnostic significance of carbam-
4. Linskey ME Trigeminal neuralgia. American College of Physicians azepine and trigger zones in trigeminal neuralgia. Oral Surg Oral Med
(ACP) Smart Medicine. <http://pier.acponline.org/physicians/ Oral Pathol Oral Radiol Endod. 2004;97:18-22.
diseases/d625/d625-pdf.htnl> <http://smartmedicine.acponline.org/ 34. Bowsher D, Miles JB, Haggett CE, et al. Trigeminal neuralgia: a
content.aspx?gbosid=299>. quantitative sensory perception threshold study in patients who had
5. Zakrzewska JM, Linskey ME. Trigeminal neuralgia. BMJ. 2015; not undergone previous invasive procedures. J Neurosurg. 1997;86:
350:h1238. doi:10.1136/bmj.h1238; PMID:2576710. 190-192.
6. Devor M, Amir R, Rappaport ZH. Pathophysiology of trigeminal 35. Nurmikko TJ. Altered cutaneous sensation in trigeminal neuralgia.
neuralgia: the ignition hypothesis. Clin J Pain. 2002;18:4-13. Arch Neurol. 1991;48:523-527.
7. Duff JM, Spinner RJ, Lindor NM, et al. Familial trigeminal neuralgia 36. Sinay VJ, Bonamico LH, Dubrovsky A. Subclinical sensory abnor-
and contralateral hemifacial spasm. Neurology. 1999;53:216-218. malities in trigeminal neuralgia. Cephalalgia. 2003;23:541-544.
8. Ebner FH, Tatagiba M, Roser F. Familial trigeminal neuralgia— 37. Bennett MH, Jannetta PJ. Evoked potentials in trigeminal neuralgia.
microsurgical experience and psychological observations. Acta Neu- Neurosurgery. 1983;13:242-247.
rochir (Wien). 2010;152:381-382. 38. Mursch K, Schafer M, Steinhoff BJ, et al. Trigeminal evoked poten-
9. El Otmani H, Moutaouakil F, Fadel H, et al. [Familial trigeminal tials and sensory deficits in atypical facial pain—a comparison with
neuralgia]. Rev Neurol (Paris). 2008;164:384-387. results in trigeminal neuralgia. Funct Neurol. 2002;17:133-136.
10. Fleetwood IG, Innes AM, Hansen SR, et al. Familial trigeminal neu- 39. Burchiel KJ, Slavin KV. On the natural history of trigeminal neural-
ralgia. Case report and review of the literature. J Neurosurg. 2001; gia. Neurosurgery. 2000;46:152-154.
95:513-517. PMID: 11565877. 40. Horsley V. Remarks on the various surgical procedures devised for
11. Hall GC, Carroll D, Parry D, et al. Epidemiology and treatment of the relief of cure of trigeminal neuralgia (tic douloureux). Br Med J.
neuropathic pain: the UK primary care perspective. Pain. 2006;122: 1891;2:1249-1252.
156-162. 41. Spiller W, Frazier C. The division of the sensory root of the trigemi-
12. Hemminki K, Li X, Sundquist K. Familial risks for nerve, nerve root nus for relief of tic douloureux: an experimental pathological and
and plexus disorders in siblings based on hospitalisations in Sweden. clinical study with a preliminary report of one surgical successful
J Epidemiol Community Health. 2007;61:80-84. case. Phila Med J. 1901;8:1039-1049.
13. Pollack IF, Jannetta PJ, Bissonette DJ. Bilateral trigeminal neuralgia: 42. Cushing H. The major trigeminal neuralgias and their surgical treat-
a 14-year experience with microvascular decompression. J Neurosurg. ment based on experiences with 332 gasserian operations. Am J Med
1988;68:559-565. Sci. 1920;160:157-184.
14. Smyth P, Greenough G, Stommel E. Familial trigeminal neuralgia: 43. Dandy WE. Trigeminal neuralgia and trigeminal tic douloureux. In:
case reports and review of the literature. Headache. 2003;43:910- Lewis D, ed. Practice of Surgery. Hagerstown, MD: WF Prior Co;
915. 1932:177-200.
15. Gupta V, Singh AK, Kumar S, et al. Familial trigeminal neuralgia. 44. Pareja JA, Cuadrado ML, Caminero AB, et al. Duration of attacks
Neurol India. 2002;50:87-89. of first division trigeminal neuralgia. Cephalalgia. 2005;25:305-
16. Youkilis AS, Sagher O. Diagnosis and non-operative management of 308.
trigeminal neuralgia. In: Winn HR, ed. Youman’s Neurosurgery. 5th 45. Rushton JG, Olafson RA. Trigeminal neuralgia associated with mul-
ed. Philadelphia: Saunders; 2004:2987-2995. tiple sclerosis. A case report. Arch Neurol. 1965;13:383-386.
17. Stookey BR. Trigeminal Neuralgia. Springfield, IL: Charles C. 46. Majoie CB, Verbeeten B Jr, Dol JA, et al. Trigeminal neuropathy:
Thomas; 1959. evaluation with MR imaging. Radiographics. 1995;15:795-811.
18. Bergouignan M. Cures hereuses de neuralgie facilaes essentielles par 47. Centre for Clinical Practice at NICE (UK). Neuropathic Pain: The
le diphenylhydantoinate de soude. Rev Laryngol Otol Rhinol (Bord). Pharmacological Management of Neuropathic Pain in Adults in Non-
1942;63:34-41. Specialist Settings. London: National Institute for Health and Care
19. Trousseau A. De la neuralgie epileptiforme. Arch Gen Med. 1853;1: Excellence; 2013.
33-44. 48. Ferrell PB Jr, McLeod HL. Carbamazepine, HLA-B*1502
20. Blom S. Trigeminal neuralgia. Its treatment with a new anticonvul- and risk of Stevens-Johnson syndrome and toxic epidermal necroly-
sant drug (G-32883). Lancet. 1962;1:839-840. sis: U.S. FDA recommendations. Pharmacogenomics. 2008;9:1543-
21. Blom S. Tic douloureux treated with a new anticonvulsant. Arch 1546.
Neurol. 1962;9:285-290. 49. National Clinical Guideline Centre (UK). The Epilepsies: The Diag-
22. Zakrzewska JM, Hamlyn PJ. Facial pain. In: Crombie IK, Croft PR, nosis and Management of the Epilepsies in Adults and Children in Primary
Linton SJ, et al., eds. Epidemiology of Pain. Seattle: IAS Press; 1999: and Secondary Care: Pharmacological Update of Clinical Guideline 20.
171-202. London: Royal College of Physicians; 2012.
23. Hall GC, Carroll D, Parry D, et al. Epidemiology and treatment of 50. Zakrzewska JM, Patsalos PN. Long-term cohort study comparing
neuropathic pain: the UK primary care perspective. Pain. 2006;122: medical (oxcarbazepine) and surgical management of intractable tri-
156-162. geminal neuralgia. Pain. 2002;95:259-266.
24. Koopman JS, et al. Incidence of facial pain in the general population. 51. Beydoun A. Safety and efficacy of oxcarbazepine: results of random-
Pain. 2009;147:122-127. ized, double-blind trials. Pharmacotherapy. 2000;20:152S-158S.
25. Mueller D, et al. Prevalence of trigeminal neuralgia and persistent 52. Moore RA, Wiffen PJ, Derry S, et al. Gabapentin for chronic neu-
idiopathic facial pain: a population-based study. Cephalalgia. 2011;31: ropathic pain and fibromyalgia in adults. Cochrane Database Syst Rev.
1542-1548. 2014;(4):CD007938.
26. Resnick DK, Levy EI, Jannetta PJ. Microvascular decompression for 53. De SL, Annunziata P. Symptomatic cranial neuralgias in multiple
pediatric onset trigeminal neuralgia. Neurosurgery. 1998;43:804-807. sclerosis: clinical features and treatment. Clin Neurol Neurosurg. 2012;
27. Bahgat D, Ray DK, Raslan AM, et al. Trigeminal neuralgia in young 114:101-107.
adults. J Neurosurg. 2011;114:1306-1311. 54. Lemos L, Flores S, Oliveira P, et al. Gabapentin supplemented with
28. Bender MT, Pradilla G, James C, et al. Surgical treatment of pedi- ropivacaine block of trigger points improves pain control and quality
atric trigeminal neuralgia: case series and review of the literature. of life in trigeminal neuralgia patients when compared with gaba­
Childs Nerv Syst. 2011;27:2123-2129. pentin alone. Clin J Pain. 2008;24:64-75.
1404.e2 SECTION 6  Pain

55. Zakrzewska JM, Palmer J, Ettlin DA, et al. Novel design for a phase 59. Zakrzewska JM, Lopez BC, Kim SE, et al. Patient reports of satisfac-
IIa placebo-controlled, double-blind randomized withdrawal study tion after microvascular decompression and partial sensory rhizotomy
to evaluate the safety and efficacy of CNV1014802 in patients with for trigeminal neuralgia. Neurosurgery. 2005;56:1304-1311.
trigeminal neuralgia. Trials. 2013;14:402. 60. Linskey ME. Four key points when it comes to pediatric trigeminal
56. Jancin B IHC: promising new drug cuts pain in trigeminal neuralgia. neuralgia. Trigeminal Neuralgia Assoc Q. Spring 2014;1:11-13.
<http://www.pm360online.com/ihc-promising-new-drug-cuts 61. MEG. Pediatric TN issues: four key points. <http://tnkidsrock.com/
-pain-in-trigeminal-neuralgia/>; 2015 Accessed 07.04.16. 2015/04/27/pediatric-tn-issues-first-issue-us-medical-healthcare
57. Zakrzewska JM. Consumer views on management of trigeminal neu- -training-structure-culture/>; 2015 Accessed 07.04.16.
ralgia. Headache. 2001;41:369-376.
58. Rushton JG, MacDonald HNA. Trigeminal neuralgia. Special con-
siderations of nonsurgical treatment. JAMA. 1957;165:437-440.
172 Percutaneous Procedures for Trigeminal Neuralgia
172

Andrew L. Ko and Kim J. Burchiel

HISTORICAL DEVELOPMENT OF PERCUTANEOUS shape of the foramen ovale is typically oval, yet it can be almond
shaped, round, or slit-like. The average length and width are
PROCEDURES FOR TRIGEMINAL NEURALGIA 7.46 mm ± 1.41 mm and 3.21 mm ± 1.02 mm, respectively.26 The
Percutaneous procedures for treatment of trigeminal neuralgia foramen may be divided into two or three components in 4.5%
were first introduced in 1853 by Patruban, who divided the maxil- of cases.27
lary nerve behind the orbit by passing a tenotome along the floor Several nerves, arteries, and veins pass through the foramen
of the orbit and cutting the nerve as far back as possible.1 In 1910, ovale: the mandibular nerve, lesser superficial petrosal nerve,
Harris applied a lateral approach to reach the foramen ovale and accessory meningeal artery, and emissary veins (from the cavern-
inject alcohol.2 In 1914, Härtel modified the approach to an ous sinus to the pterygoid plexus). The optic ganglion is situated
anterior one similar to that currently practiced,3,4 and in 1930, directly under the foramen but may also pass through the foramen
Irger used an inferior approach (both approaches used alcohol ovale.25
injection).4,5 It was Rethi who first introduced the use of electro- The gasserian ganglion originates from the cranial neural
lytic techniques for treatment of trigeminal neuralgia.6 In 1960, crest and the overlying thickened ectoderm. The three processes
radiofrequency (RF) lesioning was introduced by Sweet, and it of the ganglion can be identified at 6 weeks’ gestation; the oph-
was applied to the treatment of trigeminal neuralgia in 1974.7,8 thalmic division develops first, followed by the maxillary and
Serendipity led to the application of glycerol to treat trigeminal mandibular divisions. Meckel’s cave attains its final shape at 12
neuralgia. In 1981, Häkanson,9 used glycerol as a medium to weeks’ gestation, with the arachnoid ending around the ganglia.28
introduce tantalum dust into the trigeminal cistern as part of a The gasserian ganglion is a sensory ganglion of the trigeminal
stereotactic radiosurgical technique for the treatment of trigemi- nerve; it is crescent shaped with convexity directed forward. It is
nal neuralgia. He noticed that patients became pain free imme- located at the apex of the petrous portion of the temporal bone
diately. Later, he used glycerol alone and confirmed the pain relief and may extend to the foramen lacerum, or the posterior lip or
effect. By applying Taarnhøj and Shelden’s10-12 principle of inter- floor of the foramen ovale. The mean distance from the foramen
nal neurolysis of the root of the trigeminal nerve in the dural ovale to the gasserian ganglion is 6 mm (range, 5.8 to 6.3 mm).29
canal over the margin of the petrous ridge, Mullan and Lichtor,13,14 The anterior border of the ganglion is usually within 2 mm in
in 1983, introduced the technique of percutaneous balloon com- front of, or behind, the posterior lip of the foramen ovale. The
pression of the gasserian ganglion. ganglion’s dural covering forms Meckel’s cave and, medially, the
The development of procedural safety was necessary for the cavernous sinus.30
widespread adoption of percutaneous techniques for treatment of Meckel’s cave is confined between the outer and inner layers
trigeminal neuralgia. Visualization of needle placement to avoid of the dura but also contains a separate sleeve of meningeal dura
extratrigeminal injury was first addressed in 1916 by Pollock and formed by extension of the posterior fossa dura into the middle
Potter15 in cadaveric studies, in which the needle site during fossa. Surfaces are concave inferiorly and medially and more flat
alcohol injection was confirmed using radiography. It was not superiorly. The sensory branches of the trigeminal nerve and
until 1936 that Putnam and Hampton applied this procedure to partially the gasserian ganglion and its three divisions occupy
patients, with good results.10,16 Another important development Meckel’s cave. The subarachnoid space within Meckel’s cave
involved the use of electrophysiology to confirm needle place- extends an average of 4.9 mm medially and 1.7 mm laterally
ment and ensure adequate neurolysis, while avoiding excessive beyond the posterior edge of the gasserian ganglion. This space
damage. To this end, in 1955, Silverstone designed an insulated may extend over the divisions of the trigeminal nerve. The mean
needle with an exposed tip used as a nerve stimulator.10,17 Later, length of Meckel’s cave ranges from 6 to 16 mm.31 The gasserian
motor and sensory evoked potential monitoring was used in tri- ganglion measures 4 to 5 mm wide and 15 to 25 mm long. The
geminal destructive procedures by Karol and colleagues,18 and length of the sensory root varies from 5 to 15 mm; the length of
this was adopted by Sindou and others.19-21 Most recently, neu- the mandibular nerve from the anterosuperior margin of the
ronavigation has been used to assist with percutaneous trigeminal foramen ovale to the gasserian ganglion is 0 to 10 mm.30-32
neurolysis.22-24 The ganglion has a posterior sensory root, which joins the
brainstem about halfway between the lower and upper borders
of the pons. It starts with an oblique run upward from the lateral
TRIGEMINAL ANATOMY part of the pons toward the petrous apex, exits the posterior fossa,
Percutaneous procedures in general aim to reach the trigeminal enters the middle cranial fossa by passing forward beneath the
nerve ganglion or sensory root through the foramen ovale. An tentorial attachment, and enters Meckel’s cave to join the gan-
understanding of the relationship among the foramen, the third glion through a deep hilum on its posterior aspect. The motor
division of the nerve (mandibular), and surrounding structures is branch of the trigeminal nerve runs in front of and medial to the
vital to avoid injury to the neighboring nervous system structures sensory root and passes beneath the ganglion, leaving the skull
and vasculature. through the foramen ovale and, immediately below this foramen,
The foramen ovale is situated in the anterior part of the sphe- joining the mandibular nerve.33
noid bone. It lies lateral to the foramen lacerum, which is occluded The sensory rootlets, between the gasserian ganglion and the
at its base only by cartilage. It is also near the posterior margin root in the cerebellopontine angle, form a plexus at its entry into
of the lateral pterygoid plate. Posterolaterally lies the foramen the ganglion. This retrogasserian triangular area has been named
spinosum, which transmits the middle meningeal artery and men- the triangular plexus and has been identified as the best place to
ingeal branch of the mandibular nerve. Rarely, the foramen spi- create a lesion for the treatment of trigeminal neuralgia.34
nosum and ovale may be confluent. Posteroinferiorly lies the The trigeminal ganglion is formed by union of the three divi-
jugular foramen, and posterolaterally, the carotid canal.25 The sions of the trigeminal nerve: the mandibular nerve, maxillary
1405
1406 SECTION 6  Pain

nerve, and ophthalmic nerve, also known as V3, V2, and V1, imaging (MRI) and magnetic resonance angiography processed
respectively. The ophthalmic nerve is the smallest of the three as three-dimensional images have been used to verify vascular
trigeminal divisions. It inclines upward as it passes forward near compression.52-54 MRI may not be necessary in cases managed
the medial surface of the dura, forming the lower part of the purely by a percutaneous procedure.
lateral wall of the cavernous sinus and reaching the superior
orbital fissure. The maxillary nerve takes a more direct course
and enters the foramen rotundum. The mandibular nerve occu-
SURGICAL TREATMENT
pies most of the gasserian ganglion and takes a caudolateral There are no specific surgery guidelines for patient selection or
course from the ganglion and enters the foramen ovale.33,35 guidelines for which surgical method to select. A report by
Gronseth and colleagues details the diagnostic evaluation and
treatment of trigeminal neuralgia as an evidence-based review
DIAGNOSIS and makes some recommendations.55
The first known references to what is believed to be trigeminal Surgery is generally offered to patients who fail medical treat-
neuralgia were by Aretaeus of Cappadocia36 and later by Avi- ment, have only partial relief of pain after 1 year, or, according
cenna, who described trigeminal neuralgia in his text Canon to Garvan and Siegfried,56 still require medication after consump-
Medicinx.37,38 The International Association for the Study of Pain tion of more than 3000 tablets of a single drug. Microvascular
defined the essential features of idiopathic trigeminal neuralgia decompression (MVD) remains the surgical option of choice for
as “sudden, transient, intense bouts of superficially located pain, trigeminal neuralgia, with percutaneous procedures generally
strictly confined to the distribution of one or more divisions of reserved for patients who experience recurrent pain after MVD
the trigeminal nerve usually precipitated by light mechanical or are a high surgical risk owing to medical comorbidity. The
activation of a trigger point or area.”39 Penman, in a detailed less invasive percutaneous procedures are more often employed
description of trigeminal neuralgia, stated that the only constant in patients older than 65 to 70 years of age, although outcomes
feature is the symptom of pain that is unilateral, trigeminal, from MVD in this population are similar to those in younger
severe, paroxysmal, and precipitated.40 patients.57,58 In the case of suspected symptomatic trigeminal neu-
A diagnosis of trigeminal neuralgia is dependent on the ralgia caused by multiple sclerosis (MS), further evaluation may
patient’s history and analysis of the patient’s complaint. Pain is be needed for surgical planning and prognostication. Percutane-
usually described as spreading outward from the trigger point to ous procedures are usually offered to patients with MS.59-61
cover an area that roughly approximates the territory of distribu- Although neurovascular compression may be seen in patients
tion of one (or more) of the trigeminal divisions.41 Usually, there with MS, surgical outcomes tend to be less satisfactory than in
is no clinically evident sensory deficit, yet quantitative and quali- cases of idiopathic trigeminal neuralgia.62,63
tative tests have demonstrated deficits for tactile and warm sensa-
tions in the affected area, with no deficit for heat pain or for
pinprick.41-44 There is also an absolute, followed by a relative,
PERCUTANEOUS PREOPERATIVE CARE
refractory period after an attack, during which further paroxysms All percutaneous procedures for the management of trigeminal
cannot be elicited by trigger-point stimulation,45 a phenomenon neuralgia involve producing some injury to trigeminal afferents.
confirmed by microelectrode recordings obtained during percu- This is achieved by using heat in the case of RF rhizotomy,
taneous RF ablation of the ganglion for trigeminal neuralgia.46 chemical neurolysis in the case of glycerol rhizotomy, mechanical
A differential diagnosis of trigeminal neuralgia includes a large neurolysis in the case of balloon compression rhizotomy, and
array of facial pains. Trigeminal neuralgia itself may be due to radiation-induced neural injury in the case of stereotactic radio-
multiple causes and pathologic processes, which influence the surgery (SRS). That so many percutaneous procedures are avail-
mode of treatment and outcome. To assist in diagnosis and treat- able points to the fact that none has proved ideal. That is, no
ment of trigeminal neuralgia, a facial pain classification system one percutaneous procedure is applicable in all cases with a uni-
has been proposed by Burchiel (Table 172-1).47-49 formly high rate of long-term success and with minimal possibil-
ity of complication or recurrence. RF rhizotomy is perhaps the
most commonly used procedure, and long-term case results are
Diagnostic Tools available.61,64,65
Trigeminal neuralgia is a purely clinical diagnosis; there is no
confirmatory laboratory or imaging study for this disease.
However, there are electrophysiologic and imaging studies that
Patient Selection
may be useful as an adjunct to clinical acumen. Trigeminal evoked Percutaneous procedures are usually offered to patients with tri-
potentials and electrophysiologic studies are not widely used geminal neuralgia who have failed medical management or have
but are complementary diagnostic tools.50,51 Magnetic resonance developed complications or side effects of medical management
and either have already had or are not suitable candidates for
MVD. Although there are no surgical age guidelines or limits,
age older than 65 years can often lead to exclusion of an otherwise
TABLE 172-1  Burchiel Classification of Facial Pain eligible patient from the benefits of MVD. Some surgeons prefer
Diagnosis History
percutaneous procedures for patients older than 65 years of age.66
One could argue that it is prudent to offer MVD to the most
Spontaneous onset eligible patients: those who demonstrate vascular impingement
Trigeminal neuralgia, type 1   >50% episodic pain of the trigeminal root on the affected side and are otherwise
Trigeminal neuralgia, type 2   >50% constant pain
fit to undergo the procedure. However, a recent study indicates
Trigeminal injury
Trigeminal neuropathic pain   Unintentional, incidental trauma
that patients without neurovascular compression who undergo
Trigeminal deafferentation pain   Intentional deafferentation posterior fossa exploration and internal neurolysis have outcomes
Symptomatic trigeminal neuralgia Multiple sclerosis, tumor comparable to those undergoing MVD.67 Patients with MS,
Post-herpetic neuralgia Trigeminal herpes zoster outbreak pontine infarction that affects the trigeminal root entry zone, or
Atypical facial pain* Somatoform pain disorder brainstem white matter lesions without MS and those who have
*Cannot be diagnosed by history alone.
previously failed MVD should initially be considered for a per-
cutaneous procedure.64,68,69
CHAPTER 172  Percutaneous Procedures for Trigeminal Neuralgia 1407

Preoperative Patient Preparation 172


We recommend preoperative MRI for all trigeminal neuralgia
patients, with the exception of those with a confirmed MS diag-
nosis. Patients with trigeminal schwannoma and epidermoid
tumor have been described as presenting with trigeminal pain, 3 cm Midpupil
but this is not usually considered typical trigeminal neuralgia.
MRI serves to begin to address primary pathology and to poten- 2.5 cm
tially exclude such patients from percutaneous procedures.70-73 1 cm
Also, when a vascular compression is in doubt, high-resolution,
three-dimensional, time-of-flight magnetic resonance angiogra- Trajectory
phy may be performed to assess and demonstrate vascular
impingement (if present) with greater than 90% accuracy.74,75
Patients are advised that, although most percutaneous proce-
dures can be performed as day surgery, they may be required to
stay overnight in the hospital if circumstances dictate. Anticoagu-
lant and antiplatelet medications should be stopped before
surgery, and patients are advised to continue taking any antihy-
pertensive medications throughout the perioperative period.
Patients are advised to take nothing by mouth on the day of the
procedure.

PERCUTANEOUS SURGICAL PROCEDURES


Radiofrequency Rhizotomy
RF rhizotomy was popularized by Sweet and Wepsic, as reported
in 1974.76 The concept behind the technique is to use RF stimula-
tion (alternating electrical field with an oscillating frequency of
500,000 Hz) to cause a thermal lesion in the retrogasserian root, Figure 172-1. The radiofrequency needle is inserted into the cheek at
or ganglion.77 In an animal study, RF heating was reported to a point 2.5 cm from the corner of the mouth, 1 cm below the occlusal
cause selective injury to small myelinated and unmyelinated plane. The trajectory is the intersection of two planes: one directed to
fibers78; however, a subsequent neuropathologic clinical model the midpupil and the other to a point 3 cm anterior to the tragus.
study failed to substantiate this.79 It is now generally believed that During needle insertion, the surgeon’s index finger is inserted into the
RF heating causes a nonselective destruction of axons, regardless patient’s mouth lateral to the teeth to prevent intraoral penetration and
of fiber size.80 to ensure that the needle is not directed lateral to the maxilla.
In the operating suite, the patient is placed supine on the
operating table. Intravenous access is secured. The patient is then
placed with the head extended to about 30 degrees to obtain a occlusal plane and advances through the cheek in the submucosal
clear submental-vertex view of the foramina ovale and spinosum, layer, between the pterygoid bone and the angle of the mandible,
located on the greater wing of the sphenoid bone, of the affected guided by the surgeon’s index finger of the other hand within the
side. Biplanar fluoroscopy can be used if available. Usually a patient’s mouth (Fig. 172-1). The needle is directed along a line
portable C-arm digital fluoroscope is sufficient. Magnification representing the intersection of a vertical plane passing through
(2×) of the fluoroscopic image may be required to visualize the the medial aspect of the ipsilateral pupil and a horizontal plane
foramen clearly. passing through a point 3 cm anterior to the external auditory
Anesthesia is induced using a short-acting agent, such as pro- meatus along the inferior border of the zygoma. The needle
pofol. The goal is to reach plane 1 of stage III anesthesia, with engages the foramen ovale about 6 to 8 cm from the skin surface.
loss of eyelash reflex but without abolition of laryngeal reflexes. This can usually be evidenced by a contraction of the masseter
Bolus dosing (usually 40 mg of propofol) is most effective. Ben- muscle manifested as jaw jerk (see Fig. 172-1).
zodiazepines should be avoided because they prolong awakening Lateral fluoroscopy is used thereafter, and the needle is
time. Anticholinergics (usually 0.2 mg of glycopyrrolate) may be advanced to a point just superior to the intersection of the petrous
given to minimize oral secretions and blunt the vasovagal response part of the temporal bone and the clivus on a true lateral view.
during penetration of the needle through the foramen ovale. The needle trajectory should be at 45 degrees to the planum
Some surgeons prefer to watch for the vagal response as a guide sphenoidale on this view. The patient is now awakened from
while engaging the foramen ovale and may not use preoperative anesthesia, and the stylet of the trochar is withdrawn and replaced
anticholinergics. If anticholinergics are not used, an external with the RF electrode. The RF generator should report the
pacemaker strapped to the chest wall, set to deliver 45 beats/min patient’s temperature accurately. Electrode impedance within the
should bradycardia occur during electrode insertion or during ganglion is usually 150 to 350 ohm. These readings are helpful
stimulation, may be used.80 to confirm that the RF generator is functioning and that the
The skin of the cheek and perioral region on the ipsilateral electrode is within the ganglion rather than in cerebrospinal fluid
side is painted with antimicrobial solution after the required (CSF). A curved-tip (Tew) electrode may be necessary to target
depth of anesthesia has been obtained (eyelash reflex is lost). A V1 and V2, but typically a straight electrode suffices to produce
nasal cannula or trumpet may assist in the maintenance of the a lesion in V3 (Fig. 172-2).
airway. The surgeon stands on the same side as the patient’s pain, Once the patient is adequately awake, test stimulation of the
and cannulation of the foramen proceeds under fluoroscopic target is achieved using a square wave stimulus at 50 Hz, using a
guidance using the Härtel technique with the RF electrode 1-millisecond pulse duration, to evoke paresthesias in the stimu-
trochar.3 The needle enters the cheek through a point 2.5 cm lated division. This is usually obtained at an amplitude of 0.05 to
lateral to the corner of the mouth and 1 cm inferior to the 0.15 volts, although a somewhat higher threshold of stimulation
1408 SECTION 6  Pain

Figure 172-2. On lateral fluoroscopy, the needle is seen entering the foramen ovale, directed to a region
that is just anterior to the intersection of the petrous bone and clivus. For V3, the needle is directed at  
the 10-o’clock position, and for V2 and/or V1, the needle is directed more superiorly toward the 11- or
12-o’clock position. The latter approach will require a somewhat lower and more lateral entry point on the
cheek.

may be required in patients who have had a previous lesioning not require intraoperative patient response to guide the surgeon
procedure. When the electrode location has been confirmed, the during the lesioning), the patient can be sedated throughout
patient is again anesthetized. Lesion creation is achieved by the procedure, although some surgeons prefer that the patient
heating the electrode up to 75° to 80° C for 90 seconds. For be awakened after the needle has been placed through the
lesions in the V1 segment, a duration of 60 seconds may be used. foramen ovale. Patients often receive a short-acting barbiturate,
The initial lesion may be bracketed by making a lesion on either such as methohexital, or another short-acting agent, such as
side by moving the electrode 2 mm proximally and distally from propofol.
the initial position. After neurolysis is complete, the patient is The patient is positioned supine with the neck extended,
again awakened from anesthesia to test the procedure efficacy. similar to the RF procedure, and a 20-gauge, 3.5-inch spinal
The end point of this procedure is slight hypoesthesia, such that needle is inserted into the foramen ovale using the Härtel tech-
the patient is able to feel touch in the affected area but cannot nique, as described earlier. To ensure access to the trigeminal
differentiate between the sharp and blunt ends of a safety pin. cistern, the foramen ovale should be entered through the medial
When the desired end point has been achieved, the needle is third. The position of the needle is confirmed by fluoroscopy and
withdrawn with the electrode, and the entry point is checked for is manipulated, usually in 1-mm increments under guidance, to
bleeding. Rarely, pressure application may be required in the obtain a free flow of CSF on removing the stylet. When a free
presence of bleeding to preempt the formation of a hematoma. flow of CSF is obtained on removing the stylet, the stylet is
The patient is then transferred to the recovery room and is kept reinserted into the needle, and the patient is carefully moved into
under observation for a few hours. Typically, the patient can a sitting position with the neck flexed forward such that the
return home the same day. orbital-meatal line is past horizontal. A cisternogram is then per-
In 98% to 99% of the patients, pain relief is obtained imme- formed using a water-soluble contrast medium such as iohexol
diately after surgery.81,82 A recurrence rate of 20% at 9 years has (Omnipaque; GE Healthcare AS, Oslo, Norway). Using a tuber-
been described, with an incidence of 9% for mild dysesthesia, 2% culin syringe, the contrast medium is injected in 0.05-mL boluses
for major dysesthesia, and 0.2% for anesthesia dolorosa.81 Because and, under continuous lateral fluoroscopy, the filling of the tri-
hypoesthesia is the end point of the procedure, numbness is geminal cistern is noted. A typical pear-shaped appearance of the
present in 100% of patients after surgery.81 Other studies show a cistern is visualized. The volume of the cistern is recorded as
higher pain recurrence rate associated with lower rate of dyses- the amount of contrast required to fill up the cistern, and then
thesia and anesthesia dolorosa.83 It is generally accepted that a the contrast is allowed to drain out either through the needle by
denser lesion results in longer pain relief at the cost of a higher removing the syringe or into the posterior fossa by tilting the
sensory complication rate. The irreversible loss of long-latency head backward. The average volume of the cistern is 0.25 to 0.30
trigeminal root evoked potentials has been documented as a mL, and it uncommonly exceeds 0.5 mL (range, 0.1 to 1 mL).82,87
means of objective assessment of the effects of the rhizotomy,84 Some surgeons have reported successful procedures without
and a newer multiarray electrode method for mapping the tri- using the cisternogram for localization, instead relying on fluo-
geminal nerve may help in producing more selective lesions.85 roscopy and CSF backflow,88 although this introduces an element
of uncertainty as to the destination of the injected glycerol.
Glycerol is then slowly injected into the trigeminal cistern in
Glycerol Rhizotomy a similar manner, under continuous fluoroscopy, with the patient
The procedure for injecting anhydrous glycerol (99.5%), a mild sitting upright and the head flexed forward. Although many sur-
neurolytic, can be performed both in an operating room with geons simply fill the cistern with glycerol,89 others attempt to
fluoroscopic facilities and in a radiology suite setting with anes- produce a more selective lesion.80 Calculating the volume of
thetic support.86 Because the procedure is largely anatomic (does glycerol required for a more “selective” neurolytic effect relies on
CHAPTER 172  Percutaneous Procedures for Trigeminal Neuralgia 1409

the “floating glycerol” technique, which depends on the assump- technique, as discussed in the “Radiofrequency Rhizotomy”
tion that glycerol has a lower specific gravity than the contrast section earlier, a 14-gauge needle with a blunt obturator is intro- 172
medium and therefore floats on its surface. The ratio of glycerol duced into the foramen ovale of the affected side.3 When the
to contrast medium can be varied for directing treatment against cannula has engaged the foramen ovale, the blunt obturator is
specific targeted fibers; ratios of 30:70 for V1, 50:50 for V2, and removed, and a straight guiding stylet is inserted. Under fluoro-
70:30 for V3 are used to achieve the purpose. The emphasis is scopic guidance, the stylet is then directed toward the porus
that it is more important to drain the cistern completely of con- trigeminus (proximal entrance to Meckel’s cave), radiographically
trast medium before injecting glycerol in the case of V3 pain than denoted by the medial dip in the petrous bone on an anteropos-
in the case of V1 pain. terior image (with the petrous ridge positioned in the radio-
Another technique for injecting glycerol is that of varying the graphic center of the orbital cavity). This brings the tip of the
amounts of glycerol used to fill the trigeminal cistern, subsequent stylet between 17 and 22 mm beyond the foramen ovale, within
to drainage of all the contrast medium after cisternography. One 5 mm beyond the clival line. In this location, the tip of the stylet
half of the cistern is filled in cases of V3 pain, two thirds in cases abuts the maxillary fibers. Manipulations (1 mm) of the stylet
of V2 pain, and the whole cistern in cases of V1 pain or for pain medially or laterally bring the tip closer to V1 or V3 fibers,
in multiple trigeminal divisions. respectively.
After withdrawal of the spinal needle, the glycerol should be A No. 4 Fogarty balloon catheter is test-inflated with contrast,
kept in contact with the nerve for at least 1 hour (up to 2 hours and all air should be excluded from the balloon. The stylet is
has been recommended), by keeping the patient in a sitting posi- withdrawn, and the balloon with inner stylet is introduced. This
tion with the head flexed.80,86,87,90 The patient may then be dis- is the standard apparatus used for this procedure, although the
charged home. use of variably sized balloon catheters has also been reported.97
Most patients experience relief immediately or within 1 to 2 A pressure transducer attached to an insufflation syringe or a
days after surgery, although it can take up to 2 weeks to achieve tuberculin syringe is screwed to the proximal end of the catheter
pain relief in some cases.89 Liu and Apfelbaum have recom- using a three-way stopcock. The balloon is then inflated using
mended a repeat procedure if pain relief does not occur within 7 about 1 mL of contrast agent. If accurately positioned, the balloon
days of surgery.86 assumes a pear shape on lateral fluoroscopy. The thin neck of the
Sensory loss after glycerol retrogasserian rhizotomy is variable pear is the portion of the balloon within the porus trigeminus.
and may present as a mild numbness, usually in the perioral Adequate compression can be verified by measuring the intralu-
region; however, profound sensory loss may occur. Corneal hypo- minal pressure changes and is limited by lifting the dura off
esthesia may occur in a few patients when the V1 division or all the trigeminal ganglion. An intraluminal pressure of 1200 to
the fibers are targeted, and these patients need to be advised to 1500 mm Hg (1.3 to 1.5 atm) is considered adequate, signifying
check their eyes daily for signs of irritation. a resultant tissue compression pressure of 650 to 950 mm Hg.80,98
The balloon is kept inflated for 1 to 1.5 minutes if it is a first
surgery and for 1.5 to 2 minutes in cases of recurrent pain to
Balloon Compression minimize the risk for dysesthesia. Compression times of up to 10
Percutaneous balloon microcompression of the trigeminal gan- minutes have been reported.93,99 Limiting the compression time
glion using a balloon catheter was introduced by Mullan and to less than 2 minutes has resulted in reduction of the incidence
Lichtor.13 This technique was purportedly derived from the open of masseter muscle weakness and severe numbness.98 After com-
technique of mechanical injury to the trigeminal ganglion per- pression is completed, the balloon is deflated, and the balloon and
formed through temporal craniotomy by Shelden and Pudenz in catheter are removed, with pressure applied to the cheek for
1955.91,92 Skirving and Dan have reported performing a similar about 5 minutes to prevent hematoma formation. The patient can
technique since 1980,93 following an introduction to the method be discharged home after about 4 hours of observation. Most
by Mullan and before the detailed method published by Mullan patients obtain pain relief immediately after surgery. The remain-
and Lichtor in 1983.13 Based on a rabbit experimental study, ing patients experience pain relief by postoperative day 2.
Brown and colleagues have postulated that balloon compression
has the advantage of selectively avoiding injury to the small,
unmyelinated fibers that mediate corneal reflex, providing rela-
Postoperative Management
tive protection of the corneal sensation.60 Microcompression of After all percutaneous procedures, the entry point on the cheek
the trigeminal ganglion takes place during the procedure; it has is cleaned and dressed, if necessary. Corneal sensation should be
also been documented in anatomic study on cadavers that, when tested, and the patient should be counseled as to how to recognize
the balloon is fully inflated, there is stretching of the dura, reliev- signs of corneal irritation. If substantial loss of corneal sensation
ing what is called the dural compression of the trigeminal ganglion occurs or corneal irritation occurs, artificial tears may be advised
and its root.94 This lends some support to the hypothesis of for use every 2 to 4 hours, and an ophthalmologic evaluation may
Taarnhøj, who postulated that trigeminal neuralgia may, in part, be warranted. Dietary restrictions are not necessary; however,
be caused by the compression and angulation of the nerve root many patients prefer to restrict their intake to soft foods for a
where it crosses the apex of the petrous ridge.95,96 few days. Jaw opening exercises may be necessary, especially in
Balloon microcompression does not require the patient to be patients undergoing the balloon compression procedure. The
awake during surgery and can thus be performed under general inner cheek on the affected side may feel numb, and the patient
anesthesia, thereby limiting the anxiety and discomfort of the should be advised to avoid biting the tongue and cheek on that
patient. The procedure is performed in a surgical suite with fluo- side. Some patients may develop a maceration of the buccal
roscopic facilities. General anesthesia is administered, and either mucosa, requiring irrigation of the mouth with warm saline solu-
premedication with an anticholinergic, such as glycopyrrolate, is tion every 4 hours. Some patients may develop herpes simplex
administered, or an external pacemaker is placed to mitigate type 1 lesions a few days after surgery, which can be treated with
bradycardia that may occur during insertion of the needle through a topical application of acyclovir.80 In cases of headache after
the foramen ovale or during balloon compression of the trigemi- glycerol rhizotomy, it is necessary to differentiate between chem-
nal ganglion. A combination of both precautions against bradyar- ical meningitis, which will resolve over time, and infective
rythmia is advisable. meningitis. Anticonvulsant medications need to be continued
The patient is positioned supine with the head extended immediately after surgery but can be tapered off and discontinued
about 15 degrees. Using fluoroscopic guidance and the Härtel depending on the degree of pain relief obtained.
1410 SECTION 6  Pain

17% (average, 6%), and transient motor weakness in about


RADIOSURGERY 19%.82 After glycerol rhizotomy, the most common complica-
SRS was first used by Leksell for the treatment of trigeminal tions are corneal hypoesthesia in about 5% (range, 0% to 10%)
neuralgia in 1971.100 Since then, several groups have demon- of cases and significant dysesthesia in about 4% (range, 0% to
strated the efficacy of various radiosurgical techniques in the 13%).80 Although permanent motor root weakness is a major
management of the condition.101-105 The mechanism of action of complication of balloon rhizotomy, occurring in 3% of cases
radiosurgery is presumed to be axonal degeneration as result of (range, 0% to 12%), significant dysesthesia is also common,
radiation. This was postulated by Kondziolka and colleagues occurring in about 5% (range, 0% to 10.6%).80 The most signifi-
from their work on a primate model106 and was later confirmed cant complication of SRS is numbness, which has been reported
by histologic analysis of the cisternal segment of the trigeminal to occur in up to 21% of cases and is considered as bothersome
nerve in a patient with recurrent trigeminal neuralgia who under- or painful in 5% to 10% of patients.113,120 Significant dysesthesia
went partial sectioning of the nerve.107 Arteriolar thickening that has been reported in about 3.2% of patients after radiosurgery
occurs after radiation insult to the vessel in contact with the nerve for trigeminal neuralgia.121
has been postulated to have a possible therapeutic effect.108 Other potential, but rare, complications resulting from percu-
The procedure is indicated in selected patients of advanced taneous needle procedures through the foramen ovale include the
age or poor medical condition, patients who are on anticoagu- risks for stroke, hemorrhage, pseudomeningocele, permanent
lants, and those who are reluctant to pursue other forms of hearing loss or facial weakness, diplopia resulting from trochlear
therapy. nerve or abducens nerve palsy, and CSF rhinorrhea.80,122-125
The procedure involves placing a stereotactic head frame on
a patient who is under local anesthesia or with the help of a brief
general anesthetic. Thin-section (1- to 1.4-mm) axial MRI is
SPECIAL CONSIDERATIONS
obtained. Constructive interference in steady state images are of
great value in identifying the nerve as it passes through the tri-
Multiple Sclerosis
geminal cistern. Various radiosurgical targets, including the gas- About 2% of patients with MS suffer from trigeminal neural-
serian ganglion, the root entry zone at the pons, and the gia.126 Although the possibility of concomitant vascular impinge-
retrogasserian portion of the nerve (4 to 6 mm long, 2 to 3 mm ment on the trigeminal nerve in cases of MS with trigeminal
anterior to the root entry zone), have been used. Most centers neuralgia has been well documented63 and anecdotal reports of
now employ the posterior target (root entry zone).86,109-112 The treatment with decompression published,127 the procedure of
patient is positioned in the radiosurgery device (linear particle MVD is not routinely recommended for such cases even by the
accelerator or Leksell Gamma Knife [Elekta AB, Stockholm, staunchest of advocates,128,129 primarily because of a high failure
Sweden]), and the target is irradiated using a small collimator rate. In this instance, failures may be due to MS patients having
(4 mm) at a maximal dose of between 60 and 90 Gy for initial extension of demyelination into the brainstem, central to the area
treatment and of about 50 Gy in cases undergoing repeat treat- generally treated by MVD.130,131 MS patients generally present at
ment. A 50% isodose line is used at the nerve boundary. a younger age and are somewhat more likely to have bilateral
On completion of the radiosurgical procedure, the stereotactic facial pain.59,130 Percutaneous techniques, including glycerol rhi-
frame is removed, and the patient can be prepared for discharge zotomy, RF lesioning, and radiosurgery, have been advocated for
home. Patients should be advised to continue taking their pain management.64,132,133 Treatment failure occurs in MS-related tri-
relief medications because radiosurgery may take up to 4 to 6 geminal neuralgia independently of the type of treatment modal-
weeks to relieve pain. Depending on the degree of pain relief ity, but recent work indicates that balloon compression may have
achieved, patients may slowly taper off pain relief medications. the highest initial pain-free response rates and longest pain-free
intervals when compared to glycerol rhizotomy, SRS, RF lesion-
ing, or MVD.134
RESULTS AND COMPLICATIONS
Of the available percutaneous procedures for trigeminal neural-
gia, RF rhizotomy has been reported to provide the highest rate
Recurrent Treatment
of pain relief. In some series, results are comparable to those of In patients in whom pain has returned after MVD, a second
MVD.80 Long-term pain relief (average, 6 years; range, 1 to 9 MVD procedure is unlikely to be of any benefit unless the offend-
years) has been reported in 75% of patients (range, 63% to 89%) ing vessels from the first MVD were reported to be veins.135,136
after RF rhizotomy.80 Pain relief after glycerol rhizotomy (average, In recurrent cases, glycerol rhizotomy or RF rhizotomy has been
3 years; range, 0.5 to 5.5 years) has been reported in 55% of advocated as the procedure of choice.137 Because the trigeminal
patients (range, 22% to 70%),80 whereas in the case of balloon cistern may become scarred after a prior glycerol rhizotomy,
compression, pain relief was reported in 73% of patients (range, a repeat glycerol injection has a higher chance of technical
62% to 83%) at 4 years’ follow-up (range, 0.5 to 10.7 years).80 failure.138 It may be prudent in such cases to undertake an RF
Pain relief obtained after SRS has been reported to be as high as procedure. There is no evidence that RF rhizotomy and balloon
83.1% after the first year,113 with a decrease to 55.8% (complete compression are associated with greater technical failure rates
or partial pain relief) at 5 years.114 when repeated.80
Although graded lesions involving the different trigeminal
nerve distributions are best obtained using RF rhizotomy, in the
case of trigeminal neuralgia affecting the V1 segment, corneal
CONCLUSION
hypoesthesia is a significant complication in up to 16% of cases.115 Percutaneous procedures for trigeminal neuralgia involve pene-
The initial theory that glycerol could relieve neuralgia without tration of the foramen ovale with a cannula and then creation of
causing sensory loss has largely been disproved. Sensory loss and a destructive lesion either on the trigeminal ganglion or root.
dysesthesia continue to be significant complications of the This can be accomplished by thermal injury through RF, chemi-
procedure.116-118 Damage to the motor rootlets affects the results cal injury by injection of glycerol, or mechanical compression by
of balloon compression and has been shown to occur in up to balloon inflation in Meckel’s cave.
12% of cases.119 Currently, there are no evidence-based guidelines for when to
The most common complications of RF rhizotomy are severe apply these procedures or which one to choose. The general
dysesthesia in 6% to 9% of cases, corneal hypoesthesia in 1% to consensus has been to reserve percutaneous procedures for older
CHAPTER 172  Percutaneous Procedures for Trigeminal Neuralgia 1411

patients, for those not able to tolerate MVD, for patients with that employ standardized outcomes measures to answer these
trigeminal neuralgia caused by MS, and for recurrent disease. questions.140,142-144 172
The overall trend in the United States has been a dramatic
decline in the use of percutaneous procedures for the treatment
SUGGESTED READINGS
of trigeminal neuralgia.139 Bowsher D. Trigeminal neuralgia: an anatomically oriented review. Clin
As a group, about 90% of patients with trigeminal neuralgia Anat. 1997;10:409-415.
attain immediate pain relief from a percutaneous procedure. Burchiel K, ed. Surgical Management of Pain. 2nd ed. New York: Thieme;
Most postoperative pain recurrences occur within the first 2 2002.
years, after which the recurrence rate decreases, with a more Burchiel KJ. A new classification for facial pain. Neurosurgery. 2003;53:
than 50% recurrence after 5 years. Sensory loss after these per- 1164-1166.
cutaneous procedures occurs in almost half of patients and varies Cruccu G, Gronseth G, Alksne J, et al. AAN-EFNS guidelines on tri-
from slight hypoesthesia to troublesome dysesthesia. Other com- geminal neuralgia management. Eur J Neurol. 2008;15:1013-1028.
plications include anesthesia dolorosa, corneal numbness, and Hakanson S. Trigeminal neuralgia treated by the injection of glycerol into
the trigeminal cistern. Neurosurgery. 1981;9:638-646.
motor weakness. A low pain recurrence rate is clearly associated Kanpolat Y, Berk C, Savas A, et al. Percutaneous controlled radiofre-
with more sensory loss, but complications of deafferentation, quency rhizotomy in the management of patients with trigeminal neu-
such as anesthesia dolorosa and its more minor variants, also ralgia due to multiple sclerosis. Acta Neurochir (Wien). 2000;142:685-689,
increase in parallel with numbness. The surgeon must balance discussion 689-690.
adequate and sustained pain relief with unwanted sensory com- Leksell L. Sterotaxic radiosurgery in trigeminal neuralgia. Acta Chir
plications. These procedures provide a good alternative for man- Scand. 1971;137:311-314.
agement of trigeminal neuralgia as an initial modality, in recurrent Lopez BC, Hamlyn PJ, Zakrzewska JM. Systematic review of ablative
cases, and in patients with symptomatic trigeminal neuralgia sec- neurosurgical techniques for the treatment of trigeminal neuralgia.
ondary to MS. Neurosurgery. 2004;54:973-982.
Mullan S, Lichtor T. Percutaneous microcompression of the trigeminal
At present, there is no ideal surgical procedure for trigeminal ganglion for trigeminal neuralgia. J Neurosurg. 1983;59:1007-1012.
neuralgia—one that is minimally invasive, uniformly effective, Resnick DK, Jannetta PJ, Lunsford LD, et al. Microvascular decompres-
lacking complications, and without failures or recurrences. MVD sion for trigeminal neuralgia in patients with multiple sclerosis. Surg
still remains the standard by which all other contemporary pro- Neurol. 1996;46:358-361, discussion 361-362.
cedures are measured. MVD provides the longest pain-free inter- Sweet WH, Wepsic JG. Controlled thermocoagulation of trigeminal gan-
val, yet it is not free of morbidity and mortality. SRS provides a glion and rootlets for differential destruction of pain fibers. 1. Trigemi-
reasonable noninvasive option, but it has delayed onset of relief nal neuralgia. J Neurosurg. 1974;40:143-156.
and a recurrence interval comparable to other percutaneous pro- Taha J. Trigeminal neuralgia: percutaneous procedures. Semin Neurosurg.
cedures. Finding an answer to what procedure to use, and when 2004;15:115-134.
Tatli M, Satici O, Kanpolat Y, et al. Various surgical modalities for tri-
to use it, is difficult because there is no class I or II evidence, in geminal neuralgia: literature study of respective long-term outcomes.
part owing to a lack of uniformity in terms of both the outcome Acta Neurochir (Wien). 2008;150:243-255.
measures and the reporting methods used by most reported case
series.140,141 Clearly, randomized controlled studies are needed See a full reference list on ExpertConsult.com
CHAPTER 172  Percutaneous Procedures for Trigeminal Neuralgia 1411.e1

REFERENCES 26. Ray B, Gupta N, Ghose S. Anatomic variations of foramen ovale.


1. Harris W. A history of the treatment of trigeminal neuralgia. Post- Kathmandu Univ Med J. 2005;3:64-68. 172
grad Med J. 1951;27:18-21. 27. Reymond J, Charuta A, Wysocki J. The morphology and morphom-
2. Harris W. An analysis of 1,433 cases of paroxysmal trigeminal neu- etry of the foramina of the greater wing of the human sphenoid
ralgia (trigeminal-tic) and the end-results of gasserian alcohol injec- bone. Folia Morphol (Warsz). 2005;64:188-193.
tion. Brain. 1940;63:209-224. 28. Kehrli P, Maillot C, Wolff MJ. Anatomy and embryology of the
3. Härtel F. Über die intracranielle Injektionsbehandlung der Tri- trigeminal nerve and its branches in the parasellar area. Neurol Res.
geminusneuralgie. Med Klin. 1914;10:582-584 [in German]. 1997;19:57-65.
4. Vaughan G. Injection of the gasserian ganglion for neuralgia of the 29. Kaplan M, Erol FS, Ozveren MF, et al. Review of complications due
fifth cranial nerve. Ann Surg. 1917;66:287-289. to foramen ovale puncture. J Clin Neurosci. 2007;14:563-568.
5. Irger J. Penetrating to the gasserian ganglion. Ann Surg. 1930;92: 30. Henderson W. The anatomy of the gasserian ganglion and the
984-992. distribution of pain in relation to injections and operations for
6. Wilkins R. Trigeminal neuralgia: historical overview, with emphasis trigeminal neuralgia. Ann R Coll Surg Engl. 1965;37:346-373.
on surgical treatment. In: Burchiel K, ed. Surgical Management of 31. Janjua RM, Al-Mefty O, Densler DW, et al. Dural relationships of
Pain. New York: Thieme; 2002:228-301. Meckel cave and lateral wall of the cavernous sinus. Neurosurg Focus.
7. Sweet WH, Mark VH, Hamlin H. Radiofrequency lesions in 2008;25(6):E2.
the central nervous system of man and cat: including case reports 32. Soeira G, Abd el-Bary TH, Dujovny M, et al. Microsurgical
of eight bulbar pain-tract interruptions. J Neurosurg. 1960;17: anatomy of the trigeminal nerve. Neurol Res. 1994;16:273-283.
213-225. 33. Rhoton AL Jr. The cavernous sinus, the cavernous venous plexus,
8. Sweet WH, Wepsic JG. Controlled thermocoagulation of trigemi- and the carotid collar. Neurosurgery. 2002;51:S375-S410.
nal ganglion and rootlets for differential destruction of pain fibers. 34. Sindou M, Keravel Y. Trigeminal neuralgia. Percutaneous thermo-
1. Trigeminal neuralgia. J Neurosurg. 1974;40:143-156. coagulation of the trigeminal nerve (author’s transl). Neurochirurgie.
9. Häkanson S. Trigeminal neuralgia treated by the injection of glyc- 1979;25:166-172 [in French].
erol into the trigeminal cistern. Neurosurgery. 1981;9:638-646. 35. Rhoton AL Jr. The anterior and middle cranial base. Neurosurgery.
10. Cole CD, Liu JK, Apfelbaum RI. Historical perspectives on the 2002;51:S273-S302.
diagnosis and treatment of trigeminal neuralgia. Neurosurg Focus. 36. Rose F. Trigeminal neuralgia. Arch Neurol. 1999;56:1163-1164.
2005;18(5):1-10. 37. Aciduman A, Arda B, Gunaydin A, et al. Laqve (wry mouth) consid-
11. Taarnhøj P. Decompression of the trigeminal root and the posterior ered in Avicenna’s renowned treatise the Canon of Medicine. Neu-
part of the ganglion as treatment in trigeminal neuralgia; prelimi- rocirugia (Astur). 2008;19:267-271.
nary communication. J Neurosurg. 1952;9:288-290. 38. Ameli N. Avicenna and trigeminal neuralgia. J Neurol Sci. 1965;2:
12. Taarnhøj P. Trigeminal neuralgia and decompression of the trigemi- 105-107.
nal root. Surg Clin North Am. 1956;1145-1157. 39. Mersky H, Bogduk N. Classification of Chronic Pain: Descriptions of
13. Mullan S, Lichtor T. Percutaneous microcompression of the tri- Chronic Pain Syndromes and Definitions of Pain Terms. 2nd ed. Seattle:
geminal ganglion for trigeminal neuralgia. J Neurosurg. 1983;59: IASP Press; 1994.
1007-1012. 40. Penman J. The differential diagnosis and treatment of tic doulou-
14. Mullan S, Duda EE, Patronas NJ. Some examples of balloon tech- reux. Postgrad Med J. 1950;26:627-636.
nology in neurosurgery. J Neurosurg. 1980;52:321-329. 41. Bowsher D. Trigeminal neuralgia, an anatomically oriented review.
15. Pollock LJ, Potter HE. Experimental studies of injection of the Clin Anat. 1997;10:409-415.
gasserian ganglion controlled by fluoroscopy. JAMA. 1916;67: 42. Chan AW, MacFarlane IA, Bowsher D, et al. Weighted needle pin-
1357-1361. prick sensory thresholds: a simple test of sensory function in dia-
16. Putnam TJ, Hampton AO. A technic of injection into the gasserian betic peripheral neuropathy. J Neurol Neurosurg Psychiatry. 1992;55:
ganglion under roentgenographic control. Arch Neurol Psychiatry. 56-59.
1936;35:92-98. 43. Fruhstorfer H, Lindblom U, Schmidt WC. Method for quantitative
17. Stookey BP, Ransohoff J. Trigeminal Neuralgia; Its History and Treat- estimation of thermal thresholds in patients. J Neurol Neurosurg
ment. Springfield, IL: Thomas; 1959. Psychiatry. 1976;39:1071-1075.
18. Karol EA, Sanz OP, Rey RD. Sensory and motor trigeminal evoked 44. Nurmikko T. Altered cutaneous sensation in trigeminal neuralgia.
potentials to localize the position of trigeminal electrodes. Acta Arch Neurol. 1991;48:523-527.
Neurochir (Wien). 1991;108:110-115. 45. Kugelberg E, Lindblom U. The mechanism of the pain in trigemi-
19. Sindou M. Neurophysiological navigation in the trigeminal nerve: nal neuralgia. J Neurol Neurosurg Psychiatry. 1959;22:36-43.
use of masticatory responses and facial motor responses evoked 46. Burchiel KJ, Baumann TK. Pathophysiology of trigeminal neural-
by electrical stimulation of the trigeminal rootlets for RF- gia: new evidence from a trigeminal ganglion intraoperative micro-
thermorhizotomy guidance. Stereotact Funct Neurosurg. 1999;73: neurographic recording. J Neurosurg. 2004;101:872-873.
117-121. 47. Burchiel KJ. A new classification for facial pain. Neurosurgery. 2003;
20. Sindou M, Fobe JL, Berthier E, et al. Facial motor responses evoked 53:1164-1167, discussion 1166-1167.
by direct electrical stimulation of the trigeminal root. Localizing 48. Eller JL, Raslan AM, Burchiel KJ. Trigeminal neuralgia: definition
value for radiofrequency thermorhizotomy. Acta Neurochir (Wien). and classification. Neurosurg Focus. 2005;18(5):1-3.
1994;128:57-67. 49. Miller JP, Acar F, Burchiel KJ. Classification of trigeminal neuralgia:
21. Lin B, Lu X, Zhai X, et al. Use of sensory and motor action clinical, therapeutic, and prognostic implications in a series of
potentials to identify the position of trigeminal nerve divisions 144 patients undergoing microvascular decompression. J Neurosurg.
for radiofrequency thermocoagulation. J Neurosurg. 2014;121(6): 2009;111:1231-1234.
1497-1503. 50. Lunsford LD, Bennett MH. Percutaneous retrogasserian glycerol
22. Xu SJ, Zhang WH, Chen T, et al. Neuronavigator-guided percuta- rhizotomy for tic douloureux: Part 1. Technique and results in 112
neous radiofrequency thermocoagulation in the treatment of intrac- patients. Neurosurgery. 1984;14:424-430.
table trigeminal neuralgia. Chin Med J. 2006;119:1528-1535. 51. Szapiro J Jr, Sindou M, Szapiro J. Prognostic factors in microvas-
23. Yang Y, Shao Y, Wang H, et al. Neuronavigation-assisted percutane- cular decompression for trigeminal neuralgia. Neurosurgery. 1985;17:
ous radiofrequency thermocoagulation therapy in trigeminal neu- 920-929.
ralgia. Clin J Pain. 2007;23:159-164. 52. Fukuda H, Ishikawa M, Okumura R. Demonstration of neurovas-
24. Chen K-T, Lin MH-C, Tsai Y-H, et al. Application of MRI and cular compression in trigeminal neuralgia and hemifacial spasm
intraoperative CT fusion images with integrated neuronavigation with magnetic resonance imaging: comparison with surgical find-
in percutaneous radiofrequency trigeminal rhizotomy. Acta Neuro- ings in 60 consecutive cases. Surg Neurol. 2003;59:93-99.
chir (Wien). 2015;157(8):1443-1448. 53. Yoshino N, Akimoto H, Yamada I, et al. Trigeminal neuralgia: evalu-
25. Gray H, Standring S, Ellis H, et al. Gray’s Anatomy: The Anatomical ation of neuralgic manifestation and site of neurovascular compres-
Basis of Clinical Practice. 39th ed. Edinburgh: Elsevier Churchill sion with 3D CISS MR imaging and MR angiography. Radiology.
Livingstone; 2005. 2003;228:539-545.
1411.e2 SECTION 6  Pain

54. Miller JP, Acar F, Hamilton BE, et al. Radiographic evaluation of 77. Sluijter M, Racz G. Technical aspects of radiofrequency. Pain Pract.
trigeminal neurovascular compression in patients with and without 2002;2:195-200.
trigeminal neuralgia. J Neurosurg. 2009;110:627-632. 78. Letcher FS, Goldring S. The effect of radiofrequency current and
55. Gronseth G, Cruccu G, Alksne J, et al. Practice parameter: heat on peripheral nerve action potential in the cat. J Neurosurg.
the diagnostic evaluation and treatment of trigeminal neuralgia 1968;29:42-47.
(an evidence-based review): report of the Quality Standards Sub- 79. Smith HP, McWhorter JM, Challa VR. Radiofrequency neurolysis
committee of the American Academy of Neurology and the Euro- in a clinical model. Neuropathological correlation. J Neurosurg.
pean Federation of Neurological Societies. Neurology. 2008;71: 1981;55:246-253.
1183-1190. 80. Taha J. Trigeminal neuralgia: percutaneous procedures. Semin Neu-
56. Garvan NJ, Siegfried J. Trigeminal neuralgia—earlier referral for rosurg. 2004;15:115-134.
surgery. Postgrad Med J. 1983;59:435-437. 81. Taha JM, Tew JM Jr. Comparison of surgical treatments for trigemi-
57. Ashkan K, Marsh H. Microvascular decompression for trigeminal nal neuralgia: reevaluation of radiofrequency rhizotomy. Neurosur-
neuralgia in the elderly: a review of the safety and efficacy. Neuro- gery. 1996;38:865-871.
surgery. 2004;55:840-848, discussion 848-850. 82. Taha JM, Tew JM. Percutaneous rhizotomy in the treatment of
58. Sekula RF Jr, Frederickson AM, Jannetta PJ, et al. Microvascular intractable facial pain. In: Schmidek H, ed. Schmidek & Sweet Opera-
decompression for elderly patients with trigeminal neuralgia: a pro- tive Neurosurgical Techniques: Indications, Methods, and Results. Vol. 2.
spective study and systematic review with meta-analysis. J Neuro- 4th ed. Philadelphia: W.B. Saunders; 2000:1537-1551.
surg. 2011;114:172-179. 83. Burchiel KJ, Steege TD, Howe JF, et al. Comparison of percutane-
59. Brisman R. Trigeminal neuralgia and multiple sclerosis. Arch Neurol. ous radiofrequency gangliolysis and microvascular decompression
1987;44:379-381. for the surgical management of tic douloureux. Neurosurgery. 1981;
60. Brown JA, McDaniel MD, Weaver MT. Percutaneous trigeminal 9:111-119.
nerve compression for treatment of trigeminal neuralgia: results in 84. Macon JB, Poletti CE. Human trigeminal root evoked potentials
50 patients. Neurosurgery. 1993;32:570-573. during differential retrogasserian thermal and chemical rhizotomy.
61. Kanpolat Y, Savas A, Bekar A, et al. Percutaneous controlled radio- Pain. 1987;31:307-316.
frequency trigeminal rhizotomy for the treatment of idiopathic 85. Karol EA, Karol MN. A multiarray electrode mapping method for
trigeminal neuralgia: 25-year experience with 1,600 patients. Neu- percutaneous thermocoagulation as treatment of trigeminal neural-
rosurgery. 2001;48:524-534. gia. Technical note on a series of 178 consecutive procedures. Surg
62. Ariai MS, Mallory GW, Pollock BE. Outcomes after microvascular Neurol. 2009;71:11-17, discussion 17-18.
decompression for patients with trigeminal neuralgia and suspected 86. Liu JK, Apfelbaum RI. Treatment of trigeminal neuralgia. Neurosurg
multiple sclerosis. World Neurosurg. 2014;81:599-603. Clin N Am. 2004;15:319-334.
63. Broggi G, Ferroli P, Franzini A, et al. Operative findings and out- 87. Kondziolka D, Lunsford LD. Percutaneous retrogasserian glycerol
comes of microvascular decompression for trigeminal neuralgia in rhizotomy for trigeminal neuralgia: technique and expectations.
35 patients affected by multiple sclerosis. Neurosurgery. 2004;55:830- Neurosurg Focus. 2005;18(5):E7.
838, discussion 838-839. 88. Pickett GE, Bisnaire D, Ferguson GG. Percutaneous retrogasserian
64. Berk C, Constantoyannis C, Honey CR. The treatment of trigemi- glycerol rhizotomy in the treatment of tic douloureux associated
nal neuralgia in patients with multiple sclerosis using percutaneous with multiple sclerosis. Neurosurgery. 2005;56:537-545, discussion
radiofrequency rhizotomy. Can J Neurol Sci. 2003;30:220-223. 537-545.
65. Nugent G. Radiofrequency treatment of trigeminal neuralgia using 89. Jho HD, Lunsford LD. Percutaneous retrogasserian glycerol rhi-
a cordotomy-type electrode. A method. Neurosurg Clin N Am. zotomy. Current technique and results. Neurosurg Clin N Am. 1997;
1997;8:41-52. 8:63-74.
66. Broggi G, Franzini A, Giorgi C, et al. Trigeminal neuralgia: new 90. Brown JA. Percutaneous techniques. In: Winn HR, Youmans JR,
surgical strategies. Acta Neurochir Suppl (Wien). 1993;58:171-173. eds. Youmans Neurological Surgery. Vol. 3. 5th ed. Philadelphia: Saun-
67. Ko AL, Ozpinar A, Lee A, et al. Long-term efficacy and safety of ders; 2004:2996-3004.
internal neurolysis for trigeminal neuralgia without neurovascular 91. Park SS, Lee MK, Kim JW, et al. Percutaneous balloon compres-
compression. J Neurosurg. 2015;122:1048-1057. sion of trigeminal ganglion for the treatment of idiopathic trigemi-
68. Golby AJ, Norbash A, Silverberg GD. Trigeminal neuralgia result- nal neuralgia: experience in 50 patients. J Korean Neurosurg Soc.
ing from infarction of the root entry zone of the trigeminal nerve: 2008;43:186-189.
case report. Neurosurgery. 1998;43:620-622, discussion 622-623. 92. Shelden CH, Pudenz RH, Freshwater DB, et al. Compression
69. Arrese I, Lagares A, Alday R, et al. Typical trigeminal neuralgia rather than decompression for trigeminal neuralgia. J Neurosurg.
associated with brainstem white matter lesions on MRI in patients 1955;12:123-126.
without criteria of multiple sclerosis. Acta Neurochir (Wien). 2008; 93. Skirving DJ, Dan NG. A 20-year review of percutaneous balloon
150:1157-1161. compression of the trigeminal ganglion. J Neurosurg. 2001;94:
70. de Benedittis G, Bernasconi V, Ettorre G. Tumours of the fifth 913-917.
cranial nerve. Acta Neurochir (Wien). 1977;38:37-64. 94. Urculo E, Martinez L, Arrazola M, et al. Macroscopic effects of
71. Miller J, Acar F, Hamilton B, et al. Preoperative visualization of percutaneous trigeminal ganglion compression (Mullan’s tech-
neurovascular anatomy in trigeminal neuralgia. J Neurosurg. 2008; nique): an anatomic study. Neurosurgery. 1995;36:776-779.
108:477-482. 95. Taarnhøj P. Decompression of the trigeminal root. J Neurosurg.
72. Guttal KS, Naikmasur VG, Joshi SK, et al. Trigeminal neuralgia 1954;11:299-305.
secondary to epidermoid cyst at the cerebellopontine angle: case 96. Taarnhøj P. Decompression of the posterior trigeminal root in tri-
report and brief overview. Odontology. 2009;97:54-56. geminal neuralgia. A 30-year follow-up review. J Neurosurg. 1982;
73. Klieb HB, Freeman BV. Trigeminal neuralgia caused by intracranial 57:14-17.
epidermoid tumour: report of a case. J Can Dent Assoc. 2008;74: 97. Goerss SJ, Atkinson JL, Kallmes DF. Variable size percutaneous
63-65. balloon compression of the gasserian ganglion for trigeminal neu-
74. Anderson VC, Berryhill PC, Sandquist MA, et al. High-resolution ralgia. Surg Neurol. 2008;70:672.
three-dimensional magnetic resonance angiography and three- 98. Brown JA, Pilitsis JG. Percutaneous balloon compression for the
dimensional spoiled gradient-recalled imaging in the evaluation of treatment of trigeminal neuralgia: results in 56 patients based on
neurovascular compression in patients with trigeminal neuralgia: a balloon compression pressure monitoring. Neurosurg Focus. 2005;
double-blind pilot study. Neurosurgery. 2006;58:666-673, discussion 18(5):E10.
666-673. 99. Liu HB, Ma Y, Zou JJ, et al. Percutaneous microballoon compres-
75. Lee A, McCartney S, Burbidge C, et al. Trigeminal neuralgia occurs sion for trigeminal neuralgia. Chin Med J. 2007;120:228-230.
and recurs in the absence of neurovascular compression. J Neuro- 100. Leksell L. Sterotaxic radiosurgery in trigeminal neuralgia. Acta Chir
surg. 2014;120:1048-1054. Scand. 1971;137:311-314.
76. Sweet WH, Wepsic JG. Controlled thermocoagulation of trigemi- 101. Chen JC, Girvigian M, Greathouse H, et al. Treatment of trigemi-
nal ganglion and rootlets for differential destruction of pain fibers. nal neuralgia with linear accelerator radiosurgery: initial results.
J Neurosurg. 1974;40:143-156. J Neurosurg. 2004;101(suppl 3):346-350.
CHAPTER 172  Percutaneous Procedures for Trigeminal Neuralgia 1411.e3

102. Guo S, Chao ST, Reuther AM, et al. Review of the treatment of Report of three cases and a cadaver study. Neurosurgery. 2004;54:
trigeminal neuralgia with gamma knife radiosurgery. Stereotact Funct 1522-1524, discussion 1524-1526. 172
Neurosurg. 2008;86:135-146. 124. Harrigan MR, Chandler W. Abducens nerve palsy after radiofre-
103. Sheehan J, Pan HC, Stroila M, et al. Gamma knife surgery for quency rhizolysis for trigeminal neuralgia: case report. Neurosurgery.
trigeminal neuralgia: outcomes and prognostic factors. J Neurosurg. 1998;43:623-625.
2005;102:434-441. 125. Rath GP, Dash HH, Bithal PK, et al. Intracranial hemorrhage after
104. Kondziolka D, Lunsford LD, Flickinger JC, et al. Stereotactic percutaneous radiofrequency trigeminal rhizotomy. Pain Pract.
radiosurgery for trigeminal neuralgia: a multiinstitutional study 2009;9:82-84.
using the gamma unit. J Neurosurg. 1996;84:940-945. 126. Solaro C, Brichetto G, Amato MP, et al. The prevalence of pain in
105. Kondziolka D, Zorro O, Lobato-Polo J, et al. Gamma Knife stereo- multiple sclerosis: a multicenter cross-sectional study. Neurology.
tactic radiosurgery for idiopathic trigeminal neuralgia. J Neurosurg. 2004;63:919-921.
2010;112:758-765. 127. Rasche D, Kress B, Schwark C, et al. Treatment of trigeminal neu-
106. Kondziolka D, Lacomis D, Niranjan A, et al. Histological effects of ralgia associated with multiple sclerosis: case report. Neurology.
trigeminal nerve radiosurgery in a primate model: implications for 2004;63:1714-1715.
trigeminal neuralgia radiosurgery. Neurosurgery. 2000;46:971-976, 128. Resnick DK, Jannetta PJ, Lunsford LD, et al. Microvascular decom-
discussion 976-977. pression for trigeminal neuralgia in patients with multiple sclerosis.
107. Foy AB, Parisi JE, Be P. Histologic analysis of a human trigeminal Surg Neurol. 1996;46:358-361, discussion 361-362.
nerve after failed stereotactic radiosurgery: case report. Surg Neurol. 129. Eldridge PR, Sinha AK, Javadpour M, et al. Microvascular decom-
2007;68:655-658. pression for trigeminal neuralgia in patients with multiple sclerosis.
108. Kondziolka D, Lunsford LD, Claassen D, et al. Radiobiology of Stereotact Funct Neurosurg. 2003;81(1–4):57-64.
radiosurgery: Part I. The normal rat brain model. Neurosurgery. 130. Hooge JP, Redekop WK. Trigeminal neuralgia in multiple sclerosis.
1992;31:271-279. Neurology. 1995;45:1294-1296.
109. Fariselli L, Marras C, De Santis M, et al. CyberKnife radiosurgery 131. Kondziolka D, Lunsford LD, Bissonette DJ. Long-term results after
as a first treatment for idiopathic trigeminal neuralgia. Neurosurgery. glycerol rhizotomy for multiple sclerosis-related trigeminal neural-
2009;64(2 suppl):A96-A101. gia. Can J Neurol Sci. 1994;21:137-140.
110. Adler JR Jr, Bower R, Gupta G, et al. Nonisocentric radiosurgical 132. Huang E, Teh BS, Zeck O, et al. Gamma knife radiosurgery for
rhizotomy for trigeminal neuralgia. Neurosurgery. 2009;64(2 suppl): treatment of trigeminal neuralgia in multiple sclerosis patients. Ste-
A84-A90. reotact Funct Neurosurg. 2002;79:44-50.
111. Huang CF, Tu HT, Liu WS, et al. Gamma Knife surgery used as 133. Cheng JS, Sanchez-Mejia RO, Limbo M, et al. Management of
primary and repeated treatment for idiopathic trigeminal neuralgia. medically refractory trigeminal neuralgia in patients with multiple
J Neurosurg. 2008;109(suppl):179-184. sclerosis. Neurosurg Focus. 2005;18(5):e13.
112. Longhi M, Rizzo P, Nicolato A, et al. Gamma Knife radiosur- 134. Mohammad-Mohammadi A, Recinos PF, Lee JH, et al. Surgical
gery for trigeminal neuralgia: results and potentially predictive outcomes of trigeminal neuralgia in patients with multiple sclerosis.
parameters—part I: idiopathic trigeminal neuralgia. Neurosurgery. Neurosurgery. 2013;73:941-950, discussion 950.
2007;61:1254-1260, discussion 1260-1261. 135. Yamaki T, Hashi K, Niwa J, et al. Results of reoperation for failed
113. Dellaretti M, Reyns N, Touzet G, et al. Clinical outcomes after microvascular decompression. Acta Neurochir (Wien). 1992;115:
Gamma Knife surgery for idiopathic trigeminal neuralgia: review 1-7.
of 76 consecutive cases. J Neurosurg. 2008;109(suppl):173-178. 136. Lee SH, Levy EI, Scarrow AM, et al. Recurrent trigeminal neural-
114. Kondziolka D, Lunsford LD, Flickinger JC. Stereotactic radiosur- gia attributable to veins after microvascular decompression. Neuro-
gery for the treatment of trigeminal neuralgia. Clin J Pain. 2002; surgery. 2000;46:356-361, discussion 361-362.
18:42-47. 137. Rath SA, Klein HJ, Richter HP. Findings and long-term results
115. Steiger H. Prognostic factors in the treatment of trigeminal neural- of subsequent operations after failed microvascular decompression
gia: analysis of a differential therapeutic approach. Acta Neurochir for trigeminal neuralgia. Neurosurgery. 1996;39:933-938, discussion
(Wien). 1991;113:11-17. 938-940.
116. Saini S. Retrogasserian anhydrous glycerol injection therapy in tri- 138. Rappaport ZH, Gomori JM. Recurrent trigeminal cistern glyc-
geminal neuralgia: observations in 552 patients. J Neurol Neurosurg erol injections for tic douloureux. Acta Neurochir (Wien). 1988;90:
Psychiatry. 1987;50:1536-1538. 31-34.
117. Burchiel KJ. Percutaneous retrogasserian glycerol rhizolysis in 139. Wang DD, Ouyang D, Englot DJ, et al. Trends in surgical treatment
the management of trigeminal neuralgia. J Neurosurg. 1988;69: for trigeminal neuralgia in the United States of America from 1988
361-366. to 2008. J Clin Neurosci. 2013;20:1538-1545.
118. Young R. Glycerol rhizolysis for treatment of trigeminal neuralgia. 140. Lopez BC, Hamlyn PJ, Zakrzewska JM. Systematic review of abla-
J Neurosurg. 1988;69:39-45. tive neurosurgical techniques for the treatment of trigeminal neu-
119. Lobato RD, Rivas JJ, Sarabia R, et al. Percutaneous microcompres- ralgia. Neurosurgery. 2004;54:973-982, discussion 982-983.
sion of the gasserian ganglion for trigeminal neuralgia. J Neurosurg. 141. Zakrzewska JM, Akram H. Neurosurgical interventions for the
1990;72:546-553. treatment of classical trigeminal neuralgia. Cochrane Database Syst
120. Little AS, Shetter AG, Shetter ME, et al. Long-term pain response Rev. 2011;(9):CD007312.
and quality of life in patients with typical trigeminal neuralgia 142. Cruccu G, Gronseth G, Alksne J, et al. AAN-EFNS guidelines
treated with gamma knife stereotactic radiosurgery. Neurosurgery. on trigeminal neuralgia management. Eur J Neurol. 2008;15:
2008;63:915-923, discussion 923-924. 1013-1028.
121. Brisman R. Gamma Knife surgery with a dose of 75 to 76.8 Gray 143. Tatli M, Satici O, Kanpolat Y, et al. Various surgical modalities for
for trigeminal neuralgia. J Neurosurg. 2004;100:848-854. trigeminal neuralgia: literature study of respective long-term out-
122. Urculo E, Alfaro R, Arrazola M, et al. Trochlear nerve palsy after comes. Acta Neurochir (Wien). 2008;150:243-255.
repeated percutaneous balloon compression for recurrent trigemi- 144. Akram H, Mirza B, Kitchen N, et al. Proposal for evaluating
nal neuralgia: case report and pathogenic considerations. Neurosur- the quality of reports of surgical interventions in the treatment
gery. 2004;54:505-508, discussion 508-509. of trigeminal neuralgia: the Surgical Trigeminal Neuralgia Score.
123. Ugur HC, Savas A, Elhan A, et al. Unanticipated complication Neurosurg Focus. 2013;35(3):E3.
of percutaneous radiofrequency trigeminal rhizotomy: rhinorrhea.
173 Stereotactic Radiosurgery for Trigeminal Neuralgia
Zhiyuan Xu and Jason P. Sheehan

“Tic douloureux” or trigeminal neuralgia (TN) was described by high-resolution imaging of TGN and its root entry zone, interest
the French surgeon Nicholas André in 1756.1 TN is character- was renewed in the radiosurgical approach.
ized by a temporary paroxysmal, lancinating or electric shock–
like neuropathic pain in the trigeminal nerve (TGN) distribution RATIONALE FOR RADIOSURGERY IN TRIGEMINAL
and typically confined to one side of the face, but the pain may
be bilateral in rare cases. Pain-free intervals are common and vary NEURALGIA AND FACTORS RELATED TO
in length from weeks to decades. The prevalence of TN is 4 to FAVORABLE OUTCOMES
5 per 100,000.2 First-line treatment for TN is medical. Agents As early as 1941, Olivecrona12 described that mechanical pressure
typically employed include carbamazepine, gabapentin, pregaba- along the root or at the level of the Gasserian ganglion could be
lin, and baclofen. In particular, response to carbamazepine may the cause of TN. In their pioneering work 3 years later, Granit
help differentiate TN from other orofacial pain syndromes. Dif- and Skoglund13 demonstrated that local pressure on nerve fibers
ferent kinds of facial pain, broadly called “atypical” TN, may could result in painful afferent discharges from the injured neural
occur with deafferentation, trauma, multiple sclerosis (MS), segment,3 evidence that has been supported by more contempo-
somatoform pain disorders, and herpes zoster; more appropriate rary work from Jannetta and colleagues, who have suggested that
classification schemes exist.3 Nonetheless, treatment options for vascular compression of the TGN may be a causal agent in
these types of facial pain are fairly similar to those used for TN TN.6,14,15 In spite of these hypotheses, however, the fact that
but the treatment options are generally less effective. balloon compression of the TGN can lead to symptomatic
Because most patients with TN present in their 50s or 60s, improvement in some patients with TN underscores the void in
they typically have a life expectancy greater than 10 years for our understanding of the pathophysiology of this disease entity.16,17
which the TN must be managed. Over time, it is not uncommon Radiosurgery appears to be more effective when used as the
for a patient’s TN to become refractory to the medications. initial modality than as a salvage treatment for patients with
Those with medically refractory TN go on to neurosurgical TN.18-22 Maesawa and colleagues20 demonstrated that patients
treatment. The widely accepted cause of TN is demyelination who underwent radiosurgery after failed MVD had high rates of
resulting from compression of the nerve by a vascular structure. recurrence, with only 60% maintaining complete pain relief at 1
The first microvascular decompression (MVD) procedure was year, 53% at 2 years, and 33% at 5 years. Régis and colleagues21
described by Taarnhoj4,5 in 1952 and later refined by Barker and have reported that the probability of pain relief at 1 year in
colleagues.6 MVD is the only nonablative surgical procedure. patients without previous surgery was 88%, compared with 82%,
Ablative neurosurgical procedures treated through percutaneous 80%, and 75% in patients with one, two or three prior surgical
techniques (e.g., glycerol injection, thermocoagulation, radiofre- interventions, respectively.
quency rhizotomy) produce lesioning to the TGN and may However, all secondary treatments for TN are associated with
afford pain relief. The concept of stereotactic radiosurgery for poorer results than those after the first procedure, a factor related
TN was introduced as a minimally invasive option, but this to the fact that patients who need re-treatments (or alternative
approach necessitates a lesion of the TGN at least to some treatments) have more severe disease.23,24 Also, for image-guided
degree. surgery, prior procedures such as MVD that disturb the pristine
anatomy of the cisternal segment of the trigeminal nerve result
HISTORICAL PERSPECTIVE OF GAMMA KNIFE in more difficult targeting during radiosurgery. Such targeting
difficulty may translate to a slight reduction in overall efficacy
RADIOSURGERY IN TRIGEMINAL NEURALGIA and an increase in complication rate for those undergoing a
The initial interest in Gamma Knife radiosurgery (GKRS) for salvage procedure such as radiosurgery.
TN arose from Lars Leksell’s interest in using radiosurgery for Among patients who have not undergone previous surgery for
the treatment of pain syndromes.7 Twenty years before building TN, those in whom high-resolution MRI has visualized contact
the Gamma Knife, Leksell used an orthovoltage stereotactic between a blood vessel and the TGN have a particularly favorable
technique to treat patients with TN and achieved some relief of response to radiosurgery.25 In addition to these factors, Pollock
symptoms.8 and associates19 demonstrated that younger patient age and
Leksell also described the use of Gamma-thalamotomies, longer TGN segment irradiated were related to improved radio-
which were used not only for tremor but also for intracta- surgical outcomes. In contrast, Régis and colleagues26 noted that
ble pain.9 The gasserian ganglion was irradiated for TN and the probability of being pain-free was lower in patients younger
Gamma-capsulotomies were performed to interrupt fronto- than 60 years (56%, compared with 91% in older patients).
limbic connections in the treatment of intractable anxiety and The presence of atypical facial pain (such as tingling or
obsessive-compulsive disorders.10,11 Leksell’s first patient was burning) has also been reported to have a poorer prognosis when
treated through an x-ray tube attached to a stereotactically cen- radiosurgical treatment is used, with only 44% improvement of
tered device, and in some patients, Leksell achieved pain relief for symptoms in this group, compared with 84% improvement in
up to 17 years after treatment. The major challenge in targeting patients with typical pain, according to Maesawa and coworkers.20
TN at that time was the limited visualization of the gasserian Patients with atypical facial pain frequently have different types
ganglion on plain radiographs. This hurdle combined with the of underlying disease (e.g., post-herpetic neuralgia, pain follow-
introduction of new drugs and the emergence of nonablative ing sinus surgery, pain from facial trauma). The efficacy of
methods caused a decline in the use of GKRS for TN through GKRS for relief of atypical facial pain is not well defined by the
the 1970s and early 1980s. However, with the introduction of literature, in part owing to the heterogeneity of this patient popu-
high-quality magnetic resonance imaging (MRI), which permitted lation. Nevertheless, it is safe to conclude that the outcome of
1412
CHAPTER 173  Stereotactic Radiosurgery for Trigeminal Neuralgia 1412.e1

KEY WORDS:
173
trigeminal neuralgia
facial pain
stereotactic radiosurgery
Gamma Knife
CHAPTER 173  Stereotactic Radiosurgery for Trigeminal Neuralgia 1413

radiosurgical treatment for atypical facial pain is less favorable brainstem (Fig. 173-2). Using a Gamma Knife angle of 110
than for traditional TN.27 degrees frequently facilitates alignment of the isocenter with the 173
Treatment with GKRS in the setting of MS has been per- axis of the trigeminal nerve. However, a study performed by Xue
formed in small series,21,28-32 because the clinical outcomes of and colleagues,44 demonstrated that by virtue of the steep falloff
MVD and percutaneous glycerol or radiofrequency rhizotomy of the radiation dose, the brainstem may be capable of receiving
provide a lower rate of response and durability of pain relief in an increased dose without significant toxicity.
this cohort of patients.31,33,34 TN develops in approximately 2% Much has been written about whether or not the TGN should
of patients with MS, and this incidence is 20 times that in the be targeted more proximally or more distally (see Fig. 173-1).45-48
general population.35 No consensus has been reached regarding Régis and coworkers,26 who irradiated the cisternal (retrogasse-
the best surgical treatment modality, owing to the lack of pro- rian) portion of the trigeminal root in their patients, reported a
spective studies.36 Rogers and colleagues30 achieved a high level high rate of effective pain control, with an 87% rate of excellent
of success with GRKS in a study of 15 patients with MS-associated outcome and a 10% rate of painful relapse. Use of this more
TN. At a mean follow-up of 17 months, 80% of patients had anterior target choice involved fewer complications even with a
experienced relief from the use of an initial dose of 70 to 90 gray maximum dose of 90 Gy. Nicol and colleagues49 and Pollock and
(Gy). Five patients underwent repeat GKRS to the same treat- associates19 targeted the trigeminal REZ with a dose of 90 Gy
ment area with a mean maximum dose of 48 Gy. All 5 patients and reported a higher rate of pain control, along with a higher
attained some pain relief, and 60% were able to discontinue their rate of complications in comparison with use of lower doses.
TN pain medication. Weller and associates37 reported the clinical Massager and coworkers47 reported a higher pain control rate
outcomes in a series of 35 patients with MS-related TN treated with use of the plexus triangularis as the target. However, the
with GKRS with a median maximum dose of 90 Gy. At a median length of the nerve targeted in this study was significantly
follow-up of 39 months, 88% of patients had either complete increased than in other reports, and as a result, the rate of TGN
pain relief or reasonable pain control (grade I to III on the injury (38%) was higher than that reported by Régis and Pollock’s
Barrow Neurological Institute [BNI] facial pain intensity grading groups. Our group has analyzed 99 patients with idiopathic TN
system; see later discussion of postoperative pain control) at 3 treated with GKRS using a 80-Gy maximum dose and found that
months after GKRS, and the actuarial rates of freedom from proximal targeting provided significantly longer pain relief (BNI
relapse of BNI IV or V pain were 57%, 57%, and 52% at 1, 2, grade I or BNI I to IIIa), although some facial numbness was
and 5 years, respectively. “Non-bothersome” facial numbness sec- present in approximately 56% in this group compared with 27%
ondary to GKRS developed in 39% of patients. To date, one of in the group with more anterior targeting.50 These findings reca-
the largest series, published by the University of Pittsburgh pitulate that long-term pain relief is achievable using appropriate
group, involved 37 patients, in 78% of whom previous surgery targeting of GKRS; however, there is an acceptable tradeoff
had failed; 23 patients experienced complete pain relief (BNI I), between the pain relief and the risk of facial numbness, particu-
and 36 patients (97%) had reasonable pain control (BNI I-IIIb) larly for patients who have severe facial pain that multiple surgical
at a median follow-up of 57 months.38 Reasonable pain relief was procedures have failed to relieve. Interestingly, Gorgulho and
maintained in 83%, 74%, and 54% of patients at 1, 3, and 5 years, colleagues51 demonstrated that gadolinium-induced enhance-
respectively, following GKRS. Nondisabling paresthesias devel- ment of the brainstem at the REZ rather than of the TGN on
oped in 5.4% of patients. Although more investigation is required MRI correlated with favorable pain relief and a higher incidence
to assess the long-term success and toxicity (in particular the of facial numbness.51 This finding further underscores that
development of troublesome facial numbness) of GKRS, this patients with TN may benefit from proximal targeting, as evi-
treatment proves to be effective and safe owing to a reasonably denced by the enhancement of the brainstem rather than distal
favorable complication profile. targeting.
Length of the irradiated TGN is another contentious topic in
relation to pain relief and complications. A randomized clinical
RADIOSURGICAL TARGETING trial has proved that no treatment difference, other than more
Accurate targeting is critical to the success of radiosurgery, and severe complications including facial paresthesias, was obtained
for the indication of trigeminal neuralgia, it is usually performed when the length of TGN irradiated was increased by the use of
with stereotactic MRI (Video 173-1). The MRI is typically per- two adjacent 4-mm isocenters instead of a single 4-mm isocen-
formed with at least a 1.5-tesla (1.5 T) field strength (Fig. 173- ter.52 In addition, the Cleveland Clinic group explored the effec-
1A); 3 T imaging during GKRS may offer better resolution.39,40 tiveness and safety of the combined use of 4- and 8-mm
Localization is performed using T1-weighted and fast spin echo, collimators, which results in a larger target volume.53 No evi-
constructive interference in steady state (CISS) axial images along dence indicated that patients with TN treated with the combined
with coronal images of the nerve (Fig. 173-1B). The axial volume concentric collimators at a maximum of 75 Gy fared better than
acquisition matrices are then divided into sections of 1 mm those treated with a single 4-mm collimator.
without gaps. In many centers, further T1-weighted images are Dose selection for TN has also been studied extensively. Most
obtained after administration of gadolinium. Three-dimensional centers utilize a maximum dose of 70 to 85 Gy.18,22,27,32,54-58 Several
(3D) reconstructions are also routinely performed. When MRI is investigators have demonstrated a significant improvement in
not feasible (i.e., in patients with pacemakers), computed tomog- the rate of pain cessation and a reduction of recurrence rate when
raphy (CT) may be used for targeting,27,41,42 although it is not an elevated maximum dose is employed. Brisman and Mooij59
ideal. CT cisternography can be used to improve delineation of reported that treatments in which the brainstem received 20%
the radiosurgical target.43 or more of the maximum radiation dose to a volume of 20 mm3
With radiosurgery, the initial target for treating TN is the had better pain control than those in which smaller regions were
TGN’s root entry zone (REZ) at the level of the pons. Different targeted with lower doses. In cases in which higher doses are
locations along the affected nerve have been targeted in clinical required, the isocenter is usually placed away from the brainstem
practice. Varying lengths of the cisternal segment of the TGN to avoid injury. Because doses higher than 90 Gy have been
can affect radiosurgical targeting. Longer segments of the nerve associated with an increased risk of postradiosurgery complica-
allow for placement of a single isocenter with minimal to no tions,60 most radiosurgical teams tend to limit the maximum dose
blocking yet delivery of a still tolerable dose to the brainstem. In to 90 Gy or less.48,57,61
a patient with a shorter trigeminal nerve cisternal segment, block- Also important during planning is to ensure that the steepest
ing or beam shaping may be needed to reduce dose to the gradient index of the dose distribution (between the 50% and
1414 SECTION 6  Pain

Figure 173-1. Accurate magnetic


resonance imaging (MRI) for radiosurgical
treatment. A, T1-weighted axial (top) and
corresponding sagittal and coronal (bottom)
unenhanced MR images demonstrating a
Gamma Knife radiosurgical plan for trigeminal
neuralgia. B, Constructive interference in
A B
steady state (CISS) sequence, axial (top) and
corresponding coronal and sagittal (bottom)
images. The nerve is targeted with a single
unblocked isocenter. Yellow circles indicate
the 50% isodose line, which is receiving 40
Gy); green circles represent the 90% isodose
line. C, Proximal targeting: Axial (top) and
corresponding sagittal and coronal (bottom)
1-mm T1-weighted MR images obtained
during the procedure, illustrating the location
of 50% isodose lines (yellow circles) relative to
the trigeminal nerve root entry zone into the
pons. The green circles stand for the 90%
isodose line. D, Distal targeting: Axial (top)
and corresponding coronal and saggital
(bottom) 1-mm T1-weighted MR images
obtained during the procedure, illustrating the
location of a 20% isodose line (green circles)
touching the emergence of the pons. The
yellow circles represent the 50% isodose line,
C D
which was prescribed 40 Gy.

A B
Figure 173-2. Blocking or beam shaping to reduce brainstem radiation dose. A, T1-weighted axial (top)
and corresponding sagittal and coronal (bottom) magnetic resonance images. B, Constructive interference in
steady state (CISS) axial (top) and corresponding coronal and sagittal (bottom) images. The 50% isodose line
is depicted in yellow and the 20% isodose line in green. The isocenter has been blocked so as to alter the
normal spherical shape to a more elliptical one. This change helps decrease the brainstem dose in patients
who are undergoing re-treatment or in in whom the cisternal segment of the trigeminal nerve is short.
CHAPTER 173  Stereotactic Radiosurgery for Trigeminal Neuralgia 1415

70% isodose lines) coincides with the periphery of tissue being achieved within the first several weeks after the procedure. Régis
treated. This goal may require several overlapping fields of radia- and associates21 achieved a high rate of initial pain control, with 173
tion, each using a different collimator size and a separate stereo- 94% of their patients having either excellent or good response
tactic focal point. Changing the relative time of radiation at each within 10 days. Pollock and associates19 reported that 75% of
target may also change the isodose distribution. Finally, the radia- their patients experienced complete pain relief at some point after
tion field may be altered by blocking some of the radiation radiosurgery. Dhople and colleagues24 reviewed 112 patients with
sources, also known as “plugging” or shielding. However, con- typical TN for a median follow-up of 5.6 years. The median time
flicting reports exist regarding the benefit of blocking or shield- to response to treatment was 2 weeks with a range of 0 to
ing because some clinicians contend that blocking the nerve root 12 weeks. Among those patients with a response to treatment,
might not be beneficial.60,62 40% experienced pain relief within 1 week. Fountas and cowork-
ers69 reported that a majority of patients with no previous surgery
responded within 4 weeks after treatment. Pollock and associ-
CLINICAL FOLLOW-UP ates19 found a comparable mean latency period of about 3 weeks.
Generally patients should be followed up at 3- to 6-month inter- In other large series, however, the latency has been noted to be
vals, and new MRI studies should be obtained 6 to 9 months as long as 1 year following GKRS.20,64
postoperatively. In patients who have new-onset neurological According to the University of Pittsburgh group,64 the major-
symptoms, an MRI can be performed earlier. In addition to ity (89%) of patients with TN had response to GKRS within 6
imaging studies, follow-up examination should include a full neu- months of treatment, with a median of 1 month, achieving pain
rological assessment, examination of cranial nerve function, and relief grades BNI I to IIIb. Initial complete pain relief (BNI I)
full ophthalmologic examination as needed. was achieved in 40% of patients, with a range between 1 day and
The TGN function in particular may have significant correla- 1 year. The 11% of patients who did not experience pain control
tion to patient prognosis. Pollock and associates19 reported a within a year also had no clinical improvement following radio-
strong correlation between the development of new facial sensory surgery, indicating that 1 year may be the optimal time to judge
loss and the achievement and maintenance of pain relief after the true effectiveness of radiosurgery in patients with TN.
radiosurgery, with patients who have new trigeminal deficits From 1996 to 2003, 151 patients with TN were treated with
being 4.5 times more likely to have excellent pain relief than the GKRS at the University of Virginia and had evaluable
patients who have normal postradiosurgery TGN function. follow-up.55 Radiosurgery was performed once in 136 patients,
Experimental models of radiosurgical treatment have confirmed twice in 14 patients, and three times in 1 patient. One hundred
that the etiology for this difference may lie in a nonselective twenty-two patients presented with typical TN, 3 with atypical
destruction of myelinated and unmyelinated sensory fibers, TN, 4 with MS-associated TN, and 7 with TN secondary to a
including afferent nociceptive fibers.63 This finding remains con- cavernous sinus tumor. In each case, the chosen radiosurgical
troversial because it is not confirmed in the newest large series— target was located 2 to 4 mm anterior to the surface of the pons.
that is, the large series found no correlation between the long-term The maximum radiation doses ranged from 50 to 90 Gy. The
pain control and TGN dysfunction.21,24,64 mean time to relief of pain was 24 days (range, 1-180 days). Forty-
Some have suggested that enhancement of the TGN on post- seven percent, 45%, and 34% of patients were pain free without
radiosurgery MRI may correlate with the radiation dose used and medication at the 1-, 2-, and 3-year follow-up, respectively.
thus may be related to the accuracy of targeting.65 In clinical Ninety percent, 77%, and 70% of patients experienced some
practice, however, TGN enhancement has not routinely corre- improvement in pain at 1-, 2-, and 3-year follow-up, respectively.
lated with better clinical outcomes.51,66 Twelve patients (9%) suffered the onset of new facial numbness
after treatment.
PAIN CONTROL AFTER RADIOSURGERY FOR Currently the most commonly used classification systems, the
modified Marseille scale and BNI pain intensity score, remain
TRIGEMINAL NEURALGIA: A REVIEW OF less than ideal with respect to medication usage. Therefore,
THE LITERATURE quality of life (QOL) is a very important issue in assessing the
Although MVD remains the neurosurgical treatment of choice clinical results. In practice, many patients are hesitant to discon-
for TN, GKRS offers a reasonable alternative in patients who are tinue or taper off the pain medications in fear of possible pain
either unwilling to undergo surgery or who are not ideal surgical recurrence, further suggesting that the BNI pain intensity score
candidates. A summary of major series on stereotactic radiosur- may underestimate the actual rate at which BNI I pain status is
gery for TN is presented in Table 173-1.21,23,24,54,55,60,61,64,67-74 The achieved.24,32,60
majority of studies have utilized the BNI intensity grading The TN pain can be debilitating to those affected, and pain
system75,76 or the Marseille pain grading system with modifica- control itself may not be the only QOL measure that is related
tions.21 To date, however, no standardized grading system is avail- to patient outcomes. Petit and colleagues56 investigated QOL
able, rendering comparison of various TN studies difficult or associated with GKRS for TN. Even with partial pain relief,
impossible. Owing to variations in data presentation, not all data patients reported a median of 80% improvement in QOL, with
are available for every series, although every effort has been made 65% believing that their GKRS was successful. Patients in whom
to present a homogenous data set in Table 173-1 to allow direct pain relief was maintained at the time of analysis reported a
comparison of a wide variety of studies. Prior to 2006, very few median 100% improvement in their QOL, with a 100% success
papers used Kaplan-Meier methodology to indicate the durability rate. Even the patients with temporary treatment responses (pain
of pain relief in response to stereotactic radiosurgery77; the recurrence after a median of 8.5 months) described a median 80%
majority of papers documented the percentage of pain response improvement in QOL with an associated 60% rate of treatment
at the last follow-up. Currently the wide use of the Kaplan-Meier success. These findings suggest that temporary pain relief coupled
model allows us to appreciate the maintenance of failure-free with a low incidence of treatment morbidity is a worthwhile
treatment status given that facial pain is a time-to-event variable treatment outcome for most patients. Pan and associates80
similar to cancer survival.78 explored the changes in QOL after GKRS in 52 patients with
The effects of radiation depend on both acute and delayed TN whose median age was 71 years and who had remarkably
changes in the target tissue, so there is a latency interval for the various comorbidities, including cardiovascular disease, liver cir-
expected response, as in all radiosurgical procedures.79 In all of rhosis, and coagulopathy. Pain relief significantly contributed to
the series presented in Table 173-1, pain relief was typically the improvement in QOL during the follow-up of more than 4
CHAPTER 173  Stereotactic Radiosurgery for Trigeminal Neuralgia 1417

years. The improved aspects of QOL included physical function, collimator anterior to the portion of the TGN originally targeted
role limitation due to a physical problem, bodily pain, general so that there was a 50% overlap in the two radiosurgical volumes 173
health, vitality, social function, role limitation due to an emo- (compared with 100% overlap in most other studies). The ratio-
tional problem, and mental health.80 nale for this strategy is to reduce the radiation dose to the brain-
stem while increasing the dose to the length of trigeminal nerve
root. Kimball and associates93 have reported using a single 4-mm
RECURRENCE AND RE-TREATMENT collimator isocenter targeted 4 to 5 mm anterior to the initial
Reported rates of recurrence following radiosurgery for TN have 50% isocenter and a lower maximum dose of 70 Gy. They used
ranged from 5% to 43%, and recurrence is likely related to a modified Marseille pain scale in which favorable pain control
incomplete radiation effects on the targeted tissues.a Given the was set as more than 50% improvement with or without pain
low risk profile of GKRS, some patients may benefit from a medications; TN pain was controlled in 70% of their patients at
second GKRS procedure, in particular in those for whom mul- 1 year, in 50% at 3 years, and in 50% at 5 years. In addition, there
tiple surgical interventions failed prior to the initial GKRS. was a correlation between newly developed facial numbness and
Pollock and associates,28 Maesawa and associates,20 Herman and better pain relief. However, rates of other complications after the
associates,88 and Park and associates89 recommend re-treatment second GKRS increased, including dry eye in 10.9% of patients,
with radiosurgery in patients if they experienced a significant taste change in 8.7%, jaw weakness in 2.2%, and bothersome
reduction in pain after initial GKRS. Eight of 10 patients in the facial numbness in 8.7%.
initial study by Pollock and associates90 were pain-free after initial In an attempt to balance the benefits and risks, other clinicians
radiosurgery, and all of these patients had complete pain relief consider total radiation dose important in terms of efficacy and
with a second GKRS procedure. These findings suggest that the complications. Generally, the maximum dose of the second
response to initial GKRS may be used by physicians to predict GKRS is identical to or slightly lower than the first dose.88,89,91-93
the response to a second procedure. Other investigators have Some neurosurgeons recommend that the cumulative dose be
observed that patients who have longer remission after radiosur- kept within a range of 120 to 170 Gy.89 Dvorak and coworkers95
gery tend to have better results from re-treatment.88 The Uni- determined, from their data and analysis of data from seven other
versity of Pittsburgh group found that approximately 43% of studies, that a cumulative radiation dose lower than 140 Gy was
patients had pain recurrence at a median of 4 years after the initial appropriate. Zhang and colleagues46 used a lower dose of 75 Gy
GKRS.64 Subsequently, the investigators reviewed data from 119 in the initial GKRS and 40 Gy in the second procedure. They
patients who underwent a second GKRS procedure with a median also treated at the 40% isodose line tangential to the brainstem
follow-up of 4 years. As with the initial GKRS, 87% of patients surface. Both the lower isodose and the lower total dose would
achieved initial pain relief (BNI I-IIIb) and actuarial pain relief seem to confer a lower chance of pain control from the radiosur-
rate was 44% at 5 years. Facial sensory dysfunction developed in gery. In a total of 40 patients with a median follow-up of 28
21% of patients within 18 months of the second GKRS proce- months, 11 patients had complete pain relief, 7 had nearly com-
dure, much higher than the 10.5% rate in patients after the initial plete relief (≥ 90%), 8 had partial relief (≥ 50%), and 14 had
procedure.64 Nevertheless, a second GKRS procedure for recur- minimal or no relief.46 Ultimately, a prospective dose planning
rent TN is regarded as safe and effective.89 study will be required to determine which technique is preferred
The precise location to target during re-treatment is contro- for re-treatment.
versial. Generally speaking, re-treatment involves the delivery of In our experience, the side effects of repeat GKRS are more
radiation at91 or anterior to (Fig. 173-3)88,89,92,93 the previously common but occur at an acceptable rate when compared with
treated site. Hasegawa and colleagues94 positioned a single 4-mm that after the initial procedure, with new facial numbness being
the most common complication reported. New-onset or wors-
a
References 2, 3, 19, 20, 26, 47, 49, 57, 59, 66, 81-87. ened facial numbness, including mild to moderate trigeminal

A B
Figure 173-3. Repeat Gamma Knife radiosurgery for recurrent trigeminal neuralgia. A, T1-weighted
axial (top) and corresponding sagittal and coronal (bottom) magnetic resonance images. B, Constructive
interference in steady state (CISS) axial (top) and corresponding coronal and sagittal (bottom) images. The
50% isodose line, shown in in yellow, was prescribed anterior to the previously delivered 50% isodose line,
shown in blue.
1418 SECTION 6  Pain

dysfunction, can be found in 11% to 70% of patients after second cannot be identified as the cause of symptoms, or those unwilling
treatments.2,20,21,92 The overall effectiveness of repeat radiosur- to undergo open surgery, radiosurgery is an appropriate alterna-
gery is similar to that of the initial procedure, with rates of tive. The risks of repeat MVD are also known to be significantly
complete or partial pain relief ranging from 72% to 85%,28,49,89,91,94 higher than after the first procedure,6 and thus patients who have
although fewer patients undergoing re-treatment have enough recurrence following failed MVD may also be considered candi-
pain relief to completely discontinue their pain medications. dates for radiosurgery albeit with a lower chance of successful
remission. Nevertheless, patients treated with GKRS are gener-
ally one decade older than those who underwent MVD in prior
COMPLICATIONS FROM RADIOSURGERY publications, making the comparison less equivalent.98,100
Although stereotactic radiosurgery is not associated with some of
the immediate risks of open surgical procedures, it nonetheless
has risks. Potential complications after radiosurgery include
CONCLUSION
radiation-induced parenchymal changes (only some of which are Although MVD remains the treatment of choice for patients with
reversible), radiation-induced neoplasia, and vascular injury.96,97 A TN, stereotactic radiosurgery is an effective treatment for those
noteworthy complication in the treatment of trigeminal pain is unwilling or unable to undergo an MVD. Stereotactic radiosur-
facial numbness or dysesthesias, with anesthesia dolorosa being gery is also a reasonable treatment option for patients with recur-
the most dreaded one. The incidence of new-onset trigeminal rent TN after previous invasive surgical intervention. More than
dysfunction varies from 6% to 66%.22-24,45,55-57,64 In general, the 50% of patients with typical facial pain have relief following
placement of the isocenter proximal to the brainstem or increased radiosurgery, although a lower proportion of patients with atypi-
radiation dose is correlated with a higher risk of newly developed cal pain do so. Optimal target selection and radiation dose remain
facial numbness. Pollock and colleagues57 reported an association the subjects of ongoing debate within the neurosurgical com-
between higher radiation doses and the risk of TGN dysfunction. munity. Long-term follow-up for recurrence as well as for
In that study, 54% of patients treated with 90 Gy experienced radiation-induced complications is required in all patients.
facial numbness, whereas only 15% of patients treated with 70
Gy experienced a similar problem.57 In our series, new facial
numbness developed in 12 out of 136 patients (9%) following SUGGESTED READINGS
GKRS.55 Only 1 patient had received a dose of 90 Gy, and no Brisman R. Gamma Knife surgery with a dose of 75 to 76.8 Gray for
trigeminal neuralgia. J Neurosurg. 2004;100:848-854.
facial numbness was noted in this case. Our later study comparing Brisman R, Khandji AG, Mooij RB. Trigeminal nerve-blood vessel rela-
the proximal and the distal targeting locations indicated that with tionship as revealed by high-resolution magnetic resonance imaging
the same 80 Gy maximum dose, targeting to the proximal loca- and its effect on pain relief after Gamma Knife radiosurgery for tri-
tion achieved longer facial pain relief than targeting to the distal geminal neuralgia. Neurosurgery. 2002;50:1261-1266.
location (56% vs. 25%).50 Fortunately, cases of anesthesia dolo- Brisman R, Mooij R. Gamma knife radiosurgery for trigeminal neuralgia:
rosa and absence of the corneal reflex following radiosurgery are dose-volume histograms of the brainstem and trigeminal nerve. J Neu-
quite rare. The risk of anesthesia dolorosa secondary to GKRS rosurg. 2000;93(suppl 3):155-158.
is claimed to be less than 1%, and only a few cases have been Eller JL, Raslan AM, Burchiel KJ. Trigeminal neuralgia: definition and
reported.23,61,64 classification. Neurosurg Focus. 2005;18:E3.
Hasegawa T, Kondziolka D, Spiro R, et al. Repeat radiosurgery for refrac-
tory trigeminal neuralgia. Neurosurgery. 2002;50:494-500.
COMPARISON OF RADIOSURGERY AND OPEN Kondziolka D, Lacomis D, Niranjan A, et al. Histological effects of tri-
geminal nerve radiosurgery in a primate model: implications for tri-
SURGERY FOR TRIGEMINAL NEURALGIA geminal neuralgia radiosurgery. Neurosurgery. 2000;46:971-976.
MVD is the only nonablative treatment modality for TN that Kondziolka D, Lunsford LD, Flickinger JC, et al. Stereotactic radiosur-
addresses the underlying pathophysiology of the disease, making gery for trigeminal neuralgia: a multiinstitutional study using the
it the well-established gold standard. Barker and colleagues6 gamma unit. J Neurosurg. 1996;84:940-945.
reported that 70% of patients who underwent MVD had com- Maesawa S, Salame C, Flickinger JC, et al. Clinical outcomes after ste-
plete pain relief 10-years postoperatively. Likewise, Olson and reotactic radiosurgery for idiopathic trigeminal neuralgia. J Neurosurg.
2001;94:4-20.
associates98 reported in a retrospective study that 74% of patients Massager N, Lorenzoni J, Devriendt D, et al. Gamma knife surgery
were pain free at 10 years after treatment in a total of 156 patients for idiopathic trigeminal neuralgia performed using a far-anterior cis-
with TN. Pain recurred in 18% of patients over a 25-year ternal target and a high dose of radiation. J Neurosurg. 2004;100:
follow-up. Although this type of follow-up and sustained pain 597-605.
relief are lacking in the radiosurgical literature, GKRS provides Merrison AF, Fuller G. Treatment options for trigeminal neuralgia. BMJ.
similar favorable pain response and exceedingly low rates of com- 2003;327:1360-1361.
plications except facial numbness.64 In patients without previous Nicol B, Regine WF, Courtney C, et al. Gamma knife radiosurgery
surgery, Pollock and coworkers99 observed a 67% rate of com- using 90 Gy for trigeminal neuralgia. J Neurosurg. 2000;93(suppl 3):
plete pain relief at 1 and 3 years, whereas Maesawa and col- 152-154.
Petit JH, Herman JM, Nagda S, et al. Radiosurgical treatment of trigemi-
leagues20 observed a 70% rate of complete pain relief at 9 months nal neuralgia: evaluating quality of life and treatment outcomes. Int J
and 5 years. Given these results, radiosurgery as a primary treat- Radiat Oncol Biol Phys. 2003;56:1147-1153.
ment appears to provide a rate of pain control comparable to that Pollock BE, Foote RL, Stafford SL, et al. Results of repeated Gamma
of open surgery. Knife radiosurgery for medically unresponsive trigeminal neuralgia.
The major disadvantages of radiosurgery are the higher rate J Neurosurg. 2000;93(suppl 3):162-164.
of recurrence and the delay in onset of pain relief in comparison Pollock BE, Phuong LK, Foote RL, et al. High-dose trigeminal neuralgia
with MVD. As described in the previous section, only 27% of radiosurgery associated with increased risk of trigeminal nerve dys-
patients in the Pollock series were pain free without medication function. Neurosurgery. 2001;49:58-62.
at the time of latest follow-up.99 Therefore, it is generally accepted Pollock BE, Phuong LK, Gorman DA, et al. Stereotactic radiosurgery for
idiopathic trigeminal neuralgia. J Neurosurg. 2002;97:347-353.
that, particularly in patients who are young and have less signifi- Régis J, Bartolomei F, Metellus P, et al. Radiosurgery for trigeminal neu-
cant comorbidities, MVD should (at least for now) remain the ralgia and epilepsy. Neurosurg Clin N Am. 1999;10:359-377.
first-line treatment for those who can tolerate the operation Régis J, Metellus P, Hayashi M, et al. Prospective controlled trial of
because of the durability of pain relief.50,64 In patients who cannot Gamma Knife surgery for essential trigeminal neuralgia. J Neurosurg.
tolerate a surgical procedure, those in whom a vascular etiology 2006;104:913-924.
CHAPTER 173  Stereotactic Radiosurgery for Trigeminal Neuralgia 1419

Richards GM, Bradley KA, Tomé WA, et al. Linear accelerator radiosur- Sheehan J, Pan HC, Stroila M, et al. Gamma knife surgery for trigeminal
gery for trigeminal neuralgia. Neurosurgery. 2005;57:1193-1200. neuralgia: outcomes and prognostic factors. J Neurosurg. 2005;102: 173
Rogers CL, Shetter AG, Fiedler JA, et al. Gamma knife radiosurgery for 434-441.
trigeminal neuralgia: the initial experience of The Barrow Neurologi- Tawk RG, Duffy-Fronckowiak M, Scott BE, et al. Stereotactic Gamma
cal Institute. Int J Radiat Oncol Biol Phys. 2000;47:1013-1019. Knife surgery for trigeminal neuralgia: detailed analysis of treatment
Rogers CL, Shetter AG, Ponce FA, et al. Gamma knife radiosurgery for response. J Neurosurg. 2005;102:442-449.
trigeminal neuralgia associated with multiple sclerosis. J Neurosurg. Zhang P, Brisman R, Choi J, et al. Where to locate the isocenter? The
2002;97(suppl 5):529-532. treatment strategy for repeat trigeminal neuralgia radiosurgery. Int J
Shaya M, Jawahar A, Caldito G, et al. Gamma knife radiosurgery for Radiat Oncol Biol Phys. 2005;62:38-43.
trigeminal neuralgia: a study of predictors of success, efficacy, safety,
and outcome at LSUHSC. Surg Neurol. 2004;61:529-534. See a full reference list on ExpertConsult.com
CHAPTER 173  Stereotactic Radiosurgery for Trigeminal Neuralgia 1419.e1

REFERENCES 28. Pollock BE, Foote RL, Stafford SL, et al. Results of repeated
1. Eboli P, Stone JL, Aydin S, et al. Historical characterization of Gamma Knife radiosurgery for medically unresponsive trigeminal 173
trigeminal neuralgia. Neurosurgery. 2009;6:1183-1186, discussion neuralgia. J Neurosurg. 2000;93(suppl 3):162-164.
1186-1187. 29. Huang E, Teh BS, Zeck O, et al. Gamma knife radiosurgery for
2. Merrison AF, Fuller G. Treatment options for trigeminal neuralgia. treatment of trigeminal neuralgia in multiple sclerosis patients. Ste-
BMJ. 2003;327:1360-1361. reotact Funct Neurosurg. 2002;79:44-50.
3. Eller JL, Raslan AM, Burchiel KJ. Trigeminal neuralgia: definition 30. Rogers CL, Shetter AG, Ponce FA, et al. Gamma knife radiosurgery
and classification. Neurosurg Focus. 2005;18:E3. for trigeminal neuralgia associated with multiple sclerosis. J Neuro-
4. Taarnhoj P. Decompression of the trigeminal root and the posterior surg. 2002;97(suppl):529-532.
part of the ganglion as treatment in trigeminal neuralgia; prelimi- 31. Cheng JS, Sanchez-Mejia RO, Limbo M, et al. Management of
nary communication. J Neurosurg. 1952;9:288-290. medically refractory trigeminal neuralgia in patients with multiple
5. Taarnhoj P. A new operation for trigeminus neuralgia; decompres- sclerosis. Neurosurg Focus. 2005;18:e13.
sion of the trigeminal root and the posterior part of the ganglion; 32. Loescher AR, Radatz M, Kemeny A, et al. Stereotactic radiosurgery
preliminary report. Nord Med. 1952;47:360-364. for trigeminal neuralgia: outcomes and complications. Br J Neuro-
6. Barker FG 2nd, Jannetta PJ, Bissonette DJ, et al. The long-term surg. 2012;26:45-52.
outcome of microvascular decompression for trigeminal neuralgia. 33. Ariai MS, Mallory GW, Pollock BE. Outcomes after microvascular
N Engl J Med. 1996;334:1077-1083. decompression for patients with trigeminal neuralgia and suspected
7. Steiner L, Lindquist C, Foster D, et al., eds. Radiosurgery: Baseline multiple sclerosis. World Neurosurg. 2014;81:599-603.
and Trends. New York: Raven; 1992:263-292. 34. Resnick DK, Jannetta PJ, Lunsford LD, et al. Microvascular decom-
8. Leksell L. Brain fragments in radiosurgery. In: Steiner L, Lindquist pression for trigeminal neuralgia in patients with multiple sclerosis.
C, Foster D, et al., eds. Radiosurgery: Baseline and Trends. New York: Surg Neurol. 1996;46:358-361, discussion 361-362.
Raven Press; 1992:623-692. 35. Love S, Gradidge T, Coakham HB. Trigeminal neuralgia due to
9. Steiner L, Forster D, Leksell L, et al. Gammathalamotomy in multiple sclerosis: ultrastructural findings in trigeminal rhizotomy
intractable pain. Acta Neurochir (Wien). 1980;52:173-184. specimens. Neuropathol Appl Neurobiol. 2001;27:238-244.
10. Rylander G. Stereotactic Radiosurgery in Anxiety and Obsessive- 36. Montano N, Papacci F, Cioni B, et al. What is the best treatment
compulsive Neurosis. Amsterdam: Elsevier/North-Holland Biomedi- of drug-resistant trigeminal neuralgia in patients affected by mul-
cal Press; 1979. tiple sclerosis? A literature analysis of surgical procedures. Clin
11. Mindus P. Capsulotomy in Anxiety Disorders, a Multidiscipline Study. Neurol Neurosurg. 2013;115:567-572.
Stockholm: Neurosurgery, Karolinska Institute; 1991. 37. Weller M, Marshall K, Lovato JF, et al. Single-institution retrospec-
12. Olivecrona H. Die Trigeminusneuralgie und ihre Behandlung. Ner- tive series of Gamma Knife radiosurgery in the treatment of mul-
venarzt. 1941;14:49-57. tiple sclerosis-related trigeminal neuralgia: factors that predict
13. Granit R, Leksell L, Skoglund CR. Fibre interaction in injured or efficacy. Stereotact Funct Neurosurg. 2014;92:53-58.
compressed region of nerve. Brain. 1944;67:125-140. 38. Zorro O, Lobato-Polo J, Kano H, et al. Gamma Knife radiosurgery
14. Lovely TJ, Jannetta PJ. Microvascular decompression for trigeminal for multiple sclerosis-related trigeminal neuralgia. Neurology. 2009;
neuralgia: surgical technique and long-term results. Neurosurg Clin 73:1149-1154.
N Am. 1997;8:11-29. 39. Cassetta M, Pranno N, Pompa V, et al. High resolution 3-T MR
15. Jannetta PJ. Arterial compression of the trigeminal nerve at the pons imaging in the evaluation of the trigeminal nerve course. Eur Rev
in patients with trigeminal neuralgia. J Neurosurg. 1967;26(suppl): Med Pharmacol Sci. 2014;18:257-264.
159-162. 40. Leal PR, Hermier M, Souza MA, et al. Visualization of vascular
16. Brown JA, Chittum CJ, Sabol D, et al. Percutaneous balloon com- compression of the trigeminal nerve with high-resolution 3T MRI:
pression of the trigeminal nerve for treatment of trigeminal neural- a prospective study comparing preoperative imaging analysis to
gia. Neurosurg Focus. 1996;1:e4, discussion 1 p following e4. surgical findings in 40 consecutive patients who underwent micro-
17. Brown JA, Gouda JJ. Percutaneous balloon compression of the vascular decompression for trigeminal neuralgia. Neurosurgery.
trigeminal nerve. Neurosurg Clin N Am. 1997;8:53-62. 2011;69:15-25, discussion 26.
18. Guo S, Chao ST, Reuther AM, et al. Review of the treatment of 41. Park KJ, Kano H, Berkowitz O, et al. Computed tomography-
trigeminal neuralgia with Gamma Knife radiosurgery. Stereotact guided Gamma Knife stereotactic radiosurgery for trigeminal neu-
Funct Neurosurg. 2008;86:135-146. ralgia. Acta Neurochir (Wien). 2011;153:1601-1609.
19. Pollock BE, Phuong LK, Gorman DA, et al. Stereotactic radiosur- 42. Attia A, Tatter SB, Weller M, et al. CT-only planning for Gamma
gery for idiopathic trigeminal neuralgia. J Neurosurg. 2002;97: Knife radiosurgery in the treatment of trigeminal neuralgia: meth-
347-353. odology and outcomes from a single institution. J Med Imaging
20. Maesawa S, Salame C, Flickinger JC, et al. Clinical outcomes after Radiat Oncol. 2012;56:490-494.
stereotactic radiosurgery for idiopathic trigeminal neuralgia. J Neu- 43. Worthington C, Hutson K, Boulware R, et al. Computerized
rosurg. 2001;94:14-20. tomography cisternography of the trigeminal nerve for stereotactic
21. Régis J, Metellus P, Hayashi M, et al. Prospective controlled trial of radiosurgery: case report. J Neurosurg. 2000;93(suppl 3):169-171.
Gamma Knife surgery for essential trigeminal neuralgia. J Neuro- 44. Xue J, Goldman HW, Grimm J, et al. Dose-volume effects on brain-
surg. 2006;104:913-924. stem dose tolerance in radiosurgery. J Neurosurg. 2012;117(suppl):
22. Tawk RG, Duffy-Fronckowiak M, Scott BE, et al. Stereotactic 189-196.
Gamma Knife surgery for trigeminal neuralgia: detailed analysis of 45. Kondziolka D, Lunsford LD, Flickinger JC, et al. Stereotactic
treatment response. J Neurosurg. 2005;102:442-449. radiosurgery for trigeminal neuralgia: a multiinstitutional study
23. Verheul JB, Hanssens PE, Lie ST, et al. Gamma Knife surgery for using the gamma unit. J Neurosurg. 1996;84:940-945.
trigeminal neuralgia: a review of 450 consecutive cases. J Neurosurg. 46. Zhang P, Brisman R, Choi J, et al. Where to locate the isocenter?
2010;113(suppl):160-167. The treatment strategy for repeat trigeminal neuralgia radiosurgery.
24. Dhople AA, Adams JR, Maggio WW, et al. Long-term outcomes of Int J Radiat Oncol Biol Phys. 2005;62:38-43.
Gamma Knife radiosurgery for classic trigeminal neuralgia: impli- 47. Massager N, Lorenzoni J, Devriendt D, et al. Gamma Knife surgery
cations of treatment and critical review of the literature. Clinical for idiopathic trigeminal neuralgia performed using a far-anterior
article. J Neurosurg. 2009;111:351-358. cisternal target and a high dose of radiation. J Neurosurg. 2004;100:
25. Brisman R, Khandji AG, Mooij RB. Trigeminal nerve-blood vessel 597-605.
relationship as revealed by high-resolution magnetic resonance 48. Matsuda S, Serizawa T, Nagano O, et al. Comparison of the results
imaging and its effect on pain relief after Gamma Knife radiosur- of 2 targeting methods in Gamma Knife surgery for trigeminal
gery for trigeminal neuralgia. Neurosurgery. 2002;50:1261-1266, neuralgia. J Neurosurg. 2008;109(suppl):185-189.
discussion 1266-1267. 49. Nicol B, Regine WF, Courtney C, et al. Gamma Knife radiosurgery
26. Régis J, Bartolomei F, Metellus P, et al. Radiosurgery for tri- using 90 Gy for trigeminal neuralgia. J Neurosurg. 2000;93(suppl 3):
geminal neuralgia and epilepsy. Neurosurg Clin N Am. 1999;10: 152-154.
359-377. 50. Xu Z, Schlesinger D, Moldovan K, et al. Impact of target location
27. Brisman R. Gamma knife surgery with a dose of 75 to 76.8 Gray on the response of trigeminal neuralgia to stereotactic radiosurgery.
for trigeminal neuralgia. J Neurosurg. 2004;100:848-854. J Neurosurg. 2014;120:716-724.
1419.e2 SECTION 6  Pain

51. Gorgulho A, De Salles AA, McArthur D, et al. Brainstem and tri- 74. Longhi M, Rizzo P, Nicolato A, et al. Gamma Knife radiosurgery
geminal nerve changes after radiosurgery for trigeminal pain. Surg for trigeminal neuralgia: results and potentially predictive
Neurol. 2006;66:127-135, discussion 135. parameters—part I: idiopathic trigeminal neuralgia. Neurosurgery.
52. Flickinger JC, Pollock BE, Kondziolka D, et al. Does increased 2007;61:1254-1260, discussion 1260-1261.
nerve length within the treatment volume improve trigeminal neu- 75. Rogers CL, Shetter AG, Fiedler JA, et al. Gamma Knife radiosur-
ralgia radiosurgery? A prospective double-blind, randomized study. gery for trigeminal neuralgia: the initial experience of The Barrow
Int J Radiat Oncol Biol Phys. 2001;51:449-454. Neurological Institute. Int J Radiat Oncol Biol Phys. 2000;47:
53. Kanner AA, Neyman G, Suh JH, et al. Gamma Knife radiosurgery 1013-1019.
for trigeminal neuralgia: comparing the use of a 4-mm versus con- 76. Akram H, Mirza B, Kitchen N, et al. Proposal for evaluating the
centric 4- and 8-mm collimators. Stereotact Funct Neurosurg. 2004; quality of reports of surgical interventions in the treatment of
82:49-57. trigeminal neuralgia: the Surgical Trigeminal Neuralgia Score.
54. Richards GM, Bradley KA, Tome WA, et al. Linear accelerator Neurosurg Focus. 2013;35:E3.
radiosurgery for trigeminal neuralgia. Neurosurgery. 2005;57:1193- 77. Kondziolka D, Lunsford LD, Flickinger JC. Stereotactic radiosur-
1200, discussion 1193-1200. gery for the treatment of trigeminal neuralgia. Clin J Pain.
55. Sheehan J, Pan HC, Stroila M, et al. Gamma Knife surgery for 2002;18:42-47.
trigeminal neuralgia: outcomes and prognostic factors. J Neurosurg. 78. Régis J, Tuleasca C. Fifteen years of Gamma Knife surgery for
2005;102:434-441. trigeminal neuralgia in the Journal of Neurosurgery: history of a
56. Petit JH, Herman JM, Nagda S, et al. Radiosurgical treatment of revolution in functional neurosurgery. J Neurosurg. 2011;115(suppl):
trigeminal neuralgia: evaluating quality of life and treatment out- 2-7.
comes. Int J Radiat Oncol Biol Phys. 2003;56:1147-1153. 79. Jagannathan J, Sherman JH, Mehta GU, et al. Radiobiology of brain
57. Pollock BE, Phuong LK, Foote RL, et al. High-dose trigeminal metastasis: applications in stereotactic radiosurgery. Neurosurg Focus.
neuralgia radiosurgery associated with increased risk of trigeminal 2007;22:E4.
nerve dysfunction. Neurosurgery. 2001;49:58-62, discussion 62-64. 80. Pan HC, Sheehan J, Huang CF, et al. Quality-of-life outcomes after
58. Kim YH, Kim DG, Kim JW, et al. Is it effective to raise the irradia- Gamma Knife surgery for trigeminal neuralgia. J Neurosurg. 2010;
tion dose from 80 to 85 Gy in Gamma Knife radiosurgery for tri- 113(suppl):191-198.
geminal neuralgia? Stereotact Funct Neurosurg. 2010;88:169-176. 81. Flannery TW, Suntharalingam M, Kwok Y, et al. Gamma Knife
59. Brisman R, Mooij R. Gamma Knife radiosurgery for trigeminal stereotactic radiosurgery for synchronous versus metachronous
neuralgia: dose-volume histograms of the brainstem and trigeminal solitary brain metastases from non-small cell lung cancer. Lung
nerve. J Neurosurg. 2000;93(suppl 3):155-158. Cancer. 2003;42:327-333.
60. Young B, Shivazad A, Kryscio RJ, et al. Long-term outcome of 82. Levivier M, Lorenzoni J, Massager N, et al. Use of the Leksell
high-dose Gamma Knife surgery in treatment of trigeminal neural- Gamma Knife C with automatic positioning system for the treat-
gia. J Neurosurg. 2013;119:1166-1175. ment of meningioma and vestibular schwannoma. Neurosurg Focus.
61. Marshall K, Chan MD, McCoy TP, et al. Predictive variables for 2003;14:e8.
the successful treatment of trigeminal neuralgia with Gamma Knife 83. Levivier M, Massager N, Wikler D, et al. Use of stereotactic PET
radiosurgery. Neurosurgery. 2012;70:566-572, discussion 572-573. images in dosimetry planning of radiosurgery for brain tumors:
62. Massager N, Nissim O, Murata N, et al. Effect of beam channel clinical experience and proposed classification. J Nucl Med. 2004;45:
plugging on the outcome of Gamma Knife radiosurgery for tri- 1146-1154.
geminal neuralgia. Int J Radiat Oncol Biol Phys. 2006;65:1200-1205. 84. Levivier M, Wikler D Jr, Massager N, et al. The integration of
63. Kondziolka D, Lacomis D, Niranjan A, et al. Histological effects of metabolic imaging in stereotactic procedures including radiosur-
trigeminal nerve radiosurgery in a primate model: implications for gery: a review. J Neurosurg. 2002;97(suppl):542-550.
trigeminal neuralgia radiosurgery. Neurosurgery. 2000;46:971-976, 85. Lorenzoni J, Devriendt D, Massager N, et al. Radiosurgery for
discussion 976-977. treatment of brain metastases: estimation of patient eligibility using
64. Kondziolka D, Zorro O, Lobato-Polo J, et al. Gamma Knife stereo- three stratification systems. Int J Radiat Oncol Biol Phys. 2004;60:
tactic radiosurgery for idiopathic trigeminal neuralgia. J Neurosurg. 218-224.
2010;112:758-765. 86. Lorenzoni JG, Massager N, David P, et al. Neurovascular compres-
65. Alberico RA, Fenstermaker RA, Lobel J. Focal enhancement of sion anatomy and pain outcome in patients with classic trigeminal
cranial nerve V after radiosurgery with the Leksell Gamma Knife: neuralgia treated by radiosurgery. Neurosurgery. 2008;62:368-375,
experience in 15 patients with medically refractory trigeminal neu- discussion 375-376.
ralgia. AJNR Am J Neuroradiol. 2001;22:1944-1948. 87. Massager N, Lorenzoni J, Devriendt D, et al. Radiosurgery for
66. Massager N, Abeloos L, Devriendt D, et al. Clinical evaluation of trigeminal neuralgia. Prog Neurol Surg. 2007;20:235-243.
targeting accuracy of Gamma Knife radiosurgery in trigeminal neu- 88. Herman JM, Petit JH, Amin P, et al. Repeat Gamma Knife radio-
ralgia. Int J Radiat Oncol Biol Phys. 2007;69:1514-1520. surgery for refractory or recurrent trigeminal neuralgia: treatment
67. McNatt SA, Yu C, Giannotta SL, et al. Gamma Knife radiosurgery outcomes and quality-of-life assessment. Int J Radiat Oncol Biol Phys.
for trigeminal neuralgia. Neurosurgery. 2005;56:1295-1301, discus- 2004;59:112-116.
sion 1301-1303. 89. Park KJ, Kondziolka D, Berkowitz O, et al. Repeat Gamma Knife
68. Villavicencio AT, Lim M, Burneikiene S, et al. Cyberknife radiosur- radiosurgery for trigeminal neuralgia. Neurosurgery. 2012;70:295-
gery for trigeminal neuralgia treatment: a preliminary multicenter 305, discussion 305.
experience. Neurosurgery. 2008;62:647-655, discussion 647-655. 90. Pollock BE, Foote RL, Link MJ, et al. Repeat radiosurgery for
69. Fountas KN, Smith JR, Lee GP, et al. Gamma Knife stereotactic idiopathic trigeminal neuralgia. Int J Radiat Oncol Biol Phys. 2005;
radiosurgical treatment of idiopathic trigeminal neuralgia: long- 61:192-195.
term outcome and complications. Neurosurg Focus. 2007;23:E8. 91. Gellner V, Kurschel S, Kreil W, et al. Recurrent trigeminal neural-
70. Pusztaszeri M, Villemure JG, Regli L, et al. Radiosurgery for tri- gia: long term outcome of repeat Gamma Knife radiosurgery.
geminal neuralgia using a linear accelerator with BrainLab system: J Neurol Neurosurg Psychiatry. 2008;79:1405-1407.
report on initial experience in Lausanne, Switzerland. Swiss Med 92. Aubuchon AC, Chan MD, Lovato JF, et al. Repeat Gamma Knife
Wkly. 2007;137:682-686. radiosurgery for trigeminal neuralgia. Int J Radiat Oncol Biol Phys.
71. Adler JR Jr, Bower R, Gupta G, et al. Nonisocentric radiosurgical 2011;81:1059-1065.
rhizotomy for trigeminal neuralgia. Neurosurgery. 2009;64(suppl): 93. Kimball BY, Sorenson JM, Cunningham D. Repeat Gamma Knife
A84-A90. surgery for trigeminal neuralgia: long-term results. J Neurosurg.
72. Smith ZA, Gorgulho AA, Bezrukiy N, et al. Dedicated linear accel- 2010;113(suppl):178-183.
erator radiosurgery for trigeminal neuralgia: a single-center experi- 94. Hasegawa T, Kondziolka D, Spiro R, et al. Repeat radiosurgery for
ence in 179 patients with varied dose prescriptions and treatment refractory trigeminal neuralgia. Neurosurgery. 2002;50:494-500, dis-
plans. Int J Radiat Oncol Biol Phys. 2011;81:225-231. cussion 500-502.
73. Chen JC, Rahimian J, Rahimian R, et al. Frameless image-guided 95. Dvorak T, Finn A, Price LL, et al. Retreatment of trigeminal neu-
radiosurgery for initial treatment of typical trigeminal neuralgia. ralgia with Gamma Knife radiosurgery: is there an appropriate
World Neurosurg. 2010;74:538-543. cumulative dose? Clinical article. J Neurosurg. 2009;111:359-364.
CHAPTER 173  Stereotactic Radiosurgery for Trigeminal Neuralgia 1419.e3

96. Lim YJ, Leem W, Park JT, et al. Cerebral infarction with ICA occlu- 99. Pollock BE, Foote RL, Stafford SL. Stereotactic radiosurgery: the
sion after Gamma Knife radiosurgery for pituitary adenoma: a case preferred management for patients with nonvestibular schwanno- 173
report. Stereotact Funct Neurosurg. 1999;72(suppl 1):132-139. mas? Int J Radiat Oncol Biol Phys. 2002;52:1002-1007.
97. Ganz JC. Gamma knife radiosurgery and its possible relationship 100. Pollock BE, Schoeberl KA. Prospective comparison of posterior
to malignancy: a review. J Neurosurg. 2002;97(suppl):644-652. fossa exploration and stereotactic radiosurgery dorsal root entry
98. Olson S, Atkinson L, Weidmann M. Microvascular decompression zone target as primary surgery for patients with idiopathic trigemi-
for trigeminal neuralgia: recurrences and complications. J Clin Neu- nal neuralgia. Neurosurgery. 2010;67:633-638, discussion 638-639.
rosci. 2005;12:787-789.
174 Trigeminal
Microvascular Decompression for
Neuralgia
Jonathan P. Miller and Kim J. Burchiel

Trigeminal neuralgia (TN) is a syndrome of neuropathic pain millimeters, there is a transition to peripheral myelin (produced
characterized by severe paroxysmal lancinating pain in one or by Schwann cells). The region of this transition is called the
more distributions of the trigeminal nerve. Vascular compression Obersteiner-Redlich zone. It is thought that the area of the nerve
of the trigeminal nerve at the point of entrance into the brainstem containing the central form of myelin is especially susceptible to
has been associated with this syndrome, and surgical separation pathologic changes from vascular contact that result in demyelin-
of the vessel from the nerve is helpful for many patients. This is ation and altered conduction. Pathologic studies from patients
usually done by a retrosigmoid craniotomy or craniectomy using with TN have demonstrated severe damage to myelin as well as
intraoperative microscopic or endoscopic assistance. axon loss within the nerve adjacent to the site of compression.11
The resulting conduction abnormality may lead to nerve hyper-
activity owing to ectopic impulse discharge, spontaneous and
HISTORY triggered after discharge, and cross-excitation among neighbor-
TN has been known since ancient times. Two thousand years ing afferent fibers (ephaptic transmission).11,12
ago, Aretaeus of Cappadocia referred to headache with “spasm Separation of the nerve from the offending vessel appears to
and distortion of countenance,” and Persian scholar Avicenna immediately reverse many of the physiologic changes. In Leandri
described a similar syndrome of facial pain in the 10th century.1 and colleagues’13 study of 10 patients undergoing MVD who
In the 17th century, Fehr described the syndrome in a eulogy.2 underwent nerve root and scalp electrode recordings, 7 showed
John Locke described facial pain of the Countess of Northum- signs of immediate improvement in neurophysiologic parameters
berland, wife of the English Ambassador to France, as a “fit of after decompression. Other investigators have reported improve-
such violent and exquisite torment that it forced her to … cries ment in sensory thresholds for touch, pinprick, and temperature
and shrieks … which extended itself all over the right side of her sensations after MVD14 as well as resolution of asymmetric jaw
face and mouth.”1 In 1756, Nicolas Andre coined the term tic motion.15,16 These findings suggest that the changes associated
douloureux, describing it as “a cruel and obscure illness, which with neurovascular compression are likely to be reversible if the
causes … in the face some violent motions, some hideous gri- nerve is decompressed, at least in the early stages.
maces which are an insurmountable obstacle to the reception of Radiographic and anatomic studies have demonstrated that
food, [and] which put off sleep.”3 The first comprehensive clinical vascular contact with the trigeminal nerve is common even in
description occurred in 1773 when John Fothergill wrote to the asymptomatic individuals but tends to be more severe and more
medical society of London about 14 patients with TN, and his proximal on the nerve ipsilateral to TN symptoms.17,18 Patients
descriptions of triggerable lancinating pain are still considered with TN are more likely to have contralateral arterial compres-
accurate.4 sion than asymptomatic people even though bilateral TN is
Early surgical treatments for TN involved intentional lesion- distinctly rare.17,19 Symptomatic and asymptomatic arterial com-
ing of the trigeminal nerve. In 1934, Walter Dandy described the pression of the trigeminal nerve increases with age because of
retrosigmoid approach to the trigeminal nerve and noted fre- elongation of cisternal arteries, explaining why it is primarily a
quent vascular contact in patients with TN. He wrote, “In many disease of older adults.17 However, trigeminal neuralgia can occur
instances the nerve is grooved or bent in an angle by the artery. in the absence of neurovascular compression; in one study of 184
This I believe is the cause of tic douloureux.”5 Despite his insight, patients with TN, 28.8% had no evidence of vascular compres-
Dandy did not attempt to decompress the nerve, believing the sion.20 Because most individuals with neurovascular compression
pathologic process to be irreversible. Microvascular decompres- do not have TN and many with TN do not have neurovascular
sion (MVD) was first performed by W. James Gardner in 1959, compression, it is probable that factors other than neurovascular
who described mobilizing a vessel from the trigeminal nerve and conflict are responsible for development of TN.
placing a piece of absorbable gelatin sponge (Gelfoam) between
them without any intentional damage to the nerve itself.6 The
procedure was subsequently refined and popularized by Peter
ALTERNATIVE TREATMENTS
Jannetta, who, with the aid of the operating microscope, per- TN symptoms often improve with medications that exert a sta-
formed thousands of MVD operations and demonstrated that bilizing effect on neural conduction, such as antiepileptics. Medi-
long-term relief of pain is possible in most patients with appro- cations that have been successfully used include carbamazepine,
priate selection.7-9 MVD has been the most common procedure phenytoin, valproate, gabapentin, pregabalin, baclofen, and clon-
for TN every year since 1992 and currently accounts for more azepam. Most patients obtain good pain control initially,21,22 but
than 90% of surgical procedures performed for neuropathic the effect tends to diminish over time, and after 10 years about
facial pain.10 half no longer have a response.23 Because the clinical and patho-
logic changes associated with TN may be progressive over time,
initial failure of pharmacologic therapy may represent an indica-
PATHOPHYSIOLOGY tion to proceed with more aggressive treatment.24 Nevertheless,
The etiology of TN is believed to be related to abnormal conduc- medical therapy is recommended as a first-line treatment for
tion within the trigeminal nerve possibly due to changes in patients with TN because some patients require no further
myelin induced by pulsatile mechanical trauma from an adjacent treatment.
vessel. At the point just before it enters the brainstem, there is Surgical treatments for TN other than MVD generally involve
a short segment where nerve axons are still ensheathed in intentional production of a lesion within the trigeminal nerve.
central myelin (produced by oligodendrocytes), but after a few Many commonly performed procedures consist of percutaneous
1420
CHAPTER 174  Microvascular Decompression for Trigeminal Neuralgia 1421

access to the nerve through a needle advanced through the face long-term pain control, patients with TN type 1 are more likely
followed by direct creation of a lesion using a radiofrequency to do well than those with TN type 2.34,35 Facial pain diagnoses 174
generator, glycerol injection, or balloon compression. Stereotac- other than TN are unlikely to improve after MVD. TN with a
tic radiosurgery targeting the nerve root entry zone is also effec- history of multiple sclerosis (MS) is called symptomatic trigeminal
tive, although pain relief is delayed by months, and complete neuralgia (STN). MS is present in 1% to 3% patients with TN,
elimination of pain may occur less frequently than with other and 2% to 4% of patients with MS have TN, probably because
methods. In general, the success of each of these lesioning tech- of intrinsic demyelination within the nerve or increased sensitiv-
niques requires some degree of facial numbness, with greater ity to vascular trauma.36,37 Although patients with MS sometimes
numbness associated with a higher rate of pain control but also improve after MVD, the recurrence rate is higher, and long-term
greater likelihood of complications such as facial dysesthesia and elimination of pain is rare,38-40 so destructive procedures may be
anesthesia dolorosa. Also, all these procedures are associated with more appropriate for these patients.41 Sensory loss with burning
a high recurrence rate after a few years as facial sensation returns, pain is a sign of trigeminal neuropathic pain (TNP); if it occurs
suggesting that they work primarily by blocking triggering after a previous destructive procedure, this is called trigeminal
impulses rather than by treating the cause of the pain itself. deafferentation pain (TDP). Allodynia and dysesthesia with a
MVD differs from the other treatments in that it treats the history of herpes zoster suggest postherpetic neuralgia (PHN).
primary cause of TN so that long-term pain relief is possible. MVD is not a good option for patients with any of these disor-
Also, because there is no intentional damage to the nerve, facial ders. Atypical facial pain (AFP), which refers to pain of psychologi-
dysesthesia and numbness are rare with MVD. Although it is the cal onset, requires neuropsychological testing for the diagnosis
most invasive and expensive treatment, MVD is associated with and is unlikely to improve after MVD.
the lowest rate of pain recurrence and the highest rate of patient Pain outside the trigeminal nerve distribution is not TN,
satisfaction among all surgical treatments for TN.25 MVD can although other vascular compression syndromes may be present
also be safely performed after a lesioning procedure and appears with throat pain (glossopharyngeal neuralgia) or pain deep in the
to be no less effective so long as there is no evidence of trigeminal ear (nervus intermedius neuralgia). Other causes of facial pain are
neuropathy.26 dental disease, orbital disease, sinusitis, cluster headache, tem-
poromandibular joint disease, temporal arteritis, and posttrau-
PATIENT SELECTION AND CLASSIFICATION matic neuralgias. None of these has the clinical characteristics of
TN, nor does any respond to MVD of the trigeminal nerve.
OF FACIAL PAIN
MVD is ideal for young healthy patients with TN because no
other treatment offers a significant likelihood of long-term pain
PREOPERATIVE IMAGING
relief.27 However, advanced age is not by itself a contraindication All patients should undergo magnetic resonance imaging (MRI)
because there is no difference in complication rate or outcome before MVD to verify that there is no intracranial mass lesion
in elderly patients.28-30 The operation is in fact technically easier that may be responsible for the pain, such as a tumor, cyst, aneu-
in older patients, in whom cerebellar atrophy leads to less need rysm, or arteriovenous malformation. MRI also can identify evi-
for retraction and less risk for cerebellar swelling. If life expec- dence of demyelinating disease or inflammatory changes and may
tancy is very short or general anesthesia cannot be tolerated, a reveal anatomic characteristics that would make MVD techni-
less invasive destructive procedure may be more appropriate.31,32 cally problematic, such as an ectopic basilar artery or bony abnor-
A careful history is essential during preoperative evaluation. malities. Atrophy of the affected trigeminal nerve is also frequently
Patients generally report intense stabbing or electric shock–like seen in patients with trigeminal neuralgia and is associated with
sensation, although there may be an overlying constant pain that good prognosis after MVD.42 High-resolution MRI using steady-
may be more severe than the stabbing pain. Pain in any distribu- state precession images can be used to identify neurovascular
tion within the trigeminal nerve innervation territory may be compression preoperatively with a high degree of accuracy, and
observed. Cranial nerve (CN) V2 and V3 branches of the trigemi- fusion of these images with those from magnetic resonance angi-
nal nerve are more common sources of pain,7 especially radiating ography followed by three-dimensional reconstruction can be
out from near the mouth. V1 symptoms are sometimes associated used to simulate the intraoperative view for surgical planning
with decreased corneal sensation. The pain is often worse during (Fig. 174-1).43
the day and may be positional, with relief with a supine position,
with the affected side up, or during sleep. Trigger points are
present in most patients and are activated by light cutaneous
OPERATIVE TECHNIQUE
stimuli such as wind, eating, talking, and shaving. Often, the trig- Standard neuroanesthetic techniques are used, with chemical
gers lead to guarding of the face and refusal to be touched, wash, paralysis and controlled ventilation to prevent motion in the field.
apply makeup, shave, or brush the teeth because of fear of an Use of diuresis or spinal drainage for brain relaxation is generally
attack. Pain-free intervals lasting weeks to months are common not necessary because release of cerebrospinal fluid (CSF) from
at first but become rare as the syndrome progresses. Initial onset the trigeminal cistern relaxes the cerebellum enough to allow
of pain is frequently quite memorable. Many patients undergo adequate retraction. Intraoperative monitoring is helpful to
dental procedures without relief before a diagnosis is made. If the prevent injury to the brainstem and cranial nerves. Brainstem
patient is given antiepileptic medication, pain usually improves auditory evoked potentials are very sensitive to stretch-induced
dramatically. Physical findings are usually normal, although about injury to the eighth cranial nerve, and a delay in comparison with
one third of patients have some degree of sensory loss in the baseline readings of more than 20% or a shift in interpeak latency
affected area. of more than 1.5 to 2 milliseconds requires loosening of cerebel-
In the evaluation of a patient for surgery, it is helpful to classify lar retraction until the signals normalize. Facial nerve monitoring
facial pain according to the classification scheme reported by may also be used but is less helpful.
Burchiel.33 Patients with TN type 1 have predominantly shock- A cranial fixation device such as the three-pin Mayfield head
like pain, whereas patients with TN type 2 report that at least 50% holder is applied before positioning. There are several options
is constant pain, although there still may be a component of for patient positioning for MVD. The simplest option is to place
lancinating pain. Pain relief after MVD is more strongly corre- the patient in a flat supine position with the head rotated and
lated with the lancinating pain component than with any other flexed to the side opposite the affected side (Video 174-1). Ideally,
symptom, so although most patients with either type have no shoulder roll should be used so that the ipsilateral shoulder
1422 SECTION 6  Pain

A B

C D
Figure 174-1. Preoperative magnetic resonance imaging to identify arterial compression of the
trigeminal nerve before microvascular decompression. Steady-state precession imaging demonstrates
contact of a vessel with the nerve (A), and magnetic resonance angiography reveals that the vessel is an
artery (B). Fusion and three-dimensional reconstruction of these sequences produce an image (C) that
corresponds well with intraoperative findings (D). (Modified from Miller J, Acar F, Hamilton B, et al.
Preoperative visualization of neurovascular anatomy in trigeminal neuralgia. J Neurosurg. 2008;108:477-482.)

does not obscure the operative approach. This position requires Monopolar electrocautery is used to dissect and clear muscle and
a flexible neck and may not work for obese or short-necked soft tissue until the bone of the mastoid eminence and digastric
patients. Another option is the lateral decubitus or three-quarter groove is seen. The occipital artery is often encountered in the
prone position with the shoulder taped caudally and the neck muscular tissue and sacrificed. A large mastoid emissary vein is
flexed, ensuring the chin is at least two fingerbreadths from the usually seen at this point, which represents an important land-
sternum. Regardless of the position chosen, it is generally best to mark because it overlies the junction of the transverse and
place the vertex parallel to the floor so that the seventh and eighth sigmoid sinuses. The bony exposure is extended to the curving
cranial nerves are inferior in relation to the trigeminal nerve, portion of the suboccipital bone at the floor of the posterior fossa.
simplifying the approach.44 All pressure points are padded, and The wound is held open with a self-retaining retractor or with
an axillary roll is used if necessary. The sitting position can also sutures. A dural graft is generally not necessary, but if desired, the
be used, although it is associated with complications such as air periosteum along the superior nuchal line may be harvested, or
embolism and subdural hematoma. fascia may be collected if a C-shaped incision was used.
Preoperative MRI is used to evaluate the anatomy of the Bone removal is performed with a high-speed drill or perfora-
sinuses, cerebellum, and any anomalous veins. A small posterior tor, starting at the mastoid emissary vein, which exits the bone
fossa or Chiari malformation may necessitate alteration of surgi- just inferior and posterior to the transverse sinus–sigmoid sinus
cal technique. The position of the transverse and sigmoid sinuses junction. If there is any doubt about the location of the sinuses,
may be estimated from bony landmarks. The transverse sinus it is always better to start more inferior and posterior to avoid
generally runs along a line connecting the inion to the external unintentional damage to the sinuses. A craniectomy is safe and
auditory meatus, parallel and posterior to the zygomatic arch. well tolerated if the defect is filled with artificial bone material at
The sigmoid sinus runs along the digastric groove posterior to the end of the procedure. Alternatively, some surgeons advocate
the mastoid eminence. Alternatively, frameless stereotaxy may be elevation of a craniotomy flap inferior and posterior to the initial
used to accurately define the position of the sinuses. Hair is bur hole that can be replaced after the dura is closed. With a drill
clipped from the surgical site, and the scalp is sterilized and then and a periosteal elevator to carefully elevate the dura, the crani-
infiltrated with local anesthetic with epinephrine to reduce bleed- ectomy is expanded until the junction of the transverse and
ing. The linear incision runs longitudinally two fingerbreadths sigmoid sinuses can be seen. The sigmoid sinus is especially sus-
behind the ear (5 mm behind the hairline) and extends 3 to ceptible to injury because of its thin outer wall and curved bony
5 cm, with one fourth of the incision above the iniomeatal line. groove. In older patients, the lateral wall of the sinus is adherent
CHAPTER 174  Microvascular Decompression for Trigeminal Neuralgia 1423

to the mastoid bone and may be damaged if not carefully sepa- been advocated as a method of very thorough evaluation of the
rated from the bone before the craniectomy. Small injuries to the nerve with minimal cerebellar retraction.45-48 Some advocate use 174
sinus are managed with oxidized cellulose or absorbable gelatin of a small mirror to visualize behind the nerve.
sponge and tack-up sutures, but large rents may require further The artery that most frequently produces compression is the
bony exposure with a dural patch graft because packing with a superior cerebellar artery (SCA), which often compresses the
large amount of hemostatic material may lead to venous obstruc- nerve anteromedially from within the axilla. Decompression
tion. The mastoid air cells are visible at the lateral margin of the requires elevation of the artery into a horizontal rather than verti-
bony opening and should be carefully waxed. Hemostasis is cal orientation, displacing it upward and away from the nerve.
important at this point to prevent entry of blood into the sub- SCA often divides into two branches as it courses around
arachnoid space. the midbrain, either or both of which may compress the nerve.
The dura is opened in an inverted L shape, parallel to and 3 The anterior inferior cerebellar artery (AICA) may compress the
to 5 mm from the transverse and sigmoid sinuses. A C- or nerve from below, requiring displacement more inferiorly away
T-shaped incision can be used as well, with T-shaped relaxing from the nerve. Sometimes both the SCA and AICA are involved,
incisions as needed, although it should be remembered that surrounding the nerve like a pincer, in which case both must be
watertight closure is desired at the end of the procedure. If neces- mobilized and decompressed. Less commonly, the vertebral or
sary, the dura next to the sigmoid sinus is tacked laterally to basilar artery contacts the nerve, usually in hypertensive, elderly,
slightly rotate the sigmoid sinus and expose the lateral surface of and male patients.7 Small arterioles may be seen, more often in
the cerebellum. CSF is then allowed to drain through gentle younger patients. Compression by a persistent trigeminal artery
retraction on the cerebellum with a cotton neurosurgical patty has also been described.49
(Cottonoid; Codman Neuro, Raynham, MA). Any adhesions or Any arterial loops producing compression are carefully dis-
bridging veins along the superior margin of the cerebellum along sected, with kinking of arteries avoided at all times. Shredded
the transverse sinus are coagulated and sharply divided. polytetrafluoroethylene (Teflon) is created by grasping and
At this point, the microscope is brought into the field, and the tearing a piece of the material to create a soft substance resem-
self-retaining retractor system is set up. A flexible self-retaining bling a cotton ball. This material is then placed between the
brain retractor is placed over a neurosurgical cotton patty to vessel and nerve in a proximal-to-distal manner; it is held in place
gently retract the cerebellum up and medially and slightly ele- by the tension between the artery and nerve root and reinforced
vated toward the surgeon. This retraction allows for penetration if necessary with absorbable gelatin sponge or fibrin glue. The
of the trigeminal cistern and further drainage of CSF, greatly vessel is flipped onto the dorsal aspect of the nerve with the
reducing the need for medial cerebellar retraction for exposure polytetrafluoroethylene separating the nerve from the vessel. If
of the trigeminal nerve. Superficial cerebellar veins draining into small vessels are pushed into the nerve by the polytetrafluoroeth-
the tentorial sinus and the superior petrosal vein are easy to tear ylene, it must be replaced. Multiple pieces of polytetrafluoroeth-
even with gentle retraction, so the cerebellum should first be ylene can be used to decompress multiple arteries or looping
retracted medially (as though approaching the seventh and eighth arteries affecting more than one side of the nerve. If the artery
cranial nerves), then inferiorly under direct vision once its lateral passes through the nerve, careful dissection can be used to sepa-
extent of the cerebellum. The angle of approach is changed to rate the vessel from the nerve, although there is a greater risk of
follow the petrous bone anteriorly, keeping the retractor blade sensory loss with any nerve manipulation.50 Small veins may be
close to the tentorium along the superior surface of the cerebel- coagulated, but arteries should always be preserved. The nerve
lum to avoid injury to the seventh and eighth cranial nerves. If must be explored along its entire length, because the root entry
bleeding is encountered during retraction, a dorsal bridging vein zone may extend many millimeters from the brainstem and even
may have been torn, requiring removal of the retractor and gentle very small vessels may cause compression.
depression of the cerebellum to identify and coagulate the vein. Venous contact is frequently seen, often concomitantly with
As the retractor is advanced, the petrosal vein complex is arterial compression. The vein may be anterior (transverse
encountered, often appearing as an inverted Y that drains into pontine vein), posterior (petrosal vein), or distal at the entrance
the superior petrosal sinus. This vein is usually two thirds of the to Meckel’s cave (trigeminal vein); frequently, compression is
way from the dura to the trigeminal nerve, but there is great caused by one of the many unnamed veins that run along the
variability: the vein may be completely absent or it may lie on brainstem.51,52 Posterior venous compression may be obscured by
the nerve itself, producing venous compression. The petrosal vein the ridge of the petrous bone. Compression from draining veins
is divided sharply after thorough coagulation. It is prudent not from a venous angioma or arteriovenous malformation is some-
to coagulate the nerve or to divide it all the way to the point of times observed. If venous compression is identified, the vein is
its entry into the bone, because it tends to retract with cautery carefully dissected from the nerve and coagulated with low-
and is difficult to control if it retracts into the petrous bone. Some voltage small bipolar forceps to prevent spread of current into
surgeons recommend preserving all or at least the superior part the adjacent nerve, after which the vein is coagulated and divided.
of the petrosal venous complex if the veins do not obstruct the It is uncommon to find no evidence of compression whatso-
nerve or are easily mobilizable, but their presence usually pre- ever, but if after thorough exploration no compression is found,
vents full exploration of the nerve, and they can nearly always be the nerve may be gently manipulated to produce a lesion or the
taken with impunity. nerve partially sectioned along the anteroinferior aspect. Gentle
The arachnoid membrane over the nerve is carefully removed compression of the nerve virtually always produces immediate
to allow complete inspection of the nerve. Thin, translucent pain relief, although relapse rates are high.53 Alternatively, the
membrane can be teased off, but thick, opaque bands must be patient may be treated with a subsequent destructive procedure
sharply dissected to avoid injury to the trigeminal nerve, the such as radiofrequency rhizolysis or stereotactic radiosurgery.
trochlear nerve (a thin, delicate structure in the arachnoid above After decompression, the vessels are carefully observed, and
the trigeminal nerve), or small vascular branches in the subarach- topical papaverine–soaked absorbable gelatin sponge is used if
noid space. Vascular compression is usually seen close to the there is any evidence of vasospasm. The operative field is liberally
brainstem, often anterior to the nerve. The site of compression irrigated, retractors are removed, and a Valsalva-like maneuver is
may vary according to symptoms, with symptoms closer to CN performed with the ventilator by the anesthesiologist to ensure
V1 associated with compression of more caudolateral portions of there is no bleeding. The dura is closed in a watertight manner
the nerve. The arachnoid membrane anterior to the nerve must with continuous or interrupted braided 4-0 sutures to prevent
be dissected to allow decompression. Endoscopic assistance has subsequent leakage of CSF. It is usually possible to close the dura
1424 SECTION 6  Pain

A B C

D E F
Figure 174-2. Right-sided retromastoid craniotomy. A, The patient is positioned supine with
the head turned 90 degrees toward the opposite shoulder, slightly flexed, and slightly bent
toward the floor. The Mayfield head holder apparatus is visible on the extreme upper right and
left of the figure, and the right ear has been taped forward (visible to the right of the incision).
The 5-cm incision extends just above the iniomeatal line. B, The bone is removed with high-
speed drill and rongeurs, starting at the emissary vein and extending anterior and superior to
the junction of the transverse and sigmoid sinuses. C, The dura is opened, and a neurosurgical
cotton patty is placed in the trigeminal cistern to facilitate drainage of cerebrospinal fluid. D, The
petrosal vein is visualized and divided after coagulation. E, After dissection of the arachnoid, the
right trigeminal nerve is seen to be compressed by a branch of the superior cerebellar artery.
G F, The artery is decompressed with polytetrafluoroethylene. G, The dura is closed, and the bony
defect is filled with artificial bone material; the wound is then closed in layers.

primarily, but if necessary, a graft of cadaveric, synthetic, or


autogenous tissue may be used. Fibrin dural sealant is helpful,
COMPLICATIONS
especially in reoperations. A piece of absorbable gelatin sponge Complications from MVD are rare when it is performed by
is placed over the dura, and a cranioplasty of wire mesh and experienced hands,7,27 and morbidity is lowest at high-volume
artificial bone material is fashioned, or the bone is replaced if a centers.56 Cranial nerve damage is rare, and such complications as
craniotomy was performed (Fig. 174-2). The fascia, subcutaneous facial dysesthesia and anesthesia dolorosa are much less common
tissue, and skin are closed in standard fashion with absorbable than with the lesioning procedures. Many patients have a tran-
sutures. sient conductive hearing loss due to tracking of fluid from the
mastoid bone into the middle ear that clears spontaneously within
a few weeks. Sensorineural hearing loss tends to be permanent
POSTOPERATIVE CARE but can be prevented by gentle retraction, careful technique, and
After the operation, the patient must be observed overnight in treatment of any AICA vasospasm that occurs. Brainstem audi-
the intensive care unit or stepdown unit.54 Blood pressure is tory evoked potential monitoring is very helpful in preventing
monitored with an arterial line, and an antihypertensive (labet- this complication and has diminished its incidence from 1.3% to
alol or hydralazine) is administered if systolic blood pressure 0.6%.44 Prolonged postoperative vertigo or tinnitus is reported in
exceeds 160 mm Hg. Mild narcotics may be used for headache, about 2% of patients,57 and some facial nerve palsy occurs in up to
which may persist for several weeks. Severe bifrontal headache 5% of patients.7 Both these complications tend to improve spon-
warrants computed tomography (CT) to rule out posterior fossa taneously over a few weeks. Injury to the fourth cranial nerve pro-
hemorrhage, but postoperative imaging is otherwise not neces- duces a trochlear palsy that usually subsides after a few months.
sary. Nausea is common and usually responds to antiemetics. CSF rhinorrhea, which occurs in 1.5% of cases,7 is caused by
Steroids are not helpful. The patient is transferred to the general leakage of CSF through the dural opening, into the mastoid air
medical/surgical floor on the first postoperative day, and diet and cells, and through the pharyngotympanic tube into the nasophar-
activity are gradually increased. Patients are generally discharged ynx. This complication occurs more often with reoperations and
by the third postoperative day. After discharge, activity is gradu- may be prevented by meticulous waxing of the mastoid process,
ally increased over a week, and most patients are able to return careful dural closure, and application of fibrin glue. CSF rhinor-
to work within 2 to 4 weeks. Half of all patients undergoing rhea is often successfully treated with 3 days of CSF drainage
MVD return to baseline activity within 1 month, and 90% by through a lumbar drain. If leakage continues, reexploration of the
3 months.55 wound and direct fistula closure may be needed.
CHAPTER 174  Microvascular Decompression for Trigeminal Neuralgia 1425

Aseptic meningitis with headache and sterile CSF pleocytosis likelihood of postoperative trigeminal neuropathic symptoms
may occur in response to artificial materials such as the polytet- (burning, aching pain).7 Some investigators have noted that 174
rafluoroethylene pledget used or todural graft material. It is lesioning procedures before an initial MVD may increase the
usually self-limited but often responds to steroids or lumbar likelihood of negative exploration during subsequent MVD oper-
puncture to remove CSF. Wound infection, which manifests as ations for recurrent TN.70
swelling of the wound, fever, and purulent drainage, requires Recurrent disease after a prolonged postoperative pain-free
reoperation with removal of all foreign materials and 4 to 6 weeks interval may be effectively treated with medication, but repeat
of intravenous antibiotics. MVD has also been advocated, even though it is associated with
Cerebellar contusion may be caused during the opening and a much higher rate of negative exploration. Reoperations are
initial retraction and is prevented by avoidance of aggressive technically more difficult, and if the first MVD was unsuccessful
cerebellar retraction, initial lateral retraction, and early drainage for technical reasons, it is unlikely that repeating the operation
of CSF. Cerebellar hemorrhagic infarction may occur from arte- will yield better results. When neurovascular compression is seen
rial damage but more often results from venous insufficiency, so during a repeat MVD, it is nearly always because of growth of
it is important to minimize the number of veins sacrificed. Pos- veins or expansion of arteries rather than dislodging of the
terior fossa hemorrhage occurs early in the postoperative course polytetrafluoroethylene ball placed during the first operation,
and usually requires immediate evacuation. although scarring produced by the ball can produce neural com-
pression. Reports of operative findings during repeat MVD vary
widely; most investigators report recurrent vascular compression
RESULTS in about half of patients.71,72 Outcome after the second operation
Multiple investigators have found MVD to be an effective treat- is generally good.62
ment for TN. In one study of 1204 patients, 75% had complete
relief and 9% had partial relief after 1 year. After 10 years, 64%
had complete relief, and 4% had partial relief. The annual rate
OTHER NEUROVASCULAR FACIAL PAIN SYNDROMES
of recurrence was less than 2% by 5 years and less than 1% by
10 years.7 Although initial results are similar, MVD offers a much
Glossopharyngeal Neuralgia
higher likelihood than destructive procedures for a long-term Glossopharyngeal neuralgia manifests as sharp severe pain in the
cure. In one study comparing 378 MVD recipients with 316 throat or neck, sometimes radiating to or from the upper neck
radiofrequency (RF) rhizotomy recipients over 20 years, patients or ear. It is much less common than TN and more likely to be
undergoing RF rhizotomy had a 75% chance of pain recurrence bilateral.73 Vagal involvement can lead to bradycardia, syncope,
in the first 5 years. By contrast, 63% of patients undergoing and even asystole. Like TN, it may be triggerable, and common
MVD were pain free at 20 years.57 triggers include cold beverages, yawning, chewing, coughing,
As mentioned previously, the most significant predictor of sneezing, and touching the tragus. Because the lower cranial
outcome after MVD appears to be type of TN pain. Patients with nerves exit the brainstem as a series of rootlets, attempted decom-
TN type 1 (mostly episodic, lancinating pain) tend to have much pression with polytetrafluoroethylene may lead only to worsen-
better immediate and long-term results than those with TN type ing of the compression. Instead, the nerve is exposed through a
2 (mostly constant pain). This effect appears to be proportional retrosigmoid craniectomy (as with MVD for TN), and the glos-
to the amount of lancinating pain, so that a higher amount of sopharyngeal nerve and the upper few fascicles of the vagus nerve
lancinating pain leads to a better outcome.58 TN type is more are sectioned. Patients should be warned that postoperative tem-
predictive of outcome than any other clinical feature of TN, porary dysphagia is often seen. Potential complications include
including duration of symptoms, presence of trigger points, permanently diminished gag reflex and vocal cord paralysis.
response to antiepileptics, and type of compression (arterial There are few long-term studies, but most reports indicate suc-
versus venous) found at surgery.58 In one large study, 80% of cessful long-term outcome in most patients.74,75 In one study,
patients with TN type 1 had some degree of pain relief at 5 years, 58% of patients had complete relief of symptoms, and another
compared with 51% of those with TN type 2.59 However, 18% had some relief, after a mean follow-up time of 4 years.54
although outcomes for patients with TN type 2 are less satisfac- Isolated throat pain appears to predict a greater likelihood of
tory than for those with TN type 1, MVD still benefits most postoperative success.
patients, and no alternative interventions appear to offer a better
chance of long-term relief. Geniculate Neuralgia (Nervus
Venous compression appears to predict worse outcome, pos-
sibly because of regrowth of veins. In one study of 393 patients Intermedius Neuralgia)
treated by MVD for venous compression, the 1-year recurrence The nervus intermedius is a small branch of the facial nerve that
rate was 31%, and 28 of 32 undergoing reexploration had evi- carries the sensory and parasympathetic fibers of the facial nerve.
dence of recurrent venous contact.51 Venous compression is also Within the cerebellopontine angle, the nervus intermedius runs
more common in pediatric patients with TN. In one study, between the motor component of the facial nerve and the ves-
venous contact was found in 86% of pediatric patients with TN tibulocochlear nerve branches before joining the facial nerve in
undergoing MVD, and a vein was the sole offending vessel in the internal acoustic meatus. Nervus intermedius neuralgia is
18%. These findings may explain why pediatric patients with TN associated with sharp shooting pain deep in the ear that may
have worse results after MVD, with 10-year recurrence rates as radiate to the temple or face. Parasympathetic facial nerve func-
high as 43%.60 Other predictors of recurrence include female tion (tearing) may be affected, but patients seldom complain of
gender, prolonged preoperative symptoms (>8 years), and failure this. This condition may be treated by cutting of the nerve. After
of immediate postoperative relief.7,61-67 Severity of compression exposure of the seventh and eighth nerves by a retrosigmoid
and focal arachnoiditis have also been associated with worse approach, the facial nerve is gently retracted to expose and mobi-
outcome.68 In general, the best results occur when TN is caused lize the nervus intermedius, which is then cut.
by severe compression by a single artery.69
It is unclear whether MVD for recurrent TN after a previous
destructive procedure is associated with a worse prognosis. The
CONCLUSION
likelihood of pain relief appears to be similar,26 but patients who TN is associated with and is likely to be caused by pulsatile
have undergone a lesioning procedure before MVD have a higher mechanical compression of the trigeminal nerve by a blood vessel
1426 SECTION 6  Pain

near the dorsal root entry zone. MVD differs from other treat- Kalkanis SN, Eskandar EN, Carter BS, et al. Microvascular decompres-
ments because it fixes the underlying cause to produce long-term sion surgery in the United States, 1996 to 2000: mortality rates,
relief of symptoms. Careful patient selection is the most impor- morbidity rates, and the effects of hospital and surgeon volumes. Neu-
tant determinant of outcome, and morbidity is rare when the rosurgery. 2003;52:1251-1261.
Lee SH, Levy EI, Scarrow AM, et al. Recurrent trigeminal neuralgia
procedure is performed by an experienced surgeon. attributable to veins after microvascular decompression. Neurosurgery.
2000;46:356-361.
SUGGESTED READINGS Liao JJ, Cheng WC, Chang CN, et al. Reoperation for recurrent trigemi-
Anderson VC, Berryhill PC, Sandquist MA, et al. High-resolution three- nal neuralgia after microvascular decompression. Surg Neurol. 1997;
dimensional magnetic resonance angiography and three-dimensional 47:562-568.
spoiled gradient-recalled imaging in the evaluation of neurovascular Matsushima T, Huynh-Le P, Miyazono M. Trigeminal neuralgia caused
compression in patients with trigeminal neuralgia: a double-blind pilot by venous compression. Neurosurgery. 2004;55:334-337.
study. Neurosurgery. 2006;58:666-673. McLaughlin MR, Jannetta PJ, Clyde BL, et al. Microvascular decompres-
Apfelbaum RI. Neurovascular decompression: the procedure of choice? sion of cranial nerves: lessons learned after 4400 operations [see
Clin Neurosurg. 2000;46:473-498. comment]. J Neurosurg. 1999;90:1-8.
Ashkan K, Marsh H. Microvascular decompression for trigeminal neu- Miller J, Acar F, Hamilton B, et al. Preoperative visualization of neu-
ralgia in the elderly: a review of the safety and efficacy [see comment]. rovascular anatomy in trigeminal neuralgia. J Neurosurg. 2008;108:
Neurosurgery. 2004;55:840-848. 477-482.
Barker FG 2nd, Jannetta PJ, Bissonette DJ, et al. The long-term outcome Ogungbo BI, Kelly P, Kane PJ, et al. Microvascular decompression for
of microvascular decompression for trigeminal neuralgia [see trigeminal neuralgia: report of outcome in patients over 65 years of
comment]. N Engl J Med. 1996;334:1077-1083. age [see comment] [erratum appears in Br J Neurosurg 2000;14:504].
Bonicalzi V, Canavero S. Role of microvascular decompression in trigemi- Br J Neurosurg. 2000;14:23-27.
nal neuralgia [comment]. Lancet. 2000;355:928-929. Rath SA, Klein HJ, Richter HP. Findings and long-term results of sub-
Broggi G, Ferroli P, Franzini A, et al. Microvascular decompression for sequent operations after failed microvascular decompression for tri-
trigeminal neuralgia: comments on a series of 250 cases, including 10 geminal neuralgia. Neurosurgery. 1996;39:933-938.
patients with multiple sclerosis. J Neurol Neurosurg Psychiatry. 2000;68: Sindou M, Leston J, Decullier E, et al. Microvascular decompression for
59-64. primary trigeminal neuralgia: long-term effectiveness and prognostic
Broggi G, Ferroli P, Franzini A, et al. Operative findings and outcomes factors in a series of 362 consecutive patients with clear-cut neurovas-
of microvascular decompression for trigeminal neuralgia in 35 patients cular conflicts who underwent pure decompression. J Neurosurg. 2007;
affected by multiple sclerosis. Neurosurgery. 2004;55:830-838. 107:1144-1153.
Burchiel KJ. A new classification for facial pain. Neurosurgery. 2003;53: Tatli M, Satici O, Kanpolat Y, et al. Various surgical modalities for tri-
1164-1166. geminal neuralgia: literature study of respective long-term outcomes.
Burchiel KJ, Clarke H, Haglund M, et al. Long-term efficacy of micro- Acta Neurochir (Wien). 2008;150:243-255.
vascular decompression in trigeminal neuralgia. J Neurosurg. 1988;69: Tronnier VM, Rasche D, Hamer J, et al. Treatment of idiopathic trigemi-
35-38. nal neuralgia: comparison of long-term outcome after radiofrequency
Burchiel KJ, Slavin KV. On the natural history of trigeminal neuralgia. rhizotomy and microvascular decompression. Neurosurgery. 2001;48:
Neurosurgery. 2000;46:152-154. 1261-1267.
Cho DY, Chang CG, Wang YC, et al. Repeat operations in failed micro- Tyler-Kabara EC, Kassam AB, Horowitz MH, et al. Predictors of outcome
vascular decompression for trigeminal neuralgia. Neurosurgery. 1994; in surgically managed patients with typical and atypical trigeminal
35:665-669. neuralgia: comparison of results following microvascular decompres-
Devor M, Govrin-Lippmann R, Rappaport ZH. Mechanism of trigeminal sion [see comment]. J Neurosurg. 2002;96:527-531.
neuralgia: an ultrastructural analysis of trigeminal root specimens
obtained during microvascular decompression surgery. J Neurosurg.
2002;96:532-543.
El-Garem HF, Badr-El-Dine M, Talaat AM, et al. Endoscopy as a tool in
minimally invasive trigeminal neuralgia surgery. Otol Neurotol. 2002;23:
132-135. See a full reference list on ExpertConsult.com
CHAPTER 174  Microvascular Decompression for Trigeminal Neuralgia 1426.e1

REFERENCES 27. Apfelbaum RI. Neurovascular decompression: the procedure of


1. Stookey B, Ransohoff J. Trigeminal Neuralgia: Its History and Treat- choice? Clin Neurosurg. 2000;46:473. 174
ment. Springfield, IL: Charles C. Thomas; 1959. 28. Ashkan K, Marsh H. Microvascular decompression for trigeminal
2. Schmidt J. Medical Discoveries. Vol. 503. Springfield, IL: Charles C. neuralgia in the elderly: a review of the safety and efficacy [see
Thomas; 1959. comment]. Neurosurgery. 2004;55:840.
3. Brown JA, Coursaget C, Preul MC, et al. Mercury water and cauter- 29. Jodicke A, Winking M, Deinsberger W, et al. Microvascular decom-
izing stones: Nicolas Andre and tic douloureux. J Neurosurg. 1999; pression as treatment of trigeminal neuralgia in the elderly patient.
90:977. Minim Invasive Neurosurg. 1999;42:92.
4. Fothergill J. Of a painful affection of the face. Medical Observation and 30. Ogungbo BI, Kelly P, Kane PJ, et al. Microvascular decompression
Inquiries by a Society of Physicians in London. London: Society of Physi- for trigeminal neuralgia: report of outcome in patients over 65 years
cians; 1776:129. of age [see comment] [erratum appears in Br J Neurosurg. 2000
5. Dandy W. Concerning the cause of trigeminal neuralgia. Am J Surg. Oct;14(5):504]. Br J Neurosurg. 2000;14:23.
1934;24:447. 31. Mullan S, Lichtor T. Percutaneous microcompression of the trigemi-
6. Gardner WJ. Concerning the mechanism of trigeminal neuralgia and nal ganglion for trigeminal neuralgia. J Neurosurg. 1983;59:1007.
hemifacial spasm. J Neurosurg. 1962;19:947. 32. Sweet WH, Poletti CE. Problems with retrogasserian glycerol in the
7. Barker FG 2nd, Jannetta PJ, Bissonette DJ, et al. The long-term treatment of trigeminal neuralgia. Appl Neurophysiol. 1985;48:252.
outcome of microvascular decompression for trigeminal neuralgia 33. Burchiel KJ. A new classification for facial pain. Neurosurgery.
[see comment]. N Engl J Med. 1996;334:1077. 2003;53:1164.
8. Jannetta PJ. Arterial compression of the trigeminal nerve at the pons 34. Miller JP, Acar F, Burchiel KJ. Classification of trigeminal neuralgia:
in patients with trigeminal neuralgia. J Neurosurg. 1967;(suppl 26): clinical, therapeutic, and prognostic implications in a series of 144
159. patients undergoing microvascular decompression. J Neurosurg.
9. Jannetta PJ, Rand RW. Transtentorial retrogasserian rhizotomy 2009;111(6):1231.
in trigeminal neuralgia by microneurosurgical technique. Bull Los 35. Zhang H, Lei D, You C, et al. The long-term outcome predictors of
Angeles Neurol Soc. 1966;31:93. pure microvascular decompression for primary trigeminal neuralgia.
10. Wang DD, Ouyang D, Englot DJ, et al. Trends in surgical treatment World Neurosurg. 2013;79(5-6):756.
for trigeminal neuralgia in the United States of America from 1988 36. Jensen TS, Rasmussen P, Reske-Nielsen E. Association of trigeminal
to 2008. J Clin Neurosci. 2013;20(11):1538. neuralgia with multiple sclerosis: clinical and pathological features.
11. Devor M, Govrin-Lippmann R, Rappaport ZH. Mechanism of tri- Acta Neurol Scand. 1982;65:182.
geminal neuralgia: an ultrastructural analysis of trigeminal root 37. Love S, Gradidge T, Coakham HB. Trigeminal neuralgia due to
specimens obtained during microvascular decompression surgery. multiple sclerosis: ultrastructural findings in trigeminal rhizotomy
J Neurosurg. 2002;96:532. specimens. Neuropathol Appl Neurobiol. 2001;27:238.
12. Burchiel KJ. Abnormal impulse generation in focally demyelinated 38. Broggi G, Ferroli P, Franzini A, et al. Operative findings and out-
trigeminal roots. J Neurosurg. 1980;53:674. comes of microvascular decompression for trigeminal neuralgia in 35
13. Leandri M, Eldridge P, Miles J. Recovery of nerve conduction fol- patients affected by multiple sclerosis. Neurosurgery. 2004;55:830.
lowing microvascular decompression for trigeminal neuralgia. Neu- 39. Eldridge PR, Sinha AK, Javadpour M, et al. Microvascular decom-
rology. 1998;51:1641. pression for trigeminal neuralgia in patients with multiple sclerosis.
14. Miles JB, Eldridge PR, Haggett CE, et al. Sensory effects of micro- Stereotact Funct Neurosurg. 2003;81:57.
vascular decompression in trigeminal neuralgia. J Neurosurg. 1997; 40. Ariai MS, Mallory GW, Pollock BE. Outcomes after microvascular
86:193. decompression for patients with trigeminal neuralgia and suspected
15. Jannetta PJ. Treatment of trigeminal neuralgia by suboccipital and multiple sclerosis. World Neurosurg. 2014;81(3-4):599.
transtentorial cranial operations. Clin Neurosurg. 1977;24:538. 41. Bonicalzi V, Canavero S. Role of microvascular decompression in
16. Saunders RL, Krout R, Sachs E Jr. Masticator electromyography in trigeminal neuralgia [comment]. Lancet. 2000;355:928.
trigeminal neuralgia. Neurology. 1971;21:1221. 42. Leal PR, Barbier C, Hermier M, et al. Atrophic changes in the tri-
17. Miller JP, Acar F, Burchiel KJ. Radiographic evaluation of trigeminal geminal nerves of patients with trigeminal neuralgia due to neuro-
neurovascular compression in patients with and without trigeminal vascular compression and their association with the severity of
neuralgia. J Neurosurg. 2009;110:627. compression and clinical outcomes. J Neurosurg. 2014;120(6):1484.
18. Antonini G, Di Pasquale A, Cruccu G, et al. Magnetic resonance 43. Miller J, Acar F, Hamilton B, et al. Preoperative visualization of
imaging contribution for diagnosing symptomatic neurovascular neurovascular anatomy in trigeminal neuralgia. J Neurosurg. 2008;
contact in classical trigeminal neuralgia: a blinded case-control study 108:477.
and meta-analysis. Pain. 2014;155(8):1464. 44. McLaughlin MR, Jannetta PJ, Clyde BL, et al. Microvascular decom-
19. Anderson VC, Berryhill PC, Sandquist MA, et al. High-resolution pression of cranial nerves: lessons learned after 4400 operations [see
three-dimensional magnetic resonance angiography and three- comment]. J Neurosurg. 1999;90:1.
dimensional spoiled gradient-recalled imaging in the evaluation of 45. Abdeen K, Kato Y, Kiya N, et al. Neuroendoscopy in microvascular
neurovascular compression in patients with trigeminal neuralgia: a decompression for trigeminal neuralgia and hemifacial spasm: tech-
double-blind pilot study. Neurosurgery. 2006;58:666. nical note. Neurol Res. 2000;22:522.
20. Lee A, McCartney S, Burbidge C, et al. Trigeminal neuralgia occurs 46. El-Garem HF, Badr-El-Dine M, Talaat AM, et al. Endoscopy as a
and recurs in the absence of neurovascular compression. J Neurosurg. tool in minimally invasive trigeminal neuralgia surgery. Otol Neurotol.
2014;120(5):1048. 2002;23:132.
21. Campbell FG, Graham JG, Zilkha KJ. Clinical trial of carbazepine 47. Jarrahy R, Berci G, Shahinian HK. Endoscope-assisted microvascular
(Tegretol) in trigeminal neuralgia. J Neurol Neurosurg Psychiatry. decompression of the trigeminal nerve. Otolaryngol Head Neck Surg.
1966;29:265. 2000;123:218.
22. Rockliff BW, Davis EH. Controlled sequential trials of carbamaze- 48. Rak R, Sekhar LN, Stimac D, et al. Endoscope-assisted microsurgery
pine in trigeminal neuralgia. Arch Neurol. 1966;15:129. for microvascular compression syndromes. Neurosurgery. 2004;54:876.
23. Taylor JC, Brauer S, Espir ML. Long-term treatment of trigeminal 49. Tamura Y, Shimano H, Kuroiwa T, et al. Trigeminal neuralgia associ-
neuralgia with carbamazepine. Postgrad Med J. 1981;57:16. ated with a primitive trigeminal artery variant: case report [see
24. Burchiel KJ, Slavin KV. On the natural history of trigeminal neural- comment]. Neurosurgery. 2003;52:1217.
gia. Neurosurgery. 2000;46:152. 50. Zheng X, Feng B, Hong W, et al. Management of intraneural vessels
25. Tatli M, Satici O, Kanpolat Y, et al. Various surgical modalities for during microvascular decompression surgery for trigeminal neural-
trigeminal neuralgia: literature study of respective long-term out- gia. World Neurosurg. 2012;77(5-6):77.
comes. Acta Neurochir. 2008;150:243. 51. Lee SH, Levy EI, Scarrow AM, et al. Recurrent trigeminal neuralgia
26. Shetter AG, Zabramski JM, Speiser BL. Microvascular decompres- attributable to veins after microvascular decompression. Neurosur-
sion after gamma knife surgery for trigeminal neuralgia: intraopera- gery. 2000;46:356.
tive findings and treatment outcomes. J Neurosurg. 2005;102(suppl): 52. Matsushima T, Huynh-Le P, Miyazono M. Trigeminal neuralgia
259. caused by venous compression. Neurosurgery. 2004;55:334.
1426.e2 SECTION 6  Pain

53. Revuelta-Gutierrez R, Martinez-Anda JJ, Coll JB, et al. Efficacy and 64. Burchiel KJ, Clarke H, Haglund M, et al. Long-term efficacy of
safety of root compression of trigeminal nerve for trigeminal neural- microvascular decompression in trigeminal neuralgia. J Neurosurg.
gia without evidence of vascular compression. World Neurosurg. 1988;69:35.
2013;80(3-4):385. 65. Kolluri S, Heros RC. Microvascular decompression for trigeminal
54. Romansky K, Stoianchev N, Dinev E, et al. Results of treatment of neuralgia. A five-year follow-up study. Surg Neurol. 1984;22:235.
trigeminal neuralgia by microvascular decompression of the Vth 66. Puca A, Meglio M, Cioni B, et al. Microvascular decompression for
nerve at its root entry zone. Arch Physiol Biochem. 1998;106:392. trigeminal neuralgia: prognostic factors. Acta Neurochir Suppl. 1993;
55. Lovely TJ, Lowry DW, Jannetta PJ. Functional outcome and the 58:165.
effect of cranioplasty after retromastoid craniectomy for microvas- 67. Sun T, Saito S, Nakai O, et al. Long-term results of microvascular
cular decompression. Surg Neurol. 1999;51:191. decompression for trigeminal neuralgia with reference to probability
56. Kalkanis SN, Eskandar EN, Carter BS, et al. Microvascular decom- of recurrence. Acta Neurochir. 1994;126:144.
pression surgery in the United States, 1996 to 2000: mortality rates, 68. Sindou M, Leston J, Decullier E, et al. Microvascular decompression
morbidity rates, and the effects of hospital and surgeon volumes. for primary trigeminal neuralgia: long-term effectiveness and prog-
Neurosurgery. 2003;52:1251. nostic factors in a series of 362 consecutive patients with clear-cut
57. Tronnier VM, Rasche D, Hamer J, et al. Treatment of idiopathic neurovascular conflicts who underwent pure decompression. J Neu-
trigeminal neuralgia: comparison of long-term outcome after radio- rosurg. 2007;107:1144.
frequency rhizotomy and microvascular decompression. Neurosur- 69. Jo KW, Kong DS, Hong KS, et al. Long-term prognostic factors for
gery. 2001;48:1261. microvascular decompression for trigeminal neuralgia. J Clin Neuro-
58. Miller JP, Magill ST, Acar F, et al. Predictors of long-term success sci. 2013;20(3):440.
after microvascular decompression for trigeminal neuralgia. J Neu- 70. Rath SA, Klein HJ, Richter HP. Findings and long-term results of
rosurg. 2009;110:620. subsequent operations after failed microvascular decompression for
59. Tyler-Kabara EC, Kassam AB, Horowitz MH, et al. Predictors of trigeminal neuralgia. Neurosurgery. 1996;39:933.
outcome in surgically managed patients with typical and atypical 71. Cho DY, Chang CG, Wang YC, et al. Repeat operations in failed
trigeminal neuralgia: comparison of results following microvascular microvascular decompression for trigeminal neuralgia. Neurosurgery.
decompression [see comment]. J Neurosurg. 2002;96:527. 1994;35:665.
60. Resnick DK, Levy EI, Jannetta PJ. Microvascular decompression for 72. Liao JJ, Cheng WC, Chang CN, et al. Reoperation for recurrent
pediatric onset trigeminal neuralgia. Neurosurgery. 1998;43:804. trigeminal neuralgia after microvascular decompression. Surg Neurol.
61. Barba D, Alksne JF. Success of microvascular decompression with and 1997;47:562.
without prior surgical therapy for trigeminal neuralgia. J Neurosurg. 73. Patel NK, Aquilina K, Clarke Y, et al. How accurate is magnetic
1984;60:104. resonance angiography in predicting neurovascular compression in
62. Bederson JB, Wilson CB. Evaluation of microvascular decompres- patients with trigeminal neuralgia? A prospective, single-blinded
sion and partial sensory rhizotomy in 252 cases of trigeminal neural- comparative study. Br J Neurosurg. 2003;17:60.
gia. J Neurosurg. 1989;71:359. 74. Kondo A. Follow-up results of using microvascular decompression
63. Broggi G, Ferroli P, Franzini A, et al. Microvascular decompression for treatment of glossopharyngeal neuralgia. J Neurosurg. 1998;
for trigeminal neuralgia: comments on a series of 250 cases, including 88:221.
10 patients with multiple sclerosis. J Neurol Neurosurg Psychiatry. 75. Resnick DK, Jannetta PJ, Bissonnette D, et al. Microvascular decom-
2000;68:59. pression for glossopharyngeal neuralgia. Neurosurgery. 1995;36:64.
PART 4 Surgical Procedures for Nontrigeminal Pain 175

175 Neurosurgical Management of Intractable Pain


William S. Rosenberg, Richard B. North, and Kenneth A. Follett

Neurosurgeons have a long history of accomplishments in the analgesics, and passive physical therapy.2 In contrast, chronic pain
field of pain management, and neurosurgery, as a specialty, holds does not serve a useful physiologic purpose (unless tissue injury
an important position within this discipline. The development of is ongoing). In some cases, chronic pain is no longer a symptom
neuroablative pain therapies, which were the mainstay of the of disease, but instead is a disease itself.3 Physical deconditioning
surgical treatment of intractable pain for many years, was facili- also commonly accompanies a number of chronic pain disorders,
tated by neurosurgeons’ understanding of the physiology and such as failed back surgery syndrome (FBSS). Thus, many patients
anatomy of nociception and their ability to access the peripheral with chronic pain require physical reactivation and rehabilitation,
and central nervous systems surgically. During the past two to rather than the rest and relaxation recommended for the treat-
three decades, the treatment of intractable pain has undergone ment of acute pain. This distinction between acute pain and
evolutionary change as neuromodulatory therapies, such as spinal chronic pain is critical, because treating chronic pain as acute pain
cord stimulation (SCS), peripheral nerve stimulation (PNS), and only promotes further disuse and deconditioning.2 The differen-
intrathecal drug delivery (IDD), have augmented neuroablative tiation between acute pain and chronic pain is also important
therapies. Neurosurgeons have had significant roles in guiding because psychological and social factors that can complicate a
this change. pain complaint are often more common in patients with chronic
Neurosurgeons are unique among health care providers in the pain than in those with acute pain. In fact, in some cases, psycho-
field of pain care by virtue of their training, expertise in neuro- social factors can perpetuate or even cause chronic pain. Thus,
logical differential diagnosis, and ability to provide patients the current clinical practice emphasizes the biopsychosocial model of
full range of neuromodulatory and neuroablative therapies as well chronic pain.4
as anatomic, reconstructive procedures to address the underlying The distinction between nociceptive pain and neuropathic
condition causing the pain, as appropriate. Neurosurgeons should pain is also important, because each type of pain usually responds
take advantage of this special position but must recognize that differently to specific treatments. Nociceptive pain occurs when
successful treatment of intractable pain requires more than surgi- tissue injury or disease (e.g., a broken arm or cancer with local
cal skill—it requires the ability to properly select specific treat- tissue invasion) initiates signals that are transferred through
ments for patients and manage them throughout the course peripheral nerves to the brain via the spinal cord. Such pain, which
of therapy. The neurosurgeon is an important and integral patients commonly describe as “throbbing,” “aching,” or “dull,”5
member of the pain care team, ideally establishing collaborative is a normal, protective response of the nociceptive systems. In
relationships with physicians who coordinate the long-term care contrast, neuropathic pain is the result of a pathologic process
of patients with complex pain problems. This multidisciplinary (injury or disease) affecting the peripheral or central nervous
interaction can help the neurosurgeon better understand how the system. Such neurological pathophysiology leads to abnormal
treatments provided fit within the context of the patient’s clinical neuronal excitability, spontaneous discharges, and ephaptic trans-
and psychosocial needs and promote good long-term outcomes. mission, which might, in turn, lead to generation of pain with or
without peripheral, let alone nociceptive, input. Thus, in contrast
to nociceptive pain, neuropathic pain reflects abnormal neuronal
PRELUDE TO SURGICAL TREATMENT activity. Neuropathic pain, which patients commonly describe as
The fundamental requirement for successful pain management, “burning,” “shooting,” “tingling,” or “shock-like,”5 can be con-
whether the treatment is surgical or nonsurgical, is an under- tinuous or paroxysmal (lancinating). Whereas nociceptive pain
standing of the nature of the pain being treated. Different types often responds to opioids (e.g., the pain of a broken arm can be
of pain respond differently to treatment; thus, the etiology and treated with morphine), neuropathic pain tends to resist opioid
characteristics of a patient’s pain must be understood in order for treatment (at least at typical doses) and frequently requires treat-
a rational treatment plan to be developed. In general, pain can be ment with non-opioid medications or other modalities.
classified as acute or chronic and nociceptive or neuropathic. Pain can also be classified according to its association with
Classification of a pain complaint into these simple categories cancer or lack thereof. This classification can be useful because
facilitates formulation of a proper treatment scheme. each of these types of pain (cancer vs. noncancer) might respond
Acute pain is a signal of actual or impending tissue injury and differently to specific interventions. In addition, the etiology of
is generated by activation of nociceptors in tissue that has sus- cancer-related pain can be very complex, with simultaneous noci-
tained an injury or insult. This type of pain resolves as injured ceptive and neuropathic components and causes ranging from
tissue heals. Chronic pain, on the other hand, outlasts the typical tumor expansion into soft tissue and/or nerves to iatrogenic
period required for healing of an acute injury. Some definitions (i.e., associated with surgery, chemotherapy, radiotherapy). In
of chronic pain are based on the duration of pain (e.g., pain that evaluating such a patient, the neurosurgeon must consider the
lasts longer than 3 or 6 months). This is an arbitrary distinction, entire spectrum of factors, including the continuation and objec-
however, because different types of acute injury require differing tives of ongoing oncologic treatment as well as the patient’s
lengths of time for healing, and the transition of acute pain to psychosocial support system, personal goals, and reasonable life
chronic pain can vary according to the nature of the injury.1 expectancy. It is important that these factors be considered in
The distinction between acute pain and chronic pain is some- addition to the source of the patient’s pain in the development of
times difficult to make but important. The goal of acute pain a treatment plan.
treatment should be to provide pain relief while promoting tissue Unless a correctible anatomic problem causing pain can be
healing. Such treatment might include rest or immobilization, diagnosed with confidence, surgical treatment of intractable pain
1427
1428 SECTION 6  Pain

is not usually a first-line approach. As a general principle, treat- potential benefit of subsequent surgery, SCS might be the safest
ment of intractable pain should follow a rational process that both and most appropriate treatment.15,16
considers the least disruptive therapy with a reasonable chance It is important to determine a likely organic basis for the pain,
of success and is sensitive to the patient who is in pain. Placing especially for patients with chronic pain of nonmalignant origin,
the needs of the patient at the center of decision making can help and to identify any comorbid psychologic dysfunction, which can
achieve the desired balance between avoiding unnecessary risk have a negative impact on outcomes for any treatment plan. Thus,
and exploring all options with significant probability of success, formal psychologic evaluation might be appropriate for many (or
all evaluated through the lens of the urgency, needs, and circum- most) patients being considered for surgical treatment of intrac-
stances of the patient. The overly dogmatic approach of formally table pain. Contraindications to surgical intervention include
exploring treatments with little chance of success for the sole overt psychologic dysfunction, such as active psychosis, suicidal
reason that they are “less invasive” is rarely in the best interest of or homicidal behavior, major uncontrolled depression or anxiety,
the patient. As Hippocrates said, “Timidity betrays a want of serious alcohol or drug abuse, and serious cognitive deficits.
powers, and audacity a want of skill.”6 The World Health Orga- Other psychologic problems that may be viewed as “risk factors”
nization proposed a simple, stepwise system for pain control include somatization disorder, personality disorders (e.g., border-
designed for ease of implementation.7 There is much to recom- line or antisocial), history of serious abuse, major issues of sec-
mend this approach, as each step in the ladder represents a more ondary gain, non-organic signs on physical examination, unusual
invasive approach with presumably more risk. Although it is pain ratings (e.g., 12 on a 10-point scale), inadequate social
useful, however, there is growing evidence that such a linear support, unrealistic outcome expectations, and, in the case of
system is less than optimal.8-12 implantable augmentative devices, an inability to understand the
Medical therapy, beginning typically with non-opioid agents purpose of a device or its use. Patients with psychologic risk
(e.g., nonsteroidal anti-inflammatory medications) in conjunc- factors are not necessarily precluded from surgical treatment, but
tion with adjuvant therapies when appropriate, should generally the treatment program should incorporate management of the
precede surgical intervention. If adequate pain control is not psychologic issues to facilitate a good outcome.17
obtained with non-opioid agents, mild opioids might be con-
sidered, to be replaced by strong opioids if appropriate. In most NEUROSURGICAL THERAPIES
cases, pain relief is most readily achieved with scheduled rather
than “as needed” (prn) analgesic dosing.13,14 Neuropathic pain FOR INTRACTABLE PAIN
frequently requires treatment with non-opioid medication, Neurosurgeons can use anatomic, neuromodulatory, and neu-
although opioids can be helpful for some patients. For continu- roablative therapies (Table 175-1).18 The treatment offered
ous neuropathic pain (e.g., constant burning, dysesthetic pain), should be tailored to meet the needs of each individual patient
useful adjuvant medications include antidepressants (e.g., tricy- and the skills of the treating physician. Specific interventions vary
clic antidepressants, serotonin-norepinephrine reuptake inhibi- in their appropriateness as treatment for pain in specific body
tors), clonidine, local anesthetics (e.g., mexiletine), and capsaicin regions (Boxes 175-1 through 175-4). Patient-related factors that
cream.14 Paroxysmal, lancinating, or evoked neuropathic pain must be taken into consideration in the selection of a therapy
might improve with anticonvulsants (e.g., carbamazepine, phe- include the etiology, distribution, and characteristics (nociceptive
nytoin, gabapentin) or baclofen.14 Nonpharmacologic adjuvant or neuropathic) of the pain; life expectancy; and psychological,
therapy, including psychologic support, relaxation therapies, social, and economic issues relevant to the pain complaint. The
coping strategies, passive physical therapy (e.g., massage, heat/ relative advantages and disadvantages of anatomic, augmentative,
cold), transcutaneous electrical nerve stimulation, and ortho- and ablative therapies should be weighed in view of these factors,
ses,13,14 can be useful in the treatment of nociceptive or neuro- and a choice among these three general approaches should be
pathic pain. made before a specific intervention is chosen. Selecting the right
Simple interventional therapies (e.g., nerve blocks, peripheral
nerve ablations) might be useful supplements to medical manage-
ment. Neuromodulatory therapies (e.g., SCS and neuraxial anal- TABLE 175-1  Neurosurgical Pain Therapies
gesic infusion) are typically the next step, followed by neuroablative Anatomic Correction of structural deformity
therapies if the former approaches are unsuccessful or inappro- Augmentative Stimulation:
priate. An element of flexibility should be maintained in the Peripheral nerve
approach to patients with pain, however, and treatment should Spinal cord
be tailored to meet individual needs. For example, dorsal root Deep brain structures
entry zone (DREZ) lesioning can relieve pain associated with Motor cortex
spinal nerve root avulsion and, in some cases, is preferable to Neuraxial/drug infusion:
Intrathecal/epidural
neuroaugmentative techniques. In addition, a patient with intrac-
Intraventricular
table pain related to late-stage cancer might be treated more Ablative Neurectomy
appropriately with cordotomy than with implantation of an intra- Sympathectomy
thecal drug infusion system. Ganglionectomy
Rhizotomy
PATIENT SELECTION FOR SURGICAL Spinal DREZ lesion
Cordotomy
PAIN THERAPIES Myelotomy
In most cases, surgical intervention is reserved for patients in Nucleus caudalis DREZ lesion
Trigeminal tractotomy
whom nonsurgical therapies do not provide adequate pain relief
Mesencephalotomy
or are associated with unacceptable side effects or risks, no abso- Thalamotomy
lute contradictions to surgery exist, and treatment of the underly- Cingulotomy
ing cause of pain is not possible, practical, or appropriate.5,15 For Hypophysectomy
example, radicular leg pain from lumbar spinal stenosis can be
DREZ, dorsal root entry zone.
treated with decompressive lumbar laminectomy; however, if a
Modified from North RB. Neurosurgical procedures for chronic pain:
patient also has severe coronary artery disease that increases general neurosurgical practice. Clin Neurosurg. 1992;40:182-196.
operative risk or has had prior spinal surgery that reduces the
CHAPTER 175  Neurosurgical Management of Intractable Pain 1429

BOX 175-1  Neurosurgical Procedures for Treatment of BOX 175-4  Neurosurgical Procedures for Treatment of 175
Head/Neck Pain Diffuse Pain
AUGMENTATIVE AUGMENTATIVE
Peripheral nerve stimulation Intraspinal/intraventricular analgesic administration
Intraventricular analgesic administration ABLATIVE
Deep brain/motor cortex stimulation
Thalamotomy
ABLATIVE Cingulotomy
Cranial/cervical rhizotomy/ganglionectomy Hypophysectomy
Caudalis dorsal root entry zone lesion
Trigeminal tractotomy
Mesencephalotomy
Medial thalamotomy techniques as procedures of choice for pain management, are
Cingulotomy often preferred as initial surgical treatments because of their rela-
tive safety and reversibility and the availability of a specific prog-
nostic trial.
Ablative therapies, however, have a role in the treatment of
certain pain syndromes19,20 and can target almost every level of
BOX 175-2  Neurosurgical Procedures for Treatment of Upper the peripheral and central nervous systems. Thus, ablative thera-
Trunk/Shoulder/Arm Pain pies can be directed at preventing transmission of nociceptive
information into the central nervous system at the level of periph-
AUGMENTATIVE eral nerves (neurectomy, neurotomy), roots (ganglionectomy,
Peripheral nerve stimulation rhizotomy), and the spinal cord dorsal horn (DREZ, including
Spinal cord stimulation nucleus caudalis DREZ). Ascending nociceptive pathways can
Intraspinal/intraventricular analgesic administration be disrupted at the level of the spinal cord or brainstem (cor-
Deep brain/motor cortex stimulation dotomy, myelotomy, tractotomy) or within the brain (thalamot-
ABLATIVE omy, cingulotomy).
Sympathectomy Augmentative and ablative therapies are reviewed briefly here
Neurectomy to provide a broad perspective about the applications of neuro-
Ganglionectomy/rhizotomy surgical pain therapies. A detailed discussion of indications for,
Spinal dorsal root entry zone lesion techniques of, and outcomes of many of these techniques is avail-
Mesencephalotomy able in other chapters. Outcomes of most interventional pain
Thalamotomy therapies have been studied mainly via case series. Few therapies
Cingulotomy have been studied in rigorous fashion, so data regarding efficacy
Hypophysectomy and complications should be interpreted cautiously.

Augmentative Therapies
Augmentative therapies offer the advantages of relative safety,
BOX 175-3  Neurosurgical Procedures for Treatment of Lower reversibility, and scalability. For example, intraspinal analgesic
Trunk/Leg Pain infusion can be adjusted to meet the changing needs of a patient
who has worsening cancer pain. On the other hand, augmentative
AUGMENTATIVE therapies are generally more expensive than ablative therapies
Peripheral nerve stimulation owing to initial device cost and upkeep, require maintenance
Spinal cord stimulation (e.g., refilling of infusion pumps, device replacement because of
Intraspinal/intraventricular analgesic administration battery depletion), and have the potential for device-related com-
Deep brain/motor cortex stimulation plications. Thus, cost-effective care mandates the consideration
ABLATIVE of such factors as life expectancy in the evaluation of the appro-
Sympathectomy priateness of certain neuroaugmentative therapies.
Neurectomy Stimulation therapies approved for pain management in the
Ganglionectomy/rhizotomy United States include SCS and PNS. In general, the highest yield
Spinal dorsal root entry zone lesion for SCS is for relatively focal (e.g., localized to one or two
Cordotomy extremities or focal on the trunk) and static neuropathic pain, but
Myelotomy individual responses are highly variable, and in general, a thera-
Thalamotomy peutic trial of SCS is relatively simple. Common applications
Cingulotomy include treatment of persistent radicular pain associated with
Hypophysectomy FBSS15,16,21-26 or neuropathic pain related to complex regional
pain syndrome (“reflex sympathetic dystrophy”).27-29 In patients
with FBSS, the success rate (defined typically as 50% or greater
reduction in pain) is approximately 60% at 5 years.15,22,23 Patients
treatment for the right patient at the right time increases the with complex regional pain syndromes have similar outcomes,
likelihood of a successful outcome. although success rates as high as 70% to 100% have been
Neuromodulatory therapies fall into the following two cate- reported.28,30 SCS also can be effective for neuropathic pain
gories: neurostimulation (SCS, PNS, deep brain stimulation affecting the trunk (e.g., postherpetic neuralgia and some types
[DBS], and motor cortex stimulation [MCS]) and targeted drug of postthoracotomy pain) and for extremity pain due to periph-
delivery (intrathecal and intraventricular). These neuromodula- eral neuropathy,31 root injury, phantom limb and postamputation
tion therapies, which have frequently supplanted neuroablative stump pain, and peripheral vascular disease.26,31,32 SCS is widely
1430 SECTION 6  Pain

used in Europe as a successful treatment for refractory angina of pain characteristics. Nociceptive pain and paroxysmal, lanci-
pectoris,33,34 but this indication does not have specific U.S. Food nating, or evoked neuropathic pain (e.g., allodynia, hyperpathia)
and Drug Administration (FDA) approval. tend to respond to PVG-PAG stimulation, which might acti-
SCS and PNS parameters in use since the 1960s, in particular vate endogenous opioid systems. Continuous neuropathic pain
“frequencies” (pulse repetition rates) in 10s to 100s per second responds most consistently to paresthesia-producing stimulation
(Hz), produce paresthesias, but “burst” protocols35 and higher of the sensory thalamus (nucleus ventrocaudalis).50 The affective
frequencies (up to 10 kHz)36 have been reported to relieve pain component of pain may respond to stimulation of the anterior
without producing paresthesias. Results reportedly have been cingulate gyrus.59 Because many pain syndromes (e.g., cancer
superior to those of conventional SCS, and different subgroups pain, FBSS) have components of both nociceptive pain and neu-
of patients (e.g., those with axial low back pain or nociceptive ropathic pain, some physicians offer the patient a screening trial
pain) might experience responses. Furthermore, whereas concor- using electrodes in both regions to determine which provides
dance of paresthesia with areas of pain has been necessary for the best pain relief. Patients might also be given a morphine-
pain relief using conventional stimulation,37 requiring careful naloxone test to clarify the extent of nociceptive and neuropathic
electrode placement and postoperative adjustment in the awake pain components and to facilitate selection of the best stimulation
patient, such requirements might not be necessary for high- target.48
frequency stimulation. These novel protocols (not yet FDA DBS success rates for the treatment of intractable pain are
approved) add treatment possibilities—indeed, some patients difficult to determine because patient selection, surgical tech-
with conventional SCS systems that have lost efficacy have niques, and measurement of outcomes vary substantially among
reported restored pain relief with high-frequency stimulation. studies. Approximately 60% to 80% of patients undergoing a
The traditional goal of achieving paresthesias concordant with screening trial with DBS have pain relief sufficient to warrant
areas of pain continues to inspire new devices with growing implantation of a permanent stimulation system. Of those who
numbers of independent contacts.37 Slender insulated “paddle” receive a permanent stimulation system, 25% to 80% (typically
electrodes, which can be introduced percutaneously, have been 50-60%)47 have been reported to gain acceptable long-term pain
developed to the same end.38,39 Future devices should continue to relief.47-51 Patients with cancer pain,50 FBSS, peripheral neuropa-
address this technical goal, so that conventional as well as high- thy, and trigeminal neuropathy (not anesthesia dolorosa)47,48,50
frequency stimulation can be delivered using the same electrode(s). tend to show more favorable response to DBS than patients with
Another new approach, “peripheral nerve field stimulation” central pain syndromes (e.g., thalamic pain, spinal cord injury
via subcutaneous electrodes, has been combined with SCS and pain, anesthesia dolorosa, postherpetic neuralgia, or phantom
appears to be a useful adjunct for pain control, although it is not limb pain).47,48,50 The incidence of serious complications related
yet FDA approved.40 The subcutaneous electrodes target an area to DBS is low, but the combined incidences of morbidity, mortal-
of pain rather than specific peripheral nerves. ity, and technical complications can approach 25% to 30%.47,50 A
The indications for conventional PNS (i.e., the stimulation of retrospective series involved 85 patients who were treated with
a specific, named peripheral nerve) are similar to those for SCS, DBS for a variety of etiologies; with a mean follow-up of almost
except that the distribution of pain should be able to be covered 20 months, 66% reported good relief of pain.60
by the stimulation of discrete peripheral nerves.41 Overlap exists MCS has received attention as an alternative to thalamic and
between the applications of SCS and PNS. Extremity pain that PAG-PVG stimulation.55-58 MCS is used primarily for treatment
might be appropriate for PNS can sometimes be treated equally of neuropathic pain syndromes and might be particularly effec-
well with SCS, and many surgeons find it easier to implant a tive for certain varieties of intractable facial pain (e.g., trigeminal
percutaneous SCS electrode than to implant a PNS electrode neuropathic pain).56 Approximately 50% of patients undergoing
(which usually requires an open procedure, although ultrasound- MCS have good long-term pain relief. As with DBS, MCS
based percutaneous techniques are being developed).42,43 Con- appears most effective when there is no anesthesia in the distribu-
versely, some clinical scenarios (e.g., pain in the distal leg and tion of pain being treated. The overall clinical efficacy of MCS
foot) may benefit from stimulation of the peripheral nerve, with is similar to that of DBS, but the complications associated with
little neuroplasticity or anatomic opportunity for variation over MCS might be less serious because the electrode is placed epidur-
time, rather than the spinal cord. Some situations (e.g., treatment ally, rather than directly on or within the brain parenchyma.
of occipital neuralgia or cranial postherpetic neuralgia) clearly Because of the somatotopic distribution of leg sensation in the
require PNS rather than SCS.44 High-frequency stimulation (in interhemispheric fissure, MCS is usually restricted to face and/
the kHz range) has been demonstrated experimentally to achieve or arm pain. MCS is a promising therapy the long-term efficacy
pain relief through conduction block; it is under development for of which is under active investigation at several centers.
eventual clinical use.45 Neuraxial drug infusion has become a popular interventional
Stimulation of the dorsal root ganglion has been facilitated by treatment for intractable pain,61-66 especially for pain with a sig-
new electrode technology, and it has also become a discrete nificant nociceptive component,67-69 and can be cost effective
target. Its advantages over SCS have been reported for specific, compared to treatment with more conservative measures.70 Intra-
mostly focal pain syndromes, although this technique remains thecal analgesics for the treatment of cancer pain is well accepted.71
under investigation.46 In contrast, the use of this therapy for chronic nonmalignant pain
Intracranial stimulation therapies include DBS of the has been controversial,66 reflecting concern that neuropathic pain
somatosensory thalamus, hypothalamus, and periventricular- (common in chronic nonmalignant pain syndromes) does not
periaqueductal gray (PVG-PAG)47-54 and MCS.55-58 These thera- respond adequately to opioids and that the efficacy and cost
pies are used primarily for treating pain of nonmalignant origin, effectiveness of neuraxial drug infusion for neuropathic pain have
such as pain associated with FBSS, neuropathic pain following not been determined in controlled trials. Despite these concerns,
central or peripheral nervous system injury, trigeminal pain, and intrathecal analgesic therapy has been used to treat neuropathic
cluster headache. Neither DBS nor MCS is approved by the FDA pain conditions with favorable results,62,63,65,72 and the most
for the treatment of pain, although DBS has been used for more common indication for intrathecal analgesic administration is
than three decades. FBSS, which typically has components of nociceptive (low back)
Targets for focal electrical stimulation of the brain include and neuropathic (extremity) pain.
the ventrocaudal nucleus (nucleus ventroposterolateralis for body The key advantage of neuraxial analgesic administration is its
and nucleus ventroposteromedialis for face) and the PVG-PAG. versatility, which allows it to be applied to a wide range of indica-
Stimulation sites for DBS are generally determined on the basis tions, including nociceptive and mixed nociceptive/neuropathic
CHAPTER 175  Neurosurgical Management of Intractable Pain 1431

pain syndromes. It can be used to treat focal or diffuse axial and/ nerve is uncommon, and mixed sensory-motor nerves cannot be
or extremity pain. Neuraxial analgesia is commonly used to treat sectioned without risk of functional impairment. Some specific 175
pain below cervical levels but can be effective for head and neck exceptions to this general observation exist; for example, section
pain, especially if analgesic agents are delivered intraventricularly of the lateral femoral cutaneous nerve might provide long-lasting
or within the foramen magnum.73,74 Neuraxial analgesics can also relief of meralgia paresthetica,83 and section of the ilioinguinal
treat changing pain (e.g., in a patient with progressive cancer). and/or genitofemoral nerves might relieve some inguinal pain
Significant disadvantages of neuraxial analgesics include the high syndromes (e.g., post-herniorrhaphy pain) in properly selected
cost of the device and medication and the need for maintenance patients.84,85 However, in these situations in which pain originates
(e.g., refilling and, in the case of programmable pumps, replace- from a specific peripheral nerve, as in the sympathetically medi-
ment of depleted batteries). Approximately 50% to 80% of ated pain described previously, neurostimulation of that specific
patients with neuraxial analgesics achieve good long-term relief nerve is often less invasive, is reversible and, therefore, is prefer-
of pain, and outcomes (i.e., extent of pain relief, patient satisfac- able to neurectomy.86-88 Radiofrequency facet denervation might
tion, and dose requirements) are similar in patients with cancer provide substantial relief of neck or back pain, albeit with signifi-
and noncancer pain.61,62,67-69 Serious complications of the therapy cant rates of recurrence after 2 to 3 years.89
are uncommon. Some patients, particularly those with neuro- Dorsal rhizotomy and ganglionectomy serve similar purposes
pathic pain syndromes, may need intrathecal “polyanalgesia.”38 in denervating somatic and/or visceral tissues, but ganglionec-
The cost is higher with polyanalgesia but may be offset by a tomy might produce more complete denervation than can be
greater likelihood of pain relief.75 accomplished by dorsal rhizotomy, which does not affect the
afferent fibers that enter the spinal cord through the ventral
root.90 In contrast, ganglionectomy effectively eliminates input
Neuroablative Therapies from ventral root afferent fibers by removing their cell bodies,
Although neuromodulatory pain therapies are becoming increas- which are located within the dorsal root ganglion. Rhizotomy and
ingly common, neuroablative procedures maintain a role in the ganglionectomy can be used to treat pain in the neck, trunk, or
treatment of intractable pain. Neuroablative therapy is often con- abdomen. Neither procedure is useful for the treatment of pain
sidered the treatment of “last resort,” but in some cases it may be in the extremities unless extremity function is already lost,
the procedure of choice. For example, a patient with cancer pain because denervation removes proprioceptive as well as nocicep-
who has a short life expectancy might be treated more appropri- tive input, resulting in a functionless limb. Limited denervation
ately with cordotomy than with an implanted intrathecal analge- does not provide adequate pain relief, probably because of the
sic infusion system. In such cases, as in every case, pain therapy dermatomal overlap of segmental innervation. These procedures
must be tailored to meet the needs of individual patients. are most appropriate for the treatment of cancer pain and do not
Neuroablative therapies include peripheral techniques that consistently result in long-term improvement of noncancer
interrupt or alter nociceptive input into the spinal cord (e.g., pain,91,92 although both dorsal rhizotomy93,94 and ganglionec-
sympathectomy, neurectomy, ganglionectomy, rhizotomy), spinal tomy95 have been used successfully to treat occipital neuralgia.
interventions that alter afferent input or rostral transmission of Temporary relief by “diagnostic” blocks is nonspecific96 and
nociceptive information (e.g., DREZ lesioning, cordotomy, might not predict success. In patients with cancer, rhizotomy and
myelotomy), and supraspinal intracranial procedures that inter- ganglionectomy might be useful for thoracic or abdominal wall
fere with transmission of nociceptive information (e.g., mesen- pain; for perineal pain in patients with impaired bladder, bowel,
cephalotomy, thalamotomy) or the affective perception of painful and sex function97; or for the treatment of pain in a functionless
stimuli (e.g., cingulotomy). As with neuromodulatory techniques, extremity.98 Multiple sacral rhizotomies can be performed (e.g.,
a successful outcome requires appropriate patient and interven- to treat pelvic pain from cancer) by passing a ligature around the
tion selection. Neuroablative therapies are more successful in the thecal sac below S1.99 Alternatively, intrathecal alcohol neurolysis
treatment of nociceptive pain than of neuropathic pain; when can be performed percutaneously.100 Rhizotomy might be useful
neuropathic pain is treated with neuroablative therapies, improve- for treatment of craniofacial pain101 of nontrigeminal origin as
ment is primarily limited to intermittent, paroxysmal, or evoked well as for classic trigeminal neuralgia.
(allodynia, hyperpathia) pain, whereas continuous pain remains
relatively unchanged.76
Spinal Procedures
DREZ lesioning of the spinal cord (for trunk or extremity
Peripheral Procedures pain)102-106 or of the nucleus caudalis (for facial pain)103,106-108 can
Sympathectomy has been reported to alleviate visceral pain asso- provide significant pain relief with proper selection, with improve-
ciated with certain cancers77,78 and might also be an effective ment in pain reported in 60% 80% of patients. These techniques,
treatment for noncancer pain, such as pain associated with vaso- however, are best reserved for localized pain. Certain types of
spastic disorders or sympathetically maintained pain (in which cancer pain can be treated effectively with DREZ lesioning (e.g.,
sympathetic blocks reliably relieve the pain). Sympathectomy, neuropathic arm pain associated with Pancoast tumor), but the
however, has fallen into disfavor as a treatment for intractable most successful applications are related to treatment of neuro-
pain of nonmalignant origin because of inconsistent results and pathic pain arising from root avulsion (cervical or lumbosacral)
concerns about complication rates.77-81 Furthermore, research and “end-zone” or “boundary” pain following spinal cord injury.
suggests that SCS, which has the advantage of reversibility, pro- These pain syndromes sometimes respond adequately to SCS or
vides a better long-term outcome with lower morbidity than intrathecal drug infusion, but DREZ lesioning might provide a
sympathectomy and thus might become the treatment of choice similar result without the need for long-term maintenance of an
for sympathetically maintained noncancer pain.26 augmentative device. DREZ lesioning has been used for the
Neurectomy might be a useful treatment for some patients in treatment of other neuropathic pain syndromes (e.g., post-
whom pain develops following peripheral nerve injury, including herpetic neuralgia) but does not consistently result in good pain
that associated with limb amputation or neuroma.82 However, relief. Nucleus caudalis lesioning seems most useful for deaffer-
neurectomy is not useful for treatment of nonspecific stump pain entation pain affecting the face (including post-herpetic neural-
after amputation, nor is it generally useful for the treatment of gia) and less helpful for facial pain of peripheral origin (e.g.,
other nonmalignant peripheral pain syndromes. The utility of traumatic trigeminal neuropathy); it can also be effectively
neurectomy is limited because pain arising from a pure sensory employed for cancer pain.106,109 A CT-guided percutaneous
1432 SECTION 6  Pain

Figure 175-2. Axial computed tomography myelogram image


through C1-C2. A thin-walled, 20-gauge needle enters from the left.
A small radiofrequency electrode passes through the needle and  
into the anterolateral quadrant of the spinal cord—the lateral
spinothalamic tract.

Figure 175-1. Axial computed tomography myelogram at the


foramen magnum with the patient facing to the reader’s left. The
tip of the radiofrequency electrode is seen in the brainstem in the is lateralizing opposite the side of the cordotomy.112-117 It is used
region of the nucleus caudalis. A second needle has been placed   less frequently to manage noncancer pain but a risk-benefit analy-
in the thecal sac from directly posterior (right) through which an sis of the newer percutaneous cordotomy for this indication has
endoscope has been placed. Inset: Screen capture of the endoscopic yet to be performed.
view of the thin-walled, 20-gauge needle (silver with bevel) from which Cordotomy can improve both lancinating, paroxysmal neu-
the radiofrequency electrode (black) is seen exiting and entering the ropathic pain that sometimes arises from spinal cord injury and
brainstem. The inferior edge of the cerebellum can be seen just to the evoked (allodynic or hyperpathic) pain associated with periph-
right of the needle. eral neuropathic pain syndromes, but it is not an effective
treatment for continuous neuropathic pain.117 Cordotomy pro-
vides good pain relief in approximately 60% to 90% of studied
technique for nucleotractotomy has been developed with the patients.114,117,118 Although some data suggest a significant inci-
addition of endoscopic observation (Fig. 175-1).110 As with other dence of recurrent pain following cordotomy,98,118,119 an observa-
ablative procedures, DREZ lesioning is most effective for reliev- tion used to suggest the procedure be reserved for patients with
ing paroxysmal rather than continuous pain.104 limited life expectancy, there have also been case reports of pain
Cordotomy is a valuable procedure for pain management, relief lasting years to decades.120,121
especially for cancer pain, even though it has been largely replaced Historically, open cordotomy has been associated with
by intrathecal analgesic administration. Cordotomy offers several impaired respiratory function. For example, bilateral open cor-
advantages and is one of the most clinically useful neuroablative dotomy has been linked to Ondine’s curse (potentially fatal loss
techniques.111 Although cordotomy can be performed as an of respiratory drive during sleep).112,117 However, it is unclear
open procedure,112,113 the most common modern technique is whether the same is true for the percutaneous procedure. A
CT-guided and percutaneous, often with use of local anesthesia review of 35 patients undergoing percutaneous cordotomy did
(Fig. 175-2). In addition to requiring no long-term follow-up or not demonstrate respiratory dysfunction,122 and in a later series
maintenance, percutaneous cordotomy allows for intervention of 207 cordotomies, 12 of which were bilateral, there were no
without interruption of chemotherapy, and only very brief cessa- instances of Ondine’s curse or other severe complication.114 Some
tion of anticoagulation therapy is necessary. Percutaneous cor- neurosurgeons have advocated using a CT-guided transdiscal
dotomy, with its attendant real-time patient feedback, seems to approach for the contralateral cordotomy to allow for placement
be much safer than the open procedure,114 resulting in a general at different spinal cord levels, thereby mitigating the risk of
reconsideration of its place in the pain care algorithm. It is likely adverse outcome.123 An alternative approach is to perform a uni-
that the risks and efficacy of the open procedure112,113 are not lateral high cervical percutaneous procedure followed by a con-
relevant to percutaneous cordotomy; outcome studies of the tralateral open low cervical or thoracic cordotomy.112,115
newer procedure are necessary. Moreover, the very low cost of Like sympathectomy, myelotomy fell into disuse with the
cordotomy relative to more expensive, implanted technologies advent of intrathecal drug infusion therapy, but it can provide
should be considered. significant pain relief in properly selected patients, including
Cordotomy is used most commonly for the treatment of some who fail treatment with intrathecal analgesics.124 Commis-
cancer-related pain below mid- to low-cervical dermatomes that sural myelotomy was developed to provide the benefits of
CHAPTER 175  Neurosurgical Management of Intractable Pain 1433

175

Figure 175-3. Operating microscope image of midline myelotomy


through a T8-T9 laminectomy. The dura is tented open and the pia
has been incised in the midline. A blunt micro-instrument (right) is
creating the myelotomy up to the commissure. (A previously placed Figure 175-4. Axial computed tomography myelogram just caudal
intrathecal drug delivery catheter can be seen below the pial opening.) to the foramen magnum. The patient is prone. A thin-walled,
20-gauge needle enters thecal sac from directly posterior, through
which a radiofrequency electrode passes into the parenchyma of the
bilateral cordotomy without the inherent risks associated with spinal cord between the dorsal columns.
lesioning both anterior quadrants of the spinal cord.124-127 It is
accomplished by sectioning spinothalamic tract fibers as they
decussate in the anterior commissure. Subsequently, it was good pain relief with a lower incidence of complications. Mesen-
observed that a limited midline myelotomy (Fig. 175-3)128 or high cephalotomy, however, might be the preferred therapy for some
cervical myelotomy125,129,130 could be equally effective. Identifica- patients, for example, those with short life expectancies or for
tion of a dorsal column visceral pain pathway has led to the whom the cost of the long-term follow-up required with neur-
development of punctate midline myelotomy.129,131 axial analgesic administration becomes burdensome.
A myelotomy is indicated primarily for the treatment of Thalamotomy has been used for the treatment of cancer
cancer pain, generally in the abdomen (visceral), pelvis, perineum, and noncancer pain and can be accomplished via stereotactic
or sacrum. It is most effective for nociceptive rather than neuro- radiofrequency,76,130,135-137 radiosurgery,138,139 or magnetic reso-
pathic pain and should be of particular utility to most neurosur- nance imaging (MRI)–guided, focused ultrasound.140 As a cancer
geons because the various techniques are straightforward and the pain treatment, thalamotomy is most appropriate for patients
anatomy is familiar. Early complete pain relief is achieved in more who have widespread pain (e.g., from diffuse metastatic disease)
than 90% of patients, but pain tends to recur over time; neverthe- or midline, bilateral, or head/neck pain for which other proce-
less, approximately 50% to 60% of patients experience long-term dures might not provide relief.108 Thalamotomy can be useful
pain relief.116,130 The risk of bladder, bowel, and sexual dysfunc- for patients who are not candidates for cordotomy, for example,
tion is less than that associated with bilateral cordotomy but those with pain above the C5 dermatome.137 Thalamotomy is
remains sufficiently high, so the use of myelotomy procedures is slightly less effective than mesencephalotomy but is associated
typically restricted to patients with cancer pain who have preex- with fewer complications.137 The lateral thalamus (nucleus ven-
isting dysfunction.98 Recently, myelotomy has been performed as trocaudalis) is not considered an acceptable target for ablation
a percutaneous, CT-guided outpatient technique with good owing to the risk of dysesthesia and sensory loss,76 but the sub-
results (Fig. 175-4).132,133 jacent parvicellular ventrocaudal nucleus, which is thought to
be a relay site for spinothalamic afferents, has been used as a
target. Results of thalamotomy directed to this structure might be
Supraspinal Intracranial Techniques similar to those achieved with cordotomy.135 Medial thalamotomy
Ablative neurosurgical procedures directed at the brainstem are appears to be most effective for treating nociceptive pain (e.g.,
not widely used, in part because relatively few patients require cancer pain), with acceptable long-term pain relief obtained in
such interventions and because relatively few neurosurgeons have approximately 30% to 50% of patients.76,118,136,139 Compared with
the expertise to perform them. However, as with other ablative nociceptive pain, neuropathic pain responds less consistently to
procedures, they can provide significant benefit in carefully thalamotomy, with only about one third of recipients achieving
selected cases. Mesencephalotomy, which is indicated for the long-term improvement.76,136 Patients with paroxysmal, lancinat-
treatment of intractable pain involving the head, neck, shoulder, ing neuropathic pain or neuropathic pain with elements of allo-
and arm,76,134,135 can be viewed as a supraspinal version of cor- dynia and hyperpathia might experience significant improvement
dotomy.76 Mesencephalotomy is used for the treatment of cancer after thalamotomy, whereas those with continuous neuropathic
pain and results in long-term pain relief in 85% of patients.130 pain tend not to benefit.76
This procedure, however, does not provide consistent long-term Cingulotomy is used to treat cancer pain and has also been
relief of central neuropathic pain129 and is associated with fre- used to treat noncancer pain; however, it is most commonly used
quent side effects and complications, especially oculomotor dys- for management of psychiatric disorders.41,128,141-143 Approxi-
function.76,129,130 The utility of mesencephalotomy has diminished mately 50% to 75% of patients with cancer pain benefit from the
subsequent to the advent of neuraxial analgesic administration, procedure, at least in the short term. In these patients, pain relief
such as intraventricular morphine infusion, which can provide is generally maintained for at least 3 months. The efficacy of
1434 SECTION 6  Pain

cingulotomy for chronic noncancer pain is less clear; some inves- overlooked, especially by nonsurgical specialists unfamiliar with
tigators report relatively good long-lasting pain relief,130,141,143 but the role of such neurosurgical therapies in the management of
others indicate only 20% long-term success.134 Cingulotomy intractable pain. Neurosurgeons should remain knowledgeable
might be a reasonable option for patients, especially those with about pain therapies and should remain actively involved in the
cancer pain, in whom other treatments have not worked. One field of pain medicine to ensure the continued availability of the
report suggests that the procedure may also effectively treat the entire spectrum of pain therapies. By accepting this responsibility,
symptom of air hunger.144 The ability to perform a cingulotomy neurosurgeons will continue to fulfill their important role in the
using a noninvasive radiosurgical technique may make it more management of intractable pain.
attractive as an alternative for the fragile patients in whom it is
Acknowledgment
most often used.145 Because cingulotomy is performed for treat-
ment of psychiatric disease and carries the stigma of “psychosur- The authors gratefully acknowledge the editorial assistance provided by
Keren Price.
gery,” formal review by institutional ethics committees might be
warranted if this procedure is being considered as a treatment for
intractable pain. SUGGESTED READINGS
Hypophysectomy (surgical, chemical, or radiosurgical) can Boccard SG, Pereira EA, Moir L, et al. Long-term outcomes of deep
provide good relief of cancer pain and is thought by some to be brain stimulation for neuropathic pain. Neurosurgery. 2013;72(2):221-
most effective for hormone-responsive cancers (e.g., prostate, 230, discussion 231.
breast), but seems to relieve pain related to other tumors as well. Deer TR, Leong MS, eds. Comprehensive Treatment of Chronic Pain by
Medical, Interventional, and Integrative approaches. New York: Springer;
Hypophysectomy is most appropriate for patients with wide- 2013.
spread disease and diffuse pain, especially pain associated with Deer TR, Levy R, Prager J, et al. Polyanalgesic consensus conference–
diffuse bony metastases. Pain is alleviated in 45% to 95% of 2012: Recommendations to reduce morbidity and mortality in intra-
patients independent of tumor regression; the specific mecha- thecal drug delivery in the treatment of chronic pain. Neuromodulation.
nism of pain relief is unknown.130,135,146-148 2012;15(5):467-482, discussion 482.
Deer TR, Mekhail N, Provenzano D, et al. The appropriate use of neuro-
stimulation of the spinal cord and peripheral nervous system for the
CONCLUSION treatment of chronic pain and ischemic diseases: The Neuromodula-
tion Appropriateness Consensus Committee. Neuromodulation. 2014;
Successful treatment of intractable pain requires that the right
17(6):515-550, discussion 550.
therapy be offered to the right patient at the right time. Candi- Deer TR, Mekhail N, Provenzano D, et al. The appropriate use of neuro-
dates for neurosurgical treatment must be identified carefully, stimulation: Avoidance and treatment of complications of neurostimu-
with full recognition of chronic pain as a biopsychosocial disor- lation therapies for the treatment of chronic pain. Neuromodulation.
der. Undue emphasis on “pain generators” without consideration 2014;17(6):571-597, discussion 597-8.
of the psychosocial factors that contribute to the pain complaint Deer TR, Prager J, Levy R, et al. Polyanalgesic consensus conference–
will predispose to treatment failure. Any recommendation must 2012: Recommendations on trialing for intrathecal (intraspinal) drug
be tailored to the clinical diagnosis, the emotional needs of the delivery: Report of an interdisciplinary expert panel. Neuromodulation.
patient, and his or her relevant social circumstances. Outcomes 2012;15(5):420-435, discussion 435.
of neuromodulatory therapies after proper patient selection are Deer TR, Prager J, Levy R, et al. Polyanalgesic Consensus Conference
2012: Recommendations for the management of pain by intrathecal
good, and the risk of serious or permanent complications is low, (intraspinal) drug delivery: Report of an interdisciplinary expert panel.
factors that must be carefully considered in the recommending Neuromodulation. 2012;15(5):436-464, discussion 464-466.
of first-line neurosurgical treatment of pain. These techniques, Deer TR, Prager J, Levy R, et al. Polyanalgesic Consensus Conference–
especially the neurostimulation therapies, are superior to neu- 2012: Consensus on diagnosis, detection, and treatment of catheter-
roablative techniques for the treatment of neuropathic pain with tip granulomas (inflammatory masses). Neuromodulation. 2012;15(5):
a continuous, dysesthetic component. Neuroablation, however, 483-495, discussion 496.
might be appropriate for other patients, including those with Kanpolat Y. Percutaneous destructive pain procedures on the upper spinal
cancer pain and a short life expectancy, those with predominantly cord and brain stem in cancer pain: CT-guided techniques, indications
nociceptive pain, and those with neuropathic pain with paroxys- and results. Adv Tech Stand Neurosurg. 2007;32:147-173.
Prager J, Deer T, Levy R, et al. Best practices for intrathecal drug delivery
mal or evoked components. for pain. Neuromodulation. 2014;17(4):354-372, discussion 372.
Neurosurgeons are unique within the discipline of pain Raslan AM, Cetas JS, McCartney S, et al. Destructive procedures for
medicine by virtue of their diagnostic knowledge, technical control of cancer pain: The case for cordotomy. J Neurosurg. 2011;
skills, and access to the nervous system. These attributes enable 114(1):155-170.
neurosurgeons to provide pain treatments chosen from the entire Stearns L, Boortz-Marx R, Du Pen S, et al. Intrathecal drug delivery for
spectrum of pain therapies, including neuromodulation and the management of cancer pain: A multidisciplinary consensus of best
neuroablative techniques. As more attention is focused on neu- clinical practices. J Support Oncol. 2005;3(6):399-408.
romodulation for the treatment of intractable pain, ablative
strategies that might be appropriate for some patients may be See a full reference list on ExpertConsult.com
CHAPTER 175  Neurosurgical Management of Intractable Pain 1434.e1

REFERENCES 26. Burchiel KJ, Anderson VC, Brown FD, et al. Prospective, multi-
1. Bonica JJ. Definitions and taxonomy of pain. In: Bonica JJ, Loeser center study of spinal cord stimulation for relief of chronic back 175
JD, Chapman CR, et al., eds. The Management of Pain. 2nd ed. and extremity pain. Spine (Phila Pa 1976). 1996;21(23):2786-
Philadelphia: Lea & Febiger; 1990:18-27. 2794.
2. Loeser JD, Bigos SJ, Fordyce WE, et al. Low back pain. In: Bonica 27. Kumar K, Nath RK, Toth C. Spinal cord stimulation is effective in
JJ, Loeser JD, Chapman CR, et al, eds. The Management of Pain. 2nd the management of reflex sympathetic dystrophy. Neurosurgery.
ed. Philadelphia: Lea & Febiger; 1992:1448-1483. 1997;40(3):503-508, discussion 508-509.
3. Bonica JJ. General considerations of chronic pain. In: Bonica JJ, 28. Kemler MA, Barendse GA, van Kleef M, et al. Spinal cord stimula-
Loeser JD, Chapman CR, et al., eds. The Management of Pain. 2nd tion in patients with chronic reflex sympathetic dystrophy. N Engl
ed. Philadelphia: Lea & Febiger; 1990:180-196. J Med. 2000;343(9):618-624.
4. Loeser JD. Pain and suffering. Clin J Pain. 2000;16(suppl):S2-S6. 29. Levy RM. Evidence-based review of neuromodulation for complex
5. Krames ES. Intraspinal opioid therapy for chronic nonmalignant regional pain syndrome: a conflict between faith and science? Neu-
pain: current practice and clinical guidelines. J Pain Symptom romodulation. 2012;15(6):501-506.
Manage. 1996;11(6):333-352. 30. Kumar K, Toth C, Nath RK. Spinal cord stimulation for chronic
6. Elliott JS. Outlines of Greek and Roman Medicine. New York: William pain in peripheral neuropathy. Surg Neurol. 1996;46(4):363-369.
Wood and Company; 1914:165. 31. Kumar K, Toth C, Nath RK, et al. Improvement of limb circulation
7. World Health Organization, WHO Expert Committee on Cancer in peripheral vascular disease using epidural spinal cord stimulation:
Pain Relief and Active Supportive Care. Cancer pain relief and pallia- a prospective study. J Neurosurg. 1997;86(4):662-669.
tive care: report of a WHO Expert Committee. Geneva, Switzerland: 32. Huber SJ, Vaglienti RM, Midcap ME. Enhanced limb salvage for
WHO; 1990. peripheral vascular disease with the use of spinal cord stimulation.
8. Eisenberg E, Berkey CS, Carr DB, et al. Efficacy and safety of W V Med J. 1996;92(2):89-91.
nonsteroidal antiinflammatory drugs for cancer pain: a meta- 33. Sanderson JE, Ibrahim B, Waterhouse D, et al. Spinal electrical
analysis. J Clin Oncol. 1994;12(12):2756-2765. stimulation for intractable angina–long-term clinical outcome and
9. Zernikow B, Smale H, Michel E, et al. Paediatric cancer pain man- safety. Eur Heart J. 1994;15(6):810-814.
agement using the WHO analgesic ladder—results of a prospective 34. de Jongste MJ, Hautvast RW, Hillege HL, et al. Efficacy of spinal
analysis from 2265 treatment days during a quality improvement cord stimulation as adjuvant therapy for intractable angina pectoris:
study. Eur J Pain. 2006;10(7):587-595. a prospective, randomized clinical study. Working Group on Neu-
10. Keskinbora K, Pekel AF, Aydinli I. Gabapentin and an opioid com- rocardiology. J Am Coll Cardiol. 1994;23(7):1592-1597.
bination versus opioid alone for the management of neuropathic 35. De Ridder D, Vanneste S, Plazier M, et al. Burst spinal cord stimula-
cancer pain: a randomized open trial. J Pain Symptom Manage. tion: toward paresthesia-free pain suppression. Neurosurgery. 2010;
2007;34(2):183-189. 66(5):986-990.
11. Portenoy RK. Treatment of cancer pain. Lancet. 2011;377(9784): 36. Al-Kaisy A, Van Buyten JP, Smet I, et al. Sustained effectiveness of
2236-2247. 10 kHz high-frequency spinal cord stimulation for patients with
12. Bhaskar AK. Interventional management of cancer pain. Curr Opin chronic, low back pain: 24-month results of a prospective multi-
Support Palliat Care. 2012;6(1):1-9. center study. Pain Med. 2014;15(3):347-354.
13. American Pain Society. Principles of Analgesic Use in the Treatment 37. North RB, Ewend MG, Lawton MT, et al. Spinal cord stimulation
of Acute Pain and Cancer Pain. 6th ed. Skokie, IL: American Pain for chronic, intractable pain: superiority of “multi-channel” devices.
Society; 2008. Pain. 1991;44(2):119-130.
14. Cherny NI, Portenoy RK. The management of cancer pain. CA 38. Deer T, Bowman R, Schocket SM, et al. The prospective evaluation
Cancer J Clin. 1994;44(5):263-303. of safety and success of a new method of introducing percutaneous
15. North RB, Kidd DH, Farrokhi F, et al. Spinal cord stimulation paddle leads and complex arrays with an epidural access system.
versus repeated lumbosacral spine surgery for chronic pain: a ran- Neuromodulation. 2012;15(1):21-29, discussion 29-30.
domized, controlled trial. Neurosurgery. 2005;56(1):98-106, discus- 39. Loge D, De Coster O, Washburn S. Technological innovation in
sion 106-107. spinal cord stimulation: use of a newly developed delivery device
16. North RB, Kidd D, Shipley J, et al. Spinal cord stimulation versus for introduction of spinal cord stimulation leads. Neuromodulation.
reoperation for failed back surgery syndrome: a cost effectiveness 2012;15(4):392-401.
and cost utility analysis based on a randomized, controlled trial. 40. Mironer YE, Hutcheson JK, Satterthwaite JR, et al. Prospective,
Neurosurgery. 2007;61(2):361-368, discussion 368-369. two-part study of the interaction between spinal cord stimulation
17. Doleys DM, Olson K, eds. Psychological Assessment and Intervention in and peripheral nerve field stimulation in patients with low back
Implantable Pain Therapies. Minneapolis, MN: Medtronic, Inc.; 1997. pain: development of a new spinal-peripheral neurostimulation
18. North RB, Levy RM. Consensus conference on the neurosurgical method. Neuromodulation. 2011;14(2):151-154, discussion 155.
management of pain. Neurosurgery. 1994;34(4):756-760, discussion 41. Hassenbusch SJ, Stanton-Hicks M, Schoppa D, et al. Long-term
760-761. results of peripheral nerve stimulation for reflex sympathetic dys-
19. Raslan AM, McCartney S, Burchiel KJ. Management of chronic trophy. J Neurosurg. 1996;84(3):415-423.
severe pain: spinal neuromodulatory and neuroablative approaches. 42. Huntoon MA, Burgher AH. Ultrasound-guided permanent implan-
Acta Neurochir Suppl. 2007;97(Pt 1):33-41. tation of peripheral nerve stimulation (PNS) system for neuropathic
20. Whitworth LA, Feler CA. Application of spinal ablative techniques pain of the extremities: original cases and outcomes. Pain Med.
for the treatment of benign chronic painful conditions: history, 2009;10(8):1369-1377.
methods, and outcomes. Spine (Phila Pa 1976). 2002;27(22):2607- 43. Kent M, Upp J, Spevak C, et al. Ultrasound-guided peripheral nerve
2612, discussion 2613. stimulator placement in two soldiers with acute battlefield neuro-
21. Deer TR. The role of neuromodulation by spinal cord stimulation pathic pain. Anesth Analg. 2012;114(4):875-878.
in chronic pain syndromes: current concepts. Tech Reg Anes Pain 44. Weiner RL. Occipital neurostimulation for treatment of intrac-
Manage. 1998;2(3):161-167. table headache syndromes. Acta Neurochir Suppl. 2007;97(Pt 1):
22. North RB, Kidd DH, Zahurak M, et al. Spinal cord stimulation for 129-133.
chronic, intractable pain: experience over two decades. Neurosur- 45. Kilgore KL, Bhadra N. Reversible nerve conduction block using
gery. 1993;32(3):384-394, discussion 394-395. kilohertz frequency alternating current. Neuromodulation. 2014;17(3):
23. Turner JA, Loeser JD, Bell KG. Spinal cord stimulation for chronic 242-254, discussion 254-255.
low back pain: a systematic literature synthesis. Neurosurgery. 1995; 46. Deer TR, Grigsby E, Weiner RL, et al. A prospective study of dorsal
37(6):1088-1095, discussion 1095-1096. root ganglion stimulation for the relief of chronic pain. Neuromodu-
24. Kumar K, Taylor RS, Jacques L, et al. Spinal cord stimulation versus lation. 2013;16(1):67-71, discussion 71-72.
conventional medical management for neuropathic pain: a multi- 47. Levy RM, Lamb S, Adams JE. Treatment of chronic pain by deep
centre randomised controlled trial in patients with failed back brain stimulation: long term follow-up and review of the literature.
surgery syndrome. Pain. 2007;132(1-2):179-188. Neurosurgery. 1987;21(6):885-893.
25. North R, Shipley J, Prager J, et al. Practice parameters for the use 48. Kumar K, Toth C, Nath RK. Deep brain stimulation for intractable
of spinal cord stimulation in the treatment of chronic neuropathic pain: a 15-year experience. Neurosurgery. 1997;40(4):736-746, dis-
pain. Pain Med. 2007;8(suppl 4):S200-S275. cussion 746-747.
1434.e2 SECTION 6  Pain

49. Richardson DE. Intracranial stimulation therapies: deep brain stim- 74. Karavelis A, Foroglou G, Selviaridis P, et al. Intraventricular admin-
ulation. In: Follett KA, ed. Neurosurgical Pain Management. Phila- istration of morphine for control of intractable cancer pain in 90
delphia: Elsevier; 2004:156-159. patients. Neurosurgery. 1996;39(1):57-61, discussion 61-62.
50. Kaplitt M, Rezai AR, Lozano AM, et al. Deep brain stimulation 75. Kumar K, Rizvi S, Bishop S, et al. Cost impact of intrathecal poly-
for chronic pain. In: Winn HR, ed. Youmans Neurological Surgery. analgesia. Pain Med. 2013;14(10):1569-1584.
Philadelphia: Elsevier; 2004:3119-3131. 76. Tasker RR. Stereotactic surgery. In: Wall PD, Melzack R, eds.
51. Bittar RG, Kar-Purkayastha I, Owen SL, et al. Deep brain stimula- Textbook of Pain. 3rd ed. Edinburgh: Churchill Livingstone; 1994:
tion for pain relief: a meta-analysis. J Clin Neurosci. 2005;12(5): 1137-1158.
515-519. 77. Hardy RW, Bay JW. Surgery of the sympathetic nervous system. In:
52. Leone M, Proietti Cecchini A, Franzini A, et al. Lessons from 8 Schmidek HH, Sweet WH, eds. Operative Neurosurgical Techniques:
years’ experience of hypothalamic stimulation in cluster headache. Indications, Methods, and Results. 3rd ed. Philadelphia: WB Saunders;
Cephalalgia. 2008;28(7):787-797, discussion 798. 1995:1637-1646.
53. Coffey RJ, Lozano AM. Neurostimulation for chronic noncancer 78. De Salles A, Johnson JP. Sympathectomy for pain. In: Winn HR,
pain: an evaluation of the clinical evidence and recommendations ed. Youmans Neurological Surgery. Philadelphia: Elsevier; 2004:
for future trial designs. J Neurosurg. 2006;105(2):175-189. 3039-3105.
54. Rasche D, Rinaldi PC, Young RF, et al. Deep brain stimulation for 79. Johnson JP, Obasi C, Hahn MS. Glatleider P. Endoscopic thoracic
the treatment of various chronic pain syndromes. Neurosurg Focus. sympathectomy. J Neurosurg. 1999;91(suppl):90-97.
2006;21(6):E8. 80. Schwartzman RJ, Liu JE, Smullens SN, et al. Long-term outcome
55. Nguyen JP, Lefaucheur JP, Decq P, et al. Chronic motor cortex following sympathectomy for complex regional pain syndrome type
stimulation in the treatment of central and neuropathic pain. Cor- 1 (RSD). J Neurol Sci. 1997;150(2):149-152.
relations between clinical, electrophysiological and anatomical data. 81. Mailis A, Furlan A. Sympathectomy for neuropathic pain. Cochrane
Pain. 1999;82(3):245-251. Database Syst Rev. 2003;(2):CD002918.
56. Henderson JM, Lad SP. Motor cortex stimulation and neuropathic 82. Burchiel KJ, Johans TJ, Ochoa J. The surgical treatment of painful
facial pain. Neurosurg Focus. 2006;21(6):E6. traumatic neuromas. J Neurosurg. 1993;78(5):714-719.
57. Lima MC, Fregni F. Motor cortex stimulation for chronic pain: 83. van Eerten PV, Polder TW, Broere CA. Operative treatment of
systematic review and meta-analysis of the literature. Neurology. meralgia paresthetica: transection versus neurolysis. Neurosurgery.
2008;70(24):2329-2337. 1995;37(1):63-65.
58. Velasco F, Arguelles C, Carrillo-Ruiz JD, et al. Efficacy of motor 84. Starling JR, Harms BA. Diagnosis and treatment of genitofemoral
cortex stimulation in the treatment of neuropathic pain: a random- and ilioinguinal neuralgia. World J Surg. 1989;13(5):586-591.
ized double-blind trial. J Neurosurg. 2008;108(4):698-706. 85. Kim DH, Murovic JA, Tiel RL, et al. Surgical management of 33
59. Boccard SG, Fitzgerald JJ, Pereira EA, et al. Targeting the affective ilioinguinal and iliohypogastric neuralgias at Louisiana State Uni-
component of chronic pain: a case series of deep brain stimulation versity Health Sciences Center. Neurosurgery. 2005;56(5):1013-1020.
of the anterior cingulate cortex. Neurosurgery. 2014;74(6):628-635, 86. Reverberi C, Dario A, Barolat G, et al. Using peripheral nerve
discussion 635-637. stimulation (PNS) to treat neuropathic pain: a clinical series. Neu-
60. Boccard SG, Pereira EA, Moir L, et al. Long-term outcomes of romodulation. 2014;17(8):777-783, discussion 783.
deep brain stimulation for neuropathic pain. Neurosurgery. 2013; 87. Rasskazoff SY, Slavin KV. An update on peripheral nerve stimula-
72(2):221-230, discussion 231. tion. J Neurosurg Sci. 2012;56(4):279-285.
61. Paice JA, Penn RD, Shott S. Intraspinal morphine for chronic pain: 88. Slavin KV, Lunsford D, eds. Peripheral Nerve Stimulation. Basel,
a retrospective, multicenter study. J Pain Symptom Manage. 1996; Switzerland: Karger; 2011 Progress in Neurological Surgery; No. 24.
11(2):71-80. 89. Lord SM, Barnsley L, Wallis BJ, et al. Percutaneous radio-frequency
62. Winkelmuller M, Winkelmuller W. Long-term effects of continu- neurotomy for chronic cervical zygapophyseal-joint pain. N Engl J
ous intrathecal opioid treatment in chronic pain of nonmalignant Med. 1996;335(23):1721-1726.
etiology. J Neurosurg. 1996;85(3):458-467. 90. Hosobuchi Y. The majority of unmyelinated afferent axons in
63. Schuchard M, Krames ES, Lanning R. Intraspinal analgesia for human ventral roots probably conduct pain. Pain. 1980;8(2):
nonmalignant pain: a retrospective analysis for efficacy, safety and 167-180.
feasability in 50 patients. Neuromodulation. 1998;1(1):46-56. 91. Onofrio BM, Campa HK. Evaluation of rhizotomy. Review of 12
64. Smith HS, Deer TR, Staats PS, et al. Intrathecal drug delivery. Pain years’ experience. J Neurosurg. 1972;36(6):751-755.
Physician. 2008;11(suppl):S89-S104. 92. North RB, Kidd DH, Campbell JN, et al. Dorsal root ganglionec-
65. Hassenbusch SJ, Stanton-Hicks M, Covington EC, et al. Long- tomy for failed back surgery syndrome: a 5-year follow-up study.
term intraspinal infusions of opioids in the treatment of neuropathic J Neurosurg. 1991;74(2):236-242.
pain. J Pain Symptom Manage. 1995;10(7):527-543. 93. Lovely TJ. The treatment of chronic incisional pain and headache
66. Maron J, Loeser JD. Spinal opioid infusions in the treatment of after retromastoid craniectomy. Surg Neurol Int. 2012;3:92-7806.
chronic pain of nonmalignant origin. Clin J Pain. 1996;12(3): 94. Kapoor V, Rothfus WE, Grahovac SZ, et al. Refractory occipital
174-179. neuralgia: preoperative assessment with CT-guided nerve block
67. Hayek SM, Deer TR, Pope JE, et al. Intrathecal therapy for cancer prior to dorsal cervical rhizotomy. AJNR Am J Neuroradiol.
and non-cancer pain. Pain Physician. 2011;14(3):219-248. 2003;24(10):2105-2110.
68. Bottros MM, Christo PJ. Current perspectives on intrathecal drug 95. Acar F, Miller J, Golshani KJ, et al. Pain relief after cervical gan-
delivery. J Pain Res. 2014;7:615-626. glionectomy (C2 and C3) for the treatment of medically intractable
69. Saulino M, Kim PS, Shaw E. Practical considerations and patient occipital neuralgia. Stereotact Funct Neurosurg. 2008;86(2):106-112.
selection for intrathecal drug delivery in the management of chronic 96. North RB, Kidd DH, Zahurak M, et al. Specificity of diagnostic
pain. J Pain Res. 2014;7:627-638. nerve blocks: a prospective, randomized study of sciatica due to
70. Guillemette S, Witzke S, Leier J, et al. Medical cost impact of lumbosacral spine disease. Pain. 1996;65(1):77-85.
intrathecal drug delivery for noncancer pain. Pain Med. 2013;14(4): 97. Son BC, Yoon JH, Kim DR, et al. Dorsal rhizotomy for pain from
504-515. neoplastic lumbosacral plexopathy in advanced pelvic cancer.
71. Smith TJ, Staats PS, Deer T, et al. Randomized clinical trial of Stereotact Funct Neurosurg. 2014;92(2):109-116.
an implantable drug delivery system compared with comprehen- 98. Taha JM, Favre J, Burchiel K. Percutaneous rhizotomy. In:
sive medical management for refractory cancer pain: impact on Grossman RG, Loftus CM, eds. Principles of Neurosurgery. 2nd ed.
pain, drug-related toxicity, and survival. J Clin Oncol. 2002;20(19): Philadelphia: Lippincott-Raven; 1999:435-442.
4040-4049. 99. Saris SC, Silver JM, Vieira JF, et al. Sacrococcygeal rhizotomy for
72. Falco FJ, Patel VB, Hayek SM, et al. Intrathecal infusion systems perineal pain. Neurosurgery. 1986;19(5):789-793.
for long-term management of chronic non-cancer pain: an update 100. Williams JE. Nerve blocks: chemical and physical neurolytic agents.
of assessment of evidence. Pain Physician. 2013;16(suppl):SE185- In: Sykes N, Bennett MI, Yuan C, eds. Cancer Pain. 2nd ed. London:
SE216. Hodder Arnold; 2008:225-234.
73. Dennis GC. DeWitty RL. Long-term intraventricular infusion of 101. Tew JM, Taha JM. Percutaneous rhizotomy in the treatment of
morphine for intractable pain in cancer of the head and neck. Neu- intractable facial pain (trigeminal, glossopharyngeal, and vagal
rosurgery. 1990;26(3):404-407, discussion 407-408. nerves). In: Schmidek HH, Sweet WH, eds. Operative Neurosurgical
CHAPTER 175  Neurosurgical Management of Intractable Pain 1434.e3

Techniques: Indications, Methods, and Results. 3rd ed. Philadelphia: 124. Watling CJ, Payne R, Allen RR, et al. Commissural myelotomy
Saunders; 1995:1469-1484. for intractable cancer pain: report of two cases. Clin J Pain. 1996; 175
102. Rath SA, Seitz K, Soliman N, et al. DREZ coagulations for deaf- 12(2):151-156.
ferentation pain related to spinal and peripheral nerve lesions: indi- 125. Hitchcock E. Stereotactic cervical myelotomy. J Neurol Neurosurg
cation and results of 79 consecutive procedures. Stereotact Funct Psychiatry. 1970;33(2):224-230.
Neurosurg. 1997;68(1-4 Pt 1):161-167. 126. King RB. Anterior commissurotomy for intractable pain. J Neuro-
103. Nashold JRB, Nashold BS Jr. Microsurgical DREZotomy in treat- surg. 1977;47(1):7-11.
ment of deafferentiation pain. In: Schmidek HH, Sweet WH, eds. 127. Gildenberg PL. Myelotomy through the years. Stereotact Funct Neu-
Operative Neurosurgical Techniques: Indications, Methods, and Results. rosurg. 2001;77(1-4):169-171.
3rd ed. Philadelphia: WB Saunders; 1995:1623-1636. 128. Hirshberg RM, Al-Chaer ED, Lawand NB, et al. Is there a pathway
104. Sindou MP. Microsurgical DREZotomy. In: Schmidek HH, Sweet in the posterior funiculus that signals visceral pain? Pain. 1996;
WH, eds. Operative Neurosurgical Techniques: Indications, Methods, and 67(2-3):291-305.
Results. 3rd ed. Philadelphia: WB Saunders; 1995:1613-1621. 129. Gildenberg PL. Brainstem procedures for management of pain. In:
105. Sindou M, Mertens P, Wael M. Microsurgical DREZotomy for pain Winn HR, ed. Youmans Neurosurgical Surgery. Philadelphia: Elsevier;
due to spinal cord and/or cauda equina injuries: long-term results 2004:3073-3083.
in a series of 44 patients. Pain. 2001;92(1-2):159-171. 130. Gybels J, Sweet WH. Neurosurgical Treatment of Persistent Pain:
106. Kanpolat Y, Tuna H, Bozkurt M, et al. Spinal and nucleus caudalis Physiological and Pathological Mechanisms of Human Pain. Basel.
dorsal root entry zone operations for chronic pain. Neurosurgery. Karger; 1989:442.
2008;62(suppl 1):235-242, discussion 242-244. 131. Nauta HJ, Hewitt E, Westlund KN, et al. Surgical interruption of
107. Bullard DE, Nashold BS Jr. The caudalis DREZ for facial pain. a midline dorsal column visceral pain pathway. Case report and
Stereotact Funct Neurosurg. 1997;68(1-4 Pt 1):168-174. review of the literature. J Neurosurg. 1997;86(3):538-542.
108. Gorecki JP, Nashold BS. The Duke experience with the nucleus 132. Kanpolat Y. Percutaneous destructive pain procedures on the upper
caudalis DREZ operation. Acta Neurochir Suppl. 1995;64:128-131. spinal cord and brain stem in cancer pain: CT-guided techniques,
109. Sandwell SE, El-Naggar AO. Nucleus caudalis dorsal root entry indications and results. Adv Tech Stand Neurosurg. 2007;32:147-173.
zone lesioning for the treatment of anesthesia dolorosa. J Neurosurg. 133. Vilela Filho O, Araujo MR, Florencio RS, et al. CT-guided percu-
2013;118(3):534-538. taneous punctate midline myelotomy for the treatment of intrac-
110. Teixeira MJ, de Almeida FF, de Oliveira YS. Fonoff ET. Microen- table visceral pain: a technical note. Stereotact Funct Neurosurg.
doscopic stereotactic-guided percutaneous radiofrequency trigemi- 2001;77(1-4):177-182.
nal nucleotractotomy. J Neurosurg. 2012;116(2):331-335. 134. Fountas KN, Lane FJ, Jenkins PD, et al. MR-based stereotactic
111. Raslan AM, Cetas JS, McCartney S, et al. Destructive procedures mesencephalic tractotomy. Stereotact Funct Neurosurg. 2004;82(5-6):
for control of cancer pain: the case for cordotomy. J Neurosurg. 230-234.
2011;114(1):155-170. 135. Tasker RR. Intracranial ablative procedures for pain. In: Tindall GT,
112. Poletti CE. Open cordotomy and medullary tractotomy. In: Cooper PR, Barrow DL, eds. The Practice of Neurosurgery. Baltimore:
Schmidek HH, Sweet WH, eds. Operative Neurosurgical Technique: Williams & Wilkins; 1996:3115-3128.
Indications, Methods, and Results. 3rd ed. Philadelphia: WB Saunders; 136. Tasker RR. Thalamic stereotaxic procedures. In: Schaltenbrand G,
1995:1557-1571. Walker AE, eds. Stereotaxy of the Human Brain: Anatomical, Physio-
113. Jones B, Finlay I, Ray A, et al. Is there still a role for open cor- logical and Clinical Applications. 2nd, rev. and enl. ed. Stuttgart, West
dotomy in cancer pain management? J Pain Symptom Manage. Germany; New York: Thieme; 1982:484-497.
2003;25(2):179-184. 137. Tasker RR. Thalamotomy. Neurosurg Clin N Am. 1990;1(4):841-864.
114. Kanpolat Y, Ugur HC, Ayten M, et al. Computed tomography- 138. Young RF, Vermeulen SS, Grimm P, et al. Gamma Knife thala-
guided percutaneous cordotomy for intractable pain in malignancy. motomy for the treatment of persistent pain. Stereotact Funct Neu-
Neurosurgery. 2009;64(suppl):ons187-ons193, discussion ons193- rosurg. 1995;64(suppl 1):172-181.
ons194. 139. Young RF. Radiosurgery for pain management. In: Follett KA,
115. Nagaro T, Adachi N, Tabo E, et al. New pain following cordotomy: ed. Neurosurgical Pain Management. Philadelphia: Elsevier; 2004:
clinical features, mechanisms, and clinical importance. J Neurosurg. 237-244.
2001;95(3):425-431. 140. Jeanmonod D, Werner B, Morel A, et al. Transcranial magnetic
116. Kanpolat Y. Percutaneous destructive pain procedures on the resonance imaging-guided focused ultrasound: noninvasive central
upper spinal cord and brain stem in cancer pain: CT-guided tech- lateral thalamotomy for chronic neuropathic pain. Neurosurg Focus.
niques, indications and results. Adv Tech Stand Neurosurg. 2007;32: 2012;32(1):E1.
147-173. 141. Bouckoms AJ. Limbic surgery for pain. In: Wall PD, Melzak R, eds.
117. Tasker RR. Percutaneous cordotomy. In: Schmidek HH, Sweet Textbook of Pain. 3rd ed. Edinburgh: Churchill Livingstone; 1994:
WH, eds. Operative Neurosurgical Techniques: Indications, Methods, and 1171-1187.
Results. 3rd ed. Philadelphia: WB Saunders; 1995:1595-1611. 142. Yen CP, Kung SS, Su YF, et al. Stereotactic bilateral anterior cingu-
118. Jannetta PJ, Gildenberg PL, Loeser JD, et al. Operations on the lotomy for intractable pain. J Clin Neurosci. 2005;12(8):886-890.
brain and brain stem for chronic pain. In: Bonica JJ, ed. The 143. Wilkinson HA, Davidson KM, Davidson RI. Bilateral anterior cin-
Management of Pain. 2nd ed. Philadelphia: Lea & Febiger; 1990: gulotomy for chronic noncancer pain. Neurosurgery. 1999;45(5):1129-
2082-2103. 1134, discussion 1134-1136.
119. Rosomoff HL, Papo I, Loeser JD, et al. Neurological operation on 144. Pereira EA, Paranathala M, Hyam JA, et al. Anterior cingulotomy
the spinal cord. In: Bonica JJ, ed. The Management of Pain. 2nd ed. improves malignant mesothelioma pain and dyspnoea. Br J Neuro-
Philadelphia: Lea & Febiger; 1990:2067-2081. surg. 2014;28(4):471-474.
120. Collins KL, Taren JA, Patil PG. Four-decade maintenance of anal- 145. Kim MC, Lee TK. Stereotactic lesioning for mental illness. Acta
gesia with percutaneous cordotomy. Stereotact Funct Neurosurg. Neurochir Suppl. 2008;101:39-43.
2012;90(4):266-272. 146. Levin AB, Katz J, Benson RC, et al. Treatment of pain of diffuse
121. Meeuse JJ, Vervest AC, van der Hoeven JH, et al. Five-year metastatic cancer by stereotactic chemical hypophysectomy: long
follow-up of a cordotomy. Pain Res Manag. 2008;13(6):506-510. term results and observations on mechanism of action. Neurosurgery.
122. Price C, Pounder D, Jackson M, et al. Respiratory function after 1980;6(3):258-262.
unilateral percutaneous cervical cordotomy. J Pain Symptom Manage. 147. Ramirez LF, Levin AB. Pain relief after hypophysectomy. Neurosur-
2003;25(5):459-463. gery. 1984;14(4):499-504.
123. Raslan AM. Percutaneous computed tomography-guided trans­ 148. Hayashi M, Taira T, Chernov M, et al. Role of pituitary radiosur-
discal low cervical cordotomy for cancer pain as a method to avoid gery for the management of intractable pain and potential future
sleep apnea. Stereotact Funct Neurosurg. 2005;83(4):159-164. applications. Stereotact Funct Neurosurg. 2003;81(1-4):75-83.
Neuromodulation 176

176 Evidence-Based Neurostimulation for Pain


Feridun Acar, Athanasios K. Zisakis, and Selçuk Göçmen

Chronic pain is a major cause of physical and emotional suffering. deafferentation pain, and pain that is secondary to brachial plexus
The medical treatment of patients with chronic pain is rather avulsion.
challenging because serious side effects frequently complicate the It is extremely important to select a correct nuclear
course of medical treatment. Neurostimulation is an intervention target for satisfactory outcomes. Stimulation sites included
that has become increasingly popular lately. However, patient the periventricular/periaqueductal gray matter (PVG/PAG), the
selection is of utmost importance to outcomes, and neuromodu- internal capsule, the sensory thalamus, and the posterior hypo-
lation does have recognized risks and complications, although thalamus. It is important to note that benefit varies depending on
complications very rarely result in long-term morbidity or mor- the etiology, length of follow-up, definition of adequate pain
tality. First, neuromodulation aims to address the neuroanatomic relief, and the site of stimulation.
substrates thought to play a role in the propensity for pain. Levy and colleagues4 concluded that DBS has had its best
Second, neuromodulation offers the ability to titrate the stimula- success in treating cluster headaches and nociceptive syndromes
tion parameters. Third, the nondestructive effect of neuromodu- such as chronic low back pain. Thalamic pain syndrome (prob-
lation is important in minimizing the potential side effects of the ably owing to the frequent loss of the target cells for stimulation),
surgical procedure. Several new therapies and neuroanatomic postherpetic neuralgia, and pain due to spinal cord injury are not
targets have been developed. They include stimulation of the well treated with DBS. DBS continues to play a role in the treat-
central nervous system through intracranial deep brain stimula- ment of chronic pain when other less invasive treatment modali-
tion (DBS), spinal cord stimulation (SCS), and peripheral nerve ties have been exhausted.
stimulation (PNS).
Determination of the appropriateness of using neurostimula-
tion devices for pain is an ongoing and rather evolving process
Motor Cortex Stimulation
that is often clouded by poor data, inaccurate patient selection, Patient selection and target criteria are essential for MCS in
and difficulty in understanding treatment goals. The Interna- patients with refractory neuropathic pain and central pain. MCS
tional Neuromodulation Society (INS) has identified the need is a promising therapy for the treatment of trigeminal neuro-
for better definition of the appropriate application of the use of pathic pain and central pain syndromes. Long-term stimulation
these advanced tools for disease and pain, which has led to the of the precentral cortex for the treatment of pain was first
formation of the Neuromodulation Appropriateness Consensus reported by Tsubokawa and colleagues41 in 1991. MCS is more
Committee (NACC) to evaluate the current literature and best commonly used than DBS because it is more easily performed.
practices and to form an expert opinion on this topic. Thus, this
chapter focuses on evidence-based neurostimulation in the treat-
ment of pain.
Occipital Nerve Stimulation
Occipital nerve stimulation (ONS) is being investigated as a less
invasive and less risky alternative to deep brain intracranial stimu-
INTRACRANIAL NEUROSTIMULATION lation. Studies have reported promising results in small groups of
Intracranial neurostimulation for pain relief has been used effec- patients with cluster headaches. Some patients became pain-free,
tively in the algorithmic treatment of primary chronic headache but others had reduced frequency of headache attacks.
symptoms by stimulating the motor cortex, the sensory thalamus, ONS is the most studied neuromodulation technique for
or the periaqueductal and periventricular gray matter. The stimu- primary headaches. The overall efficacy of this procedure is
lation of these sites through motor cortex stimulation (MCS) and 59%, and it has been effective in a variety of primary headache
DBS has proved effective for treating a number of neuropathic conditions. The conclusion from these studies is that peripheral
and nociceptive pain states that are not responsive or amenable neurostimulation techniques carry an advantage over central
to other therapies or types of neurostimulation. MCS has shown neuromodulation.
particular promise in the treatment of trigeminal neuropathic
pain and central pain syndromes such as thalamic pain syndrome.
On the other hand, DBS may be employed for a number of
Spinal Cord Stimulation
nociceptive and neuropathic pain states, including cluster head- Spinal cord stimulation modifies the perception of pain by stimu-
aches, failed back surgery syndrome, peripheral neuropathic pain, lating the dorsal columns of the spinal cord and may relieve
facial differentiation pain, and pain secondary to brachial plexus neuropathic or ischemic pain.72 Relief of pain from failed back
avulsion. Additionally, SCS in the high cervical (C1-C2) region surgery syndrome (FBSS) appears to respond best to SCS. SCS
has been shown also to impact both headache and facial pain was found to be superior to conventional medical management
syndromes. and reoperation.
A study by Song and associates75 demonstrated that SCS is
significantly superior to conservative medical management and
Deep Brain Stimulation reoperation for dealing with pain from FBSS. Clinical benefit was
DBS has been performed successfully to treat a variety of neuro- also conclusively shown for complex regional pain syndrome,
pathic and nociceptive pain states that are not responsive to critical limb ischemia, and refractory angina pectoris. Percutane-
other neuromodulation techniques, including cluster headaches, ous hybrid paddle leads, peripheral nerve field stimulation,
failed back surgery syndrome, peripheral neuropathic pain, facial nerve root stimulation, dorsal root ganglion, and high-frequency
1435
1436 SECTION 6  Pain

stimulation are actively being refined to address axial low back number of evidence-based studies at level I support the use of
pain and foot pain. High-frequency stimulation is unique in pro- SCS as a basic method for treatment of pain from FBSS.
viding paresthesia-free analgesia by stimulating beyond the phys- Results of studies of PNS and ONS do not reveal strong
iologic frequency range. However, large randomized control evidence-based support of their use as methods for treating pain.
trials demonstrating clear clinical benefit are needed to gain Still, the existing literature in PNS and ONS focuses on level III
evidence-based support for the use of these newer treatments and evidence-based studies, so there is more to be done before con-
approaches. clusions can be drawn about their efficacy in treatment of pain.
On the other hand, however, the studies of DBS and MCS for
treating pain are far from achieving unambiguous evidence for
Peripheral Nerve Stimulation their use as treatment methods. Mainly the studies in these two
Peripheral nerve stimulation targets the peripheral nervous methods are at level III and thus cannot be considered the ideal
system for the control of pain through either surgical placement techniques for treating chronic pain.
of electrodes directly over a peripheral nerve or percutaneous From the literature on neurostimulation as a treatment of
placement of the electrodes sufficiently near the nerve to provide pain, it can be concluded that there are data and studies support-
an effective neuromodulating waveform. ing the use of neurostimulation as a treatment method in special
Some observational studies showed that the peripheral nerve cases of pain but that further studies are needed to support an
stimulation and peripheral nerve field stimulation, two newer epistemologic and clear conclusion concerning the efficacy and
types of neurostimulation, may be combined with SCS to improve importance of neurostimulation as a treatment method in pain.
the outcomes of patients with low back pain.
Full text of this chapter is available online at ExpertConsult.com
CONCLUSION
In areas such as spinal cord stimulation for the treatment of pain,
there clearly is strong evidence for positive results. The large
Neuromodulation 176

176 Evidence-Based Neurostimulation for Pain


Feridun Acar, Athanasios K. Zisakis, and Selçuk Göçmen

Chronic pain is a major cause of physical and emotional suffering. studies of neurostimulation for peripheral neuropathic pain, post-
The medical treatment of patients with chronic pain is rather amputation pain, postherpetic neuralgia, and other causes of
challenging because serious side effects frequently complicate the nerve injury are needed to extend the support of neumodulation
course of medical treatment and some cases may be even intrac- as chronic pain treatment.3 International guidelines recommend
table to medical treatment.1 There is a long history to nerve neuromodulation, and specifically spinal cord stimulation, as
stimulation application, a process known as neurostimulation. treatment for refractory angina and other indications, such as
Neurostimulation is an intervention that has become increasingly congestive heart failure. However, these indications are still
popular lately owing to the growing body of literature showing under investigation. Thus, this chapter focuses on evidence-based
its effectiveness in treating pain and the reversible nature of the neurostimulation in the treatment of pain.
treatment with implant removal. However, patient selection is
of utmost importance to outcomes, and neuromodulation does
have recognized risks and complications, although complications
INTRACRANIAL NEUROSTIMULATION
very rarely result in long-term morbidity or mortality.2 Neuro- Intracranial neurostimulation for pain relief has been used effec-
modulation systems are unique, and it is their treatment effective- tively in the algorithmic treatment of primary chronic headache
ness that is the major advantage for patients treated with them. symptoms by stimulating the motor cortex, the sensory thalamus,
First, neuromodulation aims to address the neuroanatomic sub- or the periaqueductal and periventricular gray matter. The stimu-
strates thought to play a role in the propensity for pain. Second, lation of these sites through motor cortex stimulation (MCS) and
neuromodulation offers the ability to control the strength of DBS has proved effective for treating a number of neuropathic
the stimulation parameters for each case, potentially providing and nociceptive pain states that are not responsive or amenable
improved benefit with minimal side effects. Third, the nonde- to other therapies or types of neurostimulation.4 DBS and MCS,
structive effect of neuromodulation is important in minimizing techniques of intracranial neurostimulation, are commonly used
the potential side effects of the surgical procedure. Neuromodu- to control pain.
lation has proved to be very well tolerated by patients. If adverse MCS has shown particular promise in the treatment of tri-
effects occur, stimulation parameters may be changed, such as geminal neuropathic pain and central pain syndromes such as
decreases in intensity or frequency, and changing the stimulated thalamic pain syndrome. On the other hand, DBS may be
contacts. Another advantage is that there is no deterioration of employed for a number of nociceptive and neuropathic pain
neurologic function. The neuroanatomic targets for these tech- states, including cluster headaches, failed back surgery syndrome
niques vary. (FBSS), peripheral neuropathic pain, facial differentiation pain,
Several new therapies and neuroanatomic targets have been and pain secondary to brachial plexus avulsion.4 Additionally, SCS
developed. They include stimulation of the central nervous in the high cervical (C1-C2) region has been shown also to
system through intracranial deep brain stimulation (DBS), spinal impact both headache and facial pain syndromes.2,3,5,6
cord stimulation (SCS), and peripheral nerve stimulation (PNS).
Many studies, mostly in the form of retrospective case series
and a few prospective series, have consistently shown the benefits
DEEP BRAIN STIMULATION
of neurostimulation therapy in treating intractable chronic pain.1 DBS has been performed successfully to treat a variety of neuro-
Determination of the appropriateness of using neurostimula- pathic and nociceptive pain states that are not responsive to other
tion devices for pain is an ongoing and rather evolving process neuromodulation techniques, including cluster headaches, FBSS,
that is often clouded by poor data, inaccurate patient selection, peripheral neuropathic pain, facial deafferentation pain, and pain
and difficulty in understanding treatment goals. Lately, the Inter- secondary to brachial plexus avulsion.
national Neuromodulation Society (INS) has identified the need It is extremely important to select a correct nuclear target for
for better definition of appropriate application of these advanced satisfactory outcomes. Stimulation sites include the periventricular/
tools for disease and pain, which has led to the formation of periaqueductal gray matter (PVG/PAG), the internal capsule, the
the Neuromodulation Appropriateness Consensus Committee sensory thalamus, and the posterior hypothalamus.7 The use of
(NACC) to evaluate the current literature and best practices and PVG/PAG stimulation is generally recommended for nociceptive
to form an expert opinion on this topic.3 According to their pain, and that of sensory thalamus DBS for deafferentation pain,
conclusions, neurostimulation is rather safe. The reason is that whereas DBS of the posterior hypothalamus is an effective
the procedure is minimally invasive and has reversible character- approach to treat refractory chronic cluster headache.7 It is
istics. It is relatively difficult to compare with medical manage- important to note that benefit varies depending on the etiology,
ment, because conservative management has already failed the length of follow-up, definition of adequate pain relief, and the
patients considered for neurostimulation. Unlike alternative site of stimulation.8
therapies, neurostimulation is not associated with medication- Long-term follow-up results of DBS for a number of indica-
related side effects and has an enduring effect. However, device- tions have been reported by large studies.4,9-32 Katayama and
related complications are still not uncommon. The incidence of associates31 reviewed the effects of DBS and MCS treatments in
complications is dropping as technology progresses and surgical patients with pain after cerebrovascular accidents. DBS of the
skills improve. Randomized controlled studies on spinal cord thalamic nuclei ventralis oralis posterior et intermedius proved
stimulation support its efficacy in treating failed back surgery to be useful in more than 70% of patients who had suffered with
syndrome and complex regional pain syndrome.3 Analogous poststroke involuntary movements.31 However, the results of
e613
e614 SECTION 6  Pain

DBS of the thalamic nucleus ventralis caudalis or internal capsule Owing to the poor overall results reported by several investi-
for poststroke pain were disappointing.31 gators, DBS is not highly recommended for thalamic pain syn-
A meta-analysis performed to determine the long-term effi- drome, postherpetic neuralgia, or pain due to spinal cord injury.4
ciency of DBS for chronic pain found that the success rates varied Implantation of two leads, one in the PAG/PVG and the other
by diagnosis.8 This study showed that in periaqueductal gray in the sensory thalamus, may optimize the chances of achieving
matter (PAG), rate of long-term pain alleviation was highest with satisfactory pain relief.
DBS of the PVG/PAG (79%) or of the PVG/PAG plus the An uncritical review of the literature indicates that DBS
sensory thalamus/internal capsule (87%). Stimulation of the appears to be more effective for certain pain states than others.5
sensory thalamus alone was proven less effective (58% long-term For example, previous studies have concluded that DBS can be
success rate) (P < .05). DBS was more effective, however, for successful in treating specific conditions such as cluster headaches
nociceptive pain than for deafferentation pain (63% versus 47% and nociceptive syndromes such as chronic low back pain.38-40
long-term success rates, respectively; P < .01). Long-term success Long-term success also appears to be achieved more frequently
was attained in more than 80% of patients with intractable low for pain resulting from cervical or brachial avulsion, peripheral
back pain (FBSS) following successful trial stimulation. Trial neuropathy, and FBSS. In contrast, the literature indicates that
stimulation was also successful in approximately 50% of those DBS appears to be less effective for the treatment of thalamic pain
with poststroke pain, and 58% of patients who received perma- syndrome and paraplegia pain. Almost all of the evidence to
nent stimulation implants achieved ongoing pain relief. support the use of DBS is derived from uncontrolled, unblinded
Levy and colleagues34 performed a meta-analysis to determine case series, so this evidence must be interpreted with considerable
the efficacy of DBS for the treatment of chronic pain. They caution.
evaluated 13 series with long-term outcome reports for a total of In 2013, Martelletti and associates1 reviewed and analyzed any
1114 patients.15,26-28,30-37 Of these patients, 561 (50%) had long- currently existing method of neuromodulation for chronic head-
term successful pain relief with DBS. The rates of long-term ache if at least two case series had been published for it. This
success ranged from 19% to 79%, and it appears that there is a paper was therefore not a conventional guideline but an interna-
falloff in success as the length of follow-up increases. Overall, 711 tional expert recommendation based strictly on published evi-
patients had neuropathic pain, of whom 296 (42%) were without dence. Their conclusions are summarized below:
pain at long-term follow-up. Of the 443 patients with nociceptive
pain, 272 (61%) experienced long-term pain relief. Of 409 • General recommendations:
patients in whom the ventral posterolateral nucleus of thalamus • From a medical standpoint, neurostimulators should be
(VPL) was stimulated for neuropathic pain, 228 patients had considered only when all appropriate alternative drug and
long-term pain relief (56%), but none of the 51 patients in whom behavioral therapies as recommended by international
sensory thalamic stimulation was used for nociceptive pain did guidelines have failed and medication overuse headache is
so. A total of 35 out of 155 patients (23%) experienced long-term excluded.
pain relief when the PVG was stimulated for neuropathic pain, • The patient has to be considered to have chronic headache,
whereas 172 out of 291 patients (59%) did so when the same site following the current International Headache Society
was used to treat nociceptive pain. These results support the (IHS) definition and the evaluation has to be done by a
hypothesis that PVG stimulation is the preferred site for nocicep- tertiary care headache center.
tive pain states and that sensory thalamic stimulation is preferable • The patient’s headache has to be medically intractable as
for neuropathic pain treatment. defined by international consensus.
Levy and colleagues4 concluded that DBS appears to be • Procedural recommendations:
more effective for certain pain states than others. Long-term • Hypothalamic stimulation (HS) as a treatment for chronic
success was achieved more frequently for pain resulting from cluster headache (CCH) is invasive and expensive, and
cervical or brachial avulsion, pain due to peripheral neuropathy, should be employed with caution.
and pain in FBSS. DBS, however, appears to be less effective • Occipital nerve stimulation (ONS) is also an expensive and
for the treatment of thalamic pain syndrome and paraplegia invasive technique. ONS may prevent CCH but has no
pain. For other pain states, outcomes reported in the literature acute effect on CCH.
are mixed. • Sphenopalatine ganglion stimulation should be considered
Martelletti and associates,1 reviewing more than 60 articles experimental because there is only one placebo-controlled
about patients undergoing hypothalamic implantation of elec- study in the literature. It may be worthwhile to use this
trodes for cluster headache and other types of trigeminal auto- method for episodic cluster headaches, given the contribu-
nomic cephalalgia for the European Headache Foundation, tion of the parasympathetic system to the origin of cluster
found the overall success rate (patients pain-free or with 50% or headaches.
more improvement) was found to be around 50% to 60%; accu- • Vagal nerve stimulation (VNS) may be employed for
mulated follow-up data made it possible to better understand the chronic headache treatment but only in the context of a
advantages and limitations of the procedure.1 randomized placebo-controlled trial.
Levy and colleagues4 concluded that intracranial stimulation • Transcranial direct current stimulation (TDS) may also be
should be considered only after more conservative therapies, a candidate for a controlled trial.
including less invasive neurostimulation methods, have failed. • Transcranial magnetic stimulation (TMS) appears to be
Like other pain treatments, MCS or DBS must be employed in safe, but further study is necessary to determine whether
light of the circumstances of individual cases and the morbidity it is potentially effective for chronic migraine. However,
associated with alternative treatments, particularly the long-term further studies are important to assess factors underlying
use of opioids. MCS appears to be an appropriate treatment for its therapeutic effect.
neuropathic facial pain, poststroke pain, and chronic pain condi- • Spinal cord stimulation should be avoided in patients with
tions that do not respond to other types of neurostimulation. primary headache syndrome.
However, there may be cases in which MCS is indicated without
being preceded by such neurostimulation, as is the case in patients Subsequently, in 2014, Deer and coworkers2 reported on a
with complete deafferentation conditions. There is no compel- further consensus statement on the clinical use of neuromodula-
ling evidence that MCS is less effective than DBS for treating tion in chronic pain based on theoretical background, clinical
chronic pain. data, and side effect of each method.
CHAPTER 176  Evidence-Based Neurostimulation for Pain e615

days was significantly lower in the ONS group than in the sham
MOTOR CORTEX STIMULATION therapy population (−27.2% vs. −14.9%, respectively). A decrease 176
Patient selection and target criteria are essential for MCS in in migraine-related disability with the application of ONS was
patients with refractory neuropathic pain and central pain. MCS also found. Interestingly, the combination of occipital and supra-
is a promising therapy for the treatment of trigeminal neuro- orbital neurostimulation in an uncontrolled series of 7 patients
pathic pain and central pain syndromes. Long-term stimulation with chronic migraine produced a headache frequency improve-
of the precentral cortex for the treatment of pain was first ment greater than 90% in all patients, but there was no significant
reported by Tsubokawa and colleagues41 in 1991.41,42 MCS is response to either stimulation alone.1
more commonly used than DBS because it is more easily ONS has also been used in drug-refractory chronic cluster
performed.2,3,5,6,43 headache (dCCH), but only open studies have been performed
Previous studies involving MCS have focused on its use in and in smaller groups of patients than for chronic migraine. In
poststroke and trigeminal neuropathic pain, for which there are the three main trials treating a range of 13 to 15 patients, the
few other treatments.4,41-51 Poststroke pain responds well to success rate was slightly more than 60%.1
MCS, with approximately two thirds of patients experiencing Jenkins and colleagues70,71 systematically reviewed the avail-
adequate relief. Some studies have shown that 75% to 100% of able data on neurostimulation for primary headache conditions.
patients with trigeminal neuropathic pain have excellent pain Their first review dealt with the pathophysiology, relative
relief.43,52-56 anatomy, theoretical mechanisms, and history of neurostimula-
tion; and the second review focused on central stimulation
and contained an overall analysis of efficacy, safety, cost, patient
OCCIPITAL NERVE STIMULATION selection, and suggestions for further study based on available
Occipital nerve stimulation (ONS) is being investigated as a less evidence. In their review of 154 articles and abstracts, the inves-
invasive and less risky alternative to deep brain intracranial stimu- tigators found that ONS is the most studied neuromodulation
lation. Studies have reported promising results in small groups of technique for primary headaches. The overall efficacy of this
patients with cluster headaches.60 Some patients became pain- procedure is 59%, and it has been effective in a variety of primary
free, whereas others had reduced frequency of headache attacks. headache conditions. The investigators concluded that peripheral
Burns and associates60 in 2009 presented a study on ONS in neurostimulation techniques carry an advantage over central
14 patients with intractable chronic cluster headache. At a median neuromodulation.
follow-up of 17.5 months (range of follow-up 4-35 months), 10
of the 14 patients reported improvement. Three patients had
improvement of more than 90%, three of them had moderate
SPINAL CORD STIMULATION
improvement (40%-60%), and four patients had mild improve- Spinal cord stimulation modifies the perception of pain by stimu-
ment (20%-30%). lating the dorsal columns of the spinal cord, and may relieve
ONS implantations for 15 patients with intractable CCH neuropathic or ischemic pain.72 Pain from FBSS appears to
were prospectively monitored for up to 5 years (mean 36.8 respond best to SCS. SCS was found to be superior to conven-
months)61. The device was infected in one patient. Of the remain- tional medical management (CMM) and reoperation.73-75
ing 14 patients, nearly 80% had a 90% or greater reduction in Liem and associates,58 in a multicenter, prospective, open-
attack frequency and 60% remained pain-free during long periods label, observational study, reported 50% reduction in back, leg,
(months to years), results that are consistent with those of other and foot pain in 57%, 70%, and 89% of patients, respectively,
studies, indicating that ONS demonstrates rather promising with use of dorsal root ganglion stimulation. Simpson and
results.1,62 Furthermore, a prospective trial of ONS in 13 patients coworkers72 performed a systematic review and economic evalu-
demonstrated that the mean attack frequency and intensity ation of SCS using 6000 studies. Eleven RCTs were included in
decreased by 68% and 49%, respectively.1,63 That study showed the clinical effectiveness: three of neuropathic pain and eight of
that 8 of the 13 patients were able to reduce or even stop their ischemic pain. These reviewers suggested that SCS was more
preventive medications. Burns and colleagues64 performed ONS effective than CMM or reoperation in reducing pain, as shown
in 6 patients with hemicrania continua (range of application was in the study by Song and associates.75 The main limitation of the
6 to 21 months)64 and reported that 4 of them experienced pain ischemic pain trials was the small sample sizes, suggesting that
reduction exceeding 80%. In two reports of the use of ONS in a most of the trials may not have been adequately powered to
total of 9 patients with drug-resistant short-lasting unilateral detect clinically meaningful differences. Trial evidence failed to
neuralgiform headache attacks with conjunctival injection and demonstrate that pain relief in critical limb ischemia (CLI) was
tearing (SUNCT) and 3 with short-lasting unilateral neuralgi- better for SCS than for CMM.72 Simpson and coworkers72 con-
form headache attacks with autonomic symptoms (SUNA), cluded that SCS was effective in reducing the chronic neuro-
the patients experienced at least 50% relief after ONS, and 4 pathic pain of FBSS and complex regional pain syndrome (CRPS)
patients were nearly pain-free after approximately 14 months of type I. In the ischemic pain disorders, development of selection
follow-up.65,66 criteria may be required for CLI, and SCS may have clinical
Only three randomized controlled trials (RCTs) of ONS in benefit for refractory angina in the short term.72
chronic migraine have been published since 2009, and their out- The favorable outcome of SCS was also presented in another
comes overall were disappointing. In the Precision Implanted study of 83 patients in whom high-frequency (10-kHz) SCS
Stimulator for Migraine (PRISM) trial, patients with drug- (HF10) was used.76 The good results in 6 weeks of follow-up were
refractory chronic migraine were treated with ONS or sham sustained at the 24-month follow-up.77 However, without a sepa-
therapy without any significant improvement.67 In the ONSTIM rate comparator, the placebo effect could not be discounted.76,77
trial, 39% of 66 patients treated with active ONS over 3 months Another double-blinded study (N = 40) reported that there was
had at least 50% reduction in headache frequency and/or a no difference between HF5 SCS (5 kHz stimulation) and sham
3-point decrease in intensity scale score, while there was no control after 2 weeks of stimulation.75,78 Sham control (no stimu-
improvement in the nonstimulated group or in those having lation after paresthesia-free stimulation was achieved) was the
preset rather than adjustable stimulation.68 Finally, in a trial control. Assessing the results from these two studies (HF10 SCS
involving 157 patients, the percentage of “responders” did not and HF5 SCS), one can conclude that different frequencies may
significantly differ between active (17.1%) and control (13.5%) have different effects. Furthermore, the study using HF10 SCS
groups (primary end point).69 However, the number of headache looked at 6-month and 24-month results, but the HF5 SCS study
e616 SECTION 6  Pain

looked only at 2-week results, making comparisons between the paresthesia-free analgesia by stimulating beyond the physiologic
two studies more challenging. HF10 SCS is in phase 3 trials in frequency range. However, large RCTs demonstrating clear
the United States and a multicenter, prospective, RCT is under clinical benefit are needed to gain evidence-based support for
way; the results may shed further light on the future of paresthesia- their use.
free, high-frequency analgesia.
Sears and colleagues102 reported better results for SCS, with
only 50% or greater pain relief in 50% of the patients with CRPS,
PERIPHERAL NERVE STIMULATION
at a mean follow-up of 4.4 years. However, Kemler and associ- Peripheral nerve stimulation targets the peripheral nervous
ates79,80 did report that 95% of the patients implanted with an system for the control of pain through either surgical placement
SCS device for RSD/CRPS reported that they would undergo of electrodes directly over a peripheral nerve91 or percutaneous
the treatment again. Ultimately, however, a higher-powered RCT placement of the electrodes sufficiently near the nerve92 to
may be needed to resolve whether SCS is a reasonable approach provide an effective neuromodulating waveform.5
to patients with CRPS, in the long term. Some observational studies showed that PNS and peripheral
Furthermore, SCS for treatment of critical limb ischemia nerve field stimulation (PNfS), two newer types of neurostimula-
appears to have mixed results for pain relief and limb salvage; tion, may be combined with SCS to improve the outcomes of
however, a meta-analysis of several RCTs demonstrated clear patients with low back pain.75,93-96 In these observational studies,
benefit of SCS in limb salvage.81 SCS was reported to be superior baseline VAS was used as the internal control, adding the possibil-
to conservative management for refractory angina pectoris but ity, as previously suggested, that the placebo effect was a con-
equivalent to coronary artery bypass graft surgery (CABG).82-85 founding variable.75
Poor patient selection and indiscriminate use of SCS for a variety The larger area of analgesia provided by PNfS may be due to
of pain conditions were identified as accounting for the low effi- inter-lead stimulation (“cross-talk”) between the two separate
cacy rates (<25%) in early SCS studies.57 Later RCTs have dem- subcutaneous leads, as demonstrated in cadavers by Falco and
onstrated that with careful patient selection, SCS can be clinically colleagues.93 The mechanism for PNfS is unknown but is specu-
efficacious and cost effective in FBSS, CRPS, refractory angina, lated to be similar to that for SCS.75
and critical limb ischemia.33,73,74,80-89 The disciplined pain physi-
cian generally selects SCS for the patient in whom conservative
treatment has failed and has the patient undergo psychological
DISCUSSION
evaluation prior to performing a SCS trial.90 Evidence-based medicine aims to optimize decision making by
Satisfactory pain relief after SCS therapy is supported also by emphasizing the use of evidence from well-designed and well-
a randomized, controlled study (N = 50) that compared the clini- conducted research. Although all medicine based on science has
cal outcomes of SCS and repeated back operations.74 In the trial, some degree of empirical support, evidence-based medicine goes
patients in whom results of the randomly assigned treatment further, classifying evidence by its epistemologic strength and
were unsatisfactory could “cross over” to undergo the other treat- requiring that only the strongest evidence (coming from meta-
ment. It was found that patients treated with SCS reported better analyses, systemic reviews, and randomized controlled trials) can
pain relief and higher satisfaction levels than those treated with yield strong recommendations; weaker evidence (such as from
surgical reoperations, and when given the choice, many patients case-control studies) can yield only weak recommendations.
who received reoperations would have chosen SCS over reopera- Evidence quality can be assessed according to the source type
tion.74,90 Among the 45 patients (90%) available for follow-up, as well as other factors, including statistical validity, clinical rel-
SCS was more successful than reoperation (9 of 19 patients versus evance, currency, and peer-reviewed acceptance. Evidence-based
3 of 26 patients; P < .01). Patients who initially were randomly medicine categorizes different types of clinical evidence and rates
allocated to undergo SCS were significantly less likely to cross or grades them according to the strength of their freedom from
over than were those allocated to reoperation (5 of 24 patients the various biases that beset medical research.
versus 14 of 26 patients; P = .02). Also, patients undergoing Several organizations have developed grading systems for
reoperation required increased opiate analgesics significantly assessing the quality of evidence. This chapter used the grading
more often than those undergoing SCS (P < .025).74 system of the U.S Preventive Services Task Force (USPSTF),
Another study compared the outcomes of CMM alone with which defines the grades or levels as follows103:
CMM plus SCS in 100 patients with FBSS and predominantly
• Level I: Evidence obtained from at least one properly designed
radicular leg pain.73 In the intention-to-treat analysis at 6 months,
randomized controlled trial.
the primary outcome (50% or more pain relief in the legs) was
• Level II-1: Evidence obtained from well-designed controlled
achieved in 24 of the 50 patients in the CMM + SCS group (48%)
trials without randomization.
and 4 of the 50 in the CMM alone group (9%) (P < .001).
• Level II-2: Evidence obtained from well-designed cohort or
Between 6 and 12 months, 5 patients in the CMM + SCS group
case-control analytic studies, preferably from more than one
crossed over to CMM alone, and 32 patients in the CMM alone
center or research group.
group crossed over to CMM + SCS. At 12 months, 27 patients
• Level II-3: Evidence obtained from multiple time series designs
in the CMM + SCS group (32%) had experienced device-related
with or without the intervention. Dramatic results in uncon-
complications. The investigators concluded that CMM plus SCS
trolled trials might also be regarded as this type of evidence.
provides better pain relief and improves health-related quality of
• Level III: Opinions of respected authorities, based on
life and functional capacity in comparison with CMM alone in
clinical experience, descriptive studies, or reports of expert
selected patients with FBSS.73,90
committees.
A study by Song and associates75 demonstrated that SCS is
significantly superior to conservative medical management and
reoperation in patients with pain from FBSS. Clinical benefit was
also conclusively shown for complex regional pain syndrome,
Intracranial Stimulation
critical limb ischemia, and refractory angina pectoris. Percutane- For general studies on intracranial stimulation, five studies were
ous hybrid paddle leads, peripheral nerve field stimulation, nerve examined, of which four can be assigned level I for quality of
root stimulation, dorsal root ganglion, and high-frequency stimu- evidence,2,3,5,6 and the fifth, level II-3.4,34 It could be said that there
lation are actively being refined to address axial low back pain is strong support for the efficacy of neurostimulation for the
and foot pain. High-frequency stimulation is unique in providing treatment of the pain, because the studies are mainly level I.
CHAPTER 176  Evidence-Based Neurostimulation for Pain e617

procedures cannot be recommended as other than an option for


Deep Brain Stimulation patients with chronic pain. 176
Of the studies examining the effect of DBS in the treatment Taking into account the studies on neurostimulation as a treat-
of pain, we found 3 with level I quality of evidence,3,6,8 3 with ment for pain, we can conclude that there are data and studies
level II-3,1,4,8 and 34 with level III.9-28,30-33,36-40,45,46,73,97,98 So it is supporting the use of neurostimulation as a treatment method
obvious that in this area, because there are only a few evidence in special cases of pain but that further studies are needed
level I studies and mainly level III studies, the support for to support an epistemologic and clear conclusion concerning
the efficacy of DBS as treatment method for pain is not strong. the efficacy and importance of neurostimulation as a treatment
DBS for the treatment of chronic pain is a technique that evolved method for pain.
more than 35 years ago, at a time when the level and quality
of medical evidence to support a procedure were not as objective
SUGGESTED READINGS
as they are currently. As a result, effectively no level I studies Bittar RG, Kar-Purkayastha I, Owen SL, et al. Deep brain stimulation
have been performed on these procedures. Thus, we must rely for pain relief: a meta-analysis. J Clin Neurosci. 2005;12:515-519.
on meta-analyses and systematic reviews as the underpinnings Deer TR, Krames E, Mekhail N, et al. The appropriate use of
of our conclusions about the effectiveness of DBS as a pain neurostimulation: new and evolving neurostimulation therapies and
treatment. applicable treatment for chronic pain and selected disease states. Neu-
romodulation. 2014;17:599-615.
Deer TR, Mekhail N, Petersen E, et al. The appropriate use of neuro-
Motor Cortex Stimulation stimulation: stimulation of the intracranial and extracranial space and
head for chronic pain. Neuromodulation. 2014;17:551-570.
A similar pattern of studies was found in the case of MCS. There
Deer TR, Mekhail N, Provenzano D, et al. The appropriate use of neuro-
were 23 studies. Three studies were level I,2,5,6 1 was level II-1,58 stimulation of the spinal cord and peripheral nervous system for
1 was level II-2,59 and two were level II-34,57; the remaining 16 the treatment of chronic pain and ischemic diseases: the neuromodula-
were level III.41-45,47-52,54,56,99,100 Strong support cannot be given to tion appropriateness consensus committee. Neuromodulation. 2014;17:
MCS as a method of treating pain because most of the evidence 515-550.
is level III quality. Deer TR, Mekhail N, Provenzano D, et al. The appropriate use of neuro-
stimulation: avoidance and treatment of complications of neurostimu-
lation therapies for the treatment of chronic pain. Neuromodulation.
Occipital Nerve Stimulation 2014;17:571-598.
Levy R, Deer TR, Henderson J. Intracranial neurostimulation for pain
In the treatment of pain by ONS, however, there were three
control: a review. Pain Physician. 2010;13:157-165.
studies with level I evidence,67-69 two with level II-2,64,70 three with Levy RM, Lamb S, Adams JE. Treatment of chronic pain by deep brain
level II-31,65,66 and four with level III.60-63 The majority of the stimulation: long term follow-up and review of the literature. Neuro-
studies were level III and there are few level I and level II studies, surgery. 1987;21:885-893.
so it could be said that ONS as treatment of pain has stronger Liem L, Russo M, Huygen FJ, et al. A multicenter, prospective trial to
support than MCS, suggesting the role of ONS as a rather effec- assess the safety and performance of the spinal modulation dorsal root
tive pain treatment. ganglion neurostimulator system in the treatment of chronic pain.
Neuromodulation. 2013;16:471-482.

Spinal Cord Stimulation


REFERENCES
Examining the studies on SCS shows that this method is clearly 1. Martelletti P, Jensen RH, Antal A, et al. Neuromodulation of
effective in the treatment of pain. Out of 21 studies, 10 were level chronic headaches: position statement from the European Head-
I in quality of evidence,73,74,78,79,81-84,86,87 2 were level II-1,58,101 4 ache Federation. J Headache Pain. 2013;14:86.
were level II-2,75-77,85 2 were level II-3,57,88 and 3 were level 2. Deer TR, Mekhail N, Petersen E, et al. The appropriate use of
III.80,90,102 These studies support SCS as an important way of neurostimulation: stimulation of the intracranial and extracranial
treating pain. space and head for chronic pain. Neuromodulation. 2014;17(6):551-
570, discussion 570.
3. Deer TR, Mekhail N, Provenzano D, et al. The appropriate use of
Peripheral Nerve Stimulation neurostimulation: avoidance and treatment of complications of
neurostimulation therapies for the treatment of chronic pain. Neu-
Finally in the area of PNS there were eight studies. Two were romodulation. 2014;17(6):571-597, discussion 597-598.
level I in quality of evidence,6,95 two were level II-2,75,96 and four 4. Levy R, Deer TR, Henderson J. Intracranial neurostimulation for
were level III,91-94 again showing rather good support for the pain control: a review. Pain Physician. 2010;13(2):157-165.
efficacy of PNS in pain treatment. 5. Deer TR, Krames E, Mekhail N, et al. The appropriate use of
neurostimulation: new and evolving neurostimulation therapies and
applicable treatment for chronic pain and selected disease states.
CONCLUSION Neuromodulation. 2014;17(6):599-615, discussion 615.
6. Deer TR, Mekhail N, Provenzano D, et al. The appropriate use of
It can be concluded that in areas such as spinal cord stimulation neurostimulation of the spinal cord and peripheral nervous system
for the treatment of pain, there clearly is strong evidence for for the treatment of chronic pain and ischemic diseases: the Neu-
positive results. The large number of evidence-based studies at romodulation Appropriateness Consensus Committee. Neuromodu-
level I support the use of SCS as a basic method for treatment of lation. 2014;17(6):515-550, discussion 550.
pain from FBSS. 7. Cheng CY, Hsing MT, Chen YH, et al. Deep brain stimulation
Results of studies of PNS and ONS do not reveal a strong for Parkinson’s disease using frameless technology. Br J Neurosurg.
evidence-based support for their use as methods for treating pain. 2014;28(3):383-386.
Still, the existing literature in PNS and ONS focuses on level 8. Bittar RG, Kar-Purkayastha I, Owen SL, et al. Deep brain stimula-
III evidence–based studies, and thus there is more to be done tion for pain relief: a meta-analysis. J Clin Neurosci. 2005;12(5):
515-519.
before conclusions can be drawn about their efficacy in treatment 9. Mazars G, Merienne L, Cioloca C. [Treatment of certain types of
of pain. pain with implantable thalamic stimulators]. Neurochirurgie. 1974;
Studies of DBS and MCS for treating pain have not been 20(2):117-124.
conducted by evidentiary standards that stand up to contempo- 10. Mazars GJ. Intermittent stimulation of nucleus ventralis postero-
rary scrutiny. Therefore, until such data is available, these lateralis for intractable pain. Surg Neurol. 1975;4(1):93-95.
e618 SECTION 6  Pain

11. Schvarcz JR. Chronic self-stimulation of the medial posterior infe- 38. Franzini A, Ferroli P, Leone M, et al. Hypothalamic deep brain
rior thalamus for the alleviation of deafferentation pain. Acta Neu- stimulation for the treatment of chronic cluster headaches: a series
rochir Suppl (Wien). 1980;30:295-301. report. Neuromodulation. 2004;7(1):1-8.
12. Turnbull IM, Shulman R, Woodhurst WB. Thalamic stimulation 39. Mogilner AY. Rezaih, A. R., Brain stimulation: current clinical
for neuropathic pain. J Neurosurg. 1980;52(4):486-493. indications and future prospects. Suppl Clin Neurophysiol. 2004;57:
13. Siegfried J. Long Term Results of Intermittent Stimulation of the Sensory 721-732.
Thalamic Nuclei in 67 Cases of Deafferent Pain. New York: Futura 40. Schoenen J, Di Clemente L, Vandenheede M, et al. Hypothalamic
Publishing; 1985:129-143. stimulation in chronic cluster headache: a pilot study of efficacy and
14. Tsubokawa T, Yamamoto T, Katayama Y, et al. Clinical results and mode of action. Brain. 2005;128(Pt 4):940-947.
physiological basis of thalamic relay nucleus stimulation for relief 41. Tsubokawa T, Katayama Y, Yamamoto T, et al. Chronic motor
of intractable pain with morphine tolerance. Appl Neurophysiol. cortex stimulation for the treatment of central pain. Acta Neurochir
1982;45(1-2):143-155. Suppl (Wien). 1991;52:137-139.
15. Young RF, Feldman RA, Kroening R, et al. Electrical Stimulation of 42. Tsubokawa T, Katayama Y, Yamamoto T, et al. Treatment of tha-
the Brain in the Treatment of Chronic Pain in Man. Vol. 6. New York: lamic pain by chronic motor cortex stimulation. Pacing Clin Electro-
Raven Press; 1984:289-303. physiol. 1991;14(1):131-134.
16. Young RF, Kroening R, Fulton W, et al. Electrical stimulation of 43. Nguyen JP, Keravel Y, Feve A, et al. Treatment of deafferentation
the brain in treatment of chronic pain. Experience over 5 years. pain by chronic stimulation of the motor cortex: report of a series
J Neurosurg. 1985;62(3):389-396. of 20 cases. Acta Neurochir Suppl. 1997;68:54-60.
17. Hosobuchi Y, Adams JE, Linchitz R. Pain relief by electrical stimu- 44. Carroll D, Joint C, Maartens N, et al. Motor cortex stimulation for
lation of the central gray matter in humans and its reversal by chronic neuropathic pain: a preliminary study of 10 cases. Pain.
naloxone. Science. 1977;197(4299):183-186. 2000;84(2-3):431-437.
18. Hosobuchi Y, Lamb S, Baskin D. Tryptophan loading may reverse 45. Katayama Y, Fukaya C, Yamamoto T. Poststroke pain control
tolerance to opiate analgesics in humans: a preliminary report. Pain. by chronic motor cortex stimulation: neurological character-
1980;9(2):161-169. istics predicting a favorable response. J Neurosurg. 1998;89(4):
19. Hosobuchi Y, Rossier J, Bloom FE. Oral loading with L-tryptophan 585-591.
may augment the simultaneous release of ACTH and beta- 46. Katayama Y, Tsubokawa T, Yamamoto T. Chronic motor cortex
endorphin that accompanies periaqueductal stimulation in humans. stimulation for central deafferentation pain: experience with bulbar
Adv Biochem Psychopharmacol. 1980;22:563-570. pain secondary to Wallenberg syndrome. Stereotact Funct Neurosurg.
20. Hosobuchi Y, Rossier J, Bloom FE, et al. Stimulation of human 1994;62(1-4):295-299.
periaqueductal gray for pain relief increases immunoreactive 47. Nguyen JP, Lefaucher JP, Le Guerinel C, et al. Motor cortex stimu-
beta-endorphin in ventricular fluid. Science. 1979;203(4377):279- lation in the treatment of central and neuropathic pain. Arch Med
281. Res. 2000;31(3):263-265.
21. Adams JE. Naloxone reversal of analgesia produced by brain stimu- 48. Saitoh Y, Hirano S, Kato A, et al. Motor cortex stimulation for
lation in the human. Pain. 1976;2(2):161-166. deafferentation pain. Neurosurg Focus. 2001;11(3):E1.
22. Akil H, Richardson DE, Hughes J, et al. Enkephalin-like material 49. Saitoh Y, Shibata M, Hirano S, et al. Motor cortex stimulation for
elevated in ventricular cerebrospinal fluid of pain patients after central and peripheral deafferentation pain. Report of eight cases.
analgetic focal stimulation. Science. 1978;201(4354):463-465. J Neurosurg. 2000;92(1):150-155.
23. Amano K, Kitamura K, Kawamura H, et al. Alterations of immu- 50. Smith H, Joint C, Schlugman D, et al. Motor cortex stimulation for
noreactive beta-endorphin in the third ventricular fluid in response neuropathic pain. Neurosurg Focus. 2001;11(3):E2.
to electrical stimulation of the human periaqueductal gray matter. 51. Tsubokawa T, Katayama Y, Yamamoto T, et al. Chronic motor
Appl Neurophysiol. 1980;43(3-5):150-158. cortex stimulation in patients with thalamic pain. J Neurosurg.
24. Richardson DE, Akil H. Long term results of periventricular gray 1993;78(3):393-401.
self-stimulation. Neurosurgery. 1977;1(2):199-202. 52. Ebel H, Rust D, Tronnier V, et al. Chronic precentral stimulation
25. Gybels J. Electrical stimulation of the brain for pain control in in trigeminal neuropathic pain. Acta Neurochir (Wien). 1996;138(11):
human. Verh Dtsch Ges Inn Med. 1980;86:1553-1559. 1300-1306.
26. Ray CD, Burton CV. Deep brain stimulation for severe, chronic 53. Meyerson BA, Boethius J, Carlsson AM. Alleviation of Malignant
pain. Acta Neurochir Suppl (Wien). 1980;30:289-293. Pain by Electrical Stimulation in the Periventricular Periaqueductal
27. Dieckmann G, Witzmann A. Initial and long-term results of deep Region: Pain Relief as Related to Stimulation Sites. Vol. 3. New York:
brain stimulation for chronic intractable pain. Appl Neurophysiol. Raven Press; 1979.
1982;45(1-2):167-172. 54. Meyerson BA, Lindblom U, Linderoth B, et al. Motor cortex stimu-
28. Plotkin R. Results in 60 cases of deep brain stimulation for chronic lation as treatment of trigeminal neuropathic pain. Acta Neurochir
intractable pain. Appl Neurophysiol. 1982;45(1-2):173-178. Suppl (Wien). 1993;58:150-153.
29. Richardson DE. Analgesia produced by stimulation of various sites 55. Nguyen JP, Lefaucheur JP, Decq P, et al. Chronic motor cortex
in the human beta-endorphin system. Appl Neurophysiol. 1982; stimulation in the treatment of central and neuropathic pain. Cor-
45(1-2):116-122. relations between clinical, electrophysiological and anatomical data.
30. Hosobuchi Y. Subcortical electrical stimulation for control of Pain. 1999;82(3):245-251.
intractable pain in humans. Report of 122 cases (1970-1984). 56. Rainov NG, Fels C, Heidecke V, et al. Epidural electrical stimula-
J Neurosurg. 1986;64(4):543-553. tion of the motor cortex in patients with facial neuralgia. Clin Neurol
31. Katayama Y, Yamamoto T, Kobayashi K, et al. Deep brain and Neurosurg. 1997;99(3):205-209.
motor cortex stimulation for poststroke movement disorders and- 57. Cameron T. Safety and efficacy of spinal cord stimulation for the
poststroke pain. Acta Neurochir Suppl. 2003;87:121-123. treatment of chronic pain: a 20-year literature review. J Neurosurg.
32. Hosobuchi Y. Dorsal periaqueductal gray-matter stimulation in 2004;100(3 suppl Spine):254-267.
humans. Pacing Clin Electrophysiol. 1987;10(1 Pt 2):213-216. 58. Liem L, Russo M, Huygen FJ, et al. A multicenter, prospective
33. Kumar K, Wyant GM, Nath R. Deep brain stimulation for control trial to assess the safety and performance of the spinal modulation
of intractable pain in humans, present and future: a ten-year dorsal root ganglion neurostimulator system in the treatment
follow-up. Neurosurgery. 1990;26(5):774-781, discussion 781-782. of chronic pain. Neuromodulation. 2013;16(5):471-482, discussion
34. Levy RM, Lamb S, Adams JE. Treatment of chronic pain by deep 482.
brain stimulation: long term follow-up and review of the literature. 59. Rosenow JM, Stanton-Hicks M, Rezai AR, et al. Failure modes
Neurosurgery. 1987;21(6):885-893. of spinal cord stimulation hardware. J Neurosurg Spine. 2006;5(3):
35. Mazars G. Comparative Study of Electrical Stimulation of Posterior 183-190.
Thalamic Nuclei, Periaqueductal Gray and Other Midline Mesencephalic 60. Burns B, Watkins L, Goadsby PJ. Treatment of intractable chronic
Structures in Man. Vol. 3. New York: Raven Press; 1979. cluster headache by occipital nerve stimulation in 14 patients. Neu-
36. Sweet W. Intracerebral Electrical Stimulation for the Relief of Chronic rology. 2009;72(4):341-345.
Pain. Philadelphia: WB Saunders; 1982:3739-3748. 61. Magis D, Gerardy PY, Remacle JM, et al. Sustained effectiveness of
37. Young RF, Rinaldi PC. Brain Stimulation in Pain. New York: occipital nerve stimulation in drug-resistant chronic cluster head-
Springer-Verlag; 1996. ache. Headache. 2011;51(8):1191-1201.
CHAPTER 176  Evidence-Based Neurostimulation for Pain e619

62. Magis D, Allena M, Bolla M, et al. Occipital nerve stimulation for 81. Ubbink DT, Vermeulen H. Spinal cord stimulation for non-
drug-resistant chronic cluster headache: a prospective pilot study. reconstructable chronic critical leg ischaemia. Cochrane Database Syst 176
Lancet Neurol. 2007;6(4):314-321. Rev. 2013;(2):CD004001.
63. Fontaine D, Christophe Sol J, Raoul S, et al. Treatment of refractory 82. de Jongste MJ, Hautvast RW, Hillege HL, et al. Efficacy of spinal
chronic cluster headache by chronic occipital nerve stimulation. cord stimulation as adjuvant therapy for intractable angina pectoris:
Cephalalgia. 2011;31(10):1101-1105. a prospective, randomized clinical study. Working Group on Neu-
64. Burns B, Watkins L, Goadsby PJ. Treatment of hemicrania conti- rocardiology. J Am Coll Cardiol. 1994;23(7):1592-1597.
nua by occipital nerve stimulation with a bion device: long-term 83. Hautvast RW, Brouwer J, DeJongste MJ, et al. Effect of spinal cord
follow-up of a crossover study. Lancet Neurol. 2008;7(11):1001-1012. stimulation on heart rate variability and myocardial ischemia in
65. Marin JC, Goadsby PJ. Response of SUNCT (Short-Lasting Uni- patients with chronic intractable angina pectoris–a prospective
lateral Neuralgiform Headache with Conjunctival Injection and ambulatory electrocardiographic study. Clin Cardiol. 1998;21(1):
Tearing), SUNA (Short-Lasting Unilateral Neuralgiform Head- 33-38.
aches with Autonomic Symptoms) and Primary Stabbing Head- 84. Hautvast RW, DeJongste MJ, Staal MJ, et al. Spinal cord stimula-
aches to Occipital Nerve Stimulation (ONS). Neurology. 2010;74: tion in chronic intractable angina pectoris: a randomized, controlled
P04.006. efficacy study. Am Heart J. 1998;136(6):1114-1120.
66. Shanahan P, Watkins L, Matharu M. Treatment of medically intrac- 85. Mannheimer C, Eliasson T, Augustinsson LE, et al. Electrical stim-
table short- lasting unilateral neuralgiform headache attacks with ulation versus coronary artery bypass surgery in severe angina pec-
conjunctival injection and tearing (SUNCT) and short-lasting uni- toris: the ESBY study. Circulation. 1998;97(12):1157-1163.
lateral neuralgiform headache attacks with autonomic symptoms 86. Jivegard LE, Augustinsson LE, Holm J, et al. Effects of spinal cord
(SUNA) with occipital nerve stimulation (ONS) in 6 patients. Ceph- stimulation (SCS) in patients with inoperable severe lower limb
alalgia. 2009;29(suppl 1):150. ischaemia: a prospective randomised controlled study. Eur J Vasc
67. Lipton RB, Goadsby PJ, Cady RK, et al. PRISM Study: occipital Endovasc Surg. 1995;9(4):421-425.
nerve stimulation for treatment-refractory migraine. Headache. 87. Klomp HM, Spincemaille GH, Steyerberg EW, et al. Design issues
2010;50:515. of a randomised controlled clinical trial on spinal cord stimulation
68. Saper JR, Dodick DW, Silberstein SD, et al. Occipital nerve stimu- in critical limb ischaemia. ESES Study Group. Eur J Vasc Endovasc
lation for the treatment of intractable chronic migraine headache: Surg. 1995;10(4):478-485.
ONSTIM feasibility study. Cephalalgia. 2011;31(3):271-285. 88. Kumar K, Rizvi S. Cost-effectiveness of spinal cord stimulation
69. Silberstein SD, Dodick DW, Saper J, et al. Safety and efficacy therapy in management of chronic pain. Pain Med. 2013;14(11):
of peripheral nerve stimulation of the occipital nerves for the man- 1631-1649.
agement of chronic migraine: results from a randomized, multi- 89. Suy R, Gybels J, Wan Damme H, et al. Spinal Cord Stimulation for
center, double-blinded, controlled study. Cephalalgia. 2012;32(16): Ischemic Rest Pain. The Belgian Randomized Study. 1st ed. New York:
1165-1179. Springer; 1994.
70. Jenkins B, Tepper SJ. Neurostimulation for primary headache dis- 90. Mekhail NA, Cheng J, Narouze S, et al. Clinical applications of
orders, part 1: pathophysiology and anatomy, history of neuro- neurostimulation: forty years later. Pain Pract. 2010;10(2):103-112.
modulation in headache treatment, and review of peripheral 91. Campbell JN, Long DM. Peripheral nerve stimulation in the treat-
neuromodulation in primary headaches. Headache. 2011;51(8): ment of intractable pain. J Neurosurg. 1976;45(6):692-699.
1254-1266. 92. Monti E. Peripheral nerve stimulation: a percutaneous minimally
71. Jenkins B, Tepper SJ. Neurostimulation for primary headache dis- invasive approach. Neuromodulation. 2004;7(3):193-196.
orders: part 2, review of central neurostimulators for primary head- 93. Falco FJ, Berger J, Vrable A, et al. Cross talk: a new method for
ache, overall therapeutic efficacy, safety, cost, patient selection, and peripheral nerve stimulation. An observational report with cadaveric
future research in headache neuromodulation. Headache. 2011;51(9): verification. Pain Physician. 2009;12(6):965-983.
1408-1418. 94. Hamm-Faber TE, Aukes HA, de Loos F, et al. Subcutaneous stimu-
72. Simpson EL, Duenas A, Holmes MW, et al. Spinal cord stimulation lation as an additional therapy to spinal cord stimulation for the
for chronic pain of neuropathic or ischaemic origin: systematic treatment of lower limb pain and/or back pain: a feasibility study.
review and economic evaluation. Health Technol Assess. 2009;13(17):iii, Neuromodulation. 2012;15(2):108-116, discussion 116-117.
ix-x, 1-154. 95. McRoberts WP, Wolkowitz R, Meyer DJ, et al. Peripheral nerve
73. Kumar K, Taylor RS, Jacques L, et al. Spinal cord stimulation versus field stimulation for the management of localized chronic intrac-
conventional medical management for neuropathic pain: a multi- table back pain: results from a randomized controlled study. Neuro-
centre randomised controlled trial in patients with failed back modulation. 2013;16(6):565-574, discussion 574-575.
surgery syndrome. Pain. 2007;132(1-2):179-188. 96. Mironer YE, Hutcheson JK, Satterthwaite JR, et al. Prospective,
74. North RB, Kidd DH, Farrokhi F, et al. Spinal cord stimulation two-part study of the interaction between spinal cord stimulation
versus repeated lumbosacral spine surgery for chronic pain: a ran- and peripheral nerve field stimulation in patients with low back
domized, controlled trial. Neurosurgery. 2005;56(1):98-106, discus- pain: development of a new spinal-peripheral neurostimulation
sion 106-107. method. Neuromodulation. 2011;14(2):151-154, discussion 155.
75. Song JJ, Popescu A, Bell RL. Present and potential use of spinal 97. Hosobuchi Y. The majority of unmyelinated afferent axons in human
cord stimulation to control chronic pain. Pain Physician. 2014;17(3): ventral roots probably conduct pain. Pain. 1980;8(2):167-180.
235-246. 98. Katayama Y, Yamamoto T, Kobayashi K, et al. Deep brain and
76. Van Buyten JP, Al-Kaisy A, Smet I, et al. High-frequency spinal cord motor cortex stimulation for post-stroke movement disorders and
stimulation for the treatment of chronic back pain patients: results post-stroke pain. Acta Neurochir Suppl. 2003;87:121-123.
of a prospective multicenter European clinical study. Neuromodula- 99. Nguyen JP, Lefaucheur JP, Le Guerinel C, et al. Treatment of
tion. 2013;16(1):59-65, discussion 65-66. central and neuropathic facial pain by chronic stimulation of the
77. Al-Kaisy A, Van Buyten JP, Smet I, et al. Sustained effectiveness of motor cortex: value of neuronavigation guidance systems for the
10 kHz high-frequency spinal cord stimulation for patients with localization of the motor cortex. Neurochirurgie. 2000;46(5):483-491.
chronic, low back pain: 24-month results of a prospective multi- 100. Nguyen JP, Nizard J, Keravel Y, et al. Invasive brain stimulation for
center study. Pain Med. 2014;15(3):347-354. the treatment of neuropathic pain. Nat Rev Neurol. 2011;7(12):
78. Perruchoud C, Eldabe S, Batterham AM, et al. Analgesic efficacy of 699-709.
high-frequency spinal cord stimulation: a randomized double-blind 101. Sun YG, Zhou ZJ, Zhang XM, et al. Muscarine receptor gene
placebo-controlled study. Neuromodulation. 2013;16(4):363-369, dis- expression in brain and peripheral blood lymphocytes of rats pre-
cussion 369. treated with dimethoate for 28 days. Zhonghua Lao Dong Wei Sheng
79. Kemler MA, Barendse GA, van Kleef M, et al. Spinal cord stimula- Zhi Ye Bing Za Zhi. 2004;22(1):7-10.
tion in patients with chronic reflex sympathetic dystrophy. N Engl 102. Sears NC, Machado AG, Nagel SJ, et al. Long-term outcomes of
J Med. 2000;343(9):618-624. spinal cord stimulation with paddle leads in the treatment of
80. Kemler MA, de Vet HC, Barendse GA, et al. Effect of spinal cord complex regional pain syndrome and failed back surgery syndrome.
stimulation for chronic complex regional pain syndrome Type I: Neuromodulation. 2011;14(4):312-318, discussion 318.
five-year final follow-up of patients in a randomized controlled trial. 103. Harris RP, Helford M, Woolf S, et al. A review of the process. Am
J Neurosurg. 2008;108(2):292-298. J Pre Med. 2001;20:21-35.
177 Peripheral Nerve Stimulation for Neuropathic Pain
177

Cristian Gragnaniello, Angela Li Ching Ng, and Andrew C. Zacest

Peripheral nerve stimulation (PNS) involves passing electrical Contraindications include patients on anticoagulants, active
current through peripheral nerve fibers to cause paresthesias in infection at the site of electrode insertion, and major cognitive
the distribution of that nerve to alleviate neuropathic pain, based problems such as refractory depression or major personality
on the gate-control theory of pain, suggested by Melzack and disorder.
Wall.1,2 The concept was initially instigated from an observation
made by Scribonius Largius that electric shocks sent by sea crea-
tures such as the torpedo fish or Mediterranean ray could attenu-
TECHNIQUE
ate pain in patients with gout and headaches.3,4 The PNS device is made up of electrodes inserted along the
The first reports of PNS with implantable devices are even desired peripheral nerve and a generator system that is buried
older than those of spinal cord stimulation (SCS) in 1966 and subcutaneously some distance away. PNS is composed of two
1967. Wall and Sweet stimulated patients with neuropathic pain, different phases—an initial trial followed by a permanent implant.
achieving pain suppression while the stimulator was in function.5 Electrodes can be placed in an open fashion or percutaneously.
Around the same time, Shelden implanted PNS electrodes with Before the development of a percutaneous technique, direct
14,000-Hz stimulation, achieving temporary pain relief.6 nerve exposure and placement of the peripheral nerve electrode
over the nerve was performed under radiologic guidance.
In open surgery the nerve is exposed and a plate-type elec-
THEORY AND APPLICATION trode is placed next to the nerve, surrounded by fascia. Major
According to the gate-control theory of pain, large A delta and peripheral nerves are exposed at their traditional exposure sites
small C nerve fibers carry normal and painful sensations, respec- (Table 177-1 and Fig. 177-1).
tively, to the substantia gelatinosa, where stimulatory and inhibi- The leads for the first part of the procedure are usually kept
tory neurons are activated to transmit sensory messages to the for 2 days, and a chart of pain scores at different stimulation
brain. Pain perception is allowed to pass through this gate only intensities is maintained.
when small nerve fibers are triggered; stimulation by signals from At present the procedure is truly minimally invasive, with a
both types of fibers suppresses the nociceptive input at the spinal sedated patient. A Tuohy needle is used to insert the percutaneous
cord level and reduces the signals to the sensory cortex. leads subcutaneously perpendicularly to the nerve from medial
Wall and Street later tested this theory on themselves by to lateral or vice versa, and the electrode is guided over the region
inserting electrodes in their own infraorbital foramina to stimu- of maximal pain. An ultrasound probe can be used for peripheral
late A delta fibers and found that they had pinprick sensation in nerves that are difficult to locate.
the area stimulated, followed by numbness or paresthesia. Shortly Over the years, delivery of electrical stimuli has improved
after this, the first surgical implantation of electrodes was per- while safety has been maintained. Initially, electrodes were
formed around the ulnar and median nerves of a patient with paddle-like or cylindrical in shape, but these were changed to
complex regional pain syndrome.5 button electrodes or fascial padding after a high rate of fibrosis
Surgical exposure of nerves for implantation of electrodes has was noted after use in surgically exposed nerves. Moreover, the
been a source of major delays in popularizing PNS surgery, and newer electrodes are designed to cover wider surface area of
only when Weiner and Reed developed the percutaneous inser- stimulation and are better insulated for patient safety. They have
tion of electrodes for occipital neuralgia and migraines did PNS also proven to have a lower risk of migration when anchored to
start to become more accepted and widely performed.7-13 Since the skin and produce more targeted stimulation of the required
then, the technique has been used for treatment of occipital nerve if the nerve is below or between two electrodes. With the
headaches, migraines, trigeminal nerve pain, truncal pain and invention of percutaneous PNS the electrodes can be quadripolar
autonomic disruption (as in diaphragmatic palsy), complex pain or octapolar.
syndrome, back pain, and sacroiliac pain and fibromyalgia.12,14-20 The implantable generators can have a primary cell or be
Supraorbital PNS has been used for postherpetic neuralgia and rechargeable. Although rechargeable batteries last longer, they
trigeminal neuralgia.21,22 are usually more expensive and should be inserted only in patients
able to recharge them frequently. Combinations of different leads
and generators are available to suit stimulation requirements.
INDICATIONS After the electrode is implanted, it is tunneled out to connect to
Patient selection is crucial for success of PNS. The primary the external generator. The device can be tested intraoperatively
indication is a description of neuropathic pain by the patient, with and the generator is buried subcutaneously, often in the gluteus
the classic symptoms of hyperalgesia, hyperpathia, hyperesthesia, region, abdominal wall or infraclavicular region, away from joints
and allodynia.23-25 The patient’s pain should also have been refrac- and locations that may reduce mobility. They should also be
tory to previous conservative treatment. The area of pain inserted deep enough to reduce the risk of pressure sores in bed-
described should correspond to nerve root distribution and neu- bound patients.
ropathy confirmed via electromyography (EMG) and somatosen-
sory evoked potentials. A psychological and cognitive assessment
should also be conducted. A test trial is also often performed 7
OUTCOME
to 10 days before implantation of the device to determine the Although studies of PNS for various peripheral nerve locations
likely response to PNS. Patients who benefit from PNS usually remain small, there is evidence to show that in select groups of
experience at least 50% improvement after trial of a temporary patients, PNS can have major effects in reducing neuropathic
device.26 A good response to transcutaneous electrical nerve stim- pain. In a study of PNS with suboccipital stimulators in chronic
ulation (TENS) may also be a good indicator of benefit. migraine by Matharu and colleagues, 50% of patients had
1437
1438 SECTION 6  Pain

3 Lead
4 2
Medial intermuscular Ulnar n.
5
septum
Brachialis m.

C7 Subclavicular
pouch
C8 Struther’s (IPG)
C6 arcade
Skin incision
Ulnar nerve
Medial epicondyle
Arcuate 2 Subcutaneous
C6 1 Cubital Suture tunnel
C8 ligament
tunnel Lead extension in
subcutaneous tunnel to IPG
Ulnar n.
C5 Lead

T1 Ulnar nerve

Lead
A B 3 C
Figure 177-1. Ulnar nerve stimulation. A, Distribution of the ulnar nerve in the right forearm and hand.
B, Surgical anatomy of the approach and placement of the ulnar nerve stimulator. 1, Skin incision, with
musculature, ligaments, and tendons shown. 2, The ulnar nerve is mobilized in the usual fashion and the lead
placed beneath it. 3, The lead is anchored to prevent its mobilization. C, Subcutaneous pacemaker placed
under the right clavicle and with a cord tunneled to the lead at the elbow. IPG, implantable pulse generator.

TABLE 177-1  Major Peripheral Nerves Are Exposed at Their 4.2 ± 2.5 pain scale points on an 11-point scale after PNS and an
Traditional Exposure Sites overall reduction in analgesic use at follow-up 1 to 23 months
later.28
Nerve Exposure
Individual complications after PNS in the aforementioned
Median and ulnar Brachial groove studies included electrode migration, breakage of parts, skin
Radial Spiral groove infection, and muscle spasm that could have required removal and
Common peroneal Deep to the biceps femoris cranial to replacement of the apparatus; but in general, rates of complica-
the popliteal space
tions are low.
Posterior tibial Proximal to the medial malleolus
Sciatic nerve Sciatic notch
FUTURE
There is a lot of positive outcome to gain from PNS in neuro-
pathic pain. There are still unanswered questions on the way PNS
excellent response, with headaches being completely suppressed works to cause analgesia. More studies should be guided toward
with rare occasions of breakthrough headaches; 25% had good patient selection, and more long-term follow-up studies should
response with headaches completely suppressed most of the time be conducted to expand the range of conditions that can be
with a pain reduction of 50% to 75% at follow-up of up to 3 managed by this noninvasive treatment. Another field of expan-
years.20 In 30 patients craniofacial pain, including supraorbital, sion is the devices used; there is increased interest in new elec-
occipital, and infraorbital pain, Slavin and colleagues showed that trodes and the nerves’ response to electrode type and positioning.
16 (53%) experienced more than 50% improvement in pain There are still devices that are not approved for clinical use, and
intensity; three patients (10%) had less than 50% pain improve- we should invest in further research to make these pieces of
ment, and three (10%) had complications from their devices hardware safe for clinical use.
including loss of effect or infection.16 Johnson and colleagues
revealed some successful treatment of trigeminal neuralgia after
SUGGESTED READINGS
postherpetic neuralgia and facial trauma—at least 50% reduction Eisenberg E, Waisbrod H, Gerbeshagen HU. Long term peripheral nerve
in pain in 7 of 10 patients, as well as decrease in medication use.21 stimulation for painful nerve injuries. Clin J Pain. 2004;20:143-146.
In a long-term study of PNS in 30 patients with reflex sympa- Freitas TD, Paiva AV, Ogliari KC, et al. Peripheral nerve stimulation to
thetic dystrophy, 60% experienced good or fair relief and there treat chronic painful syndromes. Rev Dor. 2013;14:315-319.
was an overall 60.9% pain reduction after electrode implantation Johnson MD, Burchiel KJ. Peripheral stimulation for treatment of tri-
in the ulnar, median, radial, and superficial peroneal nerves. In geminal postherpetic neuralgia and trigeminal posttraumatic neuro-
case reports of three patients with intractable postoperative pathic pain: a pilot study. Neurosurgery. 2004;55:135-142.
inguinal pain, all patients had 75% to 100% pain relief up to 12 Slavin KV. History of peripheral nerve stimulation. Prog Neurol Surg.
months after implantation. Paicius and colleagues27 reported on 2011;24:1-15.
Wall PD, Sweet WH. Temporary abolition of pain in man. Science.
six patients with chronic low back pain treated with PNS; in this 1967;155:108-109.
cohort, all patients had less pain and better quality of life. Finally, White JC, Sweet WH. Pain and the Neurosurgeon. A Forty-Year Experience.
in a 100-patient case study of PNS in chronic pain including Springfield, IL: Thomas; 1969:894-899.
craniofacial, thorax, lumbosacral, abdominal, pelvic, and groin
pain conditions, the authors showed an average pain reduction of See a full reference list on ExpertConsult.com
CHAPTER 177  Peripheral Nerve Stimulation for Neuropathic Pain 1438.e1

REFERENCES 16. Slavin KV, Burchiel KJ. Use of long-term nerve stimulation with
1. White JC, Sweet WH. Pain and the Neurosurgeon. A Forty-Year Expe- implanted electrodes in the treatment of intractable craniofacial 177
rience. Springfield, IL: Thomas; 1969:894-899. pain. J Neurosurg. 2000;92:576.
2. Melzack R, Wall PD. Pain mechanisms: a new theory. Science. 1965; 17. Freitas TD, Paiva AV, Ogliari KC, et al. Peripheral nerve stimula-
150:971-979. tion to treat chronic painful syndromes. Rev Dor. 2013;14:315-319.
3. Rode J. Scribonius (Largus), Compositiones Medicae. Padua. 1655; 18. Hassenbusch SJ, Stanton-Hicks M, Schoppa D, et al. Long-term
23-24. results of peripheral nerve stimulation for reflex sympathetic dys-
4. Kellaway P. The part played by electric fish in the early history of trophy. J Neurosurg. 1996;84:415-423.
bioelectricity and electrotherapy. Bull Hist Med. 1946;20:112-137. 19. Thimineur M, De Ridder D. C2 area neurostimulation: a surgical
5. Wall PD, Sweet WH. Temporary abolition of pain in man. Science. treatment for fibromyalgia. Pain Med. 2007;8:639-646.
1967;155:108-109. 20. Matharu MS, Bartsch T, Ward N, et al. Central neuromodulation
6. Shelden CH, Paul F, Jacques DB, et al. Electrical stimulation of the in chronic migraine patients with suboccipital stimulators: a PET
nervous system. Surg Neurol. 1975;4:127-132. study. Brain. 2004;127:220-230.
7. Sweet WH, Wepsic JG. Treatment of chronic pain by stimulation 21. Johnson MD, Burchiel KJ. Peripheral stimulation for treatment of
of fibers of primary afferent neuron. Trans Am Neurol Assoc. 1968; trigeminal postherpetic neuralgia and trigeminal posttraumatic
93:103-107. neuropathic pain: a pilot study. Neurosurgery. 2004;55:135-141.
8. Shealy CN, Mortimer JT, Reswick JB. Electrical inhibition of pain 22. Palmer SC, Jimenez AA. Peripheral nerve stimulation for treatment
by stimulation of the dorsal columns: preliminary clinical report. of postherpetic neuralgia: a case report. Acta Med Acad. 2011;40:
Anesth Analg. 1967;46:489-491. 187-191.
9. Slavin KV. History of peripheral nerve stimulation. Prog Neurol 23. Dworkin RH, Backonja M, Rowbotham MC, et al. Advances in
Surg. 2011;24:1-15. neuropathic pain: diagnosis, mechanisms, and treatment recom-
10. Hanai F. Effect of electrical stimulation of peripheral nerves on mendations. Arch Neurol. 2003;60:1524-1534.
neuropathic pain. Spine. 2000;25:1886-1892. 24. Heavner JE, Racz G, Diede JM. Peripheral nerve stimulation:
11. Ignelzi RJ, Nyquist JK. Direct effect of electrical stimulation on current concepts. In: Waldman SD, Winnie AP, eds. Interventional
peripheral nerve evoked activity: implications in pain relief. J Neu- pain management. Philadelphia: Saunders; 1996:423-425.
rosurg. 1976;45:159-165. 25. Slavin KV. Peripheral nerve stimulation for neuropathic pain. Neu-
12. Weiner RL, Reed KL. Peripheral neurostimulation for control of rotherapeutics. 2008;5:100-106.
intractable occipital neuralgia. Neuromodulation. 1999;2:217-221. 26. Eisenberg E, Waisbrod H, Gerbeshagen HU. Long term peripheral
13. Stanton-Hicks M, Salamon J. Stimulation of the central and periph- nerve stimulation for painful nerve injuries. Clin J Pain. 2004;20:
eral nervous system for the control of pain. J Clin Neurophysiol. 143-146.
1997;14:46-62. 27. Paicius RM, Bernstein CA, Lempert-Cohen C. Peripheral nerve
14. Dunteman E. Peripheral nerve stimulation for unremitting oph- field stimulation for the treatment of chronic low back pain: pre-
thalmic postherpetic neuralgia. Neuromodulation. 2002;5:32-37. liminary results of long-term follow-up: a case series. Neuromodula-
15. Stinson LW Jr, Roderer GT, Cross NE, et al. Peripheral subcuta- tion. 2007;10:279-290.
neous electrostimulation for control of intractable post-operative 28. Verrills P, Vivian D, Mitchell B, et al. Peripheral nerve field stimula-
inguinal pain: a case report series. Neuromodulation. 2001;4:99-104. tion for chronic pain: 100 cases and review of the literature. Pain
Med. 2011;12:1395-1405.
178 Spinal Cord Stimulation
178

Oren Sagher and Emily Lehmann Levin

Since its development more than 40 years ago, spinal cord stimu- data accumulated over the last three decades of the use of SCS
lation (SCS) has been increasingly utilized to treat chronic pain. in humans to further elucidate a possible mechanism.
In one of the early examples of “bench-to-bedside” research
translation, the gate control theory of pain transmission was first
put to use by the development of SCS by Shealy and collegues1,2
Neurotransmitters
in 1967, a mere 2 years after it was proposed. However, pain relief In order to better clarify the mechanism of action of SCS,
through the use of electrical stimulation had not been an entirely investigators have also examined the role of local neurotransmit-
novel idea. For centuries eels, as biologic sources of electrical ters within the spinal cord in pain conduction. Early studies
current, have been used as analgesic aids. This effect was largely explored opioid receptors as the conventional model of pharma-
written off as magical until the work of Melzack and Wall offered cologic analgesia. It was found that administration of the narcotic
a reasonable explanation. The ensuing clinical experience and antagonist naloxone had no effect on the relief of pain by
accumulated body of knowledge constitute one of the most suc- SCS.7 Other studies showed that substance P is increased in the
cessful examples of useful, translational research in the history of cerebrospinal fluid (CSF) following SCS. It has also been noted
neurosurgery. This chapter outlines the proposed mechanisms by that SCS causes a decrease in the release of excitatory amino
which spinal cord stimulation works and discusses contemporary acids, such as glutamate and aspartate, while also increasing
issues and clinical uses of this technology. the release of γ-aminobutyric acid (GABA).6 This finding sug-
gests that neuropathic pain may be a state of imbalance between
excitatory and inhibitory neurotransmitters and that SCS may
MECHANISM OF ACTION restore that balance (Fig. 178-1).5 These data are further cor-
roborated by studies that show that the GABA agonist baclofen
Gate Theory improves the effect of SCS and that GABA antagonists inhibit its
The gate control theory of pain transmission proposed by effect.8 These lines of research offer the possibility of adjunctive
Melzack and Wall3 in 1965 describes a balance between small and pharmacotherapy.
large sensory fibers, with positive and negative feedback loops
controlling the activity of the dorsal horn cells of the spinal cord.
The theory postulates that a predominance of small-diameter
Summary
sensory fiber activity opens “gates” within the dorsal horn of the The mechanisms underlying SCS-induced analgesia are still open
spinal cord and that predominance of large-diameter fiber activ- to question. However, it is clear that the effect is related to modu-
ity closes them. Shealy and associates1,2 reasoned that because lation of signals mediated by dorsally located fibers within the
large fibers have a lower threshold for depolarization by an elec- spinal cord. Neurohumoral changes in the spinal cord may be
trical field applied to a peripheral nerve, they may be recruited secondary to such activity modulation and may explain post-
selectively by an externally applied field. Moreover, because stimulation analgesia, which is quite prominent in some patients.9
large-diameter sensory fibers within peripheral nerves are segre-
gated into the dorsal columns, they may be more selectively
activated by electrical stimulation of the dorsal aspect of the
INDICATIONS AND OUTCOMES
spinal cord. It is for this reason that the primary electrical effect Early uses of SCS spanned the gamut of chronic pain etiologies,
of spinal cord stimulation has been assumed to be mediated by and outcomes were decidedly mixed. Clinicians soon learned that
the dorsal columns. electrical stimulation was well-suited for the treatment of certain
As elegant as it is, the gate control theory alone does not fully diagnoses, whereas its use in others yielded disappointing results.
explain the clinical effect of spinal cord stimulation. For example, With this fact in mind, prior to implantation of a spinal cord
many pain conditions are not effectively treated by SCS, includ- stimulator, one should establish the cause of the patient’s pain.
ing the pain of acute injury.4 On the other hand, SCS is effective There should be appropriate, objective evidence of a pain disor-
at treating hyperalgesia, which is signaled by large fibers. This der for which spinal cord stimulation has been shown to have
feature may indicate that relief of pain by electrical stimulation efficacy. In general terms, SCS has had the most favorable track
is due to frequency-related conduction block acting at primary record for pain conditions marked by nerve injury (so-called
afferent branch points where dorsal column fibers and dorsal neuropathic pain). For an exhaustive description of indications
horn collaterals diverge.5 If so, other mechanisms involving inter- for and outcomes of SCS, the reader is referred to Krames’s10
neurons in the dorsal horn or involving descending fibers or 1999 paper. In this section, we highlight a few of the most
sympathetic mechanisms may exist.6 common diagnoses treated with spinal cord stimulation.
One of the major limitations in our understanding of the
mechanisms underlying spinal cord stimulation has been the
paucity of animal models that reproduce the human chronic pain
Failed Back Surgery Syndrome
condition. Initial animal models utilizing acute tissue injury have Chronic neuropathic pain is most commonly located in the back
given way to those employing peripheral nerve injury, in an effort and legs. Of patients undergoing lumbosacral spine surgery for
to create a reliable chronic pain model. However, it has been treatment of this pain, 10% to 40% eventually have persistent or
difficult to develop a rat model of chronic pain, because rats recurrent pain.11 This postoperative pain is often referred to as
generally recover from painful insults without long-term pain.5 failed back surgery syndrome (FBSS). Patients commonly have com-
Even in humans, peripheral nerve injury does not reliably result plaints of axial low back pain and lower extremity pain. Tradition-
in neuropathic pain, and objective assessment of pain in the rat ally, SCS has been indicated in patients with FBSS in whom the
relies on behavioral changes. We therefore rely largely on the radicular leg symptoms are more severe than the component of
1439
1440 SECTION 6  Pain

GABA GABA
neuron neuron
Afferent Afferent

Ca2+

AM
DA PA Ca2+ Ca2+
NM
A Normal B Peripheral nerve injury

SCS GABA
neuron
Afferent
1

C D SCS

Figure 178-1. Schematic illustration of the postulated effect of spinal cord stimulation (SCS). A, Dorsal
horn neurons under normal conditions. Afferent inputs release excitatory neurotransmitters into the synaptic
cleft. Simultaneously, γ-aminobutyric acid (GABA) neurons are activated exerting both presynaptic and
postsynaptic inhibition, balancing the state of excitation. AMPA, α-amino-3-hydroxy-5-methyl-4-
isoxazolepropionic acid; NMDA, N-methyl-D-aspartate. B, After peripheral nerve injury, GABAergic activity is
reduced and excitatory neurotransmission is enhanced. These changes can give rise to allodynia and
hyperalgesia. C, SCS activates dorsal column fibers, and the resulting nerve activity is antidromically (2)
mediated to neurons in the dorsal horn superficial laminae (3). The paresthesias experienced during SCS are
due to orthodromic activation of the dorsal columns (1). D, SCS-evoked nerve activity induces the release of
GABA from dorsal horn interneurons, reducing hyperexcitability. (Modified from Cui JG, O’Connor WT,
Ungerstedt U, et al. Spinal cord stimulation attenuates augmented dorsal horn release of excitatory amino
acids in mononeuropathy via a GABAergic mechanism. Pain. 1997;73:87.)

axial back pain. Objective evidence for a source of their neuro- the latest data from this trial shows that, at 6 months, health care
pathic pain should be sought, including radiographic evidence of costs were significantly higher in the SCS group, likely in relation
an anatomically successful procedure for herniated disk with an to the costs of the device and implantation. Longer follow-up of
appropriate radiculopathy, matching the pain complaint. these patients is required to assess the true cost of this therapy.
Failed back surgery syndrome is the most common indication The role of SCS in the management of recurrent back and leg
for SCS and has the most evidence supporting its use. The Pro- pain following spine surgery has been examined in a randomized
spective, Randomized, Controlled, Multicenter Study of Patients controlled trial comparing this therapy with repeat lumbar
with Failed Back Surgery Syndrome (PROCESS trial), a large, spine surgery.16 In this series of 42 patients with operable spinal
multicenter, randomized controlled trial of spinal cord stimula- pathology, SCS proved more effective than repeat spine surgery.
tion, has provided some of the best evidence for the use of spinal Moreover, a significant proportion of patients who had been
cord stimulation in failed back surgery syndrome.12-14 The data randomly allocated to spine surgery and had not benefited from
emerging from this ongoing trial, first presented in 2005, reflect it subsequently had good results with SCS. Finally, the cost of
pain relief after and cost-effectiveness of SCS in comparison with therapy with SCS proved lower than that of repeat lumbar spine
conventional medical management. Pain relief was significantly surgery, despite the significant cost of the hardware utilized in
better in patients treated with spinal cord stimulators than in the former.17
those managed conventionally.13 Patients also reported signifi- The latest outcomes data evaluate patients with FBSS involved
cant improvement in health care–related quality of life. However, in workers’ compensation claims. In this challenging population,
CHAPTER 178  Spinal Cord Stimulation 1441

spinal cord stimulation decreased leg pain but failed to reduce are also data suggesting that the predictive value of psychological
opioid dependence or improve disability.18 It is likely that factors testing is low.30 In summary, evidence regarding the usefulness of 178
affecting medication use and disability in patients with chronic psychological screening of patients prior to spinal cord stimula-
pain who are involved in worker’s compensation claims are far tion is mixed. Nevertheless, this type of screening continues to
more complex and multifactorial than can be accounted for by be widely practiced.
the addition of a surgical procedure like SCS.
Early Lead Configurations
Complex Regional Pain Syndrome The initial foray into spinal cord stimulation utilized a small
Spinal cord stimulation has been in use for the treatment of laminectomy for placement of the stimulating electrode into the
complex regional pain syndrome (CPRS). Also known as reflex epidural, endodural, or subarachnoid space. The placement of the
sympathetic dystrophy, CPRS is a condition for which there are electrode with a laminectomy under local anesthesia provided
few effective treatment options. The pathophysiology is unclear, limited access to different levels of the spinal cord, yielding little
with pain, dysfunction, and trophic changes occurring in an intraoperative flexibility in tailoring the stimulation field to the
affected limb after trauma or surgery to that limb.19 Kemler and patient’s pain. The advent in the 1970s of slender electrodes that
associates20 have published 5-year follow-up results of a random- may be inserted percutaneously through a Tuohy needle improved
ized, controlled trial comparing SCS and physical therapy with the maneuverability of the electrode longitudinally along the
physical therapy alone. They found significantly better relief of spinal canal. This development, in turn, has allowed for improved
symptoms in patients undergoing SCS than in those conserva- ability to determine the optimal level for electrode placement,
tively managed during the first 3 years. However, a spontaneous maximizing stimulation in affected areas.
improvement in the control group seems to gradually diminish
the effect of SCS over longer periods, with no statistical differ-
ence between the groups at 5 years. Despite these results, 95%
MODERN-DAY LEAD TYPES
of patients with SCS reported that they would undergo implanta- Contemporary stimulator lead types consist of either insulated
tion again. wires containing anywhere from one to eight contacts at their
tips in a linear array, or paddle-shaped electrodes with two or
three columns of disk-shaped electrodes (Fig 178-2). The con-
Ischemic Pain tacts are composed of a nonferromagnetic alloy such as platinum-
It has been noted that spinal cord stimulation may alter vascular iridium. In general, the insulating material is Silastic (Dow
tone and improve tissue perfusion. SCS is thought to dilate Corning Corp., Carrollton, KY) based in order to maintain
peripheral vasculature through a combination of sympathetic flexibility and durability. The percutaneous, wire-shaped elec-
outflow modulation and antidromic activation of sensory fibers.21 trodes can be placed through a needle under fluoroscopic guid-
The use of SCS in the treatment of peripheral vascular disease ance, giving the surgeon broad access to multiple levels of the
was first proposed in the 1970s.22 Subsequently, placement of the spinal cord in a minimally invasive fashion. However, by virtue
stimulator in the low thoracic or lumbar region of the spine has of their cylindrical shape, these contacts radiate current in all
been shown to be effective in the treatment of pain caused by directions, requiring more energy and potentially stimulating
lower extremity peripheral artery occlusive disease.21,23,24 Place- dorsally located nerve fibers in addition to the ventrally located
ment of the stimulator electrode at higher thoracic levels has spinal cord. The paddle-shaped leads allow for more focused
been used in the treatment of intractable angina, with studies dispersion of current but require a more invasive implantation
showing better exercise tolerance and fewer ST-segment changes procedure.31
with SCS treatment.21 In addition, the effect of cervical SCS on
the cerebral vasculature has been explored, with focus on cerebral
vasospasm following subarachnoid hemorrhage.25 Although there
is evidence that SCS in the setting of subarachnoid hemorrhage
is safe, the growing use of endovascular therapies for ruptured
aneurysms has complicated this practice, likely owing to the fre-
quent use of systemic anticoagulation in affected patients.26,27

Psychological Screening
One of the practical issues in spinal cord stimulation is the
screening of patients. Clearly, not every pain syndrome responds
equally well to SCS. In the same vein, not every patient with
chronic pain responds well to this therapy. In an effort to sort
out who will respond well to SCS, many practitioners have turned
to psychological screening. Aside from its ability to identify
patients with major psychiatric morbidity, psychological screen-
ing has been reported to have some predictive value in identifying
patients who would benefit from the therapy.28 Some studies have
found that it is important to assess the psychological profile of
the patient in order to determine the suitability of SCS for treat-
ment of the pain. In particular, pre-implantation screening pro-
tocols have examined the patient’s overall stability, defensiveness, Figure 178-2. Electrode arrays and generators in contemporary
self-confidence, and self-efficacy. Other aspects examined are the use for spinal cord stimulation. The linear arrays on the left may be
response to and concern about the illness and the ability to cope placed through a Tuohy needle, whereas the flat, paddle-shaped
with setbacks. Family support and a willingness to be an active electrodes (center) require open surgery for placement. Several
participant in his or her care are also important.29 However, the implantable generators made by the various manufacturers are
utility of psychological screening is not uniformly accepted; there pictured on the right.
1442 SECTION 6  Pain

The percutaneous technique was initially used for temporary Surrounding layer Dorsal
trials of stimulation prior to laminectomy and placement of a Vertebral bone
permanent lead. However, the percutaneous leads have since
Lead material Electrode
become popular for long-term use, owing to ease of placement.
The earliest percutaneous electrode had a single contact and
required the placement of multiple leads to achieve bipolar stim- Epidural fat
ulation. In the 1980s, leads were developed with increasing Cerebrospinal fluid Dura
numbers of contacts, providing more maneuverability in stimula- Dorsal mater
tion. Earlier lead designs incorporated large contacts with long columns
intercontact spacing, which were believed to broaden the effect
of stimulation. However, more contemporary lead designs utilize Dorsal
Gray matter
smaller contacts with narrow intercontact spacing. Work with root
finite element modeling of the spinal canal has demonstrated that
this narrow intercontact spacing provides superior targeting of
the dorsal columns of the spinal cord (Fig. 178-3).32 In addition,
it appears that a transverse orientation of anode and cathode
minimizes uncomfortable radicular stimulation and maximizes Vertebral bone
stimulation of the dorsal columns. The “transverse tripole,” Ventral 5 mm
which involves a negative terminal flanked transversely by two A
positive terminals, is currently believed to provide for optimal
stimulation characteristics.33 Accomplishing a transverse orienta-
tion requires that multiple percutaneous leads be placed parallel
to one another or, alternatively, requires the placement of a
paddle electrode with multiple linear electrode arrays.

LAMINECTOMY VERSUS
PERCUTANEOUS PLACEMENT
The ability to implant leads for spinal cord stimulation via a
minimally invasive, percutaneous technique has resulted in a pro-
found rise in the popularity of this therapy over the last 20 years.
However, over the last decade a number of reports have surfaced
suggesting that leads implanted via laminectomy may deliver
better results than those placed percutaneously.31,34 In the early
2000s, a randomized prospective controlled trial was conducted
in order to compare both the technical and clinical results associ-
ated with the percutaneous and laminectomy lead placements. B
Initial technical data, published in 2002, touted the greater insu-
Dorsal
lation of the paddle-style electrodes for longer battery life and
the ability to use lower stimulation amplitudes.31 All patients
participating in the trial initially underwent temporary percuta-
neous lead implantation, followed by random allocation to
permanent implantation of either percutaneously placed or
laminectomy-placed leads. Patients who were able to compare
the two types of leads reported better ratings of paresthesia
coverage of pain as well as improved low back coverage with Spinal
laminectomy-placed electrodes. Clinical data, published in 2005, cord
revealed that this advantage was durable to a follow-up point of
2 years, but the statistical significance disappeared at a mean of
2.9 years of follow-up. The evidence therefore does not clearly
support the theoretical and anecdotal reports of superiority of CSF
laminectomy-placed leads.34 Still, laminectomy placement is
clearly indicated in patients in whom percutaneous implantation
cannot be accomplished because of epidural adhesions and in
patients who have experienced repeated migration of percutane- Ventral
ously placed leads. C
Figure 178-3. A, Transverse section of cervical three-dimensional
GENERATORS model of the spinal canal and surrounding tissues; the cathode is
centered at this section, and dorsal roots are not included in the
Advances in the miniaturization of electronics and battery tech- model. B, Iso-potential lines in unipolar stimulation. C, Iso-current
nology have provided significant improvement in the flexibility of density lines in unipolar stimulation. CSF, cerebrospinal fluid. (Modified
SCS systems. The earliest generators employed radiofrequency from Holsheimer J. Computer modelling of spinal cord stimulation and
transmitters worn externally to energize the stimulator electrode. its contribution to therapeutic efficacy. Spinal Cord. 1998;36:53.)
These have no implanted battery that would require replacement,
and revisions were rarely required. The major disadvantage of
such systems is the need for the patient to wear an external power-
transmitting device, which may have variable communication
with the internal component, altering the perceived stimulation
CHAPTER 178  Spinal Cord Stimulation 1443

amplitude. Radiofrequency-coupled devices are therefore used stimulation programs but do offer a glimpse into the possibilities
infrequently, but are still employed in selected patients with very offered by advances in electrical engineering. Time will tell 178
high energy requirements. whether these devices will improve the efficacy of spinal cord
Implantable pulse generators became available in the 1980s stimulators.
with the advent of lithium ion battery technology (see Fig.
178-2). These have the obvious advantage of providing more
convenience and a better cosmetic result. Battery life in older
STIMULATION PARAMETERS
models is limited, however, and requires balancing stimulation The intensity of stimulation has been shown to improve efficacy
parameters with battery life to achieve the best result. The patient in animal models of neuropathic pain.40,41 Intensity may be
must undergo surgical replacement of the implanted generator varied through changes in amplitude or frequency. Traditionally,
when the battery has been exhausted. spinal cord stimulation frequency has been relatively low, in
Although fully implanted pulse generators now make up the range of 40 to 60 Hz.5 However, exploration of higher-
the lion’s share of systems in clinical use, a new generation frequency stimulation has shown promise. Observational trials
of rechargeable generators has entered the marketplace. These of stimulation at 10 kHz has been shown to be effective in provid-
devices allow patients to enjoy the convenience of an internalized ing pain relief in patients with FBSS.42 In this series, high-
power source, without the need for frequent surgical procedures frequency SCS appeared to provide better treatment of low back
to replace the generator. In order to recharge these generators, pain, which has historically been difficult to treat with conven-
the patient wears a radiofrequency-coupled external recharging tional SCS.
device periodically for several hours. Unfortunately, compliance Unlike conventional SCS, high-frequency stimulation does
with recharging these devices is very important; if completely not cause paresthesias, which some patients find uncomfortable.
depleted, the rechargeable generator may be irreversibly damaged This lack of paresthesias allowed for a double-blinded, random-
and require surgical replacement. The price of rechargeability ized, controlled trial comparing high-frequency SCS with sham
therefore appears to be more significant than patient compliance stimulation.43 This trial was conducted in a group of patients with
in a regular maintenance schedule, interfering with the conve- a history of FBSS. All had previously been successfully treated
nience of this device.35 Therefore, the new, rechargeable genera- with conventional spinal cord stimulation. The primary outcome
tors do not necessarily replace the primary-cell, implantable measured in this study compared current pain control with pain
generators that have dominated this clinical arena. control through conventional SCS. Although it was important as
one of the first blinded trials of SCS, each phase of sham or
stimulation lasted only 2 weeks, and there was no interval between
CONSTANT CURRENT VERSUS CONSTANT VOLTAGE treatments to allow for washout of the effect. Perhaps a longer
The first stimulators to come on the market employed constant- period of evaluation may show a greater difference between the
voltage circuitry to provide stimulation. In this configuration, two SCS methods.
current amplitude may vary depending on changes of impedance
of the surrounding tissues, such as may happen with changes of
position or the development of scar tissue.36 This variation has
SURGICAL TECHNIQUE
been implicated in the unpredictability of stimulation efficacy and
paresthesias.
Electrode Placement
Stimulation efficacy is directly related to current density at the Placement of epidural stimulation electrodes may be accom-
excited tissue. With constant-voltage technology, current peaks plished in a number of ways. In the simplest technique, the
early and decays rapidly in the stimulus pulse, leaving less effec- electrode may be placed percutaneously through a Tuohy needle,
tive time per cycle than with constant-current technology.37 with the leads connected to external a generator that may be
Thus, constant-current stimulation provides more effective exci- either external (temporary) or implanted. At the other end of the
tation of tissues. range, a single-level laminectomy may be performed for place-
Animal models have demonstrated greater efficacy of constant- ment of a paddle-style electrode. All of these procedures are
current stimulation than with constant-voltage stimulation.38 A traditionally performed in a conscious patient, with use of local
randomized, double-blind trial found that patients prefer the anesthetics and mild sedation to allow the patient to assist in
sensation and pain relief obtained with constant-current pro- optimal placement of the stimulating electrode.
gramming.39 Although both treatment conditions led to improve- A trial of stimulation prior to permanent implantation is
ment, this study demonstrated significantly lower pain scores and usually performed to ensure that the effects of spinal cord stimu-
better quality of life with constant-current stimulation than with lation are therapeutic. This trial involves an initial lead placement
in constant-voltage stimulation. with connection to an external generator, allowing for evaluation
of the treatment for several days. The electrodes may then be
used permanently or changed at that time.
PATIENT PROGRAMMERS Determination of the spinal level for electrode insertion and
One of the advantages enjoyed by SCS users is their ability to placement is key to successful stimulation. The location of pain
interact with their therapy. Patient programmers allow the user treated and the type of electrode being placed guide the level of
to modify the stimulation characteristics to optimize pain man- insertion. Needle-type electrodes have more flexibility, in that
agement. Over the years, the programmers have become more they may be advanced a number of levels cranially through the
sophisticated. Initially, patients were given a magnet that simply epidural space to ensure the best stimulation coverage. On the
turned the generator on or off. Contemporary programmers are other hand, paddle-type electrodes may generally be placed only
considerably more sophisticated, enabling the patient to adjust one to two levels cranial to the location of the laminectomy. For
amplitude, frequency, pulse width, and contact polarities. Some upper extremity symptoms, cervical placement is required. One
programmers allow patients to experience multiple stimulation should be mindful of the cervical cord enlargement, and percu-
programs that alternate with one another on a millisecond basis, taneous electrode insertion should ideally be performed below
essentially becoming simultaneous in their effects. Certain gen- the T1-T2 level. A practical rule of thumb is to insert the elec-
erators now have the capability to sense patient position and alter trode at T2-T3 or T3-T4 for upper extremity pathology and at
the program to provide ideal coverage. Such advanced program- L1-L2 or L2-L3 for lower extremity targets. The target level for
ming options have not been shown to be superior to traditional the electrical contacts varies by patient and the pain region being
1444 SECTION 6  Pain

treated. Common target levels for lower extremity coverage ment allows for direct confirmation in the operating room that
range from T8 through T12, spanning the lumbar enlargement the paresthesias cover the region of the patient’s pain. Surgeons
of the cord. Similarly, common levels for upper extremity symp- who advocate for placement with general anesthesia argue that
toms range from C3 to C6, covering the cervical enlargement. certain patients have physical and mental comorbidities that pre-
Placement where the spinal cord is of small caliber may result in clude awake surgery. With trial fluoroscopy to guide localization,
unpleasant local segmental effects. Barolat’s group has published electromyography and somatosensory evoked potentials are used
an exhaustive review of stimulation levels and their physiologic to tailor placement. Testing is performed as in the awake patient,
correlates.44,45 with a standard screener box, but a significantly lower frequency,
Percutaneous placement of spinal cord stimulator electrodes in the 5- to 10-Hz range.49 Each column of the lead is activated
is an attractive option, especially for temporary trial of stimula- independently, and the electromyography response pattern is
tion. The patient is placed in the prone position and biplanar evaluated visually for symmetry. Some published reports show
fluoroscopy is established. Several centimeters of the lead should efficacy of placement with the patient asleep to be equal, and
lie in the epidural space to stabilize the electrode and minimize perhaps superior, to that of awake placement.49,50 One potential
migration. This is best accomplished by entry into the spine at drawback of this method is the lack of ability to detect unpleasant
least two segments below the target stimulation level. The tech- radicular stimulation when the patient is asleep. Unintended
nique for insertion utilizes a Tuohy needle to gain access to the radicular stimulation, which is frequently the amplitude-limiting
epidural space. factor in SCS, is typically detectable when the patient is awake
The epidural space may be identified through the loss-of- during surgery and can report it, allowing for repositioning of the
resistance method. Identification of subarachnoid placement of electrode. Use of general anesthesia prevents this adjustment.
the needle versus epidural placement is important and may be
accomplished in a number of ways. Of course, the appearance of
CSF will indicate violation of the dura. In the absence of CSF
Generator Placement
flow, insertion if a guidewire or the electrode has been inserted Internal pulse generators are implanted in a subcutaneous pocket
in the subarachnoid space, it will glide more easily and the wire in a position where they will not interfere with bony promi-
may be seen to “float” in the CSF on fluoroscopy. Finally, elec- nences. Two common locations are the lower quadrant of the
tronic stimulation within the subarachnoid space elicits stimula- abdomen and the buttock. It is important to consider the ease of
tion response at extremely low thresholds. the patient to access the generator for routine programming.
When the epidural space has been identified, the electrode Also, care should be taken to avoid placing the generator where
may be advanced through the Tuohy needle to the appropriate undue pressure would be placed on it, causing skin breakdown
stimulation position. Care should be taken to recognize ventral from clothing waistbands or sitting. Some practitioners advocate
migration of the electrode, which will be indicated on AP fluo- placing a generator in the region of the buttock, because it is
roscopy by slight lateral deviation followed by medial curvature. more convenient surgically and is cosmetically acceptable to most
Lateral imaging may be useful in determining this issue as well. patients. However, some are data show that such generator place-
The electrode should be secured with multiple points of fixa- ment increases the strain on the leads, raising the propensity for
tion to reduce the chance of dislodgement. Strain-relief loops lead migration or fracture.51
may be utilized around the insertion site to deflect tension away In preparation for tunneling and generator placement, it has
from the trajectory of the electrode. Anchors and nonabsorbable been our practice to close the midline spinal incision and put the
suture are used to fix the electrode to the interspinous ligaments patient in the lateral position under general anesthesia. Any trial
and to the fascia prior to tunneling toward the generator. leads exiting the skin are disconnected from the external genera-
Similarly, the paddle electrode is placed with the patient in the tor and are cut close to the skin. The site is surgically prepared
prone position (Videos 178-1 through 178-5). A fluoroscope is and draped and the subcutaneous pocket is created. The back
positioned anteroposteriorly, and the vertebral level is identified. incision is then reopened and an extension lead is tunneled sub-
Following generous administration of local anesthetic, a midline cutaneously between the two incisions (Videos 178-6 through
incision is made over the interspace through which the electrode 178-11).
will be placed. Unlike in the percutaneous technique, the level of
entry in paddle electrode placement is usually only one or two
segments below the level of planned stimulation. The paraspi-
COMPLICATIONS
nous muscles are cleared from the spinous processes and lamina Spinal cord stimulation has proven to be a safe technique for the
bilaterally, the inferior portion of the cranial lamina is resected, management of chronic neuropathic pain. The incidence of life-
and the ligamentum flavum is carefully removed. The epidural threatening infection is low; the most severe complications
space is explored cranially to ensure there are no adhesions, and require repeat operation, whereas others may simply affect pain
the electrode is placed under visual and anteroposterior fluoro- relief.51,52 Complications may be divided into two categories:
scopic guidance. With the advent of minimally invasive spine technical and biologic. Technical complications relate to both the
surgery techniques,46,47 some writers have advocated their use for implantation technique and long-term durability of the hardware.
electrode placement to decrease postoperative surgical pain and A review of literature found that one of the most frequent com-
recovery time.48 plications is electrode migration. Percutaneously placed elec-
Often at this point, a trial of stimulation is performed to trodes are more susceptible to migration than paddle electrodes.
ensure good coverage of the patient’s pain. Once final placement Paralysis has been reported rarely, usually in relation to the devel-
is determined, the leads are secured to the interspinous ligament opment of epidural abscess.53
and the fascia is closed. Much as with percutaneous leads, strain-
relief loops are made in the subcutaneous space, and the leads are
tightly secured to the fascia.
CAUTIONS AND CONTRAINDICATIONS
The presence of an implanted neurostimulator device warrants
special consideration in medical and day-to-day situations. The
Awake versus Asleep Placement manufacturers of these devices and the U.S. Food and Drug
Debate has emerged about whether electrodes are best placed Administration (FDA) recommend caution with any sources of
intraoperatively with awake examination of the patient or with the electrical or magnetic energy, because they may cause uncon-
use of general anesthesia and neuromonitoring.49,50 Awake place- trolled heating of the electrode, leading to tissue damage. Obvious
CHAPTER 178  Spinal Cord Stimulation 1445

sources include surgical electrocautery and magnetic resonance Holsheimer J, Wesselink WA. Optimum electrode geometry for spinal
imaging (MRI). Although most practitioners advocate avoiding cord stimulation: the narrow bipole and tripole. Med Biol Eng Comput. 178
monopolar cautery in the presence of spinal cord stimulators, it 1997;35:493.
appears that this type of cautery in the operating room may be Kemler M, De Vet H, Barendse G, et al. Effect of spinal cord stimulation
for chronic complex regional pain syndrome Type I: five-year final
used safely; it use requires placing the grounding pad to direct follow-up of patients in a randomized controlled trial. J Neurosurg.
the current field away from the electronic device and keeping 2008;108:292.
the amplitude of electrocautery current at the lowest usable Kumar K, Buchser E, Linderoth B, et al. Avoiding complications from
settings. spinal cord stimulation: practical recommendations from an interna-
tional panel of experts. Neuromodulation. 2007;10:24-33.
Kumar K, Malik S, Demeria D. Treatment of chronic pain with spinal
Compatibility with Magnetic Resonance Imaging cord stimulation versus alternative therapies: cost-effectiveness analy-
Concerns about device failure and uncontrolled electrode heating sis. Neurosurgery. 2002;51:106.
has also prompted practitioners to avoid MRI, which has not been Kumar K, North R, Taylor R, et al. Spinal cord stimulation vs. conven-
tional medical management: a Prospective, Randomized, Controlled,
recommended for patients with any neurostimulator device. A Multicenter Study of Patients with Failed Back Surgery Syndrome
study published in 2007, however, cited no adverse events in a (PROCESS Study). Neuromodulation. 2005;8:213.
series of 31 patients scanned in a 1.5-T MRI machine.54 Device Kumar K, Taylor RS, Jacques L, et al. Spinal cord stimulation versus
manufacturers have made significant efforts to improve MRI conventional medical management for neuropathic pain: a multicentre
safety and compatibility. A new generation of radiofrequency- randomised controlled trial in patients with failed back surgery syn-
shielded stimulator leads has been introduced, promising to allow drome. Pain. 2007;132:179.
whole-body MRI.55 These leads, currently available in limited Kumar K, Wilson JR, Taylor RS, et al. Complications of spinal cord
configurations, hold considerable promise for clinical use, espe- stimulation, suggestions to improve outcome, and financial impact.
cially in cases in which routine MRI is required for disease J Neurosurg Spine. 2006;5:191.
Manca A, Kumar K, Taylor R, et al. Quality of life, resource consumption
surveillance. and costs of spinal cord stimulation versus conventional medical man-
Other sources of radiofrequency energy may occasionally be agement in neuropathic pain patients with failed back surgery syn-
encountered outside the operating room or imaging suite. For drome (PROCESS trial). Eur J Pain. 2008;12:1047-1058.
example, diathermy is a commonly used method of generating Melzack R, Wall PD. Pain mechanisms: a new theory. Science. 1965;
tissue heat, usually in coordination with physical therapy. This 150:971.
modality involves depositing significant energy in soft tissue, North R. Spinal cord stimulation versus reoperation for failed back
energy that may be transmitted to electrical devices, causing surgery syndrome: a prospective, randomized study design. Acta Neu-
uncontrolled heating at the electrical contacts. There have been rochir Suppl. 1995;64:106.
no reports of injury in patients with SCS undergoing diathermy. North RB, Kidd DH, Olin JC, et al. Spinal cord stimulation electrode
design: prospective, randomized, controlled trial comparing percuta-
However, there have been a number of reports of diathermy neous and laminectomy electrodes. Part I: technical outcomes. Neuro-
injury to patients with deep brain stimulators.56,57 Nonmedical surgery. 2002;51:381.
use of radiofrequency energy may also affect spinal neurostimula- North R, Kidd D, Petrucci L, et al. Spinal cord stimulation electrode
tors; patients have been reported to experience alteration in design: a prospective, randomized, controlled trial comparing percu-
stimulation while passing through antitheft devices, and a single taneous with laminectomy electrodes. Part II: clinical outcomes. Neu-
death has been reported under this circumstance.55 It is therefore rosurgery. 2005;57:990.
recommended that patients avoid lingering near these antitheft North RB, Kidd D, Shipley J, et al. Spinal cord stimulation versus reop-
devices. eration for failed back surgery syndrome: a cost effectiveness and cost
utility analysis based on a randomized, controlled trial. Neurosurg
Online. 2007;61:361.
NEW DIRECTIONS North RB, Kidd DH, Wimberly RL, et al. Prognostic value of psycho-
logical testing in patients undergoing spinal cord stimulation: a pro-
The ability to modulate nervous system impulses with the use of spective study. Neurosurgery Online. 1996;39:301.
electrical stimulation is an exciting development that has already Oosterga M, ten Vaarwerk IA, DeJongste MJ, et al. Spinal cord stimula-
resulted in a highly effective treatment modality for many patients tion in refractory angina pectoris—clinical results and mechanisms.
with chronic pain. Present-day systems provide patients with the Z Kardiol. 1997;86(suppl 1):107.
ability to modulate their therapy on the basis of their activity and Perruchoud C, Eldabe S, Batterham AM, et al. Analgesic efficacy of high-
their perceived analgesia in increasingly sophisticated ways. frequency spinal cord stimulation: a randomized double-blind placebo-
However, these systems lack real-time feedback that can sense controlled study. Neuromodulation. 2013;16:363-369. doi:10.1111/
ner.12027.
activity or position, both of which affect stimulation parameters. Schechtmann G, Song Z, Ultenius C, et al. Cholinergic mechanisms
Future iterations of generators may incorporate such closed-loop involved in the pain relieving effect of spinal cord stimulation in a
systems, already found in cardiac pacemakers, to sense patient model of neuropathy. Pain. 2008;132:10.
activity or perhaps even neural signals characteristic of neuro- Shealy CN, Mortimer JT, Reswick JB. Electrical inhibition of pain by
pathic pain and then adjust stimulation parameters accordingly. stimulation of the dorsal columns: preliminary clinical report. Anesth
Analg. 1967;46:489.
Takanashi Y, Shinonaga M. Spinal cord stimulation for cerebral vaso-
SUGGESTED READINGS spasm as prophylaxis. Neurol Med Chir. 2000;40:352.
Barolat G. Experience with 509 plate electrodes implanted epidurally
Tulgar M, He J, Barolat G, et al. Analysis of parameters for epidural spinal
from C1 to L1. Stereotact Funct Neurosurg. 1993;61:60.
cord stimulation. 3. Topographical distribution of paresthesiae—a pre-
Cui JG, O’Connor WT, Ungerstedt U, et al. Spinal cord stimulation
liminary analysis of 266 combinations with contacts implanted in the
attenuates augmented dorsal horn release of excitatory amino acids in
midcervical and midthoracic vertebral levels. Stereotact Funct Neuro-
mononeuropathy via a GABAergic mechanism. Pain. 1997;73:87.
surg. 1993;61:146.
De Andres J, Valía JC, Cerda-Olmedo G, et al. Magnetic resonance
Turner JA, Hollingworth W, Comstock BA, et al. Spinal cord stimulation
imaging in patients with spinal neurostimulation systems. Anesthesiol-
for failed back surgery syndrome: outcomes in a workers’ compensa-
ogy. 2007;106:779.
tion setting. Pain. 2010;148:14-25. doi:10.1016/j.pain.2009.08.014.
De Vries J, De Jongste MJ, Spincemaille G, et al. Spinal cord stimulation
Wall PD, Sweet WH. Temporary abolition of pain in man. Science.
for ischemic heart disease and peripheral vascular disease. Adv Tech
1967;155:108.
Stand Neurosurg. 2007;32:63.
Holsheimer J. Computer modelling of spinal cord stimulation and its
contribution to therapeutic efficacy. Spinal Cord. 1998;36:531. See a full reference list on ExpertConsult.com
CHAPTER 178  Spinal Cord Stimulation 1445.e1

REFERENCES 24. Oosterga M, Vaarwerk ten IA, DeJongste MJ, et al. Spinal cord
1. Shealy CN, Mortimer JT, Reswick JB. Electrical inhibition of pain stimulation in refractory angina pectoris: clinical results and mecha- 178
by stimulation of the dorsal columns: preliminary clinical report. nisms. Zeitschrift für Kardiologie. 1997;86(suppl 1):107-113.
Anesth Analg. 1967;46:489-491. 25. Takanashi Y, Shinonaga M. Spinal cord stimulation for cerebral vaso-
2. Shealy CN, Taslitz N, Mortimer JT, et al. Electrical inhibition of spasm as prophylaxis. Neurol Med Chir (Tokyo). 2000;40:352-356,
pain: experimental evaluation. Anesth Analg. 1967;46:299-305. discussion 356-7.
3. Melzack R, Wall PD. Pain mechanisms: a new theory. Science. 26. Goellner E, Slavin KV. Cervical spinal cord stimulation may prevent
1965;150:971-979. cerebral vasospasm by modulating sympathetic activity of the supe-
4. Kavar B, Rosenfeld JV, Hutchinson A. The efficacy of spinal cord rior cervical ganglion at lower cervical spinal level. Med Hypotheses.
stimulation for chronic pain. J Clin Neurosci. 2000;7:409-413. 2009;73:410-413. doi:10.1016/j.mehy.2009.01.055.
doi:10.1054/jocn.1999.0225. 27. Slavin KV, Vannemreddy PSSV, Goellner E, et al. Use of cervical
5. Schechtmann G, Song Z, Ultenius C, et al. Cholinergic mechanisms spinal cord stimulation in treatment and prevention of arterial vaso-
involved in the pain relieving effect of spinal cord stimulation in a spasm after aneurysmal subarachnoid hemorrhage: technical details.
model of neuropathy. Pain. 2008;10:doi:10.1016/j.pain.2008.03.023. Neuroradiol J. 2011;24:131-135.
6. Cui JG, O’Connor WT, Ungerstedt U, et al. Spinal cord stimulation 28. Doleys DM. Psychological factors in spinal cord stimulation therapy:
attenuates augmented dorsal horn release of excitatory amino acids brief review and discussion. Neurosurg Focus. 2006;21:E1.
in mononeuropathy via a GABAergic mechanism. Pain. 1997;73: 29. Kumar K, Hunter G, Demeria D. Spinal cord stimulation in treat-
87-95. ment of chronic benign pain: challenges in treatment planning and
7. Freeman TB, Campbell JN, Long DM. Naloxone does not affect present status, a 22-year experience. Neurosurgery. 2006;58:481-496,
pain relief induced by electrical stimulation in man. Pain. 1983;17: discussion 481-96. doi:10.1227/01.NEU.0000192162.99567.96.
189-195. 30. North RB, Kidd DH, Wimberly RL, et al. Prognostic value of
8. Lind G, Meyerson B, Winter J, et al. Intrathecal baclofen as adjuvant psychological testing in patients undergoing spinal cord stimula-
therapy to enhance the effect of spinal cord stimulation in neuro- tion: a prospective study. Neurosurgery. 1996;39:301-310, discussion
pathic pain: a pilot study. Eur J Pain. 2004;8:377-383. doi:10.1016/ 310-11.
j.ejpain.2003.11.002. 31. North RB, Kidd DH, Olin JC, et al. Spinal cord stimulation elec-
9. Wall PD, Sweet WH. Temporary abolition of pain in man. Science. trode design: prospective, randomized, controlled trial comparing
1967;155(758):108-109. percutaneous and laminectomy electrodes. Part I: technical out-
10. Krames E. Spinal cord stimulation: indications, mechanism of action, comes. Neurosurgery. 2002;51:381-389, discussion 389-90.
and efficacy. Curr Rev Pain. 1999;3:419-426. 32. Holsheimer J. Computer modelling of spinal cord stimulation
11. Kumar K, Malik S, Demeria D. Treatment of chronic pain with spinal and its contribution to therapeutic efficacy. Spinal Cord. 1998;36:
cord stimulation versus alternative therapies: cost-effectiveness anal- 531-540.
ysis. Neurosurgery. 2002;51:106-115, discussion 115-116. 33. Holsheimer J, Wesselink WA. Optimum electrode geometry for
12. Kumar K, North R, Taylor R, et al. Spinal cord stimulation vs. con- spinal cord stimulation: the narrow bipole and tripole. Med Biol Eng
ventional medical management: a Prospective, Randomized, Con- Comput. 1997;35:493-497.
trolled, Multicenter Study of Patients with Failed Back Surgery 34. North RB, Kidd DH, Petrucci L, et al. Spinal cord stimulation elec-
Syndrome (PROCESS Study). Neuromodulation. 2005;8:213-218. trode design: a prospective, randomized, controlled trial comparing
13. Kumar K, Taylor RS, Jacques L, et al. Spinal cord stimulation percutaneous with laminectomy electrodes. Part II: clinical out-
versus conventional medical management for neuropathic pain: a comes. Neurosurgery. 2005;57:990-996, discussion 990-6.
multicentre randomised controlled trial in patients with failed back 35. Hornberger J, Kumar K, Verhulst E, et al. Rechargeable spinal cord
surgery syndrome. Pain. 2007;132:179-188. doi:10.1016/j.pain.2007 stimulation versus non-rechargeable system for patients with failed
.07.028. back surgery syndrome: a cost-consequences analysis. Clin J Pain.
14. Manca A, Kumar K, Taylor R, et al. Quality of life, resource con- 2008;24:244-252. doi:10.1097/AJP.0b013e318160216a.
sumption and costs of spinal cord stimulation versus conventional 36. Manola L, Holsheimer J, Veltink P. Technical performance of percu-
medical management in neuropathic pain patients with failed back taneous leads for spinal cord stimulation: a modeling study. Neuro-
surgery syndrome (PROCESS trial). Eur J Pain. 2008;12:1047-1058. modulation. 2005;8:88-99. doi:10.1111/j.1525-1403.2005.00224.x.
doi:10.1016/j.ejpain.2008.01.014. 37. Merrill DR, Bikson M, Jefferys JGR. Electrical stimulation of excit-
15. Deleted in review. able tissue: design of efficacious and safe protocols. J Neurosci
16. North RB, Kidd DH, Piantadosi S. Spinal cord stimulation versus Methods. 2005;141:171-198. doi:10.1016/j.jneumeth.2004.10.020.
reoperation for failed back surgery syndrome: a prospective, random- 38. Qin C, Martinez M, Tang R, et al. Is constant current or constant
ized study design. Acta Neurochir Suppl. 1995;64:106-108. voltage spinal cord stimulation superior for the suppression of noci-
17. North RB, Kidd D, Shipley J, et al. Spinal cord stimulation versus ceptive visceral and somatic stimuli? A rat model. Neuromodulation.
reoperation for failed back surgery syndrome. Neurosurgery. 2007;61: 2012;15:132-143. doi:10.1111/j.1525-1403.2012.00431.x.
361-369. doi:10.1227/01.NEU.0000255522.42579.EA. 39. Washburn S, Catlin R, Bethel K, et al. Patient-perceived differences
18. Turner JA, Hollingworth W, Comstock BA, et al. Spinal cord stimu- between constant current and constant voltage spinal cord stimula-
lation for failed back surgery syndrome: Outcomes in a workers’ tion systems. Neuromodulation. 2013;17:28-36. doi:10.1111/ner.12085.
compensation setting. Pain. 2010;148:14-25. doi:10.1016/j.pain.2009 40. Shechter R, Yang F, Xu Q, et al. Conventional and kilohertz-
.08.014. frequency spinal cord stimulation produces intensity- and frequency-
19. Turner JA, Loeser JD, Deyo RA, et al. Spinal cord stimulation for dependent inhibition of mechanical hypersensitivity in a rat model
patients with failed back surgery syndrome or complex regional pain of neuropathic pain. Anesthesiology. 2013;119:422-432. doi:10.1097/
syndrome: a systematic review of effectiveness and complications. ALN.0b013e31829bd9e2.
Pain. 2004;108:137-147. doi:10.1016/j.pain.2003.12.016. 41. Sato KL, Johanek LM, Sanada LS, et al. Spinal cord stimulation
20. Kemler MA, De Vet HCW, Barendse GAM, et al. Effect of spinal reduces mechanical hyperalgesia and glial cell activation in animals
cord stimulation for chronic complex regional pain syndrome with neuropathic pain. Anesth Analg. 2014;118:464-472. doi:10.1213/
Type I: five-year final follow-up of patients in a randomized con- ANE.0000000000000047.
trolled trial. J Neurosurg. 2008;108:292-298. doi:10.3171/jns.2008.108 42. Al-Kaisy A, Van Buyten J-P, Smet I, et al. Sustained effectiveness of
.issue-2. 10 kHz high-frequency spinal cord stimulation for patients with
21. Wu M, Linderoth B, Foreman RD. Putative mechanisms behind chronic, low back pain: 24-month results of a prospective multicenter
effects of spinal cord stimulation on vascular diseases: a review of study. Pain Med. 2014;15:347-354. doi:10.1111/pme.12294.
experimental studies. Auton Neurosci. 2008;138:9-23. doi:10.1016/ 43. Perruchoud C, Eldabe S, Batterham AM, et al. Analgesic efficacy
j.autneu.2007.11.001. of high-frequency spinal cord stimulation: a randomized double-
22. Falowski S, Celii A, Sharan A. Spinal cord stimulation: an update. blind placebo-controlled study. Neuromodulation. 2013;16:363-369.
Neurother. 2008;5:86-99. doi:10.1016/j.nurt.2007.10.066. doi:10.1111/ner.12027.
23. De Vries J, De Jongste MJL, Spincemaille G, et al. Spinal cord 44. Tulgar M, He J, Barolat G, et al. Analysis of parameters for epidural
stimulation for ischemic heart disease and peripheral vascular disease. spinal cord stimulation. 3. Topographical distribution of paresthesiae—
Adv Tech Stand Neurosurg. 2007;32:63-89. a preliminary analysis of 266 combinations with contacts implanted
1445.e2 SECTION 6  Pain

in the midcervical and midthoracic vertebral levels. Stereotact Funct 51. Kumar K, Buchser E, Linderoth B, et al. Avoiding complications
Neurosurg. 1993;61:146-155. from spinal cord stimulation: practical recommendations from an
45. Barolat G. Experience with 509 plate electrodes implanted epidurally international panel of experts. Neuromodulation. 2007;10:24-33.
from C1 to L1. Stereotact Funct Neurosurg. 1993;61:60-79. 52. Kumar K, Wilson JR, Taylor RS, et al. Complications of spinal cord
46. Beems T, van Dongen RTM. Minimally invasive placement of epi- stimulation, suggestions to improve outcome, and financial impact.
dural plate electrodes under local anaesthesia in spinal cord stimula- J Neurosurg Spine. 2006;5:191-203. doi:10.3171/spi.2006.5.3.191.
tion. Acta Neurochir Suppl. 2007;97:105-109. 53. Arxer A, Busquets C, Vilaplana J, et al. Subacute epidural abscess after
47. Johnson MR, Tomes DJ, Treves JS, et al. Minimally invasive implan- spinal cord stimulator implantation. Eur J Anaesthesiol. 2003;20:
tation of epidural spinal cord neurostimulator electrodes by using a 755-757.
tubular retractor system: technical note. J Neurosurg. 2004;100:1119- 54. De Andres J, Valía JC, Cerda-Olmedo G, et al. Magnetic resonance
1121. doi:10.3171/jns.2004.100.6.1119. imaging in patients with spinal neurostimulation systems. Anesthesiol-
48. Vangeneugden J. Implantation of surgical electrodes for spinal cord ogy. 2007;106:779-786. doi:10.1097/01.anes.0000264776.17533.06.
stimulation: classical midline laminotomy technique versus minimal 55. Medtronic for Healthcare Professionals: Spinal cord stimulation: MRI
invasive unilateral technique combined with spinal anaesthesia. Acta guidelines. <http://professional.medtronic.com/wcm/groups/mdtcom
Neurochir Suppl. 2007;97:111-114. _sg/@mdt/@neuro/documents/documents/scs-mri-guidelines
49. Mammis A, Mogilner AY. The use of intraoperative electrophysi- -us.pdf>. Accessed June 14, 2014.
ology for the placement of spinal cord stimulator paddle leads 56. Nutt JG, Anderson VC, Peacock JH, et al. DBS and diathermy inter-
under general anesthesia. Neurosurgery. 2012;70:ons230-ons236. action induces severe CNS damage. Neurology. 2001;56:1384-1386.
doi:10.1227/NEU.0b013e318232ff29. 57. U.S. Food and Drug Administration. FDA public health notification: dia-
50. Falowski SM, Celii A, Sestokas AK, et al. Awake vs. asleep placement thermy interactions with implanted leads and implanted systems with
of spinal cord stimulators: a cohort analysis of complications leads. <http://www.fda.gov/medicaldevices/safety/alertsandnotices/
associated with placement. Neuromodulation. 2010;14:130-135. doi:10 publichealthnotifications/ucm062167.htm>; 2002 Accessed 26.10.15.
.1111/j.1525-1403.2010.00319.x.
Destructive Procedures

179 Evidence Base for Destructive Procedures


Justin S. Cetas, Amr AlBakry, Ahmed M. Raslan, and Kim J. Burchiel

The surgical treatment of pain has been an integral part of neu- Evidence was classified according to the hierarchy of clinical
rosurgery since the early 20th century, when Harvey Cushing research design and the grading system used by the U.S. Preven-
pioneered the ganglionectomy for trigeminal neuralgia (TN). tive Services Task Force,14 as follows:
Over the ensuing years as anatomic and physiologic knowledge
I: Evidence obtained from at least one properly randomized con-
of pain systems grew, new techniques aimed at new targets were
trolled trial.
developed for an array of pain conditions.1-4 Targets have included
II-I: Evidence obtained from well-designed controlled trials
the anterolateral quadrant of the spinal cord, the sensory and
without randomization.
sympathetic ganglia, the dorsal and ventral spinal roots, the dorsal
II-2: Evidence obtained from well-designed cohort or case-
root entry zone (DREZ) and decussating extralemniscal fibers,
control analytic studies, preferably from more than one center
the hypophysis, the thalamus, and the cingulate gyrus. Careful
or research group.
detailed descriptions of these techniques and their purported
II-3: Evidence obtained from multiple time series with or without
efficacy from pioneers in the field established the importance of
intervention, comparisons between times or places with or
surgery for the treatment of pain. The invasive nature of these
without the intervention. Dramatic results in uncontrolled
techniques and associated comorbidities, as well as the high
experiments (such as the results of treatment with penicillin
degree of technical skill required for their use, led to the develop-
in the 1940s) could also be included in this category.
ment and widespread adoption of newer pharmaceuticals and
III: Opinions of respected authorities, based on clinical experi-
nondestructive treatments. As a result, the use of destructive tech-
ence, descriptive studies, or reports of expert committees.
niques in the surgical treatment of pain has dramatically declined
over the last few decades. They are being or have been replaced In this edition, we also review later peer-reviewed English litera-
with nondestructive techniques and devices, such as spinal cord ture from January 2009 to December 2014 for both malignant
stimulation (SCS), intrathecal opiate pumps, newer medications, and nonmalignant pain etiologies. Additional studies have been
and high-dose opiates. However, it is becoming increasingly cataloged and the data added to tables.
apparent that none of these techniques is free from its own set of
detriments. For example, the long-term administration of opioids
is associated with addiction, opioid-induced hyperalgesia, cogni-
tive disorders, high costs, and the suppression of the immune and
RESULTS
reproductive systems.5,6 Furthermore, it has yet to be proven that In total, 182 papers on noncancer pain and 135 papers on cancer
the long-term use of opioids improves such outcomes as pain pain were reviewed; few articles were included in both noncancer
relief, functional capacity, and health-related quality of life.7,8 and cancer pain reviews. The large majority of studies constituted
Neurostimulation and intrathecal drug administration are class III evidence. Only 25 class I or class II studies were found.
expensive and require ongoing battery and catheter maintenance Seventeen of these studies evaluated radiofrequency (RF) rhi-
and pump refills.9 Despite their widespread use and study, their zotomies for different pain etiologies, such as lumbar facet syn-
clinical efficacy has not been demonstrated according to modern drome, cervical facet pain, and type I (typical) TN, whereas three
standards of evidence-based medicine.10 examined ganglionectomies for either lumbar or cervical pain.
Given that all three broad approaches to the treatment of pain Three studies, two class I and one class II, evaluated and sup-
(destructive, nondestructive, and medical) have “matured” and ported sympathectomy of visceral cancer pain. One evaluated the
the initial wide-eyed enthusiasm of their introduction is over, it use of cordotomy in malignant cancer states, and one evaluated
now seems timely to revisit the existing literature and see how DREZotomy technique in the management of intractable bra-
the older destructive techniques compare. By so doing we can chial plexus avulsion pain.
develop and implement those techniques that are effective and
relegate to history those that are not.
In this chapter we summarize the published data on destruc-
tive procedures for both malignant and nonmalignant causes of
Rhizotomy
pain in order to determine the level of evidence supporting con- Our database search and secondary review of references
tinued use and help define areas that warrant further study. located 56 rhizotomy studies that generally concerned spinal
Therefore we reviewed human clinical studies that reported out- or facet pain and facial pain syndromes (eTables 179-1A
comes for destructive techniques used in the treatment of non- through F), and 11 papers that addressed cancer pain (eTable
malignant pain conditions. Reviewed studies were grouped 179-1G). The majority of the studies (43/56) reported data
according to the surgical target, beginning with peripheral or relevant to either trigeminal neuralgia (eTable 179-1A, 33
“first-order neuron” targets.11 studies15-46) or lumbar facet syndrome (eTable 179-1B, 15
studies47-61), whereas the remainder evaluated the effects of rhi-
zotomy on a variety of truncal and extremity neuralgias (eTable
METHODS 179-1C, 1 study62), cervical pain (eTable 179-1D, 9 studies63-71),
In the sixth edition of this book, we reviewed peer-reviewed cluster headaches (eTable 179-1E, 3 studies72-74), vagoglossopha-
English literature that encompassed January1954 to July 200812 ryngeal neuralgia (eTable 179-1F, 1study75), or cancer pain of
applied to both malignant and nonmalignant pain etiologies.13 various etiologies (eTable 179-1G76-84).
1446
CHAPTER 179  Evidence Base for Destructive Procedures 1447

Rhizotomy for Truncal or Extremity Neuralgias and Overall, these studies demonstrate a significant long-term
benefit of rhizotomy for TN. Reported recurrence rates con- 179
Lumbar Facet Syndrome curred, ranging from 40% to 60% at 5 years. Several investigators
In the studies on rhizotomy for truncal or extremity neuralgias have suggested that patients with facial pain other than typical or
and lumbar faced syndrome, success rates were initially around type I TN86 had higher recurrence rates or poorer patient satis-
60% but declined on long-term follow-up. Despite the variability faction.22,23,31 (See eTable 179-1A, 20 studies.15-31,33-46,87)
in treated pain syndromes, number of sectioned roots, outcome
measures, and follow-up, the results were discouraging enough
that the procedure was relatively abandoned and replaced with
Neurectomy for Trigeminal Neuralgia
modified rhizotomies directed at facet denervation for the treat- Five additional case series concerning the treatment of TN15-19
ment of lumbar facet syndrome.52 were found with the search word “neurectomy”; they are sum-
Four randomized, blinded controlled trials compared RF rhi- marized in eTable 179-1A.15-19
zotomies of the medial branch of the dorsal root with a sham
procedure (3 studies) or intrafacetal with extrafacetal rhizotomy Rhizotomy and Neurectomy for Cluster Headache
(1 study). Outcome measures and patient selection differed
between the groups sufficiently to preclude a meta-analysis.85 and Facial Pain
Modest long-term improvement in pain scores in the treated Two retrospective studies (19 patients)72,74 and one prospective
group was observed in all three studies comparing rhizotomy study (18 patients) on the use of rhizotomy and neurectomy for
with a sham procedure. In the last study, intrafacetal rhizotomy cluster headache or facial pain were reviewed. In the prospective
was found to be superior to extrafacetal rhizotomy. Complica- study,73 83% of patients had immediate pain relief but also dem-
tions were minimal in all four studies. Of the remaining 15 onstrated a 39% recurrence rate at long-term follow-up. (See
papers, 5 are prospective in nature. Proportions of patients with eTable 179-1E, 3 studies.72-74)
“good outcomes” varied from 41% to 75% in long-term
follow-up. The remaining 7 series are classified as class III studies
and reported long-term success rates of 40% to 60%. (See eTable
Rhizotomy for Cancer Pain
179-1B, 14 studies.47-61) Ten case series studying the use of rhizotomy for cancer pain
were identified, the majority of which evaluated the effects of
dorsal root rhizotomy for malignant pain of the extremities. The
Rhizotomy for Chronic Discogenic Back Pain largest series contained 71 patients and was one of the earliest
One randomized controlled trial62 (see eTable 179-1C) evaluated papers.52 Reported outcomes generally were favorable initially
the effects of RF lesions of the ramus communicans for treatment but faded quickly with time. The open nature of the procedure,
of single-level chronic discogenic back pain. All selected patients the fading effect, and the presence of more effective procedures
had previously undergone a failed intradiscal electrothermal all contributed to the abandonment of this procedure. One case
(IDET) procedure and had a good response to local nerve block- report was available for the management of facial pain secondary
ade of the ramus communicans. At 1 month and 4 months of to squamous cell carcinoma of the tongue. (See eTable 179-1F,
follow-up, the treated group showed significant improvement in 11 studies.76-84,88)
Visual Analog Scale (VAS) and Medical Outcomes Study (MOS)
Short Form Health Survey (SF-36) scores.
Ganglionectomy
Rhizotomy for Cervical Pain A total of 20 articles on the use of ganglionectomy for treatment
RF rhizotomies were compared with sham procedures for the of pain were found that met the selection criteria previously
treatment of cervical pain or cervicogenic headache68 in three indicated. All studies addressed benign pain conditions. Three
randomized control trials (RCTs).65,68,69 One prospective class II were randomized controlled trials: one compared RF lumbar
clinical trial64 and four case series also addressed rhizotomies for ganglionectomies with sham surgery for sciatica,89 the second
cervical pain. The number of patients in each RCT was small, compared RF lesions made at two different temperatures,90 and
ranging from 12 to 24.65 Two of the RCTs demonstrated a sig- the third compared dorsal root ganglion destruction by doxoru-
nificant treatment effect of RF lesions,65,69 although one did not bicin (Adriamycin) with injection in management of postherpetic
(was an underpowered study).68 The class II study also reported neuralgia.91 The remaining articles were case series ranging in
durable reduction in validated pain scores.64 Both studies with size from 3 to 102 patients. (See eTable 179-2, 17 studies.55,89-106)
long-term follow-up demonstrated similar median-time to recur-
rence.64,65 (See eTable 179-1D, 9 studies.63-71)
Ganglionectomy for Lumbar and Cervical Radicular Pain
In 2003, Geurts and colleagues89 reported on their study, which
Rhizotomy for Trigeminal Neuralgia randomly assigned 83 patients to either RF lesioning (45 patients)
We located no controlled trials that reviewed the treatment or needle placement without RF lesion (38 patients) of selected
of TN by rhizotomy. The best evidence consisted of two class II lumbar ganglion. Evaluators and patients were blinded to the
and four class III studies that prospectively followed patients protocol used. Despite adequate power, this study found no sta-
with TN who were treated with RF rhizotomy or glycerol rhi- tistical difference between the groups.
zolysis, as well as a large (1600-patient) case series.33 In a large Slappendel and associates,90 reported in 1997 that they found
comparative study using glycerol and RF in multiple sclerosis– no difference in outcome between patients receiving RF lesions
related TN and idiopathic TN, both procedures were effective, at 60°C and those receiving lesions at 40°C for cervicobrachial-
with a mean duration to recurrence of 26 months for idiopathic gia. In another study, a case series of 61 patients with sciatica,
TN and 20 months for multiple sclerosis–related TN.37 Three 59% of patients had “markedly reduced” postoperative pain at
studies evaluated pain recurrence rates with life-table analy- follow-up ranging between 1 month and 15 years.103 Definite
ses,22,33,74 and two of these studies prospectively compared sepa- conclusions concerning long-term outcomes are difficult given
rate cohorts of patients defined by symptomatology31 or treatment the variability in follow-up data. The remaining studies were
protocol.24 small and inconsistent.
eTABLE 179-2  Ganglionectomy

Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome


Dubuisson92 1995 Case series Occipital neuralgia 11 Unclear 71% likelihood of improvement
Fedder93 1990 Case report Minor causalgia 4 6-12 mo All had pain relief and decrease in narcotic dosage
Geurts et al89 2003 Multicenter, Chronic lumbosacral 83: 45 patients had 3 mo At 3 mo, treatment was successful in 16% treated patients and 25%
randomized, radicular pain radiofrequency lesioning, 38 control patients; lumbosacral radiofrequency lesioning of dorsal root
double-blind, patients had control treatment ganglia failed to show advantage over control treatment
controlled trial (local anesthetics)
Hosobuchi94 1980 Case series Intractable chest pain 3 3 yr Total anesthesia, including loss of dysesthesia, hyperesthesia, and
original pain, occurred in 2 of the 3 patients
Jansen95 2000 Case series Cervicogenic headache 102 Unclear 80% of patients experienced improvement
Kato et al96 1990 Case report Neuropathic pain 3 1 mo-2 yr, 1 patient had complete pain relief, 2 partial pain relief
8 mo
Lozano et al97 1998 Case series Medically refractory 39 19-48 mo 19 patients had excellent results; patients who had pain from trauma or
chronic occipital described their pain as “shocklike, electric, shooting, jabbing,
pain stabbing, sharp, or exploding” (group I) had better response to
ganglionectomy (80% good or excellent response); patients who had
nontraumatic pain or described their pain as “dull, pounding, aching,
throbbing, or pressurelike” (group II) did not have a good response
Murphy98 1969 Case series Occipital neuralgia of 30 <1 yr-5 yr 18 patients had excellent results, 7 good results, 3 fair results, and 2
various etiologies poor results; it was not clear what criteria were used to determined
outcome measurements
North et al99 1991 Case series Failed back surgery 13 Mean 5.5 yr Treatment “success” found in 2 patients at 2 yr postoperatively but in 0
syndrome at 5.5 yr; “equivocal success” found in 1 patient at 2 yr
postoperatively and in 2 at 5.5 yr
Osgood 1976 Case series Lumbosacral pain, 18 Mean 7 mo 10 patients had “good” results and 4 “fair” results
et al100 brachial plexus
avulsion, amputation
Sanders and 1997 Case series CHA 66 Mean 29 mo 60.7% had complete relief of intermittent CHA; 30% (3 patients) had
Zuurmond55 (group A) or complete relief of chronic CHA
24.1 mo
(group B)
Slappendel 1997 Prospective Cervicobrachialgia 32 patients received RF lesion at 3 mo Identical outcomes in the two groups
et al90 randomized 67°C; 29 patients received RF
double-blind lesion at 40°C (control group)
study
Smith102 1970 Case series Post-thoracotomy, 10 nonmalignant, 12 total Unclear “All had some degree of relief”
herpes
Stechison and 1994 Case series Greater occipital 5 Mean 24 mo All had immediate postoperative relief of pain; 1 had recurrence of pain
Mullin107 neuralgia at 26 mo and underwent ganglionectomy; all experienced nausea
and dizziness postoperatively, and 1 had a cerebrospinal fluid leak
Taub et al103 1995 Case series Sciatica 61 <1 yr-15 yr In 59%, pain was markedly reduced or eliminated
Wetzel et al105 1997 Case series Post-surgical chronic 51 6 mo, 2 yr At 6 mo, 55% had good or excellent outcome, the rest poor outcome
lumbar radiculopathy or treatment failure; at final follow-up (2 yr? 37 patients?), 19% had
good or excellent results
Wilkinson and 2001 Retrospective Various etiologies 19 Mean 22 mo 74% reported >50% reduction; only 1 pain free
Chan106 chart review
Pisapia et al101 2012 Retrospective Occipital neuralgia 10 patients underwent C2 24 mo Mean pain reduction (on a 1-10 scale) was 4.5 ± 4.1
case series ganglionectomy (4 unilateral
and 6 bilateral)
Chun-jing 2012 Randomized Postherpetic neuralgia 36 patients treated with dorsal 6 mo Patients showed excellent or good relief of pain without any adverse
et al91 controlled trial root ganglia destruction by reactions or serious complications
Adriamycin
Weigel et al104 2012 Case series Segmental neuropathic 7 Mean 24 mo All patients had good outcome in regard to pain relief; only 3 required
pain a second procedure
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review. J Neurosurg. 2008;109:389-404.
CHAPTER 179  Evidence Base for Destructive Procedures 1447.e1

CHA, cluster headache; RF, radiofrequency.


179
1448 SECTION 6  Pain

obtained in 16 of 17 patients. Interestingly, all case series reported


Ganglionectomy for Occipital Neuralgia good to excellent results even in patients with postherpetic neu-
The largest case series evaluated the efficacy of C2 ganglionec- ralgia. In one series, for example, deep pain was eliminated or
tomies for occipital neuralgia.95 The investigators found that 80% significantly decreased in all of six patients with postherpetic
of patients had significant relief from symptoms. Unfortunately, craniofacial dysesthesia.151 (See eTable 179-4A, eight case series
long-term follow-up data were unclear, rendering any conclu- and one case report.149-157)
sions difficult. The second largest series of patients with occipital Seven papers were found for tractotomy in cancer facial pain
neuralgia compared ganglionectomies performed in patients with (eTable 179-4B).150,152,158-162 All were cases series with one excep-
occipital pain described as either “sharp, burning, jabbing, electri- tion,162 an open-label prospective study. In this prospective class
cal, or exploding” (group I) or “dull, aching, throbbing, or II study, a standardized outcomes measure (Visual Analog Scale
pressure-like” (group II).97 Patients in group I tended to have a [VAS]), Karnofsky performance score, activities of daily living,
higher prevalence of a traumatic history (74%) and had the better and sleep duration were assessed at 6 months. The researchers
response, nearly 80%. Overall, group II patients had a poor reported sustained pain relief in 80% of patients at 6 months.
response. Follow-up ranged from 19 to 48 months. Other pub- Though the study was classified as class II, the number of trac-
lished series on ganglionectomy performed for occipital neuralgia totomy recipients included was limited (10 patients). We did not
were small and without standardized follow-up data. consider this number sufficient to assign it class II evidence for
One retrospective paper evaluated the effects of occipital neu- the efficacy of tractotomy in cancer facial pain. Current literature
rectomy for the treatment of occipital neuralgia of various supports only class III evidence for tractotomy for cancer pain.
causes.98
Cordotomy
Ganglionectomy for Pain of Other Etiologies A review of cordotomy studies in patients with noncancer pain
One study evaluated sphenopalantine ganglionectomies for produced 12 case series, published between 1958 and 1990, and
cluster headache55 and found that patients with intermittent two case reports, published in 2012.163,164 No class I or II studies
headache had a 67% response rate but those with chronic, con- were found. The number of patients in these case series ranged
tinuous headache had only a 24% response. Mean follow-up from 1 to 72, and follow-up was between 1 month and 41 years.
times were 24 and 29 months, respectively. Outcome data from these studies generally demonstrated an
Two small series, one with 4 patients93 and another with 10 overall improvement in postoperative patient-reported pain over
patients,102 reported on patients treated with thoracic ganglionec- preoperative values.
tomies for a variety of symptoms. Both claimed some success. In general, for studies with long-term follow-up, surgical ben-
(See eTable 179-2, 16 articles.55,89,90,92-97,99,100,102,103,105-107) efits declined with time. However, determining the statistical
significance of these findings was difficult as neither outcome
nor follow-up was standardized in the studies. Furthermore,
DREZ Lesions outcome measures were inconsistent among studies, complicat-
A total of 31 (26 noncancer pain, 5 cancer pain) case series were ing any attempts to make generalized recommendations. (See
obtained for DREZ lesions. No class I or II studies were found. eTable 179-5A, 12 case series.132,165-175)
These reports consisted of between 3 and 124 patients with a The longest follow-up was in a case report for a period of 41
wide range of follow-up time points. For noncancer pain, all years; the patient was completely pain free for 35 years, after
studies reported a greater than 50% relief in pain in a majority which he started to experience slowly progressive aching pain.163
of patients, and these results tended to be durable.108 Patients For cancer pain, cordotomy is the invasive procedure most
with brachial plexus avulsion (BPA) injuries and traumatic spinal often reported in the literature. Eighty-four papers qualified
cord injuries tended to have the best responses to DREZ lesions under our search criteria (eTable 179-5B).79,162,166,169,173,174,176-222
in studies that compared different presenting etiologies directly, Many reports included more than 100 patients, and many had
whereas patients with postherpetic neuralgia and peripheral neu- long-term follow-up. A surge in publications followed the intro-
ropathies had the worst.109-111 Outcomes for patients with BPA duction of percutaneous cordotomy in the mid-1960s, followed
injuries were quite consistent across series, with more than 50% by a gradual decline in published reports and number of patients
reduction in reported pain levels in from 54% to 86% of patients. treated in the late 1990s. The introduction of computed tomog-
(See eTable 179-3A, 26 articles.108-134) raphy (CT) guidance for percutaneous cordotomy by Kanpolat
Since 2008, 10 studies involving DREZ lesions for manage- in the late 1980s, was considered a major contribution to the field
ment of noncancer pain were published: three case reports, six of pain surgery. However, use of intrathecal opioids was also
case series, and one comparative study (eTables 179-3A and introduced at around the same time and gained widespread
179-3B).135-144 popularity.
Only four papers were identified that address the use of DREZ Two prospective trials on cordotomy were identified. One of
lesions for cancer pain (eTables 179-3C and 179-3D).145-148 Two them used a standardized outcome measure (VAS), Karnofsky
were case series that claimed long-lasting pain relief in patients performance score, activities of daily living, and total sleeping
with mixed cancer pain etiologies. Specific follow-ups and hours, and there was a statistically significant improvement in all
outcome measures were not reported. For facial pain from cra- outcomes measures over baseline pain levels.162 The other trial
niofacial malignancies, three of the four papers were small case showed average pain scores (numeric rating scale) immediate pre-
series papers reported some benefit from DREZ of the nucleus operatively, 2 days postoperatively, and 28 days postoperatively
caudalis.145-147 to be 7, 0, and 0, respectively.221 Three papers were retrospective
cohort studies with survival analysis of pain relief of the whole
cohort until death.84,189,190 Five papers were retrospective cohort
Trigeminal Tractotomy for Facial Pain studies with large numbers (>100) of patients and 6 months of
Nine pertinent articles were found for the use of tractotomy in follow-up.185,192,206,211,212 There were no randomized controlled
noncancer pain, including the oldest article retrieved for this trials. Three case reports were found in the literature, in which
review.149 No class I data was found for this topic. Instead, all all patients had complete pain relief postoperatively.216,218,220
papers were case series. The largest case series was reported in Only one systematic review, by France and associates,219 con-
1975 by Schvarcz,150 who found that excellent pain relief was cluded that cordotomy might be safe and effective in treatment
CHAPTER 179  Evidence Base for Destructive Procedures 1449

of cancer pain but that more reliable evidence is needed to aid in series, making definite conclusions difficult. Dougherty and col-
decision making. leagues260 and Tasker261 have provided thorough discussions of 179
Despite the heterogeneity of outcome measures, the vast thalamotomy. (See eTable 179-8A, 13 case series.165,251,259,262-271)
majority of studies reported excellent lasting relief after cordot- Sixteen papers on thalamotomy in cancer pain were found
omy within the context of cancer longevity. Reported cordotomy (eTable 179-8B),165,251,262,263,266,272-282 (some articles are included in
outcomes in patients with cancer pain contrasts with outcomes the noncancer pain tables). All were case series with variable
in those with noncancer pain, in whom pain relief was moderate, outcome measures and follow-up. Most reported lasting benefits
short-lived, and often complicated with dysesthesias. in more than 50% of patients. In some instances bilateral pain
Within the medical literature, there is limited class II evidence relief was obtained with unilateral thalamotomy. Effects tended
for cordotomy as an effective treatment modality in cancer pain to fade with time and were often accompanied by persistent
patients. psychiatric complications.278

Myelotomy Cingulotomy
Three articles examining the use of myelotomy in noncancer pain A total of 14 original case series focusing on cingulotomy for
were found. These were small case series (3 to 14 patients) with noncancer pain were identified on the basis of the inclusion cri-
variable follow-up periods and pain etiologies. The largest series teria (eTable 179-9A). Because the largest case series described
reported that 64% of patients experienced a complete or marked in the literature (n = 133) was not published as a peer-reviewed
reduction in deep “background” pain.223 (See eTable 179-6A, article, it is not included in our analysis.283 No class I or class II
three case series.223-225) studies were identified. Reported case series were small, the
For cancer pain, 20 papers were identified (eTable 179- largest one describing 36 patients. Overall, only two papers used
6B).133,153,225-242 These articles were all case series and included a validated outcome measure such as the VAS,284,285 and only two
papers about commissural as well as extraleminiscal myelotomy, papers had a common diagnosis for the series.286,287 Different
both open and percutaneous. With the exception of two papers, follow-up periods and variable outcome measures were used,
most reports involved a small number (<20) of patients.133,224,232,239 making comparisons difficult, and for most series, reported out-
Outcome measures and follow-up periods were heterogeneous. comes were also variable. Also, the majority of the reports were
In general, reported outcomes after myelotomy were less favor- published before 1980. Abdelaziz and Cosgrove133 provide a
able than those after cordotomy. However, a consistent observa- detailed description and summary of published case series. (See
tion was that myelotomy was most effective in treating midline eTable 179-9A, 14 case series.251,284-296).
visceral cancer pain syndromes, that is, pain from cervical, pan- After elimination of redundancy, seven papers qualified for the
creatic, or gastric cancer. criteria of this review and addressed cingulotomy for cancer pain
The current literature supports only class III evidence for (eTable 179-9B).251,285,289,291,293,297All were case series, with small
myelotomy for pain relief. numbers and heterogeneous patient selection. In a later report,
50% of patients had some pain relief for up to 6 months.285
Results in the other papers were variable, ranging from 10%
Mesencephalotomy to 50%.
Using the criteria described, we reviewed nine relevant articles
concerning the use of mesencephalotomy for noncancer pain. All
of these articles were case series, and most had relatively small
Sympathectomy
numbers of patients. The latest publication reported the effects A Cochrane systematic review on sympathectomy for nonmalig-
of lesions made in one of two locations in 27 patients with central nant pain syndromes; specifically for neuropathic pain syndrome,
pain following a stroke.243 Long-term pain relief was produced in was published in 2003.298 The review criteria the investigators
75% of patients with lesions created at the level of the superior used included all randomized clinical trials, controlled clinical
colliculus, although with significant ocular side effects. These side trials, observational studies (cohorts and case-control studies),
effects were reduced with lesions adjacent to the inferior collicu- and relevant retrospective reviews of patient charts, for a total of
lus; long-term pain relief was achieved in 58% of patients who 223 titles and abstracts. Only surgical sympathectomy (cervico-
received the adjacent lesions. Follow-up was between 3 months thoracic or lumbar sympathetic chain ablation or coagulation by
and 5 years. In a separate study, good pain relief was produced in open, endoscopic, laser, or radiofrequency) or chemical sympa-
23 patients (67%) with thalamic syndrome and tabes dorsalis, thectomy (phenol or alcohol solution injection of cervicothoracic
followed for 3 to 70 months.244 (See eTable 179-7A, nine case or lumbar sympathetic chain) were included. The review showed
series.243-251) that treating neuropathic pain with sympathectomy is based on
Mesencephalotomy and pontine tractotomy for cancer very limited evidence. An additional publication, published in
pain were reported in seven papers ( eTables 179-7B and 2012, reported on sympathectomy for complex regional pain
179-7C).250,252-257All were case series, with variable outcome mea- syndrome (CRPS) and described achieving adequate pain relief
sures and follow-up. Outcome varied from extreme success (in for a mean duration of 3.2 years.299
92% of patients until death)257 to very poor success (lasting pain As to sympathectomy for malignant pain, our search identified
relief in 1 of 12 patients).250 There was agreement on frequency much the same information. A meta-analysis of neurolytic celiac
of side effects, specifically those related to ocular mobility. block (the most common form of sympathectomy performed for
cancer pain) was identified,300 along with seven additional papers
(eTable 179-10299,301-306) addressing either celiac block (published
Thalamotomy in subsequent years after the meta-analysis data) or other forms
All published articles describing thalamotomy for pain were of sympathectomy. These studies generally focused on patients
limited to small case series with heterogeneous patient popula- with midline pain from visceral malignancies. The meta-analysis
tions. The largest case series (n = 85) was published not in a peer- identified two class I RCTs demonstrating the efficacy and safety
reviewed article but in book format,258 and so is not included here; of celiac block in treatment of pancreatic as well as upper abdomi-
however, the investigators did describe a subset of patients from nal visceral malignancies. Of the other seven papers, one was a
this series in an earlier report.259 Follow-up, outcome measures, class I RCT for sympathectomy for inoperable cancer pain in
target site, and pain etiology were inconsistent within or among which excellent pain relief was achieved for up to 3 months. Two
eTABLE 179-6A  Myelotomy for Pain*
Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
224
Broager 1974 Case series Unbearable pain in lower 3 with nonmalignant 3 wk-18 mo Permanent excellent results
half of body pain causes, 34 in 16, temporary excellent
total in 9, permanent good in
3, continuous poor in 5; 1 179
death
Schvarcz223 1978 Case series Pain of central origin 14 6 mo-4 yr Abolition of hyperpathia and
disappearance of, or
marked reduction in, deep
background pain achieved
in 64% cases overall
Sourek225 1969 Case series Arachnoiditis, multiple 6 Long-term follow-up not 2 patients had no recurrence
sclerosis, or unknown carried out due to of pain
bad general health;
6-36 mo in 3 patients
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review. J Neurosurg.
2008;109:389-404.
*No publications after 2008.

eTABLE 179-6B  Myelotomy for Cancer Pain


Authors Year Study Design Diagnosis Patient No. Follow-up Outcome
Hwang et al233 2004 Case series Cancer pain 6 2-18 wk Moderate pain relief, at 3 mo
according to Visual Analog Scale
score
Becker et al226 2002 Case report Cancer pain 1 5 wk Complete pain relief; patient died at
5 wk
VilelaFilho et al240 2001 Case report Cancer pain 2 Short Safe techniques achieved effective
pain relief
Nauta et al237 2000 Case series Cancer pain 6 3-31 mo Significant pain reduction
Kim and Kwon235 2000 Case series Cancer pain 8 Short Adequate pain relief in 5 patients
Watling et al241 1996 Case series Cancer pain 2 Patients died after 4 Commisural myelotomy reported;
and 6 wk good pain relief prior to death
Kanpolat et al234 1997 Case series Cancer pain 14 3-6 mo 8 of 14 patients had no pain or
satisfactory pain relief
Gildenberg and 1984 Case series Cancer pain 14 2-13 mo 91% experienced total pain relief till
Hirshberg230 death
Schvarcz239 1976 Case series Cancer pain 35 with cancer pain, 45 6 mo-4 yr 30 had pain relief initially, and 25 had
total persistent pain relief
Papo and 1976 Case series Cancer pain 9 with cancer pain, 10 Till death, 1 yr Only 3 patients experienced pain relief;
Luongo238 total commissural myelotomy was
performed
Hitchcock232 1974 Case series Cancer pain 21 with cancer pain. Till death (a few 9 had no pain, and 5 had good pain
Twenty-six procedures weeks to 1 yr) relief
in 26 patients;
procedure abandoned
in 5 cases; 2 patients
lost to follow up; 19
patients with follow-up
data
Broager224 1974 Case series Cancer pain 31 with cancer pain, 34 3 wk-18 mo Permanent excellent results in 16,
total temporary excellent in 9, permanent
good in 3, and continuous poor in
5; there was 1 death
Cook et al227 1977 Case series Cancer pain 16 with cancer pain, 24 A few weeks to Of the 16 cancer patients, 11, 3, and
total months 2 had good, fair, and poor
immediate pain relief, respectively;
in late follow up, 11 died, 2 had
good pain relief, and 1 had fair pain
relief; 1 was lost to follow up
Sourek225 1969 Case series Cancer pain 19 with cancer pain, 25 Unclear 17 had good pain control
total
236
Lippert et al 1974 Case series Cancer pain 11 with cancer pain,16 A few days to 7 mo Most had good pain relief
total
Eiras et al228 1980 Case series Cancer pain 12 3-14 mo 7 had total alleviation of pain, 3 were
lost to follow-up, and few survived
>6 mo
Fascendini 1979 Case series Cancer pain 6 3 mo Initial pain relief, but pain recurred in
et al229 all patients who survived; 2 had
neurological deficits
Goedhart et al231 1984 Case series Cancer pain 10 Up to 3 yr Not clear; pain relief did not last, and
complication rate high
Hosobuchi and 1971 Case series Mixed 4 with cancer pain, 6 4-12 mo Good
Rutkin153 total
Viswanathan 2010 Retrospective Intractable 11 Median postoperative 5 patients had excellent outcomes, 3
et al242 case series malignancy- survival was 43 patients good, and 2 fair, and 1
induced days (range 11 had a poor outcome
visceral pain days-39 mo)
1449.e2 SECTION 6  Pain

eTABLE 179-7A  Mesencephalotomy for Pain*


Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Amano 1986 Case series Thalamic syndrome 27 with pain of nonmalignant 3-70 mo Overall effectiveness: good pain
et al244 and tabes dorsalis causes, 34 total relief in 23 patients (67%)
Helfant 1965 Case series Herpes zoster, 3 60 mo; lost to Low-grade pain 3 mo
et al245 thalamic follow-up; postoperatively; unchanged;
syndrome, otitic 30 mo left-sided pain and
herpes zoster paresthesias 1.5 mo
postoperatively
Liberson 1970 Case series Intractable pain of 5 with pain of nonmalignant Not stated Improvement in all cases
et al246 face, upper or causes, 6 total
lower extremities
Shieff and 1988 Case series Pain of central origin 27; lesions at original target 3 mo-5 yr 75% with long-term pain relief
Nashold243 after stroke adjacent to superior in first group; 58% with
colliculus made in 14 long-term pain relief in
patients and surgery at second group
revised target at level of
inferior colliculus in 13
patients
Spiegel and 1953 Case series Intractable facial pain 6 6.5 wk-4.5 yr Improvement in all cases,
Wycis247 ranging from no recurrence
of pain to little facial pain
Voris and 1975 Case series Intractable pain 13 with pain of nonmalignant >3 yr 0 patients with no pain relief; 8
Whisler251 causes, 58 total patients with 1-12 mo of
pain relief; 2 patients with
1-3 yr of pain relief; 3
patients with >3 yr of pain
relief; combined procedures
not included in this paper
Wycis et al248 1958 Case series Postherpetic 4 3-6 yr Mesencephalotomy alone (3
trigeminal patients) or combined with
neuralgia mesencephalothalamotomy
(1 patient) used: 3 patients
had at least partial relief of
facial pain
Wycis and 1962 Case series Mixed 42 with pain of nonmalignant Up to 13 yr 72% had immediate relief, and
Spiegel249 causes, 54 total 31% complete or partial
permanent relief
Zapletal250 1969 Case series Mixed 4 with pain of nonmalignant Unclear 1 patient had lasting relief
causes, 18 total
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review. J Neurosurg.
2008;109:389-404.
*No publications after 2008.

eTABLE 179-7B  Mesencephalotomy for Cancer Pain*


Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
253
Bosch 1991 Case series Cancer pain 40 Long term Effective in nociceptive pain relief
Frank et al254 1989 Case series Cancer pain 202 >6 mo 81% persistent pain relief
Barbera et al252 1979 Case series Cancer pain 6 Short All patients had pain relief in
neck and upper extremities
Zalpetal250 1969 Case series Cancer pain 12 with cancer Unclear 1 patient with lasting pain relief
pain, 16 total
Whisler and 1978 Case series Cancer pain 38 Till death; most patients survived 92% had pain relief till death
Voris257 1-6 mo, only 3 living >1 yr
Nashold256 1972 Case series Cancer pain 8 9.5 mo average All patients had pain relief, and
all had side effects
From Raslan AM, Cetas JS, McCartney S, et al. Destructive procedures for control of cancer pain: the case for cordotomy. J Neurosurg. 2011;114:
155-170. Published online with permission from the American Association of Neurological Surgeons Journal of Neurosurgery.
*No publications after 2008.

eTABLE 179-7C  Pontine Tractotomy for Cancer Pain*


Authors Year Study Design Diagnosis Patient No. Follow-up Outcome
255
Hitchcock et al 1985 Small case series Cancer pain of only 6 Till death 100% had pain relief; 4 patients died less
neck and upper body than 3 mo after surgery, and 2 survived
more than 3 mo
*No publications after 2008.
CHAPTER 179  Evidence Base for Destructive Procedures 1449.e3

eTABLE 179-8A  Thalamotomy for Pain


179
Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Amano 1976 Case series Intractable pain of mixed 33 with pain of 1-24 mo Good pain relief for thalamic pain or
et al165 etiology nonmalignant causes, causalgia
47 total
Choi and 1977 Case series Intractable pain 7 with pain of Not stated 91% of all patients had excellent or
Umbach262 nonmalignant causes, good results
37 total
Hitchcock 1981 Case series Mixed 29 with pain of Up to 10 yr Bilateral center-median thalamotomy
and nonmalignant causes, is superior to basal thalamotomy
Teixeira263 43 total
Jeanmonod 1993 Case series Chronic therapy-resistant 45 Mean 14 mo VAS used; 67% of all patients
et al259 neurogenic pain; gained 50%-100% relief, and
various lesions 29% had complete relief
Kudo et al270 1968 Case series Cerebrovascular accident, 8 with pain of Up to 2 yr Complete relief of pain without
atypical facial pain, nonmalignant causes, appreciable sensory loss in 10
trigeminal neuralgia 17 total patients, fairly good effects of
pain relief in 7
Lende et al264 1971 Case series Facial pain from stroke or 2 14-20 mo Relief of pain
trigeminal neuralgia
Niizuma 1982 Case series Central pain from 18 9 mo-6 yr 56% effective, but longest duration
et al265 cerebrovascular disease of pain relief was about 6 mo
Obrador and 1960 Case series Atypical facial pain and 3 Not stated Patient with trigeminal neuralgia had
Bravo269 trigeminal neuralgia no improvement; those with
atypical facial pain had less
intense pain postoperatively
Richardson266 1967 Case series Mixed ? with pain of nonmalignant Up to 5 mo Favorable results
causes, 38 total
Shieff and 1987 Case series Central pain from 24 reviewed; 27 total 3 mo-5 yr 66.7% had long-term relief
Nashold267 cerebrovascular
accident
Voris and 1975 Case series Intractable pain 23 with pain of >3 yr 4 patients had no pain relief; 12 had
Whisler251 nonmalignant causes, 1-12 mo of pain relief, 2 patients
58 total had 1-3 yr, and 5 had >3 yr;
combined procedures were not
included in this paper
Young et al315 1995 Case series Chronic intractable pain of 19 1-22 mo 15 patients were followed for
mixed etiology >3 mo; 4 patients (27%) were
pain free and 5 patients (33%)
achieved >50% pain relief;
therefore 9/15 patients (60%) had
worthwhile benefit from medial
thalamotomy with Gamma Knife
Jeanmonod 2012 Case series Chronic therapy-resistant 11 patients, but the 1 yr Preoperative mean VAS score was
et al271 neuropathic pain outcomes of only 9 59.5/100; significant pain relief
patients were used in (mean group value 55%) was
the analysis as the reported during and at the end of
lesion made in the the procedure; mean VAS was
posterior part of 34.3/100 at 3 mo and 35.3/100
the thalamic central at 1 yr
lateral nucleus
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review.
J Neurosurg. 2008;109:389-404.
VAS, Visual Analog Scale.
1449.e4 SECTION 6  Pain

eTABLE 179-8B  Thalamotomy for Cancer Pain*


Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Whittle and 1995 Case series Cancer pain 2 Patients died 6 wk and Adequate pain relief till death
Jenkinson282 3 mo postoperatively
Steiner et al279 1980 Case series Cancer pain 52 Few mo 8 had excellent pain relief, 18 with
moderate pain relief
Hitchcock and 1981 Case series Mixed 16 with cancer Up to 10 yr Bilateral center-median is superior
Teixeira263 pain, 43 total to basal thalamotomy
Fairman and 1973 Case series Cancer pain 165 2-12 mo 70% had no or satisfactory pain
Llavallol273 relief
Richardson266 1967 Case series Mixed 38 total Up to 5 mo Favorable outcome
Richardson276 1974 Case series Mixed 24 total 2-12 mo Mixed results, but return of chronic
pain occurred in all patients
Uematsu et al281 1974 Case series Cancer pain 17 Till death; average 6 mo 3 excellent, 7 good, 3 no benefit
Sugita et al280 1972 Case series Cancer pain 44 with cancer Up to 18 mo; most had 85% had excellent or satisfactory
pain, 60 total 1-yr follow-up; patients pain relief initially; generally good
with cancer mostly did results in patients with cancer
not survive to 1 yr pain
Leksell et al274 1972 Case series Cancer pain 25 Up to 1 yr; most patients 6 were pain free and 4 had
died before 1 yr moderate pain relief; effect
noticed after 3 wk
Shimizu et al278 1968 Case series Cancer pain 8 with cancer 23 days-14 mo 6 had complete pain relief till death
pain, 17 total (3 died <1 mo postprocedure), 2
had adequate pain relief;
psychological disturbances were
common but resolved 4-20 days
after surgery
Fairman272 1967 Case series Cancer pain 15 with cancer 2-12 mo 80% had bilateral pain relief
pain, 53 total
Ramamurthi and 1966 Case series Cancer pain 7 Short All had reasonable pain relief
Kalyanaraman275
Sano et al277 1966 Case series Mixed 10 total 2 mo-2.5 yr Lasting effect in 8 patients
Amano et al165 1976 Case series Mixed 14 with cancer 1-24 mo Good pain relief only in patients
pain, 47 total with thalamic pain or causalgia
Choi and Umbach262 1977 Case series Mixed 30 with cancer Unclear 91% of all patients had excellent or
pain, 37 total good results
Voris and Whisler251 1975 Case series Mixed 35 with cancer >3 yr Pain relief in most patients; few had
pain, 58 total long-lasting pain relief
From Raslan AM, Cetas JS, McCartney S, et al. Destructive procedures for control of cancer pain: the case for cordotomy. J Neurosurg. 2011;114:
155-170. Published online with permission from the American Association of Neurological Surgeons Journal of Neurosurgery.
*No publications after 2008.
CHAPTER 179  Evidence Base for Destructive Procedures 1449.e5

eTABLE 179-9A  Cingulotomy for Pain*


179
Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Broager and 1972 Case series Benign and 17 3-13 yr 14 had good or excellent results
Olesen288 psychogenic pain (combined)
Faillace et al289 1971 Case series Benign pain 2 Unclear Pain improved in 1
Foltz and White290 1968 Case series Benign and 24 1-9 yr 18 had excellent or good results
psychogenic pain
Hurt and 1974 Case series Benign 36 1-21 yr 8 had marked to complete pain relief
Ballantine291
Laitinen and 1972 Case series Intractable phantom 3 Unclear Poor in all 3
Vilkki287 limb pain
Le Beau292 1954 Case series Intractable benign pain 5 Unclear Poor
and depression
Pillay and 1992 Case series Benign 2 1 yr 1 had excellent results
Hassenbusch293
Sharma294 1974 Case series Intractable pain 3 <1 yr All 3 patients stopped craving opiates
Turnbull295 1972 Case series Benign 13 Unclear 4 had good or excellent results
(combined)
Voris and Whisler251 1975 Case series Intractable pain 11 with pain of >3 yr 3 patients had no pain relief; 6
nonmalignant patients had 1-12 mo of pain
causes, 58 total relief, 1 had 1-3 yr, and 1 patient
had >3 yr; combined procedures
not included in this paper
Wilkinson et al284 1999 Case series Benign 23 1-15 yr 18 responded to a follow-up
questionnaire; 72% reported
5-point reduction or more on VAS
Wilson and 1974 Case series Benign 4 1-55 mo Poor in all 4
Chang296
Yen et al285 2005 Case series Benign 7 1 yr 4 had significant pain relief as
demonstrated by VAS score
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review.
J Neurosurg. 2008;109:389-404.
*No publications beyond 2008.
VAS, Visual Analog Scale.

eTABLE 179-9B  Cingulotomy for Cancer Pain*


Authors Year Study Design Diagnosis Patient No. Follow-up Outcome
Yen et al285 2005 Case series Cancer pain 15 6 mo 50% had pain relief at 6 mo
Wong et al 316 1997 Case series Cancer pain 3 Not reported Adequate pain relief but with personality
changes and delusions in 1 patient
Pillay and Hassenbusch293 1992 Case series Cancer pain 8 6 mo 50% had excellent results, 25% poor
Hurt and Ballantine291 1974 case series Mixed 32 with cancer 4 days-6 yr Helpful in 12 patients
pain, 68 total
Foltz297 1968 Case series Cancer pain 11 Unclear Helpful in 9 patients
Faillace et al289 1971 Case series Mixed 9 with cancer Unclear Helpful in 1 patient
pain, 11 total
Voris and Whisler251 1972 Case series Mixed 7 with cancer 1 mo-3 yr Helpful in 5 patients
pain, 29 total
From Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review. J Neurosurg.
2008;109:389-404. Published online with permission from the American Association of Neurological Surgeons Journal of Neurosurgery.
*No publications after 2008.
1449.e6 SECTION 6  Pain

eTABLE 179-10  Sympathectomy for Cancer Pain


Authors Year Study Design Diagnosis Patient No. Follow-up Outcome
Happak et al299 2012 Case series Complex regional 16 3.2 ± 0.4 yr Twelve patients (75%) demonstrated a
pain syndrome significant decrease in their pain scores
type II after the surgery. The mean score on
the VAS was 8.8 before the operation
and 2.7 after the operation.
Functional improvement of the limbs was
reported by 15 (94%) of the 16 patients.
Kang et al303 2007 Case series Cancer pain 21 Short Significant improvement in (Verbal)
Numerical Rating Scale, percutaneous
sympathectomy
Gunaratnam et al302 2001 Prospective Cancer pain 58 6 mo Significant reduction of VAS score for up to
open-label 6 mo, percutaneous sympathectomy
Lin et al305 1994 Case series Cancer pain 14 Short Good immediate pain relief
Fujita301 1994 Case series Cancer pain 27 Short Complete pain relief in 20 of 21 patients
who received successful bilateral
splanchnic nerve neurolysis
Lillemoe et al304 1993 Prospective Cancer pain 65 Short Significant pain relief
randomized
Eisenberg et al300 1995 Meta-analysis Cancer pain Only for celiac plexus block, which was
found effective; included many papers
Erdek et al306 2010 Retrospective case Visceral cancer 50 1 mo 28 patients showed pain relief >50% for
series pain for which >1 mo
conservative
treatment failed
Wyse et al317 2011 Randomized Painful inoperable 98 3 mo Pain scores decreased significantly at 1 mo
controlled pancreatic after endoscopic ultrasound-guided
double-blind trial cancer celiac plexus neurolysis, with a further
decrease at 3 mo
VAS, Visual Analog Scale.
1450 SECTION 6  Pain

others—a class II “prospective randomized trial” involving 65 described as steady and/or aching.22,31,97,108,132 This observation
patients and a prospective open-label “class II” study involving suggests that there may be an underlying difference between
58 patients—both demonstrated statistically significant pain these two types of pain that may require distinct interventions.
relief at 6-month follow-up (P < .05) in patients with pancreatic These observations regarding BPA, TN, pain symptomology,
cancer.302,304 visceral cancer pain, and cordotomy are just a few of the issues
in the published literature that warrant further study and assess-
ment through focused, well-designed trials. The value of such
DISCUSSION studies could potentially lead to improvements in patient selec-
Numerous studies have been published on destructive techniques tion and surgical outcomes as well as to new understandings of
for the treatment of pain, many dating from the beginnings of the pathophysiology of different pain states.
modern neurosurgery. Few, however, have detailed descriptions Anyone designing appropriate modern studies must under-
of long-term outcomes using validated outcome measures and stand that clinical trials for pain treatment are especially prone
standardized follow-up. The vast majority of published articles to such confounding factors as reporting and observer bias,10
constitute case series and uncontrolled chart reviews. Heteroge- owing to the subjective nature of pain and the complete absence
neous populations of patients, inconsistent follow-up, hugely of an objective or treatment effect marker. The value of validated
variable study groups, vastly different treatment techniques, and outcome measures, such as the VAS, as surrogates for more objec-
lack of either concurrent or historical controls are endemic to the tive measures, is now well recognized, and such measures must
majority of the reviewed studies. As accepted methods of clinical be employed in modern studies. Care must also be taken to limit
reporting have evolved, investigators have made significant such confounders as expectation by the patient and the physician
efforts to incorporate modern standards of evidence, but the that the treated pain will improve. In fact, it has been well-
absolute number of studies with such standards in the field of documented not only that invasive procedures tend to have a
ablative surgery for pain remains limited. greater placebo effect on patients’ symptoms than noninvasive
The few studies that can be classified as class I,10,14 in general, treatments but also that the effect decays with time.308 In patients
are from the anesthesia literature and address percutaneous treated for cancer pain, however, this pattern might also represent
methods for the treatment of facet or radicular pain. The results the limited survival in terminally ill patients and may mask a real
of these studies are overall mixed, highlighting the difficulty in therapeutic effect. In addition, different pain states are now
extrapolating efficacy from earlier, uncontrolled studies. known to have different underlying pathophysiologies. Modern
In many instances in the literature on neuroablation, for both studies should not group all patients together and potentially miss
malignant and nonmalignant pain etiologies, repeated observa- treatable subgroups of patients. However, identifying subgroups
tions have been made across different studies, writers, and institu- of patients make it even more difficult for single centers to main-
tions that, given their consistency, should be further explored. For tain sufficient volume for a particular procedure to adequately
example, better outcomes for DREZ lesions were reported for power a valid outcome study. Collaboration among institutions
the treatment of patients with BPA than for those with posther- and countries is imperative. The accurate evaluation of surgical
petic neuralgia.109-111 Although the observations were made in pain procedures is a difficult task owing to the subjective nature
uncontrolled studies, expectation and observer bias were likely of pain and the additional challenges in blinding patients and
consistent throughout the groups. DREZ lesions may provide evaluators to the procedure performed. Although sham surgery
considerable benefit to patients with BPA. Denervation pain from has been performed in a neurosurgical clinical trial for the treat-
BPA historically has been difficult to treat despite the treatment ment of Parkinson’s disease,309 such practice is highly controver-
modality.2,108 The efficacy of DREZ lesions in BPA should be sial.310 More realistically, it should be recognized that useful and
studied in a well-controlled and sufficiently powered study. Simi- meaningful information can be obtained from well-designed
larly, consistent beneficial long-term outcomes with similar rates studies that incorporate as many of the elements of a class I study
of recurrence for TN treated with rhizotomies have been reported as possible.10,311 Modern observational studies using case controls
in several large case series.22,31,33 There is strong anecdotal evi- and modern statistics may still demonstrate treatment effects
dence for a significant treatment effect. However, considerable within the confidence intervals of RCTs.311,312 Important ele-
controversy remains regarding patient selection, timing,86 and the ments include investigator equipoise, blinded or at least impartial
place of DREZ lesioning in light of other efficacious, nondestruc- evaluators, long follow-up times with intention-to-treat type
tive treatments, such as microvascular decompression. analyses, validated outcome measures, controls, uniform diagno-
In studies of cancer pain two consistent observations stand ses, and uniform surgical techniques. With appropriate and well-
out. First commissural myelotomy and extralemniscal myelotomy designed studies, large numbers of patients may not be needed
may be effective treatments for visceral cancer pain states and to begin to answer fundamental questions in the surgical treat-
should be studied directly. Second, a large number of studies as ment of pain.
well as one class II study support the use of cordotomy for the The field of pain surgery has suffered from a general pattern
treatment of cancer pain states, reporting sustained relief for up of early enthusiasm for new techniques followed by a shift to even
to 2 years. In this regard, cordotomy is unique among invasive newer approaches without adequate establishment of indications
procedures for the treatment of cancer pain. The procedure fell and efficacy data for existing procedures. A large paradigm shift
out of favor largely because of strong competition from intrathe- occurred that favored neuromodulation over neuroablation. This
cal opioid treatment. Now that the side effects and cost of long- shift represented a more modern perspective, in which pain is
term intrathecal opioid use are better appreciated, it is time to viewed as a primitive but fundamental protective mechanism of
revisit cordotomy by means of modern, well-designed studies for the entire organism. Pain is the result of a distributed network
the treatment of certain cases of cancer pain. rather than simple afferent inputs that can be removed. However,
Important observations have also been made throughout dif- neuromodulation as well as modern pharmaceuticals have also
ferent treatment modalities and etiologies that may contribute to failed to be all things to all patients. By the time that the surgical
a deeper understanding of the underlying pathophysiology of community realized the limitations of the new paradigm, it was
different pain states. Independently, different writers reporting too late to revive many of the older and potentially efficacious
on ablative techniques for the treatment of various pain etiolo- ablative techniques. In fact, very few practicing neurosurgeons
gies, ranging from spinal cord injury to TN, have observed that are either trained in or actively perform many of the more inva-
pains described as electrical, intermittent, and stabbing were sive ablative techniques. If neurosurgery is to maintain a leader-
more successfully treated by ablative procedures than pains ship role in the treatment of pain, we must come together to first
CHAPTER 179  Evidence Base for Destructive Procedures 1451

determine and then promote and teach those techniques that Kanpolat Y, Tuna K, Bozkurt M, et al. Spinal and nucleus caudalis dorsal
hold promise for patients with pain. root entry zone operations for chronic pain. Neurosurgery. 2008;62(ONS 179
Finally, it should be noted that the studies reviewed were suppl 1):ONS235-ONS244.
limited to peer-reviewed journals using the criteria outlined in Lahuerta J, Bowsher D, Lipton S, et al. Percutaneous cervical cordotomy:
a review of 181 operations on 146 patients with a study on the location
our methodology. As eloquently stated by two statesmen in the of “pain fibers” in the C-2 spinal cord segment of 29 cases. J Neurosurg.
field, “much information about lesioning surgery in the nervous 1994;80(6):975-985.
system for pain relief can be found in textbooks devoted to Leclaire R, Fortin L, Lambert R, et al. Radiofrequency facet joint dener-
surgery for pain.”313 For new ideas, read old books (after Pavlov). vation in the treatment of low back pain: a placebo-controlled clinical
trial to assess efficacy [see comment]. Spine. 2001;26:1411-1416, dis-
cussion 1417.
CONCLUSION Lillemoe KD, Cameron JL, Kaufman HS, et al. Chemical splanchnicec-
Destructive techniques for the treatment of pain have a long and tomy in patients with unresectable pancreatic cancer. A prospective
important history within the field of neurosurgery, but their effi- randomized trial. Ann Surg. 1993;217(5):447-455, discussion 456-457.
Lord SM, Barnsley L, Wallis BJ, et al. Percutaneous radio-frequency
cacy has not been well established through the use of contempo- neurotomy for chronic cervical zygapophyseal-joint pain [see
rary standards. This comprehensive review of the existing comment]. N Engl J Med. 1996;335:1721-1726.
literature on ablative techniques for pain was aimed at summariz- Nauta HJ, Soukup VM, Fabian RH, et al. Punctate midline myelotomy
ing the extent and quality of support for these techniques. With for the relief of visceral cancer pain. J Neurosurg. 2000;92(suppl):
very few exceptions, there is limited evidence of their efficacy. 125-130.
Modern studies are needed to define appropriate indications and Oh WS, Shim JC. A randomized controlled trial of radiofrequency dener-
methodologies in the treatment of noncancer chronic pain. vation of the ramus communicans nerve for chronic discogenic low
back pain. Clin J Pain. 2004;20:55-60.
Acknowledgments Pieper DR, Dickerson J, Hassenbusch SJ. Percutaneous retrogasserian
The authors would like to express their appreciation and thanks to Robert glycerol rhizolysis for treatment of chronic intractable cluster head-
J. Coffey, M.D., and Shirley McCartney, Ph.D., for contributions to this aches: long-term results. Neurosurgery. 2000;46:363-368, discussion
chapter. 368-370.
Pillay PK, Hassenbusch SJ. Bilateral MRI-guided stereotactic cingulot-
omy for intractable pain. Stereotact Funct Neurosurg. 1992;59(1-4):
SUGGESTED READINGS 33-38.
Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment Pollock BE, Ecker RD. A prospective cost-effectiveness study of trigemi-
of nonmalignant pain: a structured literature review. J Neurosurg. nal neuralgia surgery. Clin J Pain. 2005;21:317-322.
2008;109:389-404. Raslan AM. Percutaneous computed tomography-guided radiofrequency
Friedman AH, Bullitt E. Dorsal root entry zone lesions in the treatment ablation of upper spinal cord pain pathways for cancer-related pain.
of pain following brachial plexus avulsion, spinal cord injury and herpes Neurosurgery. 2008;62(ONS1):ONS226-ONS234.
zoster. Appl Neurophysiol. 1988;51:164-169. Sanders M, Zuurmond W. Safety of unilateral and bilateral percutaneous
Gallagher J, Petriccione Di Vadi PL, Wedley JR, et al. Radiofrequency cervical cordotomy in 80 terminally ill cancer patients. J Clin Oncol.
facet joint denervation in the treatment of low back pain: a prospective 1995;13(6):1509-1512.
controlled double-blind study to assess its efficacy. Pain Clinic. 1994;7: Sindou M. Microsurgical DREZotomy (MDT) for pain, spasticity, and
193-198. hyperactive bladder: a 20-year experience. Acta Neurochir (Wien).
Geurts JWM, van Wijk RMAW, Wynne HJ, et al. Radiofrequency lesion- 1995;137(1-2):1-5.
ing of dorsal root ganglia for chronic lumbosacral radicular pain: Stovner LJ, Kolstad F, Helde G. Radiofrequency denervation of facet
a randomised, double-blind, controlled trial [see comment]. Lancet. joints C2-C6 in cervicogenic headache: a randomized, double-blind,
2003;361:21-26. sham-controlled study.[see comment]. Cephalalgia. 2004;24:821-830.
Ischia S, Ischia A, Luzzani A, et al. Results up to death in the treatment vanKleef M, Barendse GA, Kessels A, et al. Randomized trial of radiofre-
of persistent cervico-thoracic (Pancoast) and thoracic malignant pain quency lumbar facet denervation for chronic low back pain. Spine.
by unilateral percutaneous cervical cordotomy. Pain. 1985;21(4): 1999;24:1937-1942.
339-355. Wallis BJ, Lord SM, Bogduk N. Resolution of psychological distress of
Kanpolat Y, Savas A, Batay F, et al. Computed tomography-guided tri- whiplash patients following treatment by radiofrequency neurotomy:
geminal tractotomy-nucleotomy in the management of vagoglosso- a randomised, double-blind, placebo-controlled trial [see comment].
pharyngeal and geniculate neuralgias. Neurosurgery. 1998;43:484-489, Pain. 1997;73:15-22.
discussion 490. Zapletal B. Open mesencephalotomy and thalamotomy for intractable
Kanpolat Y, Savas A, Ucar T, et al. CT-guided percutaneous selective pain. Acta Neurochir (Wien). 1969;(suppl 18):1+.
cordotomy for treatment of intractable pain in patients with malignant
pleural mesothelioma. Acta Neurochir (Wien). 2002;144(6):595-599,
discussion 599. See a full reference list on ExpertConsult.com
CHAPTER 179  Evidence Base for Destructive Procedures 1451.e1

REFERENCES trigeminal neuralgia. J Oral Maxillofac Surg. 1999;57(2):104-111,


1. Kanpolat Y. Percutaneous cordotomy, tractotomy, and midline discussion 111-102. 179
myelotomy: minimally invasive stereotactic pain procedures. Semin 26. Siegfried J. 500 Percutaneous thermocoagulations of the Gasserian
Neurosurg. 2004;15(2-3):203-219. ganglion for trigeminal pain. Surg Neurol. 1977;8(2):126-131.
2. Raslan A, McCartney S, Burchiel K. Management of chronic severe 27. Taha JM, Tew JM Jr. Comparison of surgical treatments for trigemi-
pain: cerebral neuromodulatory and neuroablative approaches. In: nal neuralgia: reevaluation of radiofrequency rhizotomy.[see
Sakas DE, Simpson B, Krames ES, eds. Operative Neuromodulation. comment]. Neurosurgery. 1996;38(5):865-871.
New York: Springer-Verlag; 2007. 28. Taha JM, Tew JM Jr, Buncher CR. A prospective 15-year follow up
3. Raslan A, McCartney S, Burchiel K. Management of chronic severe of 154 consecutive patients with trigeminal neuralgia treated
pain: spinal neuromodulatory and neuroablative approaches. In: by percutaneous stereotactic radiofrequency thermal rhizotomy.
Sakas DE, Simpson B, Krames ES, eds. Operative Neuromodulation. J Neurosurg. 1995;83(6):989-993.
New York: Springer-Verlag; 2007. 29. Walchenbach R, Voormolen JH, Hermans J. Microvascular decom-
4. Kanpolat Y, Savas A, Akyar S, et al. Percutaneous computed pression for trigeminal neuralgia: a critical reappraisal. Clin Neurol
tomography-guided spinal destructive procedures for pain control. Neurosurg. 1994;96(4):290-295.
Neurosurg Q. 2004;14(4):229-238. 30. Yoon KB, Wiles JR, Miles JB, et al. Long-term outcome of percu-
5. Angst MS, Clark JD. Opioid-induced hyperalgesia: a qualitative taneous thermocoagulation for trigeminal neuralgia. Anaesthesia.
systematic review. Anesthesiology. 2006;104(3):570-587. 1999;54(8):803-808.
6. Hojsted J, Nielsen PR, Eriksen J, et al. Breakthrough pain in opioid- 31. Zakrzewska JM, Jassim S, Bulman JS. A prospective, longitudinal
treated chronic non-malignant pain patients referred to a multidis- study on patients with trigeminal neuralgia who underwent radio-
ciplinary pain centre: a preliminary study. Acta Anaesthesiol Scand. frequency thermocoagulation of the Gasserian ganglion. Pain.
2006;50(10):1290-1296. 1999;79(1):51-58.
7. Hojsted J, Sjogren P. An update on the role of opioids in the man- 32. Zorman G, Wilson CB. Outcome following microsurgical vascular
agement of chronic pain of nonmalignant origin. Curr Opin Anaes- decompression or partial sensory rhizotomy in 125 cases of trigemi-
thesiol. 2007;20(5):451-455. nal neuralgia. Neurology. 1984;34(10):1362-1365.
8. Jensen MK, Thomsen AB, Hojsted J. 10-year follow-up of chronic 33. Kanpolat Y, Savas A, Bekar A, et al. Percutaneous controlled radio-
non-malignant pain patients: opioid use, health related quality of frequency trigeminal rhizotomy for the treatment of idiopathic
life and health care utilization. Eur J Pain. 2006;10(5):423-433. trigeminal neuralgia: 25-year experience with 1,600 patients. Neu-
9. Turner JA, Sears JM, Loeser JD. Programmable intrathecal opioid rosurgery. 2001;48(3):524-532, discussion 532-524.
delivery systems for chronic noncancer pain: a systematic review 34. Broggi G, Franzini A, Lasio G, et al. Long-term results of percuta-
of effectiveness and complications. Clin J Pain. 2007;23(2):180- neous retrogasserian thermorhizotomy for “essential” trigeminal
195. neuralgia: considerations in 1000 consecutive patients. Neurosur-
10. Coffey RJ, Lozano AM. Neurostimulation for chronic noncancer gery. 1990;26(5):783-786, discussion 786-787.
pain: an evaluation of the clinical evidence and recommendations 34a.  Ong KS, Keng SB. Evaluation of surgical procedures for trigeminal
for future trial designs. J Neurosurg. 2006;105(2):175-189. neuralgia. Anesth Prog. 2003;50:181-188.
11. Jirásek A. The Surgery of Pain. Paper presented at Czechoslovak 35. Aubuchon AC, Chan MD, Lovato JF, et al. Repeat gamma knife
Academy of Sciences and Artia, 1962, Prague. radiosurgery for trigeminal neuralgia. Int J Radiat Oncol Biol Phys.
12. Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treat- 2011;81(4):1059-1065.
ment of nonmalignant pain: a structured literature review. J Neuro- 36. Bender M, Pradilla G, Batra S, et al. Effectiveness of repeat glycerol
surg. 2008;109(3):389-404. rhizotomy in treating recurrent trigeminal neuralgia. Neurosurgery.
13. Cetas JS, Raslan AM, Burchiel K. Evidence base for destructive 2012;70(5):1125-1133, discussion 1133-1124.
procedures. In: Winn HR, ed. Youmans Neurological Surgery. 6th ed. 37. Bender MT, Pradilla G, Batra S, et al. Glycerol rhizotomy and
Philadelphia: Elsevier Saunders; 2008. radiofrequency thermocoagulation for trigeminal neuralgia in mul-
14. USPSTF. U.S. Preventive Services Task Force (USPSTF). 1996; tiple sclerosis. J Neurosurg. 2013;118(2):329-336.
<http://www.ahrq.gov/clinic/uspstfix.htm>. 38. Bozkurt M, Al-Beyati ES, Ozdemir M, et al. Management of bilat-
15. Murali R, Rovit RL. Are peripheral neurectomies of value in the eral trigeminal neuralgia with trigeminal radiofrequency rhizotomy:
treatment of trigeminal neuralgia? An analysis of new cases and a treatment strategy for the life-long disease. Acta Neurochir (Wien).
cases involving previous radiofrequency gasserian thermocoagula- 2012;154(5):785-791, discussion 791-782.
tion. J Neurosurg. 1996;85(3):435-437. 39. Kondziolka D, Zorro O, Lobato-Polo J, et al. Gamma Knife stereo-
16. Oturai AB, Jensen K, Eriksen J, et al. Neurosurgery for trigeminal tactic radiosurgery for idiopathic trigeminal neuralgia. J Neurosurg.
neuralgia: comparison of alcohol block, neurectomy, and radio- 2010;112(4):758-765.
frequency coagulation [see comment]. Clin J Pain. 1996;12(4): 40. Kouzounias K, Lind G, Schechtmann G, et al. Comparison of per-
311-315. cutaneous balloon compression and glycerol rhizotomy for the treat-
17. Quinn JH, Weil T. Trigeminal neuralgia: treatment by repetitive ment of trigeminal neuralgia. J Neurosurg. 2010;113(3):486-492.
peripheral neurectomy. Supplemental report. J Oral Surg. 1975; 41. Lazzara BM, Ortiz O, Bordia R, et al. Cyberknife radiosurgery in
33(8):591-595. treating trigeminal neuralgia. J Neurointerv Surg. 2013;5(1):81-85.
18. Tucker SM, Tarlov EC. Intraorbital surgery for trigeminal neural- 42. Marshall K, Chan MD, McCoy TP, et al. Predictive variables for
gia. Ophthal Plast Reconstr Surg. 2005;21(1):11-15. the successful treatment of trigeminal neuralgia with gamma knife
19. Ziccardi VB, Janosky JE, Patterson GT, et al. Peripheral trigeminal radiosurgery. Neurosurgery. 2012;70(3):566-572, discussion 572-563.
nerve surgery for patients with atypical facial pain. J Craniomaxillofac 43. Mathieu D, Effendi K, Blanchard J, et al. Comparative study of
Surg. 1994;22(6):355-360. Gamma Knife surgery and percutaneous retrogasserian glycerol
20. Bederson JB, Wilson CB. Evaluation of microvascular decompres- rhizotomy for trigeminal neuralgia in patients with multiple sclero-
sion and partial sensory rhizotomy in 252 cases of trigeminal neu- sis. J Neurosurg. 2012;117(suppl):175-180.
ralgia. J Neurosurg. 1989;71(3):359-367. 44. Tang CT, Chang SD, Tseng KY, et al. CyberKnife stereotactic
21. Broggi G, Ferroli P, Franzini A, et al. The role of surgery in the radiosurgical rhizotomy for refractory trigeminal neuralgia. J Clin
treatment of typical and atypical facial pain. Neurol Sci. 2005;26(suppl Neurosci. 2011;18(11):1449-1453.
2):s95-s100. 45. Udupi BP, Chouhan RS, Dash HH, et al. Comparative evaluation
22. Burchiel KJ. Percutaneous retrogasserian glycerol rhizolysis in of percutaneous retrogasserian glycerol rhizolysis and radiofre-
the management of trigeminal neuralgia. J Neurosurg. 1988;69(3): quency thermocoagulation techniques in the management of
361-366. trigeminal neuralgia. Neurosurgery. 2012;70(2):407-412, discussion
23. Pickett GE, Bisnaire D, Ferguson GG. Percutaneous retrogasserian 412-403.
glycerol rhizotomy in the treatment of tic douloureux associated 46. Weller M, Marshall K, Lovato JF, et al. Single-institution retrospec-
with multiple sclerosis. Neurosurgery. 2005;56(3):537-545, discus- tive series of gamma knife radiosurgery in the treatment of multiple
sion 537-545. sclerosis-related trigeminal neuralgia: factors that predict efficacy.
24. Pollock BE, Ecker RD. A prospective cost-effectiveness study of Stereotact Funct Neurosurg. 2013;92(1):53-58.
trigeminal neuralgia surgery. Clin J Pain. 2005;21(4):317-322. 47. Barlocher CB, Krauss JK, Seiler RW. Kryorhizotomy: an alternative
25. Scrivani SJ, Keith DA, Mathews ES, et al. Percutaneous stereotactic technique for lumbar medial branch rhizotomy in lumbar facet
differential radiofrequency thermal rhizotomy for the treatment of syndrome. J Neurosurg. 2003;98(suppl):14-20.
1451.e2 SECTION 6  Pain

48. Dreyfuss P, Halbrook B, Pauza K, et al. Efficacy and validity of neurotomy: a randomised, double-blind, placebo-controlled trial
radiofrequency neurotomy for chronic lumbar zygapophysial joint [see comment]. Pain. 1997;73(1):15-22.
pain. Spine. 2000;25(10):1270-1277. 72. Cepero R, Miller RH, Bressler KL. Long-term results of spheno-
49. Echols DH. Sensory rhizotomy following operation for ruptured palatine ganglioneurectomy for facial pain. Am J Otolaryngol. 1987;
intervertebral disc. A review of 62 cases. J Neurosurg. 1969;31(3): 8(3):171-174.
335-338. 73. Pieper DR, Dickerson J, Hassenbusch SJ. Percutaneous retrogas-
50. Gallagher J, Petriccione Di Vadi PL, Wedley JR, et al. Radiofre- serian glycerol rhizolysis for treatment of chronic intractable
quency facet joint denervation in the treatment of low back pain: a cluster headaches: long-term results. Neurosurgery. 2000;46(2):363-
prospective controlled double-blind study to assess its efficacy. Pain 368, discussion 368-370.
Clinic. 1994;7(3):193-198. 74. Taha JM, Tew JM Jr. Long-term results of radiofrequency rhizot-
51. Leclaire R, Fortin L, Lambert R, et al. Radiofrequency facet joint omy in the treatment of cluster headache. Headache. 1995;35(4):
denervation in the treatment of low back pain: a placebo-controlled 193-196.
clinical trial to assess efficacy [see comment]. Spine. 2001;26(13):1411- 75. Xiong NX, Zhao HY, Zhang FC, et al. Vagoglossopharyngeal neu-
1416, discussion 1417. ralgia treated by microvascular decompression and glossopharyn-
52. Loeser JD. Dorsal rhizotomy for the relief of chronic pain. J Neu- geal rhizotomy: clinical results of 21 cases. Stereotact Funct Neurosurg.
rosurg. 1972;36(6):745-750. 2012;90(1):45-50.
53. Onofrio BM, Campa HK. Evaluation of rhizotomy. Review of 12 76. Arbit E, Galicich JH, Burt M, et al. Modified open thoracic rhi-
years’ experience. J Neurosurg. 1972;36(6):751-755. zotomy for treatment of intractable chest wall pain of malignant
54. Pevsner Y, Shabat S, Catz A, et al. The role of radiofrequency in the etiology. Ann Thorac Surg. 1989;48(6):820-823.
treatment of mechanical pain of spinal origin. Eur Spine J. 2003; 77. Barrash JM, Leavens ME. Dorsal rhizotomy for the relief of intrac-
12(6):602-605. table pain of malignant tumor origin. J Neurosurg. 1973;38(6):
55. Sanders M, Zuurmond WW. Efficacy of sphenopalatine ganglion 755-757.
blockade in 66 patients suffering from cluster headache: a 12- to 78. El-Sayed GG. A new catheter technique for thoracic subarachnoid
70-month follow-up evaluation. J Neurosurg. 1997;87(6):876-880. neurolysis in advanced lung cancer patients. Pain Pract. 2007;7(1):
56. Scoville WB. Extradural spinal sensory rhizotomy. J Neurosurg. 27-30.
1966;25(1):94-95. 79. Esposito S, Bruni P, Delitala A, et al. Therapeutic approach to the
57. Staender M, Maerz U, Tonn JC, et al. Computerized tomography- Pancoast pain syndrome. Appl Neurophysiol. 1985;48(1-6):262-266.
guided kryorhizotomy in 76 patients with lumbar facet joint syn- 80. Giorgi C, Broggi G. Surgical treatment of glossopharyngeal neu-
drome. J Neurosurg. 2005;3(6):444-449. ralgia and pain from cancer of the nasopharynx. A 20-year experi-
58. Tzaan WC, Tasker RR. Percutaneous radiofrequency facet ence. J Neurosurg. 1984;61(5):952-955.
rhizotomy—experience with 118 procedures and reappraisal of its 81. Ischia S, Luzzani A, Ischia A, et al. Subarachnoid neurolytic block
value. Can J Neurol Sci. 2000;27(2):125-130. (L5-S1) and unilateral percutaneous cervical cordotomy in the
59. van Kleef M, Barendse GA, Kessels A, et al. Randomized trial of treatment of pain secondary to pelvic malignant disease. Pain. 1984;
radiofrequency lumbar facet denervation for chronic low back pain. 20(2):139-149.
Spine. 1999;24(18):1937-1942. 82. Pagura JR, Schnapp M, Passarelli P. Percutaneous radiofrequency
60. White JC, Kjellberg RN. Posterior spinal rhizotomy: a substitute glossopharyngeal rhizotomy for cancer pain. Appl Neurophysiol.
for cordotomy in the relief of localized pain in patients with normal 1983;46(1-4):154-159.
life-expectancy. Neurochirurgia (Stuttg). 1973;16(5):141-170. 83. Rodriguez-Bigas M, Petrelli NJ, Herrera L, et al. Intrathecal phenol
61. Tome-Bermejo F, Barriga-Martin A, Martin JL. Identifying patients rhizotomy for management of pain in recurrent unresectable carci-
with chronic low back pain likely to benefit from lumbar facet noma of the rectum. Surg Gynecol Obstet. 1991;173(1):41-44.
radiofrequency denervation: a prospective study. J Spinal Disord 84. Saris SC, Silver JM, Vieira JF, et al. Sacrococcygeal rhizotomy for
Tech. 2011;24(2):69-75. perineal pain. Neurosurgery. 1986;19(5):789-793.
62. Oh WS, Shim JC. A randomized controlled trial of radiofrequency 85. Hooten WM, Martin DP, Huntoon MA. Radiofrequency neurot-
denervation of the ramus communicans nerve for chronic disco- omy for low back pain: evidence-based procedural guidelines [see
genic low back pain. Clin J Pain. 2004;20(1):55-60. comment]. Pain Med. 2005;6(2):129-138.
63. Barnsley L. Percutaneous radiofrequency neurotomy for chronic 86. Burchiel KJ. A new classification for facial pain. Neurosurgery.
neck pain: outcomes in a series of consecutive patients. Pain Med. 2003;53(5):1164-1166, discussion 1166-1167.
2005;6(4):282-286. 87. Park YS, Kim JP, Chang WS, et al. Gamma knife radiosurgery for
64. Govind J, King W, Bailey B, et al. Radiofrequency neurotomy for idiopathic trigeminal neuralgia as primary vs. secondary treatment
the treatment of third occipital headache [see comment]. J Neurol option. Clin Neurol Neurosurg. 2011;113(6):447-452.
Neurosurg Psychiatry. 2003;74(1):88-93. 88. Mendelsohn D, Ranjan M, Hawley P, et al. Percutaneous trigeminal
65. Lord SM, Barnsley L, Wallis BJ, et al. Percutaneous radio-frequency rhizotomy for facial pain secondary to head and neck malignancy.
neurotomy for chronic cervical zygapophyseal-joint pain [see Clin J Pain. 2013;29(10):e4-e5.
comment]. N Engl J Med. 1996;335(23):1721-1726. 89. Geurts JWM, van Wijk RMAW, Wynne HJ, et al. Radiofrequency
66. McDonald GJ, Lord SM, Bogduk N. Long-term follow-up of lesioning of dorsal root ganglia for chronic lumbosacral radicular
patients treated with cervical radiofrequency neurotomy for chronic pain: a randomised, double-blind, controlled trial [see comment].
neck pain [see comment]. Neurosurgery. 1999;45(1):61-67, discus- Lancet. 2003;361(9351):21-26.
sion 67-68. 90. Slappendel R, Crul BJ, Braak GJ, et al. The efficacy of radiofre-
67. Sapir DA, Gorup JM. Radiofrequency medial branch neurotomy in quency lesioning of the cervical spinal dorsal root ganglion in a
litigant and nonlitigant patients with cervical whiplash: a prospec- double blinded randomized study: no difference between 40 degrees
tive study [see comment]. Spine. 2001;26(12):E268-E273. C and 67 degrees C treatments. Pain. 1997;73(2):159-163.
68. Stovner LJ, Kolstad F, Helde G. Radiofrequency denervation of 91. Chun-jing H, yi-ran L, hao-xiong N. Effects of dorsal root ganglion
facet joints C2-C6 in cervicogenic headache: a randomized, double- destruction by Adriamycin in patients with postherpetic neuralgia.
blind, sham-controlled study [see comment]. Cephalalgia. 2004; Acta Cir Bras. 2012;27(6):404-409.
24(10):821-830. 92. Dubuisson D. Treatment of occipital neuralgia by partial posterior
69. van Kleef M, Liem L, Lousberg R, et al. Radiofrequency lesion rhizotomy at C1-3. J Neurosurg. 1995;82(4):581-586.
adjacent to the dorsal root ganglion for cervicobrachial pain: a 93. Fedder SL. T2-ganglionectomy via limited costotransversectomy
prospective double blind randomized study. Neurosurgery. 1996; for minor causalgia. Spine. 1990;15(4):269-270.
38(6):1127-1131, discussion 1131-1122. 94. Hosobuchi Y. The majority of unmyelinated afferent axons in human
70. van Suijlekom HA, van Kleef M, Barendse GA, et al. Radiofre- ventral roots probably conduct pain. Pain. 1980;8(2):167-180.
quency cervical zygapophyseal joint neurotomy for cervicogenic 95. Jansen J. Surgical treatment of non-responsive cervicogenic head-
headache: a prospective study of 15 patients. Funct Neurol. 1998; ache. Clin Exp Rheumatol. 2000;18(suppl 19):S67-S70.
13(4):297-303. 96. Kato S, Otsuki T, Yamamoto T, et al. Retrograde Adriamycin
71. Wallis BJ, Lord SM, Bogduk N. Resolution of psychological dis- sensory ganglionectomy: novel approach for the treatment of
tress of whiplash patients following treatment by radiofrequency intractable pain. Stereotact Funct Neurosurg. 1990;54-55:86-89.
CHAPTER 179  Evidence Base for Destructive Procedures 1451.e3

97. Lozano AM, Vanderlinden G, Bachoo R, et al. Microsurgical C-2 122. Sindou M, Mertens P, Wael M. Microsurgical DREZotomy for pain
ganglionectomy for chronic intractable occipital pain. J Neurosurg. due to spinal cord and/or cauda equina injuries: long-term results 179
1998;89(3):359-365. in a series of 44 patients. Pain. 2001;92(1-2):159-171.
98. Murphy JP. Occipital neurectomy in the treatment of headache. 123. Spai M, Petkovi S, Tadi R, et al. DREZ surgery on conus medullaris
Results in 30 cases. Md State Med J. 1969;18(6):62-66. (after failed implantation of vascular omental graft) for treating
99. North RB, Kidd DH, Campbell JN, et al. Dorsal root ganglionec- chronic pain due to spine (gunshot) injuries. Acta Neurochir (Wien).
tomy for failed back surgery syndrome: a 5-year follow-up study. 1999;141(12):1309-1312.
J Neurosurg. 1991;74(2):236-242. 124. Thomas DG, Jones SJ. Dorsal root entry zone lesions (Nashold’s
100. Osgood CP, Dujovny M, Faille R, et al. Microsurgical lumbosacral procedure) in brachial plexus avulsion. Neurosurgery. 1984;15(6):
ganglionectomy, anatomic rationale, and surgical results. Acta Neu- 966-968.
rochir (Wien). 1976;35(1-3):197-204. 125. Tomas R, Haninec P. Dorsal root entry zone (DREZ) localization
101. Pisapia JM, Bhowmick DA, Farber RE, et al. Salvage C2 ganglio- using direct spinal cord stimulation can improve results of the
nectomy after C2 nerve root decompression provides similar pain DREZ thermocoagulation procedure for intractable pain relief.
relief as a single surgical procedure for intractable occipital neural- Pain. 2005;116(1-2):159-163.
gia. World Neurosurg. 2012;77(2):362-369. 126. Young RF. Clinical experience with radiofrequency and laser DREZ
102. Smith FP. Trans-spinal ganglionectomy for relief of intercostal pain. lesions. J Neurosurg. 1990;72(5):715-720.
J Neurosurg. 1970;32(5):574-577. 127. Dreval ON. Ultrasonic DREZ-operations for treatment of pain due
103. Taub A, Robinson F, Taub E. Dorsal root ganglionectomy for intrac- to brachial plexus avulsion. Acta Neurochir (Wien). 1993;122(1-2):
table monoradicular sciatica. A series of 61 patients. Stereotact Funct 76-81.
Neurosurg. 1995;65(1-4):106-110. 128. Ishijima B, Shimoji K, Shimizu H, et al. Lesions of spinal and tri-
104. Weigel R, Capelle HH, Schmelz M, et al. Selective thoracic gan- geminal dorsal root entry zone for deafferentation pain. Experience
glionectomy for the treatment of segmental neuropathic pain. Eur of 35 cases. Appl Neurophysiol. 1988;51(2-5):175-187.
J Pain. 2012;16(10):1398-1402. 129. Kumagai Y, Shimoji K, Honma T, et al. Problems related to dorsal
105. Wetzel FT, Phillips FM, Aprill CN, et al. Extradural sensory rhi- root entry zone lesions. Acta Neurochir (Wien). 1992;115(3-4):
zotomy in the management of chronic lumbar radiculopathy: a 71-78.
minimum 2-year follow-up study. Spine. 1997;22(19):2283-2291, 130. Prestor B. Microsurgical junctional DREZ coagulation for treat-
discussion 2291-2282. ment of deafferentation pain syndromes. Surg Neurol. 2001;56(4):
106. Wilkinson HA, Chan AS. Sensory ganglionectomy: theory, techni- 259-265.
cal aspects, and clinical experience [see comment]. J Neurosurg. 131. Samii M, Bear-Henney S, Ludemann W, et al. Treatment of refrac-
2001;95(1):61-66. tory pain after brachial plexus avulsion with dorsal root entry zone
107. Stechison MT, Mullin BB. Surgical treatment of greater occipital lesions. Neurosurgery. 2001;48(6):1269-1275, discussion 1275-1267.
neuralgia: an appraisal of strategies. Acta Neurochir (Wien). 1994; 132. Tasker RR, DeCarvalho GT, Dolan EJ. Intractable pain of spinal
131(3-4):236-240. cord origin: clinical features and implications for surgery. J Neuro-
108. Sindou MP, Blondet E, Emery E, et al. Microsurgical lesioning surg. 1992;77(3):373-378.
in the dorsal root entry zone for pain due to brachial plexus avul- 133. Abdelaziz OS, Cosgrove GR. Stereotactic cingulotomy for the
sion: a prospective series of 55 patients. J Neurosurg. 2005;102(6): treatment of chronic pain. In: Burchiel KJ, ed. Surgical Management
1018-1028. of Pain. New York: Thieme Medical Publishers; 2002:812-820.
109. Friedman AH, Bullitt E. Dorsal root entry zone lesions in the treat- 134. Thomas DG, Kitchen ND. Long-term follow up of dorsal root
ment of pain following brachial plexus avulsion, spinal cord injury entry zone lesions in brachial plexus avulsion. J Neurol Neurosurg
and herpes zoster. Appl Neurophysiol. 1988;51(2-5):164-169. Psychiatry. 1994;57(6):737-738.
110. Powers SK, Barbaro NM, Levy RM. Pain control with laser- 135. Aichaoui F, Mertens P, Sindou M. Dorsal root entry zone lesioning
produced dorsal root entry zone lesions. Appl Neurophysiol. 1988; for pain after brachial plexus avulsion: results with special emphasis
51(2-5):243-254. on differential effects on the paroxysmal versus the continuous
111. Rath SA, Braun V, Soliman N, et al. Results of DREZ coagulations components. A prospective study in a 29-patient consecutive series.
for pain related to plexus lesions, spinal cord injuries and posther- Pain. 2011;152(8):1923-1930.
petic neuralgia. Acta Neurochir (Wien). 1996;138(4):364-369. 136. Ali M, Saitoh Y, Oshino S, et al. Differential efficacy of electric
112. Campbell JN, Solomon CT, James CS. The Hopkins experience motor cortex stimulation and lesioning of the dorsal root entry zone
with lesions of the dorsal horn (Nashold’s operation) for pain from for continuous vs paroxysmal pain after brachial plexus avulsion.
avulsion of the brachial plexus. Appl Neurophysiol. 1988;51(2-5): Neurosurgery. 2011;68(5):1252-1257, discussion 1257-1258.
170-174. 137. Chun HJ, Kim YS, Yi HJ. A modified microsurgical DREZotomy
113. Edgar RE, Best LG, Quail PA, et al. Computer-assisted DREZ procedure for refractory neuropathic pain. World Neurosurg. 2011;
microcoagulation: posttraumatic spinal deafferentation pain. 75(3-4):551-557.
J Spinal Disord. 1993;6(1):48-56. 138. Dong S, Hu YS, Du W, et al. Changes in spontaneous dorsal horn
114. Friedman AH, Nashold BS Jr. DREZ lesions for relief of pain potentials after dorsal root entry zone lesioning in patients with pain
related to spinal cord injury. J Neurosurg. 1986;65(4):465-469. after brachial plexus avulsion. J Int Med Res. 2012;40(4):1499-1506.
115. Levy WJ, Nutkiewicz A, Ditmore QM, et al. Laser-induced dorsal 139. Kanpolat Y, Al-Beyati E, Ugur HC, et al. A curative treatment
root entry zone lesions for pain control. Report of three cases. option for complex regional pain syndrome (CRPS) type I: dorsal
J Neurosurg. 1983;59(5):884-886. root entry zone operation (report of two cases). Turk Neurosurg.
116. Nashold BS Jr, Bullitt E. Dorsal root entry zone lesions to 2014;24(1):127-130.
control central pain in paraplegics. J Neurosurg. 1981;55(3): 140. Ruiz-Juretschke F, Garcia-Salazar F, Garcia-Leal R, et al. Treatment
414-419. of neuropathic deafferentation pain using DREZ lesions; long-term
117. Nashold BS Jr, Ostdahl RH. Dorsal root entry zone lesions for pain results. Neurologia. 2011;26(1):26-31.
relief. J Neurosurg. 1979;51(1):59-69. 141. Sandwell SE, El-Naggar AO. Nucleus caudalis dorsal root entry
118. Nashold BS Jr, Vieira J, el-Naggar AO. Pain and spinal cysts in zone lesioning for the treatment of anesthesia dolorosa. J Neurosurg.
paraplegia: treatment by drainage and DREZ operation. Br J Neu- 2013;118(3):534-538.
rosurg. 1990;4(4):327-335. 142. Tomycz ND, Moossy JJ. Follow-up 26 years after dorsal root entry
119. Powers SK, Adams JE, Edwards MS, et al. Pain relief from dorsal zone thermocoagulation for brachial plexus avulsion and phantom
root entry zone lesions made with argon and carbon dioxide micro- limb pain. J Neurosurg. 2011;114(1):196-199.
surgical lasers. J Neurosurg. 1984;61(5):841-847. 143. Park SH, Hwang SK, Kang DH, et al. The retrogasserian zone
120. Richter HP, Seitz K. Dorsal root entry zone lesions for the control versus dorsal root entry zone: comparison of two targeting tech-
of deafferentation pain: experiences in ten patients. Neurosurgery. niques of gamma knife radiosurgery for trigeminal neuralgia. Acta
1984;15(6):956-959. Neurochir (Wien). 2010;152(7):1165-1170.
121. Samii M, Moringlane JR. Thermocoagulation of the dorsal root 144. Tao W, Hu Y, Chen F, et al. Microsurgical dorsal root entry zone
entry zone for the treatment of intractable pain. Neurosurgery. coagulation for chronic neuropathic pain due to spinal cord and/or
1984;15(6):953-955. cauda equina injuries. Chin Med J (Engl). 2014;127(1):182-184.
1451.e4 SECTION 6  Pain

145. Bullard DE, Nashold BS Jr. The caudalis DREZ for facial pain. 173. Raskind R. Analytical review of open cordotomy. Int Surg.
Stereotact Funct Neurosurg. 1997;68(1-4 Pt 1):168-174. 1969;51(3):226-231.
146. Kanpolat Y, Tuna H, Bozkurt M, et al. Spinal and nucleus caudalis 174. Rosomoff HL. Bilateral percutaneous cervical radiofrequency cor-
dorsal root entry zone operations for chronic pain. Neurosurgery. dotomy. J Neurosurg. 1969;31(1):41-46.
2008;62(suppl 1):235-242, discussion 242-234. 175. Muke R, Correia A. Potentials and limits of percutaneous cer-
147. Rossitch E Jr, Zeidman SM, Nashold BS Jr. Nucleus caudalis DREZ vical cordotomy. Deutsche Gesellschaft fur Neurochirurgie. 1975;3:
for facial pain due to cancer [see comment]. Br J Neurosurg. 195-198.
1989;3(1):45-49. 176. Acosta C, Grossman RG. Relief of intractable pain by percutane-
148. Sindou M. Microsurgical DREZotomy (MDT) for pain, spasticity, ous anterolateral radiofrequency cordotomy. Tex Med. 1969;65(7):
and hyperactive bladder: a 20-year experience. Acta Neurochir 36-40.
(Wien). 1995;137(1-2):1-5. 177. Amano K, Kawamura H, Tanikawa T, et al. Bilateral versus unilat-
149. Brodal A. Central course of afferent fibers for pain in facial glos- eral percutaneous high cervical cordotomy as a surgical method of
sopharyngeal and vagus nerves clinical observation. Arch Neurol pain relief. Acta Neurochir Suppl (Wien). 1991;52:143-145.
Psychiatry. 1947;57:292-306. 178. Batzdorf U, Weingarten SM. Percutaneous cordotomy. A simplified
150. Schvarcz JR. Stereotactic trigeminal tractotomy. Confin Neurol. approach to the management of intractable pain. Calif Med. 1970;
1975;37(1-3):73-77. 112(1):21-26.
151. Schvarcz JR. Postherpetic craniofacial dysaesthesiae: their manage- 179. Crue BL, Felsoory A. Transcutaneous high cervical “electrical cor-
ment by stereotaxic trigeminal nucleotomy. Acta Neurochir (Wien). dotomy”. Minn Med. 1974;57(3):204-209.
1977;38(1-2):65-72. 180. Crul BJ, Blok LM, van Egmond J, et al. The present role of percu-
152. Crue BL, Carregal EJA, Felsoory A. Percutaneous stereotactic taneous cervical cordotomy for the treatment of cancer pain. J Head
radiofrequency trigeminal tractotomy with neurophysiological and Pain. 2005;6(1):24-29.
recordings. Stereotact Funct Neurosurg. 1972;34(6):389-397. 181. Fenstermaker RA, Sternau LL, Takaoka Y. CT-assisted percutane-
153. Hosobuchi Y, Rutkin B. Descending trigeminal tractotomy. Neuro- ous anterior cordotomy: technical note. Surg Neurol. 1995;43(2):147-
physiological approach. Arch Neurol. 1971;25(2):115-125. 149, discussion 149-150.
154. Kanpolat Y, Savas A, Batay F, et al. Computed tomography-guided 182. Foer WH. Percutaneous cervical radiofrequency cordotomy. A
trigeminal tractotomy-nucleotomy in the management of vagoglos- modern method for relief of intractable pain. J Med Soc N J. 1971;
sopharyngeal and geniculate neuralgias. Neurosurgery. 1998;43(3): 68(9):737-741.
484-489, discussion 490. 183. Fox JL. Percutaneous trigeminal tractotomy for facial pain. Acta
155. Kunc Z. Treatment of essential neuralgia of the 9th nerve by selec- Neurochir (Wien). 1973;29(1):83-88.
tive tractotomy. J Neurosurg. 1965;23(5):494-500. 184. Fox JL. A new method of percutaneous chordotomy for relief
156. Hitchock ER, Schvarcz JR. Stereotaxic trigeminal tractotomy for of intractable pain. Med Ann Dist Columbia. 1969;38(3):131-134.
post-herpetic facial pain. J Neurosurg. 1972;37(4):412-417. passim.
157. Thompson EM, Burchiel KJ, Raslan AM. Percutaneous trigeminal 185. Gildenberg PL. Percutaneous cervical cordotomy. Appl Neuro-
tractotomy-nucleotomy with use of intraoperative computed physiol. 1976;39(2):97-113.
tomography and general anesthesia: report of 2 cases. Neurosurg 186. Grote W, Roosen K, Bock WJ. High cervical percutaneous cor-
Focus. 2013;35(3):E5. dotomy in intractable pain. Neurochirurgia (Stuttg). 1978;21(6):
158. Fox JL. Intractable facial pain relieved by percutaneous trigeminal 209-212.
tractotomy. JAMA. 1971;218(13):1940-1941. 187. Hogberg T, Rabow L, Rosenberg P, et al. The use of chordotomy
159. Hitchcock E. Stereotactic trigeminal tractotomy. Farmatsiia. 1970; to treat pain from gynecologic cancer. Eur J Gynaecol Oncol.
19(3):131-135. 1989;10(5):337-340.
160. Kanpolat Y, Caglar S, Akyar S, et al. CT-guided pain procedures for 188. Ischia S, Ischia A, Luzzani A, et al. Results up to death in the treat-
intractable pain in malignancy. Acta Neurochir Suppl. 1995;64:88-91. ment of persistent cervico-thoracic (Pancoast) and thoracic malig-
161. Plangger CA, Fischer J, Grunert V, et al. Tractotomy and partial nant pain by unilateral percutaneous cervical cordotomy. Pain.
vertical nucleotomy—for treatment of special forms of trigeminal 1985;21(4):339-355.
neuralgia and cancer pain of face and neck. Acta Neurochir Suppl 189. Ischia S, Luzzani A, Ischia A, et al. Bilateral percutaneous cervical
(Wien). 1987;39:147-150. cordotomy: immediate and long-term results in 36 patients with
162. Raslan AM. Percutaneous computed tomography-guided radiofre- neoplastic disease. J Neurol Neurosurg Psychiatry. 1984;47(2):
quency ablation of upper spinal cord pain pathways for cancer- 141-147.
related pain. Neurosurgery. 2008;62(suppl 1):226-233, discussion 190. Ischia S, Luzzani A, Ischia A, et al. Role of unilateral percutaneous
233-224. cervical cordotomy in the treatment of neoplastic vertebral pain.
163. Collins KL, Taren JA, Patil PG. Four-decade maintenance of anal- Pain. 1984;19(2):123-131.
gesia with percutaneous cordotomy. Stereotact Funct Neurosurg. 191. Jackson MB, Pounder D, Price C, et al. Percutaneous cervical cor-
2012;90(4):266-272. dotomy for the control of pain in patients with pleural mesotheli-
164. Lewin J, Hugel H, Sharma ML. Percutaneous cervical cordotomy oma. Thorax. 1999;54(3):238-241.
for non-cancer pain in a patient with terminal esophageal carci- 192. Kanpolat Y, Savas A, Ucar T, et al. CT-guided percutaneous selec-
noma. J Pain Symptom Manage. 2012;43(3):e8-e9. tive cordotomy for treatment of intractable pain in patients with
165. Amano K, Kitamura K, Sano K, et al. Relief of intractable pain from malignant pleural mesothelioma. Acta Neurochir (Wien). 2002;
neurosurgical point of view with reference to present limits and 144(6):595-599, discussion 599.
clinical indications—a review of 100 consecutive cases. Neurol Med 193. Kelly DL Jr, Alexander E Jr. Radiofrequency cordotomy for intrac-
Chir (Tokyo). 1976;16:141-153. table pain. N C Med J. 1966;27(10):457-462.
166. Cowie RA, Hitchcock ER. The late results of anterolateral cor- 194. Krol G, Arbit E. Percutaneous lateral cervical cordotomy: target
dotomy for pain relief. Acta Neurochir (Wien). 1982;64(1-2):39-50. localization with water-soluble contrast medium. J Neurosurg. 1993;
167. Diemath HE, Heppner F, Walker AE. Anterolateral chordotomy for 79(3):390-392.
relief of pain. Postgrad Med. 1961;29:485-495. 195. Lahuerta J, Bowsher D, Lipton S, et al. Percutaneous cervical cor-
168. Frankel SA, Prokop JD. Value of cordotomy for the relief of pain. dotomy: a review of 181 operations on 146 patients with a study on
N Engl J Med. 1961;264:971-974. the location of “pain fibers” in the C-2 spinal cord segment of 29
169. French LA. High cervical tractotomy: technique and results. Clin cases. J Neurosurg. 1994;80(6):975-985.
Neurosurg. 1974;21:239-245. 196. Lin PM, Gildenberg PL, Polakoff PP. An anterior approach to
170. Grant FC, Wood FA. Experiences with cordotomy. Clin Neurosurg. percutaneous lower cervical cordotomy. J Neurosurg. 1966;25(5):
1957;5:38-65. 553-560.
171. Porter RW, Hohmann GW, Bors E, et al. Cordotomy for pain fol- 197. Lipton S. Percutaneous cervical cordotomy and the injection of the
lowing cauda equina injury. Arch Surg. 1966;92(5):765-770. pituitary with alcohol. Anaesthesia. 1978;33(10):953-957.
172. Probst C. Microsurgical cordotomy in 20 patients with epi-/intra- 198. McGirt MJ, Villavicencio AT, Bulsara KR, et al. MRI-guided frame-
dural fibrosis following operation for lumbar disc herniation. Acta less stereotactic percutaneous cordotomy. Stereotact Funct Neurosurg.
Neurochir (Wien). 1990;107(1-2):30-36. 2002;78(2):53-63.
CHAPTER 179  Evidence Base for Destructive Procedures 1451.e5

199. Meglio M, Cioni B. The role of percutaneous cordotomy in the 227. Cook AW, Kawakami Y. Commissural myelotomy. J Neurosurg.
treatment of chronic cancer pain. Acta Neurochir (Wien). 1981; 1977;47(1):1-6. 179
59(1-2):111-121. 228. Eiras J, Garcia J, Gomez J, et al. First results with extralemniscal
200. Mullan S, Hekmatpanah J, Dobben G, et al. Percutaneous, intra- myelotomy. Acta Neurochir Suppl (Wien). 1980;30:377-381.
medullary cordotomy utilizing the unipolar anodal electrolytic 229. Fascendini A, Biroli F, Cassinari V. Critical evaluation of commis-
lesion. J Neurosurg. 1965;22(6):548-553. sural myelotomy in the treatment of intractable pain. J Neurosurg
201. Nagaro T, Amakawa K, Yamauchi Y, et al. Percutaneous cervical Sci. 1979;23(4):265-272.
cordotomy and subarachnoid phenol block using fluoroscopy in 230. Gildenberg PL, Hirshberg RM. Limited myelotomy for the treat-
pain control of costopleural syndrome [see comment][erratum ment of intractable cancer pain. J Neurol Neurosurg Psychiatry.
appears in Pain 1995 Mar;60(3):355-6]. Pain. 1994;58(3):325-330. 1984;47(1):94-96.
202. O’Connell JE. Anterolateral chordotomy for intractable pain in 231. Goedhart ZD, Francaviglia N, Feirabend HK, et al. Technical pit-
carcinoma of the rectum. Proc R Soc Med. 1969;62(12):1223-1225. falls in median commissural myelotomy for malignant sacral pain.
203. Palma A, Holzer J, Cuadra O, et al. Lateral percutaneous spinotha- Appl Neurophysiol. 1984;47(4-6):216-222.
lamic tractotomy. Acta Neurochir (Wien). 1988;93(3-4):100-103. 232. Hitchcock E. Stereotactic myelotomy. Proc R Soc Med. 1974;67(8):
204. Rand RW, Bauer RO, Smart CR, et al. Experiences with percutane- 771-772.
ous stereotaxic cryocordotomy. Bull Los Angeles Neurol Soc. 1965; 233. Hwang SL, Lin CL, Lieu AS, et al. Punctate midline myelotomy
30(3):142-147. for intractable visceral pain caused by hepatobiliary or pancreatic
205. Raslan AM. Percutaneous computed tomography-guided transdis- cancer. J Pain Symptom Manage. 2004;27(1):79-84.
cal low cervical cordotomy for cancer pain as a method to avoid 234. Kanpolat Y, Savas A, Caglar S, et al. Computerized tomography-
sleep apnea. Stereotact Funct Neurosurg. 2005;83(4):159-164. guided percutaneous extralemniscal myelotomy. Neurosurg Focus.
206. Rosomoff HL, Sheptak P, Carroll F. Modern pain relief: percutane- 1997;2(1):e5.
ous chordotomy. JAMA. 1966;196(6):482-486. 235. Kim YS, Kwon SJ. High thoracic midline dorsal column myelotomy
207. Rothbard MJ, Kotsilimbas DG, Jacobson SA, et al. Relief of intrac- for severe visceral pain due to advanced stomach cancer. Neurosur-
table pain in cervical carcinoma with percutaneous radiofrequency gery. 2000;46(1):85-90, discussion 90-82.
cordotomy. Obstet Gynecol. 1972;40(1):50-55. 236. Lippert RG, Hosobuchi Y, Nielsen SL. Spinal commissurotomy.
208. Salmon JH. Percutaneous cordotomy for relief of cancer pain. Ohio Surg Neurol. 1974;2(6):373-377.
State Med J. 1969;65(12):1237-1239. 237. Nauta HJ, Soukup VM, Fabian RH, et al. Punctate midline myelot-
209. Sanders M, Zuurmond W. Safety of unilateral and bilateral percu- omy for the relief of visceral cancer pain. J Neurosurg. 2000;
taneous cervical cordotomy in 80 terminally ill cancer patients. 92(suppl):125-130.
J Clin Oncol. 1995;13(6):1509-1512. 238. Papo I, Luongo A. High cervical commissural myelotomy in
210. Smith RA. Cervical percutaneous cordotomy. J Med Assoc Ga. 1972; the treatment of pain. J Neurol Neurosurg Psychiatry. 1976;39(7):
61(2):44-46. 705-710.
211. Stuart G, Cramond T. Role of percutaneous cervical cordotomy for 239. Schvarcz JR. Stereotactic extralemniscal myelotomy. J Neurol Neu-
pain of malignant origin. Med J Aust. 1993;158(10):667-670. rosurg Psychiatry. 1976;39(1):53-57.
212. Tasker RR. Percutaneous cervical cordotomy. Appl Neurophysiol. 240. Vilela Filho O, Araujo MR, Florencio RS, et al. CT-guided percu-
1976;39(2):114-121. taneous punctate midline myelotomy for the treatment of intrac-
213. Tasker RR, Organ LW. Percutaneous cordotomy. Physiological table visceral pain: a technical note. Stereotact Funct Neurosurg.
identification of target site. Confin Neurol. 1973;35(2):110-117. 2001;77(1-4):177-182.
214. Uihlein A, Weerasooriya LA, Holman CB. Percutaneous electric 241. Watling CJ, Payne R, Allen RR, et al. Commissural myelotomy for
cervical cordotomy for the relief of intractable pain. Mayo Clin Proc. intractable cancer pain: report of two cases. Clin J Pain. 1996;12(2):
1969;44(3):176-183. 151-156.
215. Yegul I, Erhan E. Bilateral CT-guided percutaneous cordotomy for 242. Viswanathan A, Burton AW, Rekito A, et al. Commissural myelot-
cancer pain relief. Clin Radiol. 2003;58(11):886-889. omy in the treatment of intractable visceral pain: technique and
216. Atkin N, Jackson KA, Danks RA. Bilateral open thoracic cordotomy outcomes. Stereotact Funct Neurosurg. 2010;88(6):374-382.
for refractory cancer pain: a neglected technique? J Pain Symptom 243. Shieff C, Nashold BS Jr. Thalamic pain and stereotactic mesen-
Manage. 2010;39(5):924-929. cephalotomy. Acta Neurochir Suppl (Wien). 1988;42:239-242.
217. Collins KL, Patil PG. Flat-panel fluoroscopy O-arm-guided percu- 244. Amano K, Kawamura H, Tanikawa T, et al. Long-term follow-up
taneous radiofrequency cordotomy: a new technique for the study of rostral mesencephalic reticulotomy for pain relief: report
treatment of unilateral cancer pain. Neurosurgery. 2013;72(suppl of 34 cases. Appl Neurophysiol. 1986;49(3):105-111.
Operative):27-34, discussion 34. 245. Helfant MH, Leksell L, Strang RR. Experiences with Intractable
218. Fonoff ET, de Oliveira YS, Lopez WO, et al. Endoscopic-guided Pain Treated by Stereotaxic Mesencephalotomy. Acta Chir Scand.
percutaneous radiofrequency cordotomy. J Neurosurg. 2010;113(3): 1965;129:573-580.
524-527. 246. Liberson WT, Voris HC, Uematsu S. Recording of somatosensory
219. France BD, Lewis RA, Sharma ML, et al. Cordotomy in evoked potentials during mesencephalotomy for intractable pain.
mesothelioma-related pain: a systematic review. BMJ Support Palliat Confin Neurol. 1970;32(2):185-194.
Care. 2014;4(1):19-29. 247. Spiegel EA, Wycis HT. Mesencephalotomy in treatment of intrac-
220. Reddy GD, Okhuysen-Cawley R, Harsh V, et al. Percutaneous table facial pain. AMA Arch Neurol Psychiatry. 1953;69(1):1-13.
CT-guided cordotomy for the treatment of pediatric cancer pain. 248. Wycis HT, Folkerts JF, Spiegel EA. Treatment of post-herpetic
J Neurosurg Pediatr. 2013;12(1):93-96. trigeminal neuralgia by mesencephalotomy or combined mesen-
221. Bain E, Hugel H, Sharma M. Percutaneous cervical cordotomy for cephalothalamotomy. Zentralbl Neurochir. 1958;18(4):188-201.
the management of pain from cancer: a prospective review of 45 249. Wycis HT, Spiegel EA. Long-range results in the treatment of
cases. J Palliat Med. 2013;16(8):901-907. intractable pain by stereotaxic midbrain surgery. J Neurosurg. 1962;
222. Kanpolat Y, Ozdemir M, Al-Beyati E. CT-guided percutaneous cor- 19:101-107.
dotomy for intractable pain in what is more than a disease: lung 250. Zapletal B. Open mesencephalotomy and thalamotomy for intrac-
malignancies. Turk Neurosurg. 2013;23(1):81-87. table pain. Acta Neurochir (Wien). 1969;(suppl 18):1+.
223. Schvarcz JR. Spinal cord stereotactic techniques re trigeminal 251. Voris HC, Whisler WW. Results of stereotaxic surgery for intrac-
nucleotomy and extralemniscal myelotomy. Appl Neurophysiol. 1978; table pain. Stereotact Funct Neurosurg. 1975;37(1-3):86-96.
41(1-4):99-112. 252. Barbera J, Barcia-Salorio JL, Broseta J. Stereotaxic pontine spino-
224. Broager B. Commissural myelotomy. Surg Neurol. 1974;2(2): thalamic tractotomy. Surg Neurol. 1979;11(2):111-114.
71-74. 253. Bosch DA. Stereotactic rostral mesencephalotomy in cancer pain
225. Sourek K. Commissural myelotomy. J Neurosurg. 1969;31(5): and deafferentation pain. A series of 40 cases with follow-up results
524-527. [see comment]. J Neurosurg. 1991;75(5):747-751.
226. Becker R, Gatscher S, Sure U, et al. The punctate midline myelot- 254. Frank F, Fabrizi AP, Gaist G. Stereotactic mesencephalic tractotomy
omy concept for visceral cancer pain control: case report and review in the treatment of chronic cancer pain. Acta Neurochir (Wien).
of the literature. Acta Neurochir Suppl. 2002;79:77-78. 1989;99(1-2):38-40.
1451.e6 SECTION 6  Pain

255. Hitchcock E, Kim MC, Sotelo M. Further experience in stereotactic 284. Wilkinson HA, Davidson KM, Davidson RI. Bilateral anterior cin-
pontine tractotomy. Appl Neurophysiol. 1985;48(1-6):242-246. gulotomy for chronic noncancer pain [see comment]. Neurosurgery.
256. Nashold BS Jr. Extensive cephalic and oral pain relieved by mid- 1999;45(5):1129-1134, discussion 1134-1126.
brain tractotomy. Stereotact Funct Neurosurg. 1972;34(6):382-388. 285. Yen CP, Kung SS, Su YF, et al. Stereotactic bilateral anterior cingu-
257. Whisler WW, Voris HC. Mesencephalotomy for intractable pain lotomy for intractable pain. J Clin Neurol. 2005;12(8):886-890.
due to malignant disease. Appl Neurophysiol. 1978;41(1-4):52-56. 286. Gutierrez-Lara F. Stereotactic cingulotomy, a rational and effective
258. Jeanmonod D, Maguin M, Morel A. Medial thalamotomy com- approach for causalgia (report 14 cases). Excerpta Med. 1973;293(38):
mentary. In: Burchiel K, ed. Surgical Management of Pain. New York: 233-238.
Thieme Medical Publishers; 2002:802-804. 287. Laitinen L, Vilkki J. Stereotactic ventral anterior cingulotomy
259. Jeanmonod D, Magnin M, Morel A. Thalamus and neurogenic pain: in some psychological disorders. In: Hitchcock E, Laitinen L,
physiological, anatomical and clinical data [erratum appears in Neu- Vaernet K, eds. Psychosurgery. Springfield: Charles C Thomas;
roreport. 1993;4:1066]. Neuroreport. 1993;4(5):475-478. 1972:242-252.
260. Dougherty PM, Lee J, Dimitrou T, et al. Medial thalamotomy. In: 288. Broager B, Olesen K. Psychosurgery in sixty-three cases of open
Burchiel KJ, ed. Surgical Management of Pain. New York: Thieme cingulectomy and fourteen cases of bifrontal prehypothalamic
Medical Publishers; 2002:812-820. cryolesion. In: Hitchcock E, Laitinen L, Vaernet K, eds. Psychosur-
261. Tasker RR. Stereotactic medial thalamotomy for chronic pain: Is it gery. Springfield: Charles C Thomas; 1972:253-257.
an effective procedure? In: Burchiel KJ, ed. Surgical Management of 289. Faillace LA, Allen RP, McQueen JD, et al. Cognitive deficits from
Pain. New York: Thieme Medical Publishers; 2002:812-820. bilateral cingulotomy for intractable pain in man. Dis Nerv Syst.
262. Choi CR, Umbach W. Combined stereotaxic surgery for relief of 1971;32(3):171-175.
intractable pain. Neurochirurgia (Stuttg). 1977;20(3):84-87. 290. Foltz EL, White LE. The role of rostral cingulumotomy in “pain”
263. Hitchcock ER, Teixeira MJ. A comparison of results from center- relief. Int J Neurol. 1968;6(3-4):353-373.
median and basal thalamotomies for pain. Surg Neurol. 1981;15(5): 291. Hurt RW, Ballantine HT Jr. Stereotactic anterior cingulate lesions
341-351. for persistent pain: a report on 68 cases. Clin Neurosurg. 1974;21:
264. Lende RA, Kirsch WM, Druckman R. Relief of facial pain after 334-351.
combined removal of precentral and postcentral cortex. J Neurosurg. 292. Le Beau J. Anterior cingulectomy in man. J Neurosurg. 1954;11(3):
1971;34(4):537-543. 268-276.
265. Niizuma H, Kwak R, Ikeda S, et al. Follow-up results of centrome- 293. Pillay PK, Hassenbusch SJ. Bilateral MRI-guided stereotactic cin-
dian thalamotomy for central pain. Appl Neurophysiol. 1982;45(3): gulotomy for intractable pain. Stereotact Funct Neurosurg.
324-325. 1992;59(1-4):33-38.
266. Richardson DE. Thalamotomy for intractable pain. Confin Neurol. 294. Sharma T. Abolition of opiate hunger in humans following bilateral
1967;29(2):139-145. anterior cingulotomy. Tex Med. 1974;70(10):49-52.
267. Shieff C, Nashold BS Jr. Stereotactic mesencephalic tractotomy for 295. Turnbull IM. Bilateral cingulumotomy combined with thalamot-
thalamic pain. Neurol Res. 1987;9(2):101-104. omy or mesencephalic tractotomy for pain. Surg Gynecol Obstet.
268. Young RF, Vermeulen SS, Grimm P, et al. Gamma Knife thala- 1972;134(6):958-962.
motomy for the treatment of persistent pain. Stereotact Funct Neu- 296. Wilson DH, Chang AE. Bilateral anterior cingulectomy for the
rosurg. 1995;64(suppl 1):172-181. relief of intractable pain. Report of 23 patients. Confin Neurol. 1974;
269. Obrador S, Bravo G. Thalamic lesions for the treatment of facial 36(1):61-68.
neuralgias [abstract]. J Neurol Neurosurg Psychiatry. 1960;23(Sympo- 297. Foltz EL. Current status and use of rostral cingulumotomy. South
sium on Myelopathy):351-352. Med J. 1968;61(9):899-908.
270. Kudo T, Yoshii N, Shimizu S, et al. [Stereotaxic thalamotomy for 298. Mailis A, Furlan A. Sympathectomy for neuropathic pain. Cochrane
pain relief.]. Seishin Igaku Kenkyusho Gyosekishu—Bulletin of the Database Syst Rev. 2003;(2):CD002918.
Seishin-Igaku Institute. 1968;15-30. 299. Happak W, Sator-Katzenschlager S, Kriechbaumer LK. Surgical
271. Jeanmonod D, Werner B, Morel A, et al. Transcranial magnetic treatment of complex regional pain syndrome type II with regional
resonance imaging-guided focused ultrasound: noninvasive central subcutaneous venous sympathectomy. J Trauma Acute Care Surg.
lateral thalamotomy for chronic neuropathic pain. Neurosurg Focus. 2012;72(6):1647-1653.
2012;32(1):E1. 300. Eisenberg E, Carr DB, Chalmers TC. Neurolytic celiac plexus
272. Fairman D. Unilateral thalamic tractotomy for the relief of bilateral block for treatment of cancer pain: a meta-analysis [erratum
pain in malignant tumors. Confin Neurol. 1967;29(2):146-152. appears in Anesth Analg. 1995;81:213]. Anesth Analg. 1995;80(2):
273. Fairman D, Llavallol MA. Thalamic tractotomy for the alleviation 290-295.
of intractable pain in cancer. Cancer. 1973;31(3):700-707. 301. Fujita Y. CT-guided neurolytic splanchnic nerve block with alcohol
274. Leksell L, Meyerson BA, Forster DM. Radiosurgical thalamotomy [see comment]. Pain. 1993;55(3):363-366.
for intractable pain. Confin Neurol. 1972;34(2):264. 302. Gunaratnam NT, Sarma AV, Norton ID, et al. A prospective study
275. Ramamurthi B, Kalyanaraman S. Stereotactic thalamotomy in pain of EUS-guided celiac plexus neurolysis for pancreatic cancer pain.
relief. J R Coll Surg Edinb. 1966;12(1):46-48. Gastrointest Endosc. 2001;54(3):316-324.
276. Richardson DE. Thalamotomy for control of chronic pain. Acta 303. Kang CM, Lee HY, Yang HJ, et al. Bilateral thoracoscopic splanch-
Neurochir (Wien). 1974;(suppl 21):77-88. nicectomy with sympathectomy for managing abdominal pain in
277. Sano K, Yoshioka M, Ogashiwa M, et al. Thalamolaminotomy. A cancer patients. Am J Surg. 2007;194(1):23-29.
new operation for relief of intractable pain. Confin Neurol. 304. Lillemoe KD, Cameron JL, Kaufman HS, et al. Chemical splanch-
1966;27(1):63-66. nicectomy in patients with unresectable pancreatic cancer. A
278. Shimizu S, Aikawa S, Nishioka S, et al. Stereotaxic thalamotomy for prospective randomized trial. Ann Surg. 1993;217(5):447-455, dis-
pain relief. Tohoku J Exp Med. 1968;96(3):219-234. cussion 456-447.
279. Steiner L, Forster D, Leksell L, et al. Gammathalamotomy in 305. Lin CC, Mo LR, Lin YW, et al. Bilateral thoracoscopic lower
intractable pain. Acta Neurochir (Wien). 1980;52(3-4):173-184. sympathetic-splanchnicectomy for upper abdominal cancer pain.
280. Sugita K, Mutsuga N, Takaoka Y, et al. Results of stereotaxic thala- Eur J Surg Suppl. 1994(572):59-62.
motomy for pain. Confin Neurol. 1972;34(2):265-274. 306. Erdek MA, Halpert DE, Gonzalez Fernandez M, et al. Assessment
281. Uematsu S, Konigsmark B, Walker AE. Thalamotomy for allevia- of celiac plexus block and neurolysis outcomes and technique in the
tion of intractable pain. Confin Neurol. 1974;36(2):88-96. management of refractory visceral cancer pain. Pain Med. 2010;11(1):
282. Whittle IR, Jenkinson JL. CT-guided stereotactic anteromedial 92-100.
pulvinotomy and centromedian-parafascicular thalamotomy for 307. Deleted in review.
intractable malignant pain. Br J Neurosurg. 1995;9(2):195-200. 308. Freeman TB, Vawter DE, Leaverton PE, et al. Use of placebo
283. Ballantine HT, Cosgrove GR, Giriunsa IE. Surgical treatment of surgery in controlled trials of a cellular-based therapy for Parkin-
intractable psychiatric illness and chronic pain by stereotactic cin- son’s disease [see comment]. N Engl J Med. 1999;341(13):988-992.
gulotomy. In: Schmidek HH, Sweet WH, eds. Operative Neurosurgi- 309. Freed CR, Greene PE, Breeze RE, et al. Transplantation of embry-
cal Techniques: Indications, Methods, and Results. Philadelphia: WB onic dopamine neurons for severe Parkinson’s disease [see comment].
Saunders; 1995:1423-1430. N Engl J Med. 2001;344(10):710-719.
CHAPTER 179  Evidence Base for Destructive Procedures 1451.e7

310. Frank S, Kieburtz K, Holloway R, et al. What is the risk of sham 315. Young RF, Jacques DS, Rand RW, et al. Technique of stereotactic
surgery in Parkinson disease clinical trials? A review of published medial thalamotomy with the Leksell Gamma Knife for treatment 179
reports [see comment]. Neurology. 2005;65(7):1101-1103. of chronic pain. Neurol Res. 1995;17(1):59-65.
311. Concato J, Shah N, Horwitz RI. Randomized, controlled trials, 316. Wong ET, Gunes S, Gaughan E, et al. Palliation of intractable
observational studies, and the hierarchy of research designs [see cancer pain by MRI-guided cingulotomy. Clin J Pain. 1997;13(3):
comment]. N Engl J Med. 2000;342(25):1887-1892. 260-263.
312. Benson K, Hartz AJ. A comparison of observational studies and 317. Wyse JM, Carone M, Paquin SC, et al. Randomized, double-blind,
randomized, controlled trials [see comment]. N Engl J Med. 2000; controlled trial of early endoscopic ultrasound-guided celiac plexus
342(25):1878-1886. neurolysis to prevent pain progression in patients with newly diag-
313. Gybels J, Tasker R. Supratentorial neurosurgery for the treatment nosed, painful, inoperable pancreatic cancer. J Clin Oncol. 2011;
of pain. In: McMahon S, Koltzenberg M, eds. Wall and Melzack’s 29(26):3541-3546.
Textbook of Pain. 5th ed. London: Elsevier; 2006:553-561.
314. Jones B, Finlay I, Ray A, et al. Is there still a role for open cor-
dotomy in cancer pain management? J Pain Symptom Manage.
2003;25(2):179-184.
eTABLE 179-3A  Dorsal Root Entry Zone Lesioning for Pain—cont’d
Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Tomas and 2005 Case series BPA 2; group 1 (localization of DREZ Mean 44.1 mo All patients: 62% had good results, 38% fair, and 0% poor; group
Haninec125 by evoked potentials was not 1: 33% had good results, 67% fair, and 0% poor; group 2: 83%
performed), 9 patients; group had good results, 17% fair, and 0% poor
2 (direct spinal cord bipolar
stimulation and registration
with the goal to localize
DREZ), 12 patients
Young126 1990 Case series Deafferentation pain 78 Up to 5 yr 61.5% had satisfactory results
1448.e2 SECTION 6  Pain

(mixed)
Kanpolat et al222 2013 Case report Complex regional pain 2 1 case followed up The first patient experienced postoperative ataxia, with recovery
syndrome for 60 mo, the after 2 mo, and was followed up for 60 mo without any
other 33 mo recurrence or complications; the second patient experienced
ataxia that improved after 3 mo, was pain free for 6 mo then
had a recurrence, received psychiatric consultation reassurance
and antidepressants, and then was followed up for 33 mo with
partial satisfactory pain relief
Aichaoui et al135 2011 Prospective Pain following BPA 29 Mean 60 mo (range On discharge: 23 patients showed excellent global pain relief, and
case series 10-122) for 26 the other 6 showed good pain relief with the residual pain
patients controlled by class 1 analgesics and/or anticonvulsants or
antidepressants; at the first outpatient visit 3 mo later: 21 had
excellent pain relief, 7 good, and 1 poor; at last evaluation: 14
had excellent pain relief, 6 good, and 6 poor
Ruiz-Juretschke 2011 Case series Neuropathic 18 Mean 28 mo (range Mean level of pain decreased according to VAS score, which fell
et al140 deafferentiation pain 6-108) from 8.6 significantly to 2.9; at last f/u the mean VAS score was
syndromes 4.7
Chun et al137 2011 Retrospective Refractory neuropathic 38 Mean 42 mo (range Good pain relief was achieved in 30 patients (79%), fair in 4
case series pain following SCI 19-84) (10.5%), and poor in 4 (10.5%)
Tomycz and 2011 Case report BPA and phantom 1 26 yr The patient had significant relief of phantom limb pain and no
Moossy142 limb pain recurrence for 26 yr
Ali et al136 2011 Comparative Intractable BPA pain 15 patients, of whom 7 Of the EMCS patients EMCS: the percentage of patients with favorable outcomes (>50%
study underwent DREZotomy 7 were followed for reduction in VAS score) was higher for those with continuous
alone, 4 underwent EMCS average of 47 mo. pain than for those with paroxysmal pain, both during the trial
alone, and 4 underwent both Of the DREZ patients and with long-term stimulation; during the trial, 4 of 8 patients
procedures 10 were followed (50%) with continuous pain had favorable outcomes, compared
for an average of with 2 of 6 patients (33%) with paroxysmal pain; at the latest f/u,
31 mo (range 3 of 7 patients (42%) with continuous pain had favorable
12-61) outcomes, compared with 0 of the 5 patients (0%) with
paroxysmal pain, and the mean percentage of VAS reduction
was greater in those with continuous pain than in those with
paroxysmal pain (28% vs. 7%)
DREZotomy: in immediate postoperative period, 10 of 11 patients
(91%) with paroxysmal pain had favorable outcomes compared
with 8 of 11 patients (72%) with continuous pain; at the latest
follow-up, 7 of 10 patients (70%) with paroxysmal pain had
favorable outcomes compared with 2 of 10 patients (20%) with
continuous pain, and the mean percentage of VAS reduction
was greater in those with paroxysmal pain than in those with
continuous pain (63% vs. 26%)
Tao et al144 2014 Retrospective Intractable chronic 35 Mean 36.1 yr in 33 At 36.1 mo follow up, excellent pain relief was reported in 24
case series neuropathic pain patients (range patients (69%), good results in 6 patients (17%), and poor
due to SCI and/or 1-6 yr) results in 5 patients (14%)
cauda equina injury
Dong et al138 2012 Case series Pain after BPA 7 12 mo All patients showed good pain relief postoperatively as follows:
mean ± SD VAS pain scores were 8.9 ± 0.9 preoperatively and
0.9 ± 0.7 at 12 mo postoperatively
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review. J Neurosurg. 2008;109:389-404.
BPA, brachial plexus avulsion; DREZ, dorsal root entry zone; EMCS, electric motor cortex stimulation; f/u, follow-up; SCI, spinal cord injury; SD, standard deviation; VAS, Visual Analog Scale.
CHAPTER 179  Evidence Base for Destructive Procedures 1448.e3

eTABLE 179-3B  Dorsal Root Entry Zone Lesioning for Facial Pain
179
Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Sandwell and 2013 Case report Anesthesia 1 1 yr Immediate postoperatively the patient
El-Naggar141 dolorosa was free of facial pain, and at 7th
postoperative week he stopped
medication completely
Park et al143 2010 Retrospective Refractory 39 patients; 16 Mean 26.1 mo 15 (93.8%) RGZ patients and 20 (87.0%)
case series trigeminal underwent GKRS (range 12-53 mo) DREZ patients experienced treatment
neuralgia to the RGZ, and success (BNI score I-IIIb); 7 (43.8%)
23 underwent RGZ patients and 4 (17.4%) DREZ
GKRS to the DREZ patients reported complete pain relief
without medications (BNI score I)
BNI (score), Barrow Neurological Institute Pain Intensity (score); DREZ, dorsal root entry zone; GKRS, gamma knife radiosurgery; RGZ, retrogasserian
zone.

eTABLE 179-3C  Dorsal Root Entry Zone Lesioning for Cancer Pain*
Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Sindou148 1995 Case series Cancer pain 81 Long ?
Kanpolat et al146 2008 Case series cancer pain 7 Long Significant pain relief
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review.
J Neurosurg. 2008;109:389-404.
*No publications after 2008.

eTABLE 179-3D  Caudalis Dorsal Root Entry Zone Lesioning for Cancer Pain*
Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Bullard and 1997 Case series Cancer pain among 1/25 Unclear in the cancer No definitive f/u time, no outcome for the
Nashold145 noncancer facial pain patient cancer patient; yet initially 24/25 had
pain pain relief, but only 67% did so at 1 yr
Kanpolat et al146 2008 Case series Cancer pain 3 Long Significant pain relief
Rossitch et al147 1989 Case series Cancer pain 5 Mean 14.4 mo 3 patients had significant improvement
From Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review. J Neurosurg.
2008;109:389-404. Published online with permission from the American Association of Neurological Surgeons Journal of Neurosurgery.
*No publications after 2008.

eTABLE 179-4A  Tractotomy for Pain


Study
Authors Year Design Diagnosis Patient No. Follow-up Outcome
Brodal149 1947 Case series Facial pain 4 Unclear Good pain relief
Crue et al152 1972 Case series Trigeminal neuralgia 4 with pain of Unclear Unclear
nonmalignant
causes, 12 total
Hitchcock and 1972 Case series Postherpetic facial pain 3 >1 yr Good
Schvarcz156
Hosobuchi and 1971 Case series Intractable facial pain of different 2 with pain of 4-12 mo Good
Rutkin153 etiologies nonmalignant
causes, 6 total
Kanpolat et al154 1998 Case series Idiopathic vagoglossopharyngeal 9 2-24 mo Excellent in 6, good in 3
neuralgia and geniculate neuralgia
Kunc155 1965 Case series Cranial nerve IX essential neuralgia 15 Unclear Reported case histories
of last 6 patients with
“successful” operations
Schvarcz150 1975 Case series Mixed 17 with pain of 4-18 mo Excellent in 16
nonmalignant
causes, 25 total
Thompson et al157 2013 Case report Postherpetic neuralgia and 2 3 and 6 mo Both patients described
postoperative hemicranial excellent pain relief
neuralgia postoperatively
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review.
J Neurosurg. 2008;109:389-404.
1448.e4 SECTION 6  Pain

eTABLE 179-4B  Trigeminal Tractotomy for Cancer Pain*


Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
Raslan162 2008 Prospective Cancer pain 10 6 mo Significant improvement in Visual Analog Scale
open-label score, Karnofsky performance score,
activities of daily living, sleeping hours
Kanpolat et al160 1995 Case series Cancer pain 5 6 mo Satisfactory pain relief
Plangger et al161 1987 Case series Cancer pain 6 ? 50% had complete pain relief and 50% had
excellent pain control
Schvarcz150 1975 Case series Cancer pain 8 of 25 treated for 4-18 mo Excellent in all 8 cases
neoplasms
158
Fox 1971 Case series Cancer pain 8 of 12 treated for pain Unclear 6 had excellent pain relief, 1 had 50% relief,
and 1 had no relief
Hitchcock159 1970 Case series Cancer pain 5 of 7 treated for pain 6-8 mo 3 had complete pain relief, 2 partial relief
Crue et al152 1972 Case series Cancer pain 8 of 12 treated for pain Till death 6 of the 8 had excellent or good pain relief
From Raslan AM, Cetas JS, McCartney S, et al. Destructive procedures for control of cancer pain: the case for cordotomy. J Neurosurg. 2011;114:
155-170. Published online with permission from the American Association of Neurological Surgeons Journal of Neurosurgery.
*No publications after 2008.

eTABLE 179-5A  Cordotomy for Pain


Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
165
Amano et al 1976 Case series Intractable pain of 4 with pain of nonmalignant 2 wk Reduced pain without medication
mixed etiology causes, 30 total in 2 patients, persistent pain
with medication in 2 patients
Cowie and 1982 Case series Neuropathic pain of 13 with pain of nonmalignant >3 yr 85% relief at 6 mo, 35% at 1 yr,
Hitchcock166 various etiologies causes, 56 total 20% at 3 yr
Diemath et al167 1961 Case series Neuropathic pain of 33 with pain of nonmalignant >2 yr 67% complete relief immediately
various etiologies causes, 121 total postoperatively, 58% complete
relief at 2 yr
Frankel and 1961 Case series Neuropathic pain of 16 with pain of nonmalignant Up to 31% (non)complete relief
Prokop168 various etiologies causes, 75 total 10 yr
French 1974 Case series Neuropathic pain of 22 with pain of nonmalignant >6 mo Not stated for patients with pain of
various etiologies causes, 185 total nonmalignant causes
Grant and 1957 Case series Neuropathic pain of 72 with pain of nonmalignant >1 mo 32/72 complete relief, 52/72 at
Wood170 various etiologies causes, 312 total least partial relief
Porter et al171 1966 Case series Cauda equina injury 34 >18 mo 61.7% good or fair results
Probst172 1990 Case series Epidural/intradural 22 6-132 mo 65% good long-term results
fibrosis (complete and partial pain relief)
Raskind173 1969 Case series Neuropathic pain of 30 with pain of nonmalignant Not stated 60% good, not requiring narcotics
various etiologies causes, 237 total
Rosomoff174 1969 Case series Pain of various 29 with pain of nonmalignant ≥2 yr 10/17 or 60% of survivors at 2 yr
etiologies causes, 100 total with satisfactory results; unclear
how many survivors had pain of
nonmalignant causes
Tasker et al132 1992 Case series Pain due to spinal 34 underwent cordotomy, Not stated 54% for cordotomy and 60% for
cord injury of and 12 cordectomy cordectomy in patients whose
various etiologies pain had been intermittent or
evoked
Lewin et al164 2012 Case report Low back pain, 1 11 mo Patient was free of pain from
right hip and immediately postoperatively until
buttock pain his death 11 mo later
Collins et al163 2012 Case report Pain in the left leg, 1 41 yr Patient remained completely free
calf, and foot of pain for 35 yr then started to
following pelvic experience slowly progressive
bullet injury and descending recurrence of
aching pain in the left subcostal
region
Modified from Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment of nonmalignant pain: a structured literature review.
J Neurosurg. 2008;109:389-404.
eTABLE 179-5B  Cordotomy for Cancer Pain—cont’d
Author(s) Year Study Design Diagnosis Patient No. Follow-up Outcome
189
Ischia et al 1984 Retrospective cohort Cancer pain 36 Till death Bilateral cases only; 47% had complete pain relief
of prospectively and 12.5% partial pain relief; patients might be
collected data included in other Ischia studies cited in table
Cowie and 1982 Case series Cancer pain 33 Up to 1 yr 95%, 73%, and 55% of patients had pain relief
Hitchcock166 immediately postoperatively, at 6 mo, and at 1 yr,
1448.e6 SECTION 6  Pain

respectively
Meglio and 1981 Case series Cancer pain 52 15 wk 73% and 63% of patients had excellent results after
Cioni199 1 wk and 15 wk, respectively
Lipton S197 1978 Case series Cancer pain 650 Till death 95% of patients had initial pain relief, but only 75%
near death; 6.2% mortality
207
Rothbard et al 1972 Case series Cancer pain (gynecologic) 10 Up to 3 yr Pain-free interval 6-29 mo
Batzdorf and 1970 Case series Cancer pain 41 with cancer pain, 47 Up to 12 mo 27 had excellent or good pain relief, and 11 fair pain
Weingarten178 total relief
O’Connell202 1969 Case series Cancer pain (rectal) 56 6 wk-2 yr 63% had complete pain relief till death
French169 1974 Case series Cancer pain 177 with cancer pain, 200 Up to 1 yr 90.6% had no pain or good pain relief for up to
total, underwent open >1 yr, open surgical cordotomy
surgical cordotomy
Rosomoff174 1969 Case series Cancer pain 71 with cancer pain, 100 Till death Initial 93% pain relief that faded to about 60%
total, underwent based on survival at 2 yr; patients were followed
bilateral cordotomy up till death
Lin et al196 1966 Case series Cancer pain 38 2 wk-7 mo 74%-86% had adequate results (no pain or good
pain relief), depending on whether approach was
anterior or posterior
Mullan et al200 1965 Case series Cancer pain 47 Short 36 had good outcome; 1 death
Esposito et al79 1985 Case series Cancer pain 8 6 mo Cordotomy worked for 2-3 mo only
Grote et al186 1978 Case series Cancer pain 138 Unclear Initial relief in 114 (106 complete, 8 partial),
dysesthesia in 9, anesthesia dolorosa in 1
Tasker212 1976 Case series Mostly cancer pain 264 mixed Unclear Initial results, 96% and 83% pain relief with unilateral
and bilateral, respectively; 89% and 66%,
respectively, at the time of latest follow-up; 3%
permanent complications
Crue and 1974 Case series Mixed 29 with cancer pain, 48 Short 31% pain relief even in the short term
Felsoory179 total
Fox183 1973 Case series Mixed 14 with cancer pain, 18 Unclear 11 had adequate pain relief
total
Tasker and 1973 Case series Mixed, mostly cancer pain 78 Till death; survival 84% had pain relief till death
Organ213 generally 3-6 mo
Smith210 1972 Case series Mixed, mostly cancer pain 19 Around 6 mo 100% of patients with cancer pain had complete or
satisfactory pain relief
Foer182 1971 Case series Cancer pain 21 with cancer pain, 30 Unclear 90% had partial to complete pain relief; 1 death
total
Salmon208 1969 Case series Mixed 34 Unclear 25 of 34 had good pain relief, 3 had minor transient
ataxia; no deaths
Fox184 1969 Case series Mixed 50 Not reported Not reported
Uihlein et al214 1969 Case series Cancer pain 45 with cancer pain, 50 Unclear 80% complete pain relief, 52% satisfactory pain
total relief, and 28% persistent pain relief
Acosta and 1969 Case series Cancer pain 12 with cancer pain,15 3 mo-2 yr Adequate pain relief in all patients; 2 required repeat
Grossman176 total procedure
Kelly and 1966 Case series Cancer pain 14 with cancer pain, 17 1-10 mo 13 had pain relief
Alexander193 total
180 Dorsal Rhizotomy and Dorsal Root Ganglionectomy
Feridun Acar, Selçuk Göçmen, and Athanasios K. Zisakis

The treatment of pain remains a major interest of neurosurgery.


Since the beginning of 20th century, destructive procedures tar-
SURGERY
geting different locations on the pain pathway have been used
widely.1,2 As a result of new neuromodulation techniques and
Cervical Ganglionectomy and Rhizotomy
medications, there is a tendency to abandon the use of destructive For C2 ganglionectomy, the C2 ganglion is exposed through a
procedures such as dorsal rhizotomy and dorsal root ganglionec- midline incision between the inion and the transverse process of
tomy despite their value and efficacy. However, in some cases, the C3 and dissection of the paravertebral musculature to expose the
modern modalities lack the sufficiency to control pain, and more- C1-2 interspace.9 The venous complex is cauterized using bipolar
over these modalities are expensive and need maintenance. With electrocautery. The root exiting from the C2 space is identified
this perspective, a pain surgeon should know the indications and followed in both directions, and the ganglion is identified.
for destructive procedures and have the skills to perform these The nerve should be cut just proximal to the ganglion, allowing
procedures.3,4 it to retract slightly, and the proximal stump is cauterized to
prevent bleeding. The nerve is then followed distally, and the end
INDICATIONS FOR DORSAL RHIZOTOMY is cut distal to the ganglion and again cauterized while maintain-
ing hemostasis (Fig. 180-2). The C3 ganglionectomy is per-
AND GANGLIONECTOMY formed in a similar fashion with the addition of a foraminotomy
The indications for dorsal rhizotomy and ganglionectomy can to expose the C3 nerve root and ganglion (Fig. 180-3). When the
be reviewed under two major pain groups: cancer pain and ganglion is identified, it is cut and removed as in C2 surgery.
noncancer pain. The determination of level can vary from a
single root or ganglion to multilevel surgery. The level of surgery
also depends on intradural root anastomosis, rich anastomoses
Thoracic Ganglionectomy and Rhizotomy
of the sympathetic system, and ventral root sensory fibers The dorsal rhizotomy is performed similar to cervical rhizotomy.
(Fig. 180-1). The feeding segmental arteries that can enter along the posterior
Local preoperative blocks for testing can help to determine roots must be protected.10 Dorsal rhizotomy is performed
the levels of surgery. However, diagnostic blocks can also be through an intradural or extradural approach. A multilevel lami-
misleading because of inappropriate application or a placebo nectomy is a standard for intradural rhizotomy. In general, the
effect. Placebo control is important, and the blocks can be roots of at least two segments should be destroyed to accom-
repeated. The determination, however, should always be a clinical modate overlap innervations.11 In this approach, the correct
decision, and the outcome of diagnostic blocks cannot be used as sensory roots are identified by opening dura at their respective
a sole and major factor on the way to surgery. intervertebral foramina. They then are cut after coagulating. The
procedure for extradural rhizotomy is comparable to that of
extradural ganglionectomy without resection of ganglion.
Cancer Pain A posterior paraspinal approach is used for a radiofrequency
In patients with malignancies involving the brachial plexus, such rhizotomy in the thoracic region. The theory behind radiofre-
as invasive breast carcinoma, multilevel cervical dorsal rhizotomy quency lesions of the dorsal roots and dorsal root ganglia was
to denervate a functionally impaired limb can be successful in developed by Letcher and Goldring,12 who noted that the action
controlling pain.5 In patients with an intact upper extremity potentials of nociceptive fibers (A delta and C fibers) are blocked
motor function, this procedure can result in severe function loss at lower temperatures than those of the larger tactile fibers (A
of the involved limb. alpha and A-beta fibers). In the thoracic region, a posterior
In patients with malignancies of the thorax and chest wall, approach is performed percutaneously, and intravenous sedation
thoracic ganglionectomy with rhizotomy can be a good surgical and local anesthesia are given to the patient. With fluoroscopic
option.5 There is a segmental overlap of dermatomes in the tho- guidance, a 2-mm uninsulated-tip electrode is placed within the
racic area, which leads to the need for multilevel surgeries in neural foramen. Proper placement is confirmed, and sensory
these patients. A three- to five-level ganglionectomy with or stimulation is then performed at less than 1 V, with 50-Hz stimu-
without rhizotomy should be considered. lation to ascertain paresthesia in the region of patient’s pain.
The most important disadvantage of this surgery is the Motor stimulation is then tested at 2 Hz to elicit contraction in
destruction of S2 and S3 sensory innervation, which will result the paraspinal muscles. A test lesion is made at 42 degrees for 15
in neurogenic bladder, neurogenic bowel, and impotence. seconds. A permanent radiofrequency lesioning is performed at
65 to 90 degrees for 60 to 90 seconds.5
Noncancer Pain
The most common indication for ganglionectomy and rhizotomy
Lumbar Ganglionectomy and Rhizotomy
in noncancer pain is the treatment of occipital neuralgia.6-8 For L5 ganglionectomy, the L5 ganglion is exposed within the
Occipital neuralgia can be successfully treated with C2 and C3 intervertebral foramen. However, S1 ganglion usually lies within
ganglionectomies.6-8 C2 and C3 ganglionectomies can also be spinal canal.13 The resection of the lateral margin of the facet
considered for medically resistant cervicogenic headache. joint and a little bit of the inferolateral margin of the pars
1452
CHAPTER 180  Dorsal Rhizotomy and Dorsal Root Ganglionectomy 1453

180

Figure 180-1. The anatomic organization of the spinal nerve. Each spinal nerve is attached to the spinal
medulla by two roots: posterior (afferent) and anterior (efferent). The afferent fibers carry the sensory input and
efferent fibers innervate the skeletal muscles. (From Acar F, Göçmen S. Dorsal rhizotomy and dorsal root
ganglionectomy. In: Burchiel KJ, ed. Surgical Management of Pain. 2nd ed. New York: Thieme; 2015.)

A B C
Figure 180-2. Illustration of C2 ganglion. A-C, No foraminotomy is needed to expose ganglion. Note that
the ganglion is covered by venous plexus. (From Acar F, Göçmen S. Dorsal rhizotomy and dorsal root
ganglionectomy. In: Burchiel KJ, ed. Surgical Management of Pain. 2nd ed. New York: Thieme; 2015.)

interarticularis is required. For S1 ganglionectomy, the standard


L5-S1 hemilaminectomy and foraminotomy are performed. After
the dural sheath of the S1 root is identified and longitudinally
incised, the dorsal root is sectioned proximal and distal to the
ganglion. The ganglion is elevated and bluntly dissected off the
fibrous septum, which separates it from the ventral motor root.
Finally, the ganglion is cut distally after coagulating and proxi-
mally ligaturing its distal connection to the spinal nerve.
B The rhizotomy is performed in a similar manner to cervical
rhizotomy. Crue and Todd described the technique of extradural
rhizotomy in the sacral region.14 In brief, the S1 and S2 roots are
exposed after a bilateral upper sacral laminectomy. The thecal sac
is then ligated with 0-silk suture. One tie is passed around the
thecal sac just caudal to the S1 nerve root axilla, and a second tie
is passed around the thecal sac just rostral to the S2 nerve root
axilla. The thecal sac is sharply cut with its contents between the
two ties, preserving the S1 nerve roots.
A C
Figure 180-3. Illustration of C3 ganglion. A-C, Note that a CONCLUSION
foraminotomy is needed to expose the ganglion. This ganglion is also
Patients with intractable pain are difficult to treat. Destructive
covered by venous plexus. Careful coagulation is necessary before
surgeries such as ganglionectomy and rhizotomy still have an
the ganglion excision. (From Acar F, Göçmen S. Dorsal rhizotomy and
important role in the treatment of some pain syndromes men-
dorsal root ganglionectomy. In: Burchiel KJ, ed. Surgical Management
tioned in this chapter. Destructive procedures for the treatment
of Pain. 2nd ed. New York: Thieme; 2015.)
of pain have played an important role in the history of neurosur-
gery, but their efficacy has not been well established based on
contemporary standards.
1454 SECTION 6  Pain

How should future studies be designed and conducted if they Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treatment
have to determine accurately and reliably the effectiveness of of nonmalignant pain: a structured literature review. J Neurosurg.
these surgical procedures in the treatment of pain? The answer 2008;109:389-404.
is easy: double-blinded random controlled trials in patients with Gallagher J, Petriccione di Vadi PL, Wedley JR, et al. Radiofrequency
facet joint denervation in the treatment of low back pain: a prospective
standardized and well-documented diagnoses without ethical, controlled double-blind study to assess its efficacy. Pain Clin. 1994;7:
financial, and practical considerations. However, it is almost 193-198.
impossible to do such trials on destructive procedures for pain Oh WS, Shim JC. A randomized controlled trial of radiofrequency dener-
because many of the earlier studies have failed to distinguish vation of the ramus communicans nerve for chronic discogenic low
between different pain states and a single-center rarely has suf- back pain. Clin J Pain. 2004;20:55-60.
ficient volume of a particular procedure to power a valid outcome Oh WS, Shim JC. A randomized controlled trial of radiofrequency dener-
study adequately. The last one is the accurate evaluation of surgi- vation of the ramus communicans nerve for chronic discogenic low
cal pain relief procedures. Well-designed studies with appropriate back pain. Clin J Pain. 2004;20:55-60.
control groups will answer fundamental questions in the surgical Taha JM. Dorsal root ganglionectomy and dorsal rhizotomy. In: Burchiel
KJ, ed. Surgical Management of Pain. New York: Thieme; 2002:
treatment of pain. 677-685.
Young RF. Dorsal rhizotomy and dorsal root ganglionectomy. In: Youmans
SUGGESTED READINGS JR, ed. Neurological Surgery, Vol. 6. 3rd ed. Philadelphia: WB Saunders;
Arguelles J, Burchiel K. Ablative neurosurgical procedures for the treat- 1990:4026-4035.
ment of pain: peripheral. In: Tindall G, ed. The Practice of Neurosurgery.
Baltimore: Williams & Wilkins; 1996:3153-3175. See a full reference list on ExpertConsult.com
CHAPTER 180  Dorsal Rhizotomy and Dorsal Root Ganglionectomy 1454.e1

REFERENCES 8. Arguelles J, Burchiel K. Ablative neurosurgical procedures for the


1. Bell C. Idea of a New Anatomy of the Brain. London: Strahan & treatment of pain: peripheral. In: Tindall G, ed. The Practice of Neu- 180
Preston; 1811:17-19. rosurgery. Baltimore: Williams & Wilkins; 1996:3153-3175.
2. Abbe R. Intradural section of the spinal nerves for neuralgia. Boston 9. Acar F, Miller J, Golshani KJ, et al. Pain relief after cervical ganglio-
Med Surg J. 1896;135:329-335. nectomy (C2 and C3) for the treatment of medically intractable
3. Harris AB. Dorsal rhizotomy for pain relief. In: Tindall G, ed. occipital neuralgia. Stereotact Funct Neurosurg. 2008;86:106-112.
The Practice of Neurosurgery. Baltimore: Williams and Wilkins; 1996: 10. Loeser JD, Sweet WH, Tew JM, et al. Neurosurgical operations
4029-4032. involving peripheral nerves. In: Bonica JJ, ed. The Management of
4. White JC, Kjellberg RN. Posterior spinal rhizotomy: a substitute for Pain. Philadelphia: Lea & Febiger; 1990:2044-2066.
cordotomy in the relief of localized pain in patients with normal life 11. Loeser JD. Dorsal rhizotomy for the relief of chronic pain. J Neuro-
expectancy. Neurochirurgia (Stuttg). 1973;16:141-170. surg. 1972;36:745-750.
5. Taha JM. Dorsal root ganglionectomy and dorsal rhizotomy. In: 12. Letcher FS, Goldring S. The effect of radiofrequency current and
Burchiel KJ, ed. Surgical Management of Pain. New York: Thieme; heat on peripheral nerve action potential in the cat. J Neurosurg.
2002:677-685. 1968;29:42-47.
6. Dubuisson D. Treatment of occipital neuralgia by partial posterior 13. Hasue M, Kunogi J, Konno S, et al. Classification by position
rhizotomy at C1-3. J Neurosurg. 1995;82:581-586. of dorsal root ganglia in the lumbosacral region. Spine. 1989;14:
7. Stechison MT, Mullin BB. Surgical treatment of greater occipital 1261-1264.
neuralgia: an appraisal of strategies. Acta Neurochir (Wien). 1994;131: 14. Crue BL, Todd EM. A simplified technique of sacral rhizotomy for
236-240. pelvic pain. J Neurosurg. 1964;21:835-837.
181 Diagnosis and Management of Painful Neuromas
181

Brett E. Youngerman, Taylor B. Nelp, Deepa Danan, and Christopher J. Winfree

When a nerve is acutely injured, it may be sensitive to touch or


PATHOPHYSIOLOGY pressure immediately after injury. Sometimes this sensitivity per-
The capacity of peripheral nerves to regenerate following injury sists until a neuroma forms. Sometimes nerves are not particu-
enables functional recovery in many cases. Unfortunately, this larly painful in the early period after injury but become sensitive
regenerative capability, when it goes awry, can produce a painful later when the neuroma forms.
scar at the site of injury. Neuromas can affect any nerve: large, Traumatic neuromas are firm, slow-growing, and sometimes
small, or microscopic. The key process that contributes to palpable nodules that can be associated with pain and paresthesias
neuroma formation is the regrowth of injured axons into impen- in the affected area. The hallmark finding for a painful neuroma
etrable scar. A traumatic neuroma is a dense, minimally vascular is an exquisitely sensitive, focal area along the course of a previ-
fibrous mass containing many small nerve fibers originating from ously injured nerve. Pain is evoked by palpation of the region and
an otherwise functional nerve.1 The fibrous connective tissue that may remain local or radiate along the course of the peripheral
comprises the outer layer of the neuroma is continuous with the nerve sensory distribution, a positive Tinel’s sign.7 Patients can
perineurium of the involved nerve.2 also have tenderness and hypersensitivity to normal tactile
There are two categories of neuromas: terminal neuromas and stimuli2 and temperature sensitivity, more commonly cold intol-
spindle neuromas. Terminal neuromas typically form after neu- erance, in the surrounding regions.10 If the injured nerve is non-
rotmesis, or complete severing of the nerve, such as occurs in functional, there will often be loss of sensation in the distribution
laceration injuries.3 In neurotmesis, the severed ends often retract of that nerve.
apart. While the distal axons undergo wallerian degeneration and
the nerve withers into scar, the proximal end may form a bulbous
neuroma as axons regenerate haphazardly without finding the
DIAGNOSIS
fibronectin and laminin in the basal lamina of the distal segment The diagnosis of a neuroma can be made clinically: pain in the
epineurium (Fig. 181-1). Because the cut ends are physically region of a scar, altered sensation in the distribution of the
separate, without repair or conduit, the probability of meaningful affected nerve, and a Tinel’s sign. Confirmatory testing may
recovery of axons across the injured segment is vanishingly low.4 include injection of local anesthetic into the vicinity of the
Spindle neuromas, or neuromas-in-continuity, typically form neuroma11 or more proximally along the course of the affected
after axonotmesis, injury of axons with intact epineurium.3 Axo- nerve.12 The presence of temporary pain relief helps make the
notmesis may follow injuries related to compression, stretch, or diagnosis of a painful neuroma and additionally may help
gunshot wounds, which most commonly cause injury through to identify the exact nerve involved. This may be helpful if mul-
shock waves. A fusiform neuroma may form along the injured tiple candidate nerves are in the vicinity, such as in the inguinal
segment as axons regenerate and local scar tissue forms (Fig. region. Some practitioners use control injections of saline or local
181-2). Because the epineurium is intact, meaningful recovery of anesthetics of differing durations, one long-acting and one short-
axons across the injured segment often occurs, leading to a regen- acting, in an effort to increase reliability of this diagnostic modal-
erating neuroma-in-continuity.5 The presence of scar, however, ity. Such a strategy may help reduce unwanted placebo effects,
may prevent recovery in some cases, leading to a nonconducting which can complicate the treatment of any pain disorder.
neuroma-in-continuity. These forms are distinguished most reli- Imaging may be used to identify larger neuromas. Magnetic
ably by using intraoperative nerve action potential recordings. resonance imaging and magnetic resonance neurography may
Recovering nerve action potentials are seen with a conducting delineate the neuroma quite clearly in some cases but are perhaps
neuroma-in-continuity, but not with a nonconducting neuroma- most useful for excluding other pathology.13 Ultrasonography is
in-continuity (Fig. 181-3). also used routinely to diagnosis and localize neuromas.14,15 It can
Pain in neuromas relates to compression, traction, and isch- be used in in real time to guide anesthetic injections as well. A
emia of nerve fibers by scar tissue. Spontaneous firing of nocicep- method for electrophysiologic testing has also been described as
tors sending impulses centrally may lead to neuropathic pain.6 an alternative to imaging in patients with Morton’s neuroma.16
Additionally, nerve fibers in close proximity to each other may Perhaps the most reliable way to identify a neuroma is with
stimulate impulse transmission in the absence of a synapse by surgical exploration in the awake patient. After external neuroly-
ephatic transmission.7 Further stimulation may occur through the sis of the neuroma, when palpation of the neuroma reproduces
abnormal release of chemical pain mediators.8 More recent find- the patient’s exact pain, little diagnostic uncertainty remains. This
ings suggest that upregulation of ion channels (with ectopic foci), strategy is essentially identical, in theory, to the conscious pain-
nerve growth factor, and receptors (TRPA1, CGRP, alpha1C) con- mapping procedures used by gynecologists during laparoscopic
tribute to neuropathic pain in neuromas.9 procedures used to identify pelvic pain generators.17

CLINICAL PRESENTATION TREATMENT


Neuromas form after nerve trauma. Often the patient is able to After a painful neuroma is diagnosed, a comprehensive, pain
give a clear history of the traumatic event. In many cases, the management−oriented approach may be helpful. Initial efforts
nerve injury is obvious. Sometimes neuromas form unexpectedly may involve over-the-counter pain medications and protecting
after surgery following an unrecognized nerve injury. In other the painful area from additional trauma. Anticonvulsant18 and
cases, neuromas form following a prolonged period of mild, antidepressant19 medications may provide relief for neuropathic
repetitive injury. At times, a large, named nerve is injured, and in pain; however, opiates are often used clinically,20 and a study in
other cases, a small, unnamed sensory nerve forms the neuroma. a rat model comparing morphine, pregabalin, gabapentin, and
1455
CHAPTER 181  Diagnosis and Management of Painful Neuromas 1455.e1

ABSTRACT KEY WORDS:


181
Painful neuroma formation is a complex and frustrating problem
nerve injection
for physicians and patients. These neuromas may appear after
obvious injuries to nerves, or they may appear quite unexpectedly nerve injury
after procedures varying from nerve biopsies and limb amputa- neuroma
tions to tooth extractions. As the nerve regenerates, sprouting peripheral nerve
neural fibers and connective tissue may develop haphazardly,
forming neuromas and potentially producing severe neuropathic
pain. The reported incidence of neuropathic pain resulting from
traumatic neuromas ranges from 20% to 30%. The pain caused
by traumatic neuromas may be vexingly resistant to pharmaco-
logic treatments, including those typically used for neuropathic
pain, such as anticonvulsants and antidepressants. When such
pain occurs after an otherwise unremarkable surgical procedure,
patients and physicians alike may experience significant distress.
Although there is no single best treatment for the pain caused by
traumatic neuromas, invasive procedures have been developed to
help treat this condition. Refractory cases may be amenable to
treatment with neurostimulation.
1456 SECTION 6  Pain

duloxetine showed that only morphine was effective.21 Comple-


mentary and alternative therapies such as acupuncture may be
useful.22
P When these noninvasive efforts are insufficient, a more
aggressive strategy may be required. Steroid injections, some-
times coupled with a local anesthetic, may provide temporary
and, in some cases, durable pain relief.23 Ablative techniques such
as radiofrequency ablation, pulsed radiofrequency ablation,
D
chemical neurectomy,24 or cryoneurolysis25 may destroy the asso-
ciated nerve or neuroma, providing the necessary pain relief.
There are a paucity of high-quality clinical data supporting the
use of these techniques.
Surgical neurectomy is considered after noninvasive treat-
Figure 181-1. Intraoperative photograph taken after exposure of ment measures fail and percutaneous ablative techniques are
a median neuroma at a midhumeral level. This patient sustained a deemed ineffective or inappropriate. For superficial neuromas
median nerve transection by glass from a broken window 5 months along named peripheral nerves, an attempt is made in the mildly
previously. The proximal nerve stump (P) has formed a bulbous sedated patient to subject the nerve to an external neurolysis and
neuroma, whereas the distal end (D) has thinned and scarred down to definitively identify the nerve. Palpation of the suspected neuroma
adjacent tissues. Note the significant retraction that has occurred, should reproduce the patient’s pain (Fig. 181-4). When located,
producing a substantial gap between the proximal and distal ends. the nerve is sectioned proximal to the neuroma, and the neuroma
is removed.
For superficial neuromas along unnamed, cutaneous sensory
nerves, a conscious pain-mapping procedure is used. First, the
painful area is marked in the preoperative area with the patient
fully awake. In the operating room, an incision is made over the
painful area using local anesthetic in the skin. Progressively
deeper dissection through nonanesthetized tissues enables iden-
tification of the discrete painful area in the awake patient. The
tissue is excised with the patient temporarily rendered uncon-
scious by the anesthesiologist. Repeat probing of the surrounding
tissues is performed with the patient awake to find and remove
any additional painful areas, which presumably contain the
responsible mechanosensitive tissue.

Figure 181-2. Intraoperative photograph taken after excision of a


sural neuroma at a midcalf level. This patient underwent a
PREVENTION OF SECONDARY NEUROMA
partial-thickness sural nerve biopsy 1 year previously. A fusiform A major problem in neuroma excision is the recurrence of the
neuroma has formed at the biopsy site, leaving the nerve in-continuity. neuroma, and prevention of growth of a secondary neuroma is a

Tibial S NAP

Peroneal

S NAP

Figure 181-3. Intraoperative photograph taken after external neurolysis of a sciatic nerve that
sustained a blunt injury several months previously. The nerve has been subjected to an internal neurolysis
as well, dividing it into its tibial and peroneal divisions. Hook electrodes are applied proximally and distally to
the injured area, a stimulus (S) is applied, and intraoperative nerve action potential recordings (NAP) are
obtained. The tibial division (top) shows a recovering nerve action potential, indicative of a conducting
neuroma-in-continuity, whereas the peroneal division (bottom) shows a flat tracing, indicative of a
nonconducting neuroma-in-continuity.
CHAPTER 181  Diagnosis and Management of Painful Neuromas 1457

181
N

B
Figure 181-5. Intraoperative photographs of a patient who
sustained an iatrogenic injury to the superficial radial sensory
nerve during an orthopedic surgical procedure a few months
previously. A shows a typical bulbous sensory neuroma (N) at
B the site of nerve transection. B shows the result after neuroma
excision and implantation (I) of the distal end of the nerve into the
Figure 181-4. Intraoperative photographs taken during surgical brachioradialis muscle. The sutures anchor the nerve into the muscle
excision of a painful sensory neuroma that formed along the to prevent pull-out with arm movement during the postoperative
iliohypogastric nerve following a laparoscopic procedure. With the period.
patient under mild sedation, a conscious pain-mapping procedure is
performed. Local anesthetic is infiltrated into the skin only. Palpation
of the deeper tissues reveals the painful area, subjacent to the fascia, may be transposed to a nearby muscle, implanted through a small
which is marked with an X (A). After the fascia is opened, the nerve incision in the muscle belly, and sutured into place (Fig. 181-5).
and a fusiform neuroma become apparent (B). Pinching the neuroma When in muscle, the likelihood of neuroma recurrence is
with forceps reproduces the patient’s pain exactly. With the nerve reduced. In a primate model of neuroma, transection of the radial
under traction, the nerve is transected proximal to the neuroma, and sensory nerve at the wrist, followed by turning the sensory nerves
the proximal end of the nerve is allowed to retract into the muscle proximally and implanting them into a forearm muscle, effec-
belly. This reduces the likelihood of neuroma recurrence. The distal tively prevented classical neuroma formation. Specifically, the
end is then cut, enabling excision of the entire neuroma. The distal sensory nerve showed no signs of regeneration when implanted
side is cut second so that the patient only experiences a single nerve into the muscle.12 Meyer and colleagues produced superficial
transection sensation. radial nerve neuromas in a group of baboons and found that
neuromas produce spontaneous activity of pain fibers and are
sensitive to mechanical stimulation.30 Thus muscle reimplanta-
tion may also reduce pain caused by mechanohypersensitivity by
key part of surgical treatment. In the past, treatment of neuromas decreasing the mechanical stimulation encountered by the
involved neuroma excision followed by capping the nerve with neuroma.31 In a study by Burchiel and colleagues of 18 patients
many substances, including silicone and surplus epineurium, to with distal sensory neuromas, reimplantation of nerve endings
prevent new neuroma formation.26 Additionally, chemical treat- showed a success rate of 44%, and an average drop in Visual
ment of the cut proximal end of the nerve has been used.27 One Analog Scale–registered pain of 32%.32 Dellon and associates
study reported that oblique transection of peripheral nerves, as found that 80% of patients had excellent pain relief after neuroma
opposed to perpendicular transection, greatly reduces the rate of resection and reimplantation into muscle.33 They suggest that the
classical neuroma development.28 Additionally, this method of nerve must be directed into muscle without tension in a distal-
oblique transection was thought to significantly reduce neuro- to-proximal direction to accommodate the minimal amount of
pathic pain compared with that following the traditional form of nerve regeneration that will inevitably occur and that it should
nerve transection. be implanted in muscles with minimal excursion to keep the
A different surgical strategy emphasizes placement of the cut implanted nerve in place. Additionally, if the nerve is implanted
end of nerve into an appropriate environment and does not near the deep surface of the muscle, regeneration into the overly-
attempt to physically restrain axonal outgrowth.29 In cases in ing skin is prevented, thus reducing mechanical stimulation of
which the nerve arises from muscle proximally, including the the nerve.34
plantar digital nerves, the lateral femoral cutaneous nerve, or the In addition to muscle, nerve implantation in veins, artificial
abdominal nerves such as the iliohypogastric and ilioinguinal, conduits, and autologous fat grafts have yielded positive results.
sectioning the nerve under tension allows it to retract into the In a randomized controlled trial of 20 subjects, Balcin and col-
muscle. If the nerve does not arise from nearby muscle, the nerve leagues reported a statistically significant greater reduction in
1458 SECTION 6  Pain

pain when the nerve end was implanted in vein compared with
muscle.35 Multiple retrospective case series also demonstrated
pain relief in patients who underwent nerve resection with
the use of collagen conduits.36,37 In a retrospective study of 30
patients, Gould and associates found that 43% of patients treated
with a collagen conduit during neuroma resection (“graft to
nowhere”) reported no pain at follow-up, 33% reported pain
scores of 1 to 4 (on a scale from 1 to 10), while 15% reported
pain scores of 8 to 10.36 Autologous fat grafting is another tech-
nique that has yielded positive results in small case series.38,39
Baptista and colleagues report reduction of pain in 9 of 11
patients.38

POOR SURGICAL CANDIDATES


The practicing neurosurgeon may encounter cases in which
neuroma excision surgery is inappropriate. For example, patients
who have already undergone one or more failed neuroma excision
surgeries, in our experience, do not experience pain relief with
additional neurectomy surgery. We can only speculate as to the Figure 181-6. Intraoperative photograph of a patient who
reason at this point, but centralization of the pain generators and developed a sensory neuroma near her incision following a hip
psychological factors may play a role. Postamputation neuromas replacement. Her painful area was marked (hatched area) using a
of large, mixed nerves such as the sciatic nerve may be especially conscious pain-mapping procedure intraoperatively. Two
problematic for the surgeon. Wound healing in these regions may subcutaneous stimulator electrodes were placed within the painful
be fraught with difficulties. In most cases, unless there is a clear region. Intraoperative testing revealed good overlap of the stimulation
structural abnormality in a stump, such as a sharp end of bone paresthesias with her painful area. During her 1-week external trial,
cutting into the nerve, local excision surgery should probably be she experienced significant pain relief, and she later underwent
avoided.40 permanent placement of the stimulator system.
When surgical excision of neuromas is thought to be inap-
propriate, and patients have failed less invasive measures, then
interventional pain management techniques may be used. Specifi-
cally, neurostimulation can be quite helpful, depending on the of the exposure and effect on surrounding nerves in some cases,46
distribution of pain.41 Neuroma pain in the extremities can gener- but a clear role remains to be established.
ally be treated with spinal cord stimulation. Spinal nerve root Ultimately, prevention of neuroma formation by inhibiting
stimulation, which targets pain-relieving stimulation paresthesias axonal growth into the nerve scar would be ideal. Perhaps nerve
into the distribution of single or multiple nerve roots, may growth inhibitors could be injected into injured nerves before
provide relief to patients with pain in the hands, feet, inguinal neuroma formation, or perhaps a method of pharmacologically
regions, pelvic regions, and intercostal distributions.42 These silencing mechanosensitive nerves subsequent to neuroma for-
areas are often unreachable with spinal cord stimulation. Periph- mation would be a comparably effective strategy. The role of
eral nerve stimulation may be used to treat pain confined to the conduits in prevention of primary and secondary neuroma forma-
distribution of the stimulated nerve. Small regions of neuroma tion remains to be further elucidated.
pain, inaccessible to other forms of neurostimulation, may be
targeted effectively with subcutaneous peripheral nerve stimula-
SUGGESTED READINGS
tion,43 in which the electrodes are placed under the skin at the Burchiel KJ, Johans TJ, Ochoa J. The surgical treatment of painful trau-
site of pain (Fig. 181-6). Transcutaneous magnetic stimulation44 matic neuromas. J Neurosurg. 1993;78:714-719.
and extracorporeal shock-wave therapy45 have also been used Dellon AL, Mackinnon SE. Treatment of the painful neuroma by neuroma
successfully to reduce neuroma-related pain. resection and muscle implantation. Plast Reconstr Surg. 1986;77:
427-438.
Gould JS, Naranje SM, McGwin G Jr, et al. Use of collagen conduits in
FUTURE DIRECTIONS management of painful neuromas of the foot and ankle. Foot Ankle Int.
Currently the surgical treatment of painful neuromas remains 2013;34:932-940.
crude, and the field has been advanced little in recent years Mackinnon SE. Evaluation and treatment of the painful neuroma. Tech
Hand Up Extrem Surg. 1997;1:195-212.
regarding definitive treatments. The most notable advancements Mackinnon SE, Dellon AL, Hudson AR, et al. Alteration of neuroma
have been in the field of neurostimulation, with novel applica- formation by manipulation of its microenvironment. Plast Reconstr
tions of electrical stimulation targeting pain relief to specific Surg. 1985;76:345-353.
painful areas and limiting unwanted paresthesias in adjacent, non- Seddon HJ. Three types of nerve injury. Brain. 1943;66:236-288.
painful areas. Future studies will hopefully establish whether Stuart RM, Winfree CJ. Neurostimulation techniques for painful periph-
these less invasive techniques, as well as the percutaneous ablative eral nerve disorders. Neurosurg Clin N Am. 2009;20:111-120, vii-viii.
techniques such as radiofrequency ablation, are effective. Robotic
techniques have been described and may help to reduce the size See a full reference list on ExpertConsult.com
CHAPTER 181  Diagnosis and Management of Painful Neuromas 1458.e1

REFERENCES 26. Swanson AB, Boeve NR, Lumsden RM. The prevention and treat-
1. Scadding JW. Development of ongoing activity, mechanosensitivity, ment of amputation neuromata by silicone capping. J Hand Surg 181
and adrenaline sensitivity in severed peripheral nerve axons. Exp [Am]. 1977;2:70-78.
Neurol. 1981;73:345-364. 27. Battista AF, Cravioto HM, Budzilovich GN. Painful neuroma:
2. Foltan R, Klima K, Spackova J, et al. Mechanism of traumatic changes produced in peripheral nerve after fascicle ligation. Neuro-
neuroma development. Med Hypotheses. 2008;71:572-576. surgery. 1981;9:589-600.
3. Seddon HJ. Three types of nerve injury. Brain. 1943;66:236-288. 28. Marcol W, Kotulska K, Larysz-Brysz M, et al. Prevention of painful
4. Steward JD. Pathologic processes producing focal peripheral neu- neuromas by oblique transection of peripheral nerves. J Neurosurg.
ropathies. In: Stewart JD, ed. Focal Peripheral Neuropathies. 3rd ed. 2006;104:285-289.
Philadelphia: Lippincott, Williams & Wilkins; 2000:11-35. 29. Mackinnon SE, Dellon AL, Hudson AR, et al. Alteration of neuroma
5. Kline DG. Timing for exploration of nerve lesions and evaluation of formation by manipulation of its microenvironment. Plast Reconstr
the neuroma-in-continuity. Clin Orthop Relat Res. 1982;42-49. Surg. 1985;76:345-353.
6. Wall PD, Gutnick M. Properties of afferent nerve impulses originat- 30. Meyer RA, Raja SN, Campbell JN, et al. Neural activity originating
ing from a neuroma. Nature. 1974;248:740-743. from a neuroma in the baboon. Brain Res. 1985;325:255-260.
7. Seltzer Z, Devor M. Ephaptic transmission in chronically damaged 31. Mackinnon SE. Algorithm for management of painful neuroma.
peripheral nerves. Neurology. 1979;29:1061-1064. Contemp Orthop. 1986;13:15-27.
8. Mackinnon SE. Evaluation and treatment of the painful neuroma. 32. Burchiel KJ, Johans TJ, Ochoa J. The surgical treatment of painful
Tech Hand Up Extrem Surg. 1997;1:195-212. traumatic neuromas. J Neurosurg. 1993;78:714-719.
9. Kanishka R, Sapna R, Hariharan S. Painful neuromas. Clin J Pain. 33. Dellon AL, Mackinnon SE, Pestronk A. Implantation of sensory
2012;28:639-645. nerve into muscle: preliminary clinical and experimental observa-
10. Stokvis A, Ruijs AC, van Neck JW, et al. Cold intolerance in surgi- tions on neuroma formation. Ann Plast Surg. 1984;12:30-40.
cally treated neuroma patients: a prospective follow-up study. J Hand 34. Dellon AL, Mackinnon SE. Treatment of the painful neuroma by
Surg [Am]. 2009;34:1689-1695. neuroma resection and muscle implantation. Plast Reconstr Surg.
11. Defalque RJ. Painful trigger points in surgical scars. Anesth Analg. 1986;77:427-438.
1982;61:518-520. 35. Balcin H, Erba P, Wettstein R, et al. A comparative study of two
12. Mackinnon SE, Dellon AL. Results of treatment of recurrent dorso- methods of surgical treatment for painful neuroma. J Bone Joint Surg
radial wrist neuromas. Ann Plast Surg. 1987;19:54-61. Br. 2009;91:803-808.
13. Boutin RD, Pathria MN, Resnick D. Disorders in the stumps of 36. Gould JS, Naranje SM, McGwin G Jr, et al. Use of collagen conduits
amputee patients: MR imaging. AJR Am J Roentgenol. 1998;171: in management of painful neuromas of the foot and ankle. Foot Ankle
497-501. Int. 2013;34:932-940.
14. Lee MJ, Kim S, Huh YM, et al. Morton neuroma: evaluated with 37. Thomsen L, Bellemere P, Loubersac T, et al. Treatment by collagen
ultrasonography and MR imaging. Korean J Radiol. 2007;8:148-155. conduit of painful post-traumatic neuromas of the sensitive digital
15. Kaminsky S, Griffin L, Milsap J, et al. Is ultrasonography a reliable nerve: a retrospective study of 10 cases. Chir Main. 2010;29:
way to confirm the diagnosis of Morton’s neuroma? Orthopedics. 255-262.
1997;20:37-39. 38. Baptista C, Iniesta A, Nguyen P, et al. [Autologous fat grafting in the
16. Pardal-Fernandez JM, Palazon-Garcia E, Hernandez-Fernandez F, surgical management of painful scar: preliminary results]. Chir Main.
et al. Contribution of a new electrophysiologic test to Morton’s 2013;32:329-334.
neuroma diagnosis. Foot Ankle Surg. 2014;20:109-114. 39. Vaienti L, Merle M, Battiston B, et al. Perineural fat grafting in the
17. Almeida OD Jr, Val-Gallas JM. Conscious pain mapping. J Am Assoc treatment of painful end-neuromas of the upper limb: a pilot study.
Gynecol Laparosc. 1997;4:587-590. J Hand Surg Eur Vol. 2013;38:36-42.
18. Wiffen P, Collins S, McQuay H, et al. Anticonvulsant drugs for acute 40. Wellons JC, Gorecki JP, Freidman AH. Stump, phantom, and avul-
and chronic pain. Cochrane Database Syst Rev. 2005;CD001133. sion pain. In: Burchiel K, ed. Surgical Management of Pain. New York:
19. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Thieme; 2002:422-442.
Database Syst Rev. 2007;CD005454. 41. Stuart RM, Winfree CJ. Neurostimulation techniques for painful
20. Przewlocki R, Przewlocka B. Opioids in neuropathic pain. Curr peripheral nerve disorders. Neurosurg Clin N Am. 2009;20:111-120,
Pharm Des. 2005;11:3013-3025. vii-viii.
21. Miyazaki R, Yamamoto T. The efficacy of morphine, pregabalin, 42. Haque R, Winfree CJ. Spinal nerve root stimulation. Neurosurg Focus.
gabapentin, and duloxetine on mechanical allodynia is different from 2006;21:E4.
that on neuroma pain in the rat neuropathic pain model. Anesth 43. Stinson LW Jr, Roderer GT, Cross NE, et al. Peripheral subcutane-
Analg. 2012;115:182-188. ous electrostimulation for control of intractable post-operative ingui-
22. Braverman DL, Ericken JJ, Shah RV, et al. Interventions in chronic nal pain: a case report series. Neuromodulation. 2001;4:99-104.
pain management. 3. New frontiers in pain management: comple- 44. Leung A, Fallah A, Shukla S. Transcutaneous magnetic stimulation
mentary techniques. Arch Phys Med Rehabil. 2003;84:S45-S49. (tMS) in alleviating post-traumatic peripheral neuropathic pain
23. Hassouna H, Singh D, Taylor H, et al. Ultrasound guided steroid states: a case series. Pain Med. 2014;15:1196-1199.
injection in the treatment of interdigital neuralgia. Acta Orthop Belg. 45. Fridman R, Cain JD, Weil L Jr. Extracorporeal shockwave therapy
2007;73:224-229. for interdigital neuroma: a randomized, placebo-controlled, double-
24. Mozena JD, Clifford JT. Efficacy of chemical neurolysis for the treat- blind trial. J Am Podiatr Med Assoc. 2009;99:191-193.
ment of interdigital nerve compression of the foot: a retrospective 46. Tigan L, Miyamoto H, Hendriks S, et al. Interest of telemicrosur-
study. J Am Podiatr Med Assoc. 2007;97:203-206. gery in peripheral nerve tumors: about a series of seven cases. Chir
25. Friedman T, Richman D, Adler R. Sonographically guided cryoneu- Main. 2014;33:13-16.
rolysis: preliminary experience and clinical outcomes. J Ultrasound
Med. 2012;31:2025-2034.
182 Dorsal Root Entry Zone Lesions for Pain
182

Marc Sindou, Andrei Brînzeu, and Kim J. Burchiel

Integration of sensory information is a multistage process leading When lesions occur at the level of the DREZ, several pro-
to perception with several levels of modulation along pathways. cesses can develop that upset the normal balance between inhibi-
The first level of modulation, located at the entry into the central tory and excitatory controls exerted on the second-order neurons
nervous system, is the dorsal root entry zone (DREZ) and dorsal situated in the DH. Lesions and deafferentation can occur as a
horn (DH). The role of this region was popularized in 1965 in result of a number of pathologic processes. These may lead to
the gate-control theory by Melzack and Wall.1,2 scar formation within the DREZ, and/or interruption of first-
DREZ was defined as an entity including the central portion neuron afferents with dendritic sprouting at the level of the
of the dorsal rootlet, the medial part of the tract of Lissauer, and second neuron, and thus complete interruption of inputs such as
layers I to V of the DH in which the afferent fibers terminate in root avulsion lesions.
and synapse3,4 (Fig. 182-1). In patients with lesions of the DREZ, microelectrode record-
At the level of the DREZ, dysfunction or failure of modulation ings of the DH have been performed showing spontaneous
or spontaneous generation of pain signals can occur as a result paroxystic activity at the level of DH neurons. This suggests that
of pathologic lesions, especially those leading to the loss of affer- one of the pain generators in such patients is at the level of the
ent stimuli, damage to neurons, or gliosis formation.5,6 This DREZ.9,26
makes the DREZ a logical surgical target for treating some types The microanatomy of the DREZ (see Fig. 182-1), with the
of intractable pain, either by modulating pain processes with pain fibers situated laterally and the localization of pain genera-
methods such as spinal cord stimulation7 or by creating surgical tors at the level of the DH, makes the DREZ a target for lesion-
lesions in the nociceptive system that limit signal transmission or ing in an attempt to control pain. The procedure intends to
destroy pain generators located at the DREZ.5,8-9 preferentially interrupt the (nociceptive) fibers grouped in the
Attempts at DREZ surgery were first made in 1972 at Lyon lateral bundle of the dorsal rootlet and the (excitatory) medial
University by Sindou.4,10 The technique of microcoagulation part of the tract of Lissauer. The dorsalmost layers of the DH,
of the dorsal rootlets entry was successfully used in a patient which harbor hyperactive neurons in the cases with deafferenta-
with Pancoast’s syndrome. Further attempts were successful tion (Fig. 182-2), are destroyed if microcoagulations are per-
for neuropathic pain after spinal cord injury (SCI) in Decem- formed deeply inside the DH. The procedure is presumed to
ber 1972, amputation of limb in July 1973, and brachial plexus partially preserve the (inhibitory) medially located structures of
avulsion (BPA) in January 1974, all performed in Lyon. Sub- the DREZ, namely the fibers reaching the dorsal column and
sequently, lesions at the DREZ were made by Nashold and their recurrent collaterals to the DH. Microsurgical DREZot-
colleagues in 1974 using radiofrequency (RF) thermocoagula- omy (MDT) was conceived to avoid total abolition of tactile and
tion.11-13 They coined the term DREZ operation and championed proprioceptive sensations when preoperatively present, as well as
its merits. Later, lesions were made with the laser beam by further deafferentation.27 The depth and extent of the lesion are
Levy and coworkers, Powers and associates, and Young and col- tailored depending on the degree of the desired therapeutic
leagues14-18 and with the ultrasonic probe by Dreval and Kandel effects and on the patient’s preoperative sensory and functional
and associates.19,20 In recent years, Spaic and coauthors designed status.28,29
a microsurgical technique of incising the dorsolateral sulcus and
suctioning the DH.21,22
INDICATIONS
RATIONALE
The DREZ serves as the interface between the peripheral nervous
Cancer Pain
system and the central nervous system at the level of the spinal The first use of DREZ lesioning for control of pain was to the
cord. Although in the peripheral nerves afferent fibers are ran- benefit of a patient with a neoplastic lesion, namely a Pancoast-
domly organized, at the level of the DREZ they become segre- Tobias tumor invading the brachial plexus.30,31 The procedure was
gated according to size and destination. The small myelinated greatly effective in controlling the pain of the patient, and it has
and unmyelinated fibers carrying nociceptive impulses course to been used since in a number of patients with pain related to
the lateral side, which makes them appropriately situated to enter cancer.32
into the tract of Lissauer, where they run for one or two segments It is estimated that 75% of patients with advanced cancer
before penetrating the gray matter of the spinal cord. The large suffer from chronic pain. Intricate mechanisms can lead to severe
myelinated fibers course medially to enter directly into the dorsal refractory pain. Somatogenic pain due to cancer invasion is
column.3,4 certainly an important component; however, neurogenic pain
The second neuron in the nociceptive pathway is located in linked to compression and destruction of neural structures or
the DH both superficially (Rexed laminae I and the substantia iatrogenic pain (after surgery or radiation treatment) can often
gelatinosa), receiving afferences from unmyelinated fibers, and be encountered.
deep in the DH (Rexed laminae V) for small myelinated fibers of In recent years, progress in oncology has greatly prolonged
the A delta type.23,24 These will project through the spinothalamic survival of patients with cancer, and the long-term use of opioids
tracts to the third neuron situated in the thalamus. can, even when administered intraspinally, have side effects
Activity of the second neuron is influenced both by upper infringing on the quality of life. The use of ablative procedures
levels and by peripheral afferents. Collaterals of large myelinated may be considered, therefore, in patients with refractory pain
fibers of the dorsal columns give inhibitory inputs that modulate and in those requiring high doses of morphine. Options are spi-
the passage of pain stimuli at the level of the DH.2,25 This has nothalamic tractotomy, open high cervical anterolateral cordot-
been popularized as the gate-control of Melzack and Wall.1 omy, percutaneous computed tomography−guided cordotomy or
1459
1460 SECTION 6  Pain

was from 1 month to 4 years, with a mean survival of 13 months.


For surgeries performed in the cervicothoracic region, control of
pain was good in 87% of patients; and for surgeries performed
in the lumbosacral region, control of pain was good in 78%.
Complication rates remained similar to those for DREZ per-
Cervical
DC T L formed for other types of pain; however, the authors reported
that in 2 patients, the procedure may have precipitated death.
Other series report a relatively small number of patients, and
I
results are somewhat heterogeneous.12,32,35-41 A composite of all
II series can be found in Table 182-1.
III
Thoracic

IV-VII Neuropathic Pain


IN
Root Avulsion Pain (Brachial and Lumbosacral Plexus)
MN
Brachial plexus injuries occur after major trauma, typically
Lumbar motorcycle crashes in young adults. Trauma results in stretching
of the roots up to their complete avulsion from the spinal cord.
Pain occurs in 30% to 90% of patients, and its incidence is related
to the localization of the lesion.42 Preganglionic lesions with true
deafferentation lead to severe chronic pain in 90% of patients,
A B Sacral whereas only 33% of patients with postganglionic lesions develop
chronic neuropathic pain.43
Figure 182-1. Anatomy of the dorsal root entry zone (DREZ). Description of the pain is quite stereotypical: continuous
A, Organization of fibers on transverse section. Most of the fine background pain described as burning or throbbing, associated
(nociceptive) afferents, which convey excitatory input, enter the to paroxysms of shooting electrical pain. Both pain components
dorsolateral sulcus as a lateral bundle of the DREZ, then penetrate the are described by two thirds of patients; only the background pain
dorsal horn (DH) ventrolaterally through the medial part of the tract of is noted in 20%, and solely paroxysms appear in only 10%. Most
Lissauer (TL) and the dorsal aspect of the substantia gelatinosa (SG). frequently, the distal part of the limb (hand and forearm) is con-
The Ramón y Cajal’s recurrent collaterals of the large primary afferent cerned. Sensory deficits are the rule; they are complete in two
fibers25 approach the DH through the ventromedial aspect of the SG thirds of patients. Major motor deficits are usually associated and
to exert inhibitory effects on the DH neurons.24 Because a number of more often situated on the C7 to C8 territories.44
dendrites of the cells of origin of the spinoreticulothalamic tract, which Pain is constantly described as unbearable and resistant to
will form the contralateral anterolateral pathways, make synaptic various types of medication, including opioids and antiepileptics.
connections with the primary afferents inside the SG layers, the SG Its onset can be immediately after the trauma or delayed even
exerts a strong segmental modulating effect on the nociceptive years after the initial event. In the period following the avulsion,
input.23 Large pink arrowhead: The microsurgical DREZotomy (MDT) most patients underwent direct nerve suturing, grafting, or neu-
target includes the lateral bundle of the (nociceptive) fine fibers, the rotization, all theoretically known to prevent the installation of
medial (excitatory) part of the TL, and the five dorsalmost layers of the chronic pain. For these patients facing severe uncontrollable pain,
DH where the primary afferents terminate and whose neurons become various surgical techniques have been tried with limited effect.
hyperactive if deafferentated. MDT attempts to spare the maximum Spinal cord stimulation is generally ineffective as a result of fiber
number of the large fibers of the medial bundle that reach the DC. degeneration up to the brainstem.45 Trials of deep brain stimula-
B, Variations of shape, width, and depth of the DREZ area according tion and motor cortex stimulation were revealed to be scarcely
to spinal cord level (from top to bottom, seventh cervical, fifth successful.46 The several other lesional techniques—anterolateral
thoracic, fourth lumbar, and third sacral medullary segments). cordotomy, spinothalamic tractotomy, medial thalamotomy—
Note how at the thoracic level, the TL is narrow and the DH is when tried, were not only ineffective but also accompanied by
deep, so DREZ lesioning, especially at that level, can be dangerous serious harmful side effects.
for the corticospinal tract and the dorsal column. IN, interneurons; DREZ lesioning was first attempted for the control of BPA
MN, motorneurons. pain, in 1974 in Lyon, using the MDT technique.3,29,47 Soon after,
Nashold used RF thermocoagulation as the lesion maker11,12;
and later on, laser14-17 and ultrasound18,19 were also used.12,29
midline myelotomy, and DREZ lesioning, which is particularly The efficacy of DREZ lesioning in controlling BPA pain has
adapted when the topography of the pain is limited.33,34 made it a recognized technique, and several case series have
Situations in which DREZ lesioning could be useful are pul- been published, although no randomized controlled trial was ever
monary apex tumors with invasion of the brachial plexus, thoracic attempted.
tumors with invasion of the wall nerves, and pelvic tumors with Preoperative evaluation must include magnetic resonance
invasion of the perineum and parietal nerves. Principles for indi- imaging (MRI) of the cervical spinal cord. Fine images are able
cations are a limited, controlled cancer in a patient with good to show the extent of the avulsions. Root meningocele is generally
enough general status to withstand major surgery and well- associated with root avulsion, but avulsion can be present even in
identified and well-characterized pain due to a clearly defined the absence of meningocele. In addition, MRI can show associ-
lesion. An early surgical decision can be made with benefit to the ated lesions of the spinal cord as well as eventually its rotation
patient when it provides the possibility to avoid extremely dis- because of arachnoiditis. This information can be useful for the
turbing and heavy pain medication. identification of the dorsolateral sulcus when attempting DREZ
Gadgil and Viswanathan recently reviewed results of DREZ surgery.44
lesioning in cancer pain.32 The largest series, to our knowledge, Findings during surgery include the avulsion of the ventral
is the one published by Sindou in 1995 describing results in 367 and dorsal rootlets, which can be partial or complete. Several
patients in whom DREZ lesioning was performed.28 In 81 levels are usually affected, but all roots of the brachial plexus are
patients, the pain was a consequence of a neoplasm. The follow-up avulsed in about 40% of patients. Scarring of the dorsolateral
CHAPTER 182  Dorsal Root Entry Zone Lesions for Pain 1461

182

A B

Figure 182-2. Microsurgical


DREZotomy (MDT) for brachial C D
plexus avulsion (BPA). MDT at the
cervical level for C6 to T1 brachial
plexus avulsion on the left side.
A-B, T2-weighted magnetic
resonance imaging shows complete
avulsion of both ventral and dorsal
roots and also of part of the lateral
cord (A), and a pseudomeningocele
at the lower cervical spine on the left
side with a hyperintensity of the
ipsilateral dorsal column subsequent
to the trauma and the stretching
with avulsion of the dorsal root
(B). C, Schematic drawing of the
floating microelectrode (arrow)
implanted into the dorsal horn
following its axis. D, Trace of the
dorsal horn microelectrode recording
E F
of so-called deafferentation
hyperactivity, manifested by
spontaneous regular high-frequency
discharges after BPA. E-H, Operative
views (rostral at bottom, caudal at
top) show total avulsion of cervical
roots on the left side. The
dorsolateral sulcus can be easily
identified in this case (E); incision into
the dorsolateral sulcus is made with
a microknife (F); the dorsolateral
sulcus is opened (G); and dotted
microcoagulations into the dorsal
horn, which has a gliotic aspect, are
performed 3 to 5 mm deep at a
35-degree angle inside the sulcus
with a specially designed, graduated G H
bipolar microforceps (H).
1462 SECTION 6  Pain

TABLE 182-1  Dorsal Root Entry Zone Lesioning Studies and Outcomes
Follow-Up in Years: Results: Percentage of Patients
Technique References No. of Patients Range (mean) Having >75% Relief (mean)
CANCER PAIN
Microsurgery Sindou unpublished data 94 0.1-4 78-90
RF-Th 10, 13, 31, 34, 36-38, 40, 41 37 0.1-4 33-100
BRACHIAL PLEXUS AVULSION
Microsurgery 44, 48, 51, 56; Taira personal 198 1-27 (4.47) 65-82.4 (77.01)
communication
RF-Th 10, 36, 38, 49-50, 52-55, 57-60 290 1-18 (5.67) 42, 7-87, 4 (70.64)
Ultrasound 20 124 1-5 (4) 87
CO2 laser 17 4 1-5 (4) 50
SPINAL CORD INJURY
Microsurgery 65, 67, 71, 73 112 0.5-20 (4.15) Segmental 68-100 (79.8); below lesion, 0
RF-Th 17, 34, 62, 63, 69, 70, 74 261 0.1-13 (3.11) 20-92; segmental, 78
PERIPHERAL NERVE PATHOLOGIES AND COMPLEX REGIONAL PAIN SYNDROME
Microsurgery Taira personal communication 7 (complex regional >1 57
pain syndrome)
Microsurgery Sindou 42 1 to 15 Paroxysmal, allodynic: good outcome
Continuous, deep: poor outcome
POST-HERPETIC PAIN
Microsurgery Sindou; Taira personal communication 6 1 to 3 50-66
RF-Th 34, 61, 77 20 0.5-5 14-67
POSTAMPUTATION PAIN
Microsurgery Sindou; Taira personal communication 14 1-15 Paroxysmal, allodynic: good outcome
Continuous, deep: poor outcome
RF-Th 34, 74, 75 45 0.5-6 25-100
RF-Th, radiofrequency thermocoagulation.

sulcus is often evident and is considered a major factor in pain irritative free bony fragments or to the fixation devices.64 Fre-
generation. Arachnoiditis, atrophy, and rotation of the spinal cord quently observed (up to 30%) at the conus medullaris and cauda
are also often seen. Absence of rootlets and major scarring make equina level, an important component of pain is neuropathic,
identification of the sulcus difficult. It is therefore essential to described as being of two types: segmental (at the level of the
expose at least one level above and one level below the avulsion injury) and infralesional (below the level of neural structure
to identify the sulcus in a healthy region.44 damage). Distinction between the two types is of importance for
Multiple groups have reported extensive series with long-term establishing an appropriate treatment strategy.65
results and with different types of lesion makers. Regardless of The segmental pain resides in the territories corresponding
the technique, immediate and short-term results are often spec- to the injury but can extend to several adjacent levels. It is pre-
tacular, with major improvement in up to 86% to 96% of patients. dominantly characterized as sharp electrical paroxystic attacks
However, relapses are not a rarity. A summary of all published lasting several minutes confined to dermatomes corresponding to
series, including DREZ lesioning for BPA pain, is presented in the lesional level. Mechanisms are related to the contusion of the
Table 182-1.10,16,19,36,38,44,48-60 spinal cord and of the nerve roots, with local changes leading to
Sindou reported several series with long-term follow-up to 28 entrapment and scarring. Intraoperative recordings have shown
years. Two thirds of patients had good (no more opioids) to excel- paroxystic hyperactivity in the DH neurons.8 Adjacent, especially
lent (no pain, no medication) outcome, and 71% noticed improve- rostral, levels can be involved because the scarring usually extends
ment in their daily activities at a mean follow-up of 6 years. At to levels above the level of injury and the distribution of pain
12 years, the Kaplan-Meyer statistic for excellent outcome (no fibers after their entry into the spinal cord is over several levels
pain, no medication) was 59.8%, testifying to the efficacy of (Fig. 182-3).
MDT for BPA pain44; moreover, another 25% of patients had a The infralesional pain is usually burning, continuous pain in
significant decrease in pain, and the result was considered good the anesthetic territory below the injury level. Several mecha-
(no use of opioids). Noteworthy in another series of 29 patients nisms have been described, but for practical purposes one should
operated by Sindou’s team, the results of MDT on the two com- assume that the principal one is deafferentation of the third-order
ponents of pain were analyzed. Kaplan-Meyer statistics at 10 neurons due to interruption of spinothalamic tracts. This locates
years showed complete relief in 76.2% of the patients for the the most probable pain generator above the spinal cord, leading
paroxysmal pain and 43.1% for the background pain; this was to the gross inefficacy of procedures targeted at the spinal cord.
statistically significant.48 The two types of pain are often present together in a hetero-
Although less frequent than brachial plexus injuries, lumbosa- geneous mix. In a series of 44 patients reported by Sindou
cral injuries with avulsion of the roots at the conus medullaris and colleagues,65,66 mostly affected at the conus medullaris level,
level should be searched and documented by MRI in patients the pain was segmental in 84% and infralesional in 16%. In a
with severe pelvic dislocation fractures. MDT is similarly effec- more recent series of 38 patients reported by Chun and associ-
tive for controlling pain in these cases.61-63 ates, the distribution of pain was 60% segmental and 40%
infralesional.67
Neuropathic pain after SCI is notoriously difficult to treat.
Spinal Cord Injury Pain Spinal neuromodulation techniques can be useful only in the
Chronic pain after SCI is frequent and can be of many types; situation of partial lesions when sensory loss is incomplete. Func-
it may have a mechanical origin related to the fractures and tional integrity of the dorsal columns must be checked before
CHAPTER 182  Dorsal Root Entry Zone Lesions for Pain 1463

C2 C3 182
C4
C3+C2
C4 C4
C5 T2
T2
T2 C5
T4 T4
T5 T5
T6 T6
T7 T1

C5 –
T1 T7

7
–C
T10
T10

C7
T12

C5
T12

C6+
T1

C8+
S4+S5
L1

+
C8

C7
7

T1
+C
Cyst L2 S3 L2

C6
L3
L3
S2

L4

L5+S1
L4 S1

S1
L5
A L5 D
B C
Figure 182-3. Microsurgical DREZotomy (MDT) for spinal cord and cauda equina injury. MDT is
performed bilaterally in a paraplegic patient after spinal cord injury at the conus medullaris (A), with a crush of
T12 to L4 spinal segments presenting with “segmental pain” in the corresponding territory (C). The drawing
(B) illustrates the intraoperative findings: myelomalacic cavity and gliosis in the conus medullaris. MDT will be
performed at the T12 to L4 spinal cord segments, bilaterally. The operative view (D) shows fibrotic arachnoids
surrounding the conus medullaris and cauda equina roots as well as the contused spinal cord. Arrowheads
designate the dorsolateral sulcus, which was difficult to identify within the fibrotic arachnoid and gliotic cord.

implantation.45 The use of intrathecal opioids in SCI patients neuropathic pain. Overall results are summarized in Table 182-1.
brings all the disadvantages of long-term morphine use, with Again, segmental pain was shown to be responsive to DREZ
frequent hyperpathy and therefore low efficacy in the long-term. lesions, whereas infralesional pain tends to be resistant. Other
Other intrathecal analgesics, such as ziconontide,68 have yet to be positive prognostic factors are the paroxystic character of the
proved effective; intrathecal therapy faces the challenge of drug pain, conus lesions, and incomplete lesions. Recently, a modified
delivery in patients with extensive arachnoiditis and poor cere- MDT procedure has been proposed by Chun and associates,67
brospinal fluid (CSF) circulation. with the aim of controlling infralesional pain; some results,
Segmental pain is likely to be sensitive to DREZ lesioning, however, remain unverified.
whereas pain below the injury level would probably be refractory
to the lesion. This has been verified in practice, and in the now
classic series of Sindou and colleagues, patients with mostly
Other Pain Indications
infralesional, burning, continuous pain had no benefit from the More rarely, DREZ lesioning has been applied to some other
procedure at long-term follow-up, whereas segmental pain was indications, such as complex regional pain syndrome (CRPS),
cured in two thirds of patients.65 peripheral nerve injury, postamputation pain, occipital neuralgia,
Preoperative assessment should therefore focus on the types post-herpetic neuralgia, and radiation-induced plexopathy, when
of pain, the residual functions below the level of injury that would pain did not respond to spinal cord stimulation. Published series
be at risk during the procedure, and evaluation of recent imaging are short and results mitigated over the long-term; therefore,
studies. DREZ lesioning cannot be considered a first-line indication in
Technically, delivering lesioning in the DREZ of an SCI is these pathologies.
generally demanding because the morphology of the spinal cord CRPS is most often responsive to spinal cord stimulation, and
is most often greatly altered, which makes it difficult to identify ablative surgery is rarely indicated. DREZ lesioning can be con-
the dorsolateral sulcus. Stepwise the procedure must expose the sidered in disabling and refractory forms.28
appropriate bony levels that need to be identified, sometimes For pain after peripheral nerve injuries or for postamputation
through complex fixation devices and reossification. Dural expo- pain, the first option is spinal cord stimulation. DREZ lesioning
sure is rendered difficult and often bloody by local fibrosis. Intra- may be considered when the predominant component of pain
durally, arachnoiditis and intrinsic lesions make it difficult to is of the paroxysmal type (electrical flashes) or corresponds
identify and liberate the rootlets. Surgeries performed in two to severe allodynia and hyperalgesia. Saris and Nashold reported
sittings have been described, the first being the approach up to good pain relief in short-term follow-up, but good pain relief
liberation of the rootlets. Whatever exposure not only the injured persisted in long-term follow-up in only 25% of patients.11,75
segments but also of the rostral healthy levels is most useful to Severe refractory occipital neuralgia can be effectively
identify the sulcus. The classical MDT technique for SCI seg- treated by MDT at the C2 to C3 level performed through a
mental pain implies a lesion from one level above and to one level hemilaminectomy.76
below the pain levels. The method thus far used for SCI DREZ For post-herpetic pain, there is a general agreement that
lesioning has been either microsurgery with microcoagulations DREZ surgery may alleviate paroxysmal and allodynic compo-
or RF thermocoagulation.16,38,55,62,63,66,67,69-72 nents, but the deep aching pain is not as well relieved and may
Mehta and colleagues have recently systematically reviewed even be aggravated. Because herpes zoster neuralgias occur
results of DREZ lesions for SCI injury.64 They concluded that mostly at the thoracic level, the procedure entails a significant
this type of procedure is effective for control of especially risk for neurological complications.38,61,62,77
1464 SECTION 6  Pain

Exposure should allow access to the targeted levels and also


Hyperspastic States with Pain the adjacent ones not only because of the necessity to extend
DREZ lesions have been noted to diminish muscle tone in the lesion to them but also for accurate identification of normal
the corresponding territory. This is explained by the fact that levels as help for precise lesion placement. A hemilaminectomy
MDT interrupts the afferents of both the myotactic and the with conservation of the spinous processes, if transversally
nociceptive reflexes and thus deprives the somatosensory relays large enough, is sufficient when surgery is performed unilaterally.
of the ventral horn from most of their excitatory inputs (see With proper rongeuring underneath the base of the spinous
Fig. 182-1). process the roots and dorsolateral sulcus on the other side
Hyperspastic states with pain can be encountered in the lower are visible. After dural incision is made, the dura should be sus-
limbs of paraplegic patients and even more in the upper limb pended in order to keep the intradural space clean of blood
of hemiplegic patients. Besides neuropathic components, pain contamination.
in these hyperspastic states can be due primarily to severe Working in the DREZ requires knowledge of microsurgical
tendon and muscle contractures and joint deformities. Also, asso- anatomy of the spinal roots and cord29,30 as well as of the internal
ciated algodystrophic reflex mechanisms may lead to so-called morphology of the cord to avoid damaging the neighboring ana-
CRPS. tomic structures79-81 (Fig. 182-4).
DREZ lesioning is used to reduce the excess of spasticity and For MDT, access to the dorsolateral sulcus should be per-
related painful manifestations. Technique and indications are formed ventrolaterally to the dorsal rootlet entry after the dorsal
described in Chapter 103.72 For hemiplegic patients with severe rootlets are displaced dorsally and medially (see Fig. 182-4). Then
spasticity in the upper limb, MDT is performed from C5 to T1 the sulcus is opened using a microknife of the ophthalmology
segments through a C3 to C7 hemilaminectomy. For patients type, until the pink-gray color of the DH can be seen (see Figs.
presenting with hyperspastic paraplegia, MDT is performed 182-1 and 182-4). Coagulation is performed using a fine-tipped
bilaterally through a T11 to L1 laminectomy from L2 down to bipolar forceps with millimetric graduations. Coagulations are
S2, and additionally down to S5 when there is a hyperactive performed at an angle depending on the level of the spinal cord:
bladder with urine leakage around the catheter. 35 degrees at the cervical level and 45 degrees at the lumbosacral
level (see Fig. 182-1), at a depth of approximately 3 to 5 mm. We
perform continuous coagulations along the dorsolateral sulcus at
TECHNIQUE every selected root. The objective of a DREZ lesion is to coagu-
Several methods for creating DREZ lesions have been late strictly within the DH, down to the level of the nucleus
described. Bipolar coagulation under direct vision of the DH proprius (laminae IV to V included). Accurate placement of the
was described by Sindou in 1972 and is commonly referred to as lesion is critical to avoid injury to the dorsal column medially and
MDT.3,10 Nashold and coworkers described in 1974 the use the corticospinal tract laterally.
of RF current delivered through a needle electrode piercing For RF thermocoagulation lesioning, detailed technique can
the dorsolateral sulcus.12 In 1983, Levy and colleagues intro- be found in the 2009 article by El-Naggar and colleagues.82 A
duced the use of carbon dioxide laser as the lesion generator in specially designed electrode is in use (Cosman Medical Inc., Bur-
an attempt to increase precision of the lesion.14,15,82,83 Dreval and lington, MA). This has a lesion tip of 2 mm and a diameter of
associates reported in 1993 on the use of ultrasound19,20 for the 0.25 mm. The electrode is inserted into the dorsolateral sulcus
same purpose, and finally Spaic and colleagues described direct at millimetric intervals, an the RF lesion is made with a current
aspiration of the DH in 2005.21,22,71,72 strength of 35 to 40 mA at a temperature of less than 75 degrees
Regardless of the method for creating the lesion, the key to for 15 seconds. The lesion usually extends 1 to 2 mm beyond the
the surgery is accurate identification of the appropriate levels to tip of the electrode. Nashold and associates also recommended
be targeted. This has to take into account the localization of the measuring impedance during surgery after they determined that
pain but also its nature and etiology in the context of the patient. the impedance of damaged spinal cord was less than 1200 Ω,
In patients with complete loss of function at and below the levels whereas that of a normal spinal cord reaches 1500 Ω.
concerned, lesions can be extended in terms of levels and depth Carbon dioxide, argon, and neodymium:yttrium aluminum
of lesioning. As an example, in SCI pain after trauma to the conus, garnet (Nd-YAG) lasers have all been used to create DREZ
many metameres might be concerned and therefore targeted. On lesions.14,15,18 Levy and colleagues advocated a pulse duration of
the other hand, in delicate regions, such as the thoracic cord 0.1 second, 20-W power, and one to two pulses to create a 2-mm
where the DREZ area is narrow and deep, lesioning must be kept deep lesion at an angle of 45 degrees to the DREZ.18 For SCI
to a minimum. pain, Stranjalis and Torrens used an Nd:YAG laser to create
When taking decision the most likely location of the pain lesions at 1-mm intervals to a depth of 2 mm at a power output
generators should be kept in mind and pain that is not likely of 10 W/sec, with the lesions extending 3 cm above and below
generated in the DH should not be targeted; DREZ lesions the involved segment.18 In his analysis of lesions made by RF and
mostly work for paroxystic shooting pain and only to a lesser lasers, Young reported that lasers produced a V-shaped lesion,
extent for continuous burning pain. whereas RF lesions were mostly spherical.16 Young also noted that
Patients are operated on under general anesthesia; muscle RF resulted in better pain control, but at the cost of a higher
relaxants are used only at the induction of surgery, which allows complication rate.
for electromyographic monitoring and, if necessary, motor Dreval, at the Moscow Central Institute, reported on the use
evoked potentials monitoring throughout the surgery. Monitor- of ultrasound at 44 KHz and an amplitude of 15 to 50 µm to
ing sensory evoked potentials may be useful for intraoperative create a “sulcomyelotomy” along the dorsolateral sulcus at an
identification of the metameric levels and sometimes determining angle of projection of 25 degrees medially and ventrally.19 This
the extent and depth of the lesion.84 Positioning should take into procedure keeps vessels crossing the sulcus intact.
account the CSF loss that can be important in long surgeries, and Spaic and coauthors reported a technique of microsurgical
therefore the dural opening should be kept higher than the head suctioning of the DH in place of the coagulation procedure.72
of the patient. They advocated use of a 0.8-mm suction tip to suction the DH
Identification of levels is of paramount importance and under microscopic visualization after opening the dorsolateral
requires identification of the vertebrae, after which intraoperative sulcus. According to these authors, the DH can be visualized at
stimulation of the anterior roots with electromyographic moni- 20 times magnification as a band of gray substance that gradually
toring of corresponding muscles will be used.85 reaches the massive central gray matter of the cord.
CHAPTER 182  Dorsal Root Entry Zone Lesions for Pain 1465

182

A B D

E F G
Figure 182-4. Microsurgical DREZotomy (MDT) for cancer pain caused by a pulmonary tumor invading
the right brachial plexus. Positron emission tomography in coronal (A), sagittal (B), and axial (C) planes
showing a well-limited tumor at the right pulmonary apex. T1-weighted magnetic resonance imaging with
gadolinium in axial section (D) shows invasion of spine and roots at the cervicothoracic junction. The patient
presented with complete functional loss of the upper limb, shooting pain along the arm down to the hand,
and allodynia and dysesthesia distributed from the shoulder down to T4 territory. The prone position with the
head and neck flexed in the Concorde position with three-pin head holder has the advantage of avoiding
brain collapse caused by cerebrospinal fluid depletion. The level of laminectomy is determined after
identification of the prominent spinous process of C2 by palpation. For unilateral dorsal root entry zone
operation, a hemilaminectomy with preservation of the spinous processes is sufficient to access the
dorsolateral aspect of the spinal cord. After opening the dura and arachnoid, identification of roots can be
verified by electrical stimulation at their corresponding foramen and their functional value checked. Stimulated
ventral roots have a motor threshold at least 3 times lower than the dorsal roots. Responses are in the
diaphragm for C4 (the response is palpable below the lower ribs), in the shoulder abductors for C5, in the
elbow flexors for C6, in the elbow and wrist extensors for C7, and in the muscles intrinsic of the hand for C8
and T1. Microsurgical lesioning is performed at the selected levels according to the preoperative program. As
shown in E and F, the dorsal rootlets are displaced dorsally and medially with a hook or a microsucker to
access to the ventrolateral aspect of the dorsolateral sulcus. Then an incision, 2 mm in depth at a 35-degree
angle, is made with a microknife, currently an ophthalmologic microscalpel, at the ventrolateral border of the
dorsolateral sulcus (F). Microcoagulations are made in a “chain” (i.e., dotted manner), 3 to 5 mm deep into
the dorsal horn (G). Each microcoagulation is performed, under direct magnified vision, by short-duration (a
few seconds), low-intensity, bipolar electrocoagulation, with a specially designed graduated sharp bipolar
forceps incremented in millimeters (graduated bipolar forceps ref: 12-30179, DREZotomy Set, Stryker
Leibinger GmbH & Co. KG, Freiburg, Germany]. The depth and extent of the lesion depend on the desired
therapeutic effect and the preoperative functional status of the limb. If the laxity of the root is sufficient, the
incision is performed, continuously, in the dorsolateral sulcus, thus accomplishing a sulcomyelotomy. If not,
successive incisions are made ventrolaterally at entry of each of the rootlets of the root after the surgeon has
isolated each one by separating the tiny arachnoid membranes that hold them together.
1466 SECTION 6  Pain

Burchiel KJ. Surgical Management of Pain. 2nd ed. New York: Thieme;
HOW TO AVOID COMPLICATIONS 2015.
Avoiding complications while achieving maximal results is a key Cahill K, Belzberg AJ, Anderson WS. Dorsal root entry zone lesions. In:
balance problem as for all functional neurosurgical procedures. Quinones-Hinotosa A, ed. Schmidek and Sweet Operative Neurosurgical
Techniques, Vol. 2. 6th ed. Philadelphia: Elsevier-Saunders; 2012:1481-
Although local or general complications are of the usual type 1485 [Chapter 141].
for a spine surgery (CSF leaks, hematoma, infection, thrombosis), Chun HJ, Kim YS, Yi HJ. Modified microsurgical DREZotomy proce-
their frequency may be higher in some groups of patients, such dure for refractory neuropathic pain. World Neurosurg. 2011;75:
as those with cancer or SCI, in whom status can be greatly 551-557.
altered. Dreval ON. Ultrasonic DREZ-operations for treatment of pain due to
Neurological complications occur as a result of the extension brachial plexus avulsion. Acta Neurochir (Wien). 1993;122:76-81.
of the surgical lesion to structures adjacent to the DH, namely Gadgil N, Viswanathan A. DREZotomy in the treatment of cancer pain:
the ipsilateral dorsal column or pyramidal tract or tracts impli- a review. Stereotact Funct Neurosurg. 2012;90:356-360.
cated in visceral function. Cited complications are dysesthesia, Guenot M, Bullier J, Rospars J, et al. Single-Unit analysis of the spinal
dorsal horn in patients with neuropathic pain. J Clin Neurophysiol.
hypoesthesia, ataxia, motor weakness, and urogenital dysfunction, 2003;20:142-150.
all of which may be either temporary or permanent. Jeanmonod D, Sindou M. Somatosensory function following dorsal root
Typically, patients in whom the cervical cord is operated on entry zone lesions in patients with neurogenic pain or spasticity.
are at risk for ipsilateral lower limb ataxia, and this has been J Neurosurg. 1991;74:916-932.
described in 4% to 25% of patients according to series. Although Kanpolat Y, Tuna H, Bozkurt M, et al. Spinal and nucleus caudalis dorsal
the depth of the lesion is a factor, its trajectory has, in our view, root entry zone operations for chronic pain. Neurosurgery. 2008;62:
a more important role, and in this aspect, MDT has an advantage 235-244.
because it implies opening the dorsolateral sulcus and controlling Mehta S, Orenczuk K, McIntire A. Neuropathic pain post spinal cord
the lesion through direct magnified vision of the DH and adja- injury part 2: systematic review of dorsal root entry zone procedure.
Top Spinal Cord Inj Rehabil. 2013;19(1):78-86.
cent tracts. Melzach R, Wall PD. Pain mechanism. A new theory. Science. 1965;150:
DREZ lesioning in the conus medullaris entails a higher risk 971-979.
for sphincter dysfunction in cases of preserved function and for Nashold BS, Ostdahl PH. Dorsal root entry zone lesions for pain relief.
disturbances of automatism in cases of complete lesions. Also, J Neurosurg. 1979;51:59-69.
coagulation of vessels in the dorsolateral sulcus, known to harbor Nuti C, Peyron R, Garcia-Larrea L. Motor cortex stimulation for refrac-
important arteries, may be the cause of uncontrolled extension of tory neuropathic pain: four year outcome and predictors of efficacy.
the lesion volume, with subsequent deficits. Pain. 2005;118:43-52.
Lesions performed in the thoracic cord are at a higher risk for Sanford M. Intrathecal ziconotide: a review of its use in patients with
neurological complications because of the narrowness of the DH chronic pain refractory to other systemic or intrathecal analgesics. CNS
Drugs. 2013;27:989-1002.
(see Fig. 182-1). Sindou M. Study of the dorsal root entry zone. Implications for pain
DREZ surgery may generate new pain at the borders of the surgery. M.D. Thesis, Lyon, France: University of Lyon Press; 1972.
operated territory. When reported, such pain was most often Sindou M. Microsurgical DREZotomy (MDT) for pain, spasticity and
considered bearable compared with the original pain that led to hyperactive bladder: a 20 year experience. Acta Neurochir (Wien). 1995;
the surgical indication. Mechanisms remain putative and may 137:1-5.
include unmasking of previous pain or damage to the DH cells Sindou M, Blondet E, Emery E, et al. Microsurgical lesioning in the
at the origin of the spinoreticulothalamic tract. dorsal root entry zone for pain due to brachial plexus avulsion: a pro-
Direct comparisons between the different techniques have not spective series of 55 patients. J Neurosurg. 2005;102:1018-1028.
been made in terms of their complications, with the exception of Sindou M, Mertens P. Surgery in the dorsal root entry zone for spasticity.
In: Lozano A, Gildenberg PL, Tasker RR, eds. Textbook of Stereotactic
those by Young and colleagues, who found more deficits with RF and Functional Neurosurgery, Vol. 2. 2nd ed. Heidelberg, Germany:
when performed on an area of 0.5 × 2 mm than with laser or RF Springer-Verlag; 2009:1959-1972 [Chapter 116].
on an area of 0.25 × 2 mm.16,17 MDT performed under direct Sindou M, Mertens P, Bendavid U, et al. Predictive value of somatosen-
vision of the intramedullary structures seems to generate less sory evoked potentials for long-lasting pain relief after spinal cord
severe neurological side effects. stimulation: practical use for patient selection. Neurosurgery. 2003;52:
1374-1383.
Sindou M, Mertens P, Wael M. Microsurgical DREZotomy for pain due
CONCLUSION to spinal cord and/or cauda equina injuries. Long-term results in a
DREZ lesioning can be helpful to relieve pain in strictly selected series of 44 patients. Pain. 2001;92:159-171.
Sindou M, Quoex C, Baleydier C. Fiber organization at the posterior
patients, especially those in whom pain generators are likely to spinal cord-rootlet junction in man. J Comp Neurol. 1974;153:15-26.
be at the level of the DH, the more so because in such situations Schirmer CM, Shils JL, Arle JE, et al. Heuristic map of myotomal inner-
other methods are not generally effective. Surgery in the DREZ vation in humans using direct intraoperative nerve root stimulation.
requires training in microsurgical techniques for safety and accu- J Neurosurg Spine. 2011;15:64-70.
racy. Indications should be discussed within the framework a Spaic M, Houlden DA, Schwartz ML. A novel method for making dorsal
multidisciplinary team to choose the most appropriate method horn lesions. Br J Neurosurg. 2012;26:531-536.
among the wide neurosurgical armamentarium.33 Taira T. Ablative neurosurgical procedure for pain after spinal cord injury.
World Neurosurg. 2011;75:449-450.
Wall PD, Sweet WH. Temporary abolition of pain in man. Science.
SUGGESTED READINGS 1967;108-109.
Aichaoui F, Mertens P, Sindou M. Dorsal root entry zone lesioning for Young RF. Clinical experience with radio-frequency and Laser DREZ
pain after brachial plexus avulsion: results with special emphasis on lesions. J Neurosurg. 1990;72:715-720.
differential effects on the paroxysmal versus continuous components.
A prospective study in a 29 patients consecutive series. Pain. 2011;
152:1923-1930. See a full reference list on ExpertConsult.com
CHAPTER 182  Dorsal Root Entry Zone Lesions for Pain 1466.e1

REFERENCES WH, eds. Progress in Neurological Surgery. Vol. 7. Basel, Switzerland:


1. Melzach R, Wall PD. Pain mechanism. A new theory. Science. Karger; 1993:201-250. 182
1965;150:971-979. 31. Sindou M, Lapras C. Neurosurgical treatment of pain in the
2. Wall PD. Presynaptic control of impulses at the first central synapse Pancoast-Tobias syndrome: selective posterior rhizotomy and open
in the cutaneous pathway. In: Eccles JC, Schade JP, eds. Physiology of antero-lateral C2-cordotomy. In: Bonica JJ, Ventafrida V, Pagni CA,
Spinal Neurons. Amsterdam: Elsevier; 1964:92-118. eds. Advances in Pain Research and Therapy, Vol. 4. New York: Raven
3. Sindou M. Study of the dorsal root entry zone. Implications for pain Press; 1982:199-209.
surgery. M.D. Thesis, Lyon, France: University of Lyon Press; 1972. 32. Gadgil N, Viswanathan A. DREZotomy in the treatment of cancer
4. Sindou M, Quoex C, Baleydier C. Fiber organization at the posterior pain: a review. Stereotact Funct Neurosurg. 2012;90:356-360.
spinal cord-rootlet junction in man. J Comp Neurol. 1974;153: 33. Burchiel KJ. Surgical Management of Pain. New York: Thieme; 2002.
15-26. 34. Kanpolat Y, Caglar S, Akayar S, et al. CT-guided pain procedures for
5. Loeser JD, Ward AA Jr, White LE Jr. Chronic deafferentation of intractable pain in malignancy. Acta Neurochir Suppl. 1995;64:88-91.
human spinal cord neurons. J Neurosurg. 1968;29:48-50. 35. Esposito S, Delitala A, Nardi PV. Microsurgery DREZ-lesion in
6. Jeanmonod D, Sindou M, Magnin M, et al. Intraoperative unit the treatment of deafferentation pain. J Neurosurg Sci. 1998;32:
recordings in the human dorsal horn with a simplified floating micro- 113-115.
electrode. Electroencephalogr Clin Neurophysiol. 1989;72:450-454. 36. Kumagai Y, Shimoji K, Honma T. Problems related to dorsal root
7. Wall PD, Sweet WH. Temporary abolition of pain in man. Science. entry zone lesions. Acta Neurochir (Wien). 1992;115:71-78.
1967;108-109. 37. Nashold BS, Bullitt E. Dorsal root entry zone lesions to control
8. Guenot M, Bullier J, Sindou M. Clinical and electrophysiological central pain in paraplegics. J Neurosurg. 1981;55:414-419.
expression of deafferentation pain alleviated by dorsal root entry 38. Rath SA, Braun V, Soliman N. Results of DREZ-coagulations for
zone lesions in rats. J Neurosurg. 2002;97:1402-1409. pain related to plexus lesions, spinal cord injuries and post-herpetic
9. Guenot M, Bullier J, Rospars J, et al. Single-unit analysis of the spinal neuralgia. Acta Neurochir (Wien). 1996;138:364-369.
dorsal horn in patients with neuropathic pain. J Clin Neurophysiol. 39. Ruiz Juretschke E, Garcia-Salazar F, Garcia-Leal R, et al. Treatment
2003;20:142-150. of neuropathic deafferentation pain using DREZ lesions: long-term
10. Sindou M. Dorsal root entry zone lesions. In: Burchiel KJ, ed. Surgi- results. Neurologia. 2011;26:26-31.
cal Management of Pain. New York: Thieme; 2002:701-713. 40. Samii M, Moringlane JR. Thermocoagulation of the dorsal root
11. Nashold BS, Urban B, Zorub DS. Phantom pain relief by focal entry zone for the treatment of intractable pain. Neurosurgery.
destruction of substantia gelatinosa of Rolando. In: Bonica JJ, Albe- 1984;15:953-955.
Fessard D, eds. Advances in Pain Research and Therapy, Vol. 1. New 41. Teixeira MJ, Fonoff ET, Montenegro MC. Dorsal root entry zone
York: Raven Press; 1976:959-963. lesions for treatment of pain related to radiation-induced plexopathy.
12. Nashold BS, Ostdahl PH. Dorsal root entry zone lesions for pain Spine. 2007;32:E316-E319.
relief. J Neurosurg. 1979;51:59-69. 42. Wynn-Parry CB. Pain in avulsion of the brachial plexus. Neurosur-
13. Nashold BS, Pearlstein R. The DREZ Operation. Park Ridge, IL: The gery. 1984;15:960-965.
American Association of Neurological Surgeons; 1996. 43. Narakas A. Les syndromes douloureux dans les arrachements du
14. Levy WJ, Nutkiewicz A, Ditmore M, et al. Laser induced dorsal root plexus brachial. Douleur et Analgésie. 1992;3:83-101.
entry zone lesions for pain control. Report of three cases. J Neuro- 44. Sindou M, Blondet E, Emery E, et al. Microsurgical lesioning in the
surg. 1983;59:884-886. dorsal root entry zone for pain due to brachial plexus avulsion: a
15. Powers SK, Barbaro NM, Levy RM. Pain control with laser-produced prospective series of 55 patients. J Neurosurg. 2005;102:1018-1028.
dorsal root entry zone lesions. Appl Neurophysiol. 1988;51:243-254. 45. Sindou M, Mertens P, Bendavid U, et al. Predictive value of somato-
16. Young RF. Clinical experience with radio-frequency and Laser sensory evoked potentials for long-lasting pain relief after spinal cord
DREZ lesions. J Neurosurg. 1990;72:715-720. stimulation: practical use for patient selection. Neurosurgery. 2003;52:
17. Young RF. Laser versus radiofrequency lesions of the DREZ. J Neu- 1374-1383.
rosurg. 1986;64:341. 46. Nuti C, Peyron R, Garcia-Larrea L. Motor cortex stimulation for
18. Stranjalis G, Torrens M. Dorsal root entry zone lesion performed refractory neuropathic pain: four year outcome and predictors of
with Nd:YAG laser. Br J Neurosurg. 1997;11:238-240. efficacy. Pain. 2005;118:43-52.
19. Kandel EL, Ogleznev KJA, Dreval ON. Destruction of posterior root 47. Sindou M, Fischer G, Goutelle A, et al. La radicellotomie postérieure
entry zone as a method for treating chronic pain in traumatic injury sélective. Premiers résultats dans la chirurgie de la douleur. Neuro-
to the brachial plexus. Vopr Neurochir. 1987;6:20-27. chirurgie. 1974;20:391-408.
20. Dreval ON. Ultrasonic DREZ-operations for treatment of pain due 48. Aichaoui F, Mertens P, Sindou M. Dorsal root entry zone lesioning
to brachial plexus avulsion. Acta Neurochir (Wien). 1993;122:76-81. for pain after brachial plexus avulsion: results with special emphasis
21. Spaic M. Dorsal horn grey substance elasticity: a guide to dorsal horn on differential effects on the paroxysmal versus continuous compo-
aspiration—a modality of the DREZ surgery lesioning technique. nents. A prospective study in a 29 patients consecutive series. Pain.
Acta Neurochir Suppl. 2002;144:1092. 2011;152:1923-1930.
22. Spaic M, Houlden DA, Schwartz ML. A novel method for making 49. Ali M, Saitoh Y, Oshino S, et al. Differential efficacy of electric motor
dorsal horn lesions. Br J Neurosurg. 2012;26:531-536. cortex stimulation and lesioning of the dorsal root entry zone for
23. Denny-Brown D, Kirk EJ, Yanagisawa N. The tract of Lissauer in continuous vs paroxysmal pain after brachial plexus avulsion. Neuro-
relation to sensory transmission in the dorsal horn of spinal cord in surgery. 2011;68:1252-1258.
the macaque monkey. J Comp Neurol. 1973;151:175-200. 50. Campbell JN, Solomon CT, James CS. The Hopkins experience with
24. Szentagothai J. Neuronal and synaptic arrangement in the substantia lesions of the dorsal horn (Nashold’s operation) for pain from avul-
gelatinosa rolandi. J Comp Neurol. 1964;122:219-239. sion of the brachial plexus. Appl Neurophysiol. 1988;51:170-174.
25. Ramon y Cajal S. Histologie du système nerveux, Vol. 1. Paris: Maloine; 51. Chen HJ, Tu YK. Long-term follow-up results of dorsal root entry
1901:986. zone lesions for intractable pain after brachial plexus avulsion inju-
26. Guenot M, Hupe JM, Mertens P, et al. A new type of microelectrode ries. Acta Neurochir Suppl. 2006;99:2738-2745.
for obtaining unitary recordings in the human spinal cord. J Neuro- 52. Friedman AH, Nashold BS, Bronec PR. Dorsal root entry zone
surg Spine. 1999;91:25-32. lesions for the treatment of brachial plexus avulsion injuries: a
27. Jeanmonod D, Sindou M. Somatosensory function following dorsal following-up study. Neurosurgery. 1988;22:369-373.
root entry zone lesions in patients with neurogenic pain or spasticity. 53. Garcia-March G, Sanchez-Ledesma MJ, Diaz P. Dorsal root entry
J Neurosurg. 1991;74:916-932. zone lesion versus spinal cord stimulation in the management of pain
28. Sindou M. Microsurgical DREZotomy (MDT) for pain, spasticity from brachial plexus avulsion. Acta Neurochir Suppl (Wien). 1987;39:
and hyperactive bladder: a 20 year experience. Acta Neurochir (Wien). 155-158.
1995;137:1-5. 54. Ishijima B, Shimoji K, Shimizu H. Lesions of spinal and trigeminal
29. Sindou M, Daher A. Spinal cord ablation procedures for pain. In: dorsal root entry zone for deafferentation pain. Appl Neurophysiol.
Dubner A, Gebhart GF, Bond MR, eds. Proceedings for the Fifth World 1988;50:175-187.
Congress on Pain. Amsterdam: Elsevier; 1988:477-495. 55. Kanpolat Y, Tuna H, Bozkurt M, et al. Spinal and nucleus caudalis
30. Sindou M, Fischer G, Mansuy L. Posterior spinal rhizotomy and dorsal root entry zone operations for chronic pain. Neurosurgery.
selective posterior rhizidiotomy. In: Krayenbül H, Maspes PE, Sweet 2008;62:235-244.
1466.e2 SECTION 6  Pain

56. Prestor B. Microsurgical junctional DREZ-coagulation for treat- of pain and implications for surgery in a series of 26 patients. Acta
ment of deafferentation pain syndromes. Surg Neurol. 2001;56: Neurochir (Wien). 2002;144:453-462.
259-265. 72. Spaic M, Markovic N, Mikicic D, et al. The DREZ surgical treat-
57. Samii M, Bear-Henney S, Ludemann W. Treatment of refractory ment of chronic pain in traumatic paraplegia. Indian J Neurotrauma.
pain after brachial plexus avulsion with dorsal root entry zone lesions. 2005;2:111-116.
Neurosurgery. 2001;48:1269-1277. 73. Taira T. Ablative neurosurgical procedure for pain after spinal cord
58. Thiebault JB, Bruxelle J, Thurel C. Pain in avulsion lesions of the injury. World Neurosurg. 2011;75:449-450.
brachial plexus: relief by dorsal horn lesions. In: Brunelli G, ed. 74. Weigand H, Winkelmuller W. Gehandlung des Deafferentierungss-
Textbook of Microsurgery. Paris: Masson; 1988:817-819. chmerzes durch Hochfrequenzlasion der Hinterwurzeleintrittzone.
59. Thomas DGT, Kitchen ND. Long-term follow-up of dorsal root Dtsch Med Wochenschr. 1985;110:216-220.
entry zone lesions in brachial plexus avulsion. J Neurol Neurosurg 75. Saris SC, Iacono RP, Nashold BS Jr. Successful treatment of phantom
Psychiatry. 1994;57:737-738. pain with dorsal root entry zone coagulation. Appl Neurophysiol.
60. Tomas R, Haninec P. Dorsal root entry zone (DREZ) localization 1988;51:188-197.
using direct spinal cord stimulation can improve results of the DREZ 76. Dubuisson D. Treatment of occipital neuralgia by partial posterior
thermocoagulation procedure for intractable pain relief. Pain. 2005; rhizotomy at C1-C3. J Neurosurg. 1995;82:581-586.
116:159-163. 77. Friedman AH, Nashold BS, Ovelmen-Levitt J. DREZ-lesions for the
61. Friedman AH, Bullitt E. Dorsal root entry zone lesions in the treat- treatment of post-herpetic neuralgia. J Neurosurg. 1984;60:1258-1261.
ment of pain following brachial plexus avulsion, spinal cord injury 78. Sindou M, Fischer G, Goutelle A, et al. La radicellotomie postérieure
and herpes zoster. Appl Neurophysiol. 1988;51:164-169. sélective dans le traitement des spasticités. Rev Neurol. 1974;130:
62. Friedman AH, Nashold BS. DREZ lesions for relief of pain related 201-215.
to spinal cord injury. J Neurosurg. 1986;65:465-469. 79. Sindou M, Mertens P. Surgery for intractable spasticity. In: Quinones-
63. Sampson JH, Chasman RE, Nashold BS, et al. Dorsal root entry Hinotosa A, ed. Schmidek and Sweet Operative Neurosurgical Techniques,
zone lesions for intractable pain after trauma to the conus medullaris Vol. 2. 6th ed. Philadelphia: Elsevier-Saunders; 2012:1377-1392
and cauda equina. J Neurosurg. 1995;82:28-34. [Chapter 122].
64. Mehta S, Orenczuk K, McIntire A. Neuropathic pain post spinal cord 80. Sindou M, Mertens P. Surgery in the dorsal root entry zone for
injury part 2: systematic review of dorsal root entry zone procedure. spasticity. In: Lozano A, Gildenberg PL, Tasker RR, eds. Textbook of
Top Spinal Cord Inj Rehabil. 2013;19(1):78-86. Stereotactic and Functional Neurosurgery, Vol. 2. 2nd ed. Heidelberg,
65. Sindou M, Mertens P, Wael M. Microsurgical DREZotomy for pain Germany: Springer-Verlag; 2009:1959-1972 [Chapter 116].
due to spinal cord and/or cauda equina injuries. Long-term results 81. Sindou M, Mertens P. Surgical interventions for spasticity: ablative
in a series of 44 patients. Pain. 2001;92:159-171. surgery for spasticity. In: Winn HR, ed. Youmans Neurological Surgery,
66. Sindou M. Surgery in the DREZ for refractory neuropathic pain Vol. 1. 6th ed. Philadelphia: Saunders; 2011:1026-1037.
after spinal cord/cauda equina injury. World Neurosurg. 2011;75: 82. Levy WJ, Gallo C, Watts C. Comparison of laser and radiofre-
447-448. quency dorsal root entry zone lesions in cats. Neurosurgery. 1985;16:
67. Chun HJ, Kim YS, Yi HJ. Modified microsurgical DREZotomy pro- 327-330.
cedure for refractory neuropathic pain. World Neurosurg. 2011;75: 83. Powers SK, Adams JE, Edwards MS, et al. Pain relief from dorsal
551-557. root entry zone lesions made with argon and carbon dioxide micro-
68. Sanford M. Intrathecal ziconotide: a review of its use in patients with surgical lasers. J Neurosurg. 1984;61:841-847.
chronic pain refractory to other systemic or intrathecal analgesics. 84. Jeanmonod D, Sindou M, Mauguiere F. The human cervical and
CNS Drugs. 2013;27:989-1002. lumbosacral evoked spinogram: data from intraoperative spinal cord
69. Edgar RE, Best LG, Quail PA. Computer-assisted DREZ microco- surface recordings. Electroencephalogr Clin Neurophysiol. 1991;80:
agulation: post-traumatic spinal deafferentation pain. J Spinal Disord. 477-482.
1993;6:48-56. 85. Schirmer CM, Shils JL, Arle JE, et al. Heuristic map of myotomal
70. Falci S, Best L, Bayles R. Dorsal root entry zone microcoagulation innervation in humans using direct intraoperative nerve root stimula-
for spinal cord injury-related central pain: operative intramedullary tion. J Neurosurg Spine. 2011;15:64-70.
electrophysiological guidance and clinical outcome. J Neurosurg. 86. El-Naggar OA. Dorsal root entry zone lesioning for neuropathic
2002;97:193-200. pain. In: Starr PA, Barbaro NA, Larson PS, eds. Neurosurgical Opera-
71. Spaic M, Markovic N, Tadic R. Microsurgical DREZotomy for pain tive Atlas: Functional Neurosurgery. 2nd ed. New York: Thieme;
of spinal cord and cauda equina injury origin: clinical characteristics 2009:103-111.
183 Tractotomy-Nucleotomy
183
Percutaneous Cordotomy and Trigeminal

Ahmed M. Raslan

was used for both cancerous and noncancerous pain, and finally it
HISTORY settled as a procedure that is mostly used for cancer-related pain.
Spinal ablation to control pain is not a new concept. In general, The ideal candidate is a cancer patient with unilateral somatic
physicians have been progressively moving away from procedures pain and a life expectancy of approximately 1 year. However,
that cause irreversible neural tissue destruction for pain control, extreme cases of frail patients with intractable noncancerous pain
albeit with a few exceptions. Cordotomy and trigeminal tractot- can be treated by cordotomy. Bilateral pain is much more difficult
omy are two such procedure exceptions. to treat, and midline pain is not a candidate for cordotomy. Pain
The underlying principle is the creation of a discrete lesion above the dermatome of C5 is generally not managed by cordot-
in a rather compact bundle of nerve fibers or cell bodies that omy. Pulmonary function test results higher than 50% in forced
transmit or process pain from a defined area of the body. This is expiratory volume and forced vital capacity in 1 second are gener-
usually undertaken in cases of intractable pain with or without a ally the minimal cutoffs for cordotomy candidacy.6
terminal or debilitating condition when other methods have Mesothelioma is a particularly attractive malignancy to treat
failed or survival time is not sufficient to offer the luxury of using cordotomy given its unilateral nature and somatic type of
testing alternatives. Cordotomy and trigeminal tractotomy pro- pain resulting from involvement of the pleura and ribs, together
cedures share a common history that historically represents pain with late involvement of pulmonary parenchyma to the point of
surgery itself. reduction of pulmonary functions beyond the minimal cutoff for
Early procedures were open and involved a knife section of cordotomy.7
either the anterolateral cord or a transverse stab incision dorsal
to the olive, which afforded a high-risk status with associated
morbidity and mortality.1,2
Technique
In the 1960s, a second iteration of these procedures was intro-
duced in two different places; a common thread was that they
Equipment
were both percutaneous. Cordotomy was made percutaneous An LCE kit for CT-guided spinal cord radiofrequency lesioning
through the pioneering efforts of Mullan3 and Rosomoff,4 by (Cosman Medical, Burlington, MA) is used to perform the
whereas trigeminal tractotomy was made percutaneous by Hitch- procedure. A radiofrequency lesion generator that is capable of
cock,5 who developed a stereotactic apparatus specifically for stimulation and impedance monitoring is needed and is manu-
trigeminal tractotomy and myelotomy. A third iteration was factured by multiple companies.
introduced by Kanpolat, who introduced computed tomography The procedure is typically performed in the CT scanner in a
(CT) guidance to both procedures. This latest procedural itera- radiology suite; however, with the recent availability of wide-bore
tion is performed using intraoperative CT guidance with neuro- mobile intraoperative CT scanners, the procedure could be done
monitoring and possibly general anesthesia. in the operating room.

CORDOTOMY Procedure
Cordotomy is essentially a lateral spinothalamic tractotomy; the Thirty minutes before the procedure, a lumbar injection of
spinothalamic tract carries nociceptive signals, temperature, and 12 mL of Omnipaque 300 mg/mL solution is performed, and the
nondiscriminative touch from the contralateral side of the body. patient is kept in the Trendelenburg position to allow contrast to
There is somatotopic organization of axons within the spinotha- diffuse up to the cervical region.
lamic tract; fibers entering from rostral and caudal segments are Cordotomy is performed in a supine position, and the head is
located in the medial and lateral parts of the tract, respectively. immobilized by taping to the CT gantry. Maintenance of an
The spinothalamic tract terminates mainly in the ventral poste- orthogonal neutral position to help the three-dimensional orien-
rior lateral nucleus, ventral posterior medial nucleus, intralaminar tation during needle placement is critical. The shoulders are
nuclei (mainly the central lateral nucleus), and posterior complex. pulled down to ensure adequate access to the side of the neck to
The corticospinal tract lies posterior to the lateral spinothalamic allow placement of the electrodes. The area around the mastoid
tract (LST), with white matter (the safety zone) in between. The tip is prepared and draped.
ventral spinocerebellar tract overlies the LST, and a lesion that Parallel, preferably zero gantry angle, 1- to 1.25-mm cuts are
eliminates the spinocerebellar tract may cause ipsilateral ataxia scanned spanning the foramen magnum down to the bottom of
of the arm. Human autonomic pathways for vasomotor and geni- C2, with a wide field of view to allow imaging of the skin. The
tourinary control, in addition to the reticulospinal tract that field of view can be adjusted to avoid denture artifact. The slice
controls ipsilateral automatic respiration, are also part of the of choice to perform the procedure is between C1 and C2, where
anterolateral quadrant of the spinal cord. Therefore, sleep apnea there is a lateral space between C1 and C2 to allow introduction
(Ondine’s curse), incontinence, and hypotension are possible of the needle. The laser beam of the scanner is used to guide the
undesirable cordotomy side effects (Fig. 183-1).6 entry point into the skin.
The skin dura thickness and anteroposterior and lateral diam-
eters of the cord are determined from the slice through which
Indications skin entry will be done.
Patient candidacy for cordotomy has evolved over time: origi- After local infiltration with 2% lidocaine, the LCE cannula is
nally it was widely used for chronic noncancerous pain, then it introduced through the skin to a length that is shorter than the
1467
1468 SECTION 6  Pain

Posterior tract (more anteromedial = more toward the arm side; more
funiculus posterolateral = more toward the leg side) is required.
Posterior column

Clinical.  Usually, cord penetration produces pain in the area


Lateral supposed to be ablated of pain sensation if a lesion is made in
Lateral corticospinal that particular target; also, electric stimulation confirms this
funiculus tract phenomenon.
Spinothalamic
tract Electrophysiologic
Impedance Monitoring.  Impedance monitoring is mainly
beneficial in identifying cord penetration but is of very limited
significance in regard to verification of the target inside the cord.
Anterior funiculus Macrostimulation.  The electrodes used for lesioning allow
stimulation of the spinothalamic tract (the target) in both sensory
Figure 183-1. Schematic representation of somatotopic organization
and motor frequencies to verify the target.
of spinothalamic and corticospinal tracts in the cervical spinal cord.
All patients are stimulated with a sensory frequency (100 Hz),
as well as motor frequency (2 Hz); pulse duration of both fre-
quencies is 0.1 msec. The expected response to sensory stimula-
tion is a painful or hot temperature sensation in the contralateral
half of the body, preferably covering the painful area at a thresh-
old not exceeding 0.15 V. The motor stimulation should be
carried out with no motor response in the ipsilateral side of the
body up to 1.0 V with exception of neck muscle response, which
could result from C2 anterior horn cell activation. If both condi-
tions are met, lesioning could be carried out; if not, then elec-
A B trode position needs to be adjusted.
The procedure begins with one test lesion of 60 seconds’
duration at 60° C; the other half of the body is then tested for
hypothesia and pain relief. The ipsilateral leg of the patient is
elevated to detect any subtle change of motor power during
lesioning. If adequate hypothesia or pain relief is not obtained,
which is the case in most patients, a second lesion of 75° C
for 90 seconds is performed. The benchmark of successful lesion-
ing is abolishment of the distinction between sharp pinprick
C D and dull sensation. The cannula is removed from the patient,
and the puncture site is sterilized and covered. Overnight obser-
Figure 183-2. A, Computed tomographic myelography reveals the vation with continuous pulse oximetry is needed for cordotomy
dentate ligament and the upper cervical spinal cord, the cord patients to monitor for possible sleep apnea. Generally speaking,
diameter, and skin dura thickness. B, The needle is introduced to a weaning off opioids could be started the next day after cordot-
trajectory anterior to the cord. C, The needle trajectory is adjusted to omy, but in most cases, it is delayed for about 1 week after the
proper targeting of the anterolateral quadrant. D, The electrode is procedure.
inserted through the needle into the spinal cord.

Results and Complications


skin dura thickness in a direction mostly perpendicular to the skin Most cordotomy case results pertain to cancer pain only, and
in the level of the mastoid tip in the anteroposterior plane. With results of cordotomy in noncancer pain are from relatively older
frequent CT checks, the position of the cannula is adjusted until studies; a recent review of all destructive procedures for noncan-
the dura is punctured. Before puncture of the dura, 2 mL of 2% cer pain lists the results of all cases series since 1966.9 However,
lidocaine can be injected to avoid pain due to contact with C2 this general perception had been challenged by case reports of
ganglion and the sensitive dura. After free flow of cerebrospinal prolonged pain relief after cordotomy for nonmalignant pain.10
fluid is obtained, a CT check is always performed, then according CT-guided cordotomy is categorically different from fluoro-
to the previously known lateral diameter of the cord, the length scopic cordotomy in that it allows direct visualization of the
of exposed tip of the electrode is adjusted on the hub of the target-electrode relationship, leading to higher safety and preci-
electrode and the electrode introduced through the cannula. sion. In a series of 41 patients,11 the immediate postoperative
Cord penetration is known when there is a rapid rise of the results of CT-guided cordotomy ranged from 92% to 98% pain
impedance from cerebrospinal fluid values, which are between relief or satisfactory pain relief, which declined to 80% at 6
400 and 1000 Ω; also either mild pain or electric sensation is months, with a persistent clinically and statistically significant
experienced by the patient. reduction of Visual Analog Scale (Fig. 183-3). Kanpolat reported
A low transdiscal route for cordotomy below C4 had been initial success of 97%, which declined to 84% at 6 months’
described by Raslan elsewhere to avoid sleep apnea (Fig. 183-2).8 follow-up.12
Cordotomy stands alone among all other ablative procedures
in that it is the procedure most often performed and is associated
Target Verification with a great degree of confidence in results despite a lack of high
Radiologic.  Frequent CT checks on the position of the elec- level of evidence. In a recent structured review of all ablative
trode in relation to the geometry of the cord (the opposite procedures, cordotomy was graded as 1C using the Grading of
anterolateral quadrant of the cord in relation to the side of pain) Recommendations Assessment, Development and Evaluation
and also in relation to the area of maximal pain intensity and the (GRADE) system,13 which equates to a strong recommendation
somatotopic arrangement of pain fiber within the spinothalamic of use based on low-level quality evidence.14
CHAPTER 183  Percutaneous Cordotomy and Trigeminal Tractotomy-Nucleotomy 1469

CT scanners, cordotomy will move back to the surgical suite


Visual Analog Scale
rather than the radiology suite.17 183
Endoscopic cordotomy was introduced in 2010 and presents
7
the potential for improved visualization of the lesioning
6 device−spinal cord relationship.18
Mean
5 TRIGEMINAL TRACTOTOMY-NUCLEOTOMY
4 Intractable facial pain can be one of the most devastating pains
in human experience. Sensory input from the head, through
3 cranial nerves V, VII, IX, and X, are carried by the trigeminal tract
to the spinal nucleus of the trigeminal nerve.
2
The compact sensory information from the head and face
1 carried by the trigeminal tract made it an ideal target for surgical
interruption. This kind of surgical interruption dates back to
0 1937 when Sjogvist surgically severed the tract through a trans-
VASpre VASpost VAS1 VAS3 VAS6 verse incision in the dorsal medulla.1 The immediate juxtaposi-
tion of the trigeminal nucleus to the trigeminal tract would
render any trigeminal tractotomy an effective tractotomy and
Figure 183-3. Persistence of statistically and clinically significant nucleotomy, hence the name trigeminal tractotomy-nucleotomy
reduction in a Visual Analog Scale (VAS) after cordotomy at 6 months. (TR-NC) (Fig. 183-4).
Percutaneous rhizotomy of the gasserian ganglion was subse-
quently used primarily for facial pain treatment along with open
rhizotomies to treat “typical” trigeminal neuralgia. Trigeminal
tractotomy was reserved for intractable terminal pain and because
CT-guided cordotomy is a safe procedure, and there are no of the extreme morbidity of the open procedures; therefore, it
reposted mortalities in the literature. This compares with a was not used often.19
6.25% mortality rate reported for the older fluoroscopic cor- When Crue and Hitchcock developed a stereotactic technique
dotomy technique.15 and device to lesion the trigeminal tract percutaneously, the pro-
The famous Ondine’s curse is an epitome of cordotomy risk. cedure realized resurrection in the late 1960s.5,20,21
A true Ondine’s curse is universally fatal; however, no cases The invention by Hitchcock in the 1960s5 is what led to the
of Ondine’s curse have been reported so far using CT-guided present-day version of the TR-NC, which was introduced by
cordotomy.11,12 Kanpolat in the early1990s. Kanpolat described and performed a
Postcordotomy painful sensory loss (dysthesia) affects 5% of CT- guided TR-NC, which allowed a direct visualization of the
patients. This can develop immediately but commonly develops electrode-target relationship.22
in long-term survivors who had a significant component of neu- Simultaneously, Nashold described and performed an open
ropathic pain and had received a large lesion.15,16 procedure to destroy the nucleus caudalis dorsal root entry zone
With direct visualization of the electrode in the spinal cord (DREZ) for deafferentation facial pain23; the TR-NC is a minia-
and proper electrophysiologic verification, ipsilateral weakness, ture of the caudalis DREZ operation.
ataxia, and urinary incontinence are very rare occurrences.
One rare troubling and unpredictable consequence of cor-
dotomy is what is called mirror pain, which could be due either
Indications
to a bilateral pain syndrome that is masked by marked severity Trigeminal neuropathic pain with varying degrees of trigeminal
on one side or to bilateral projection of dorsal roots in the both nerve damage and neuropathy are the main candidates for
spinothalamic tracts. TR-NC. Conditions such as traumatic trigeminal neuropathy,
Unintentional injection of local anesthetic in the cerebrospi- post-herpetic neuralgia, cancer pain of the head or face, glosso-
nal fluid could lead to respiratory cessation, especially if a cister- pharyngeal or geniculate neuralgia, bilateral trigeminal neuralgia,
nal injection is contemplated, and therefore clearly marking the and possibly anesthesia dolorosa are the typical indications for
injection syringes and completely discarding local anesthetics TR-NC.24
from the field are necessary.
Technique
Future Directions
Cordotomy is poised to be used once again as part of the neuro-
Equipment
surgical armamentarium, essentially returning full circle, for The needle, stylet, and electrode are from the LCE kit provided
several reasons: (1) costs to maintain and manage the complica- for CT-guided spinal cord radiofrequency lesioning.
tions of intrathecal opioid systems are increasing; (2) pharmaco- The procedure is generally performed in the CT scanner
logic neuromodulation is not always practically reversible; (3) in the radiology suite, but with the recent availability of wide-
recognition of opioid-induced hyperalgesia is increased; (4) bore mobile intraoperative CT scanners, the procedure could
development of cost-effective procedures is needed throughout be undertaken in the operative suite. To use an intraoperative
society; and (5) there is the potential for increased safety, accu- scanner for TR-NC, a wide-bore aperture of the CT scan is
racy, and precision of a cordotomy procedure using modern tech- recommended to allow for scanning while the needle and elec-
nology, such as intraoperative monitoring and neuronavigation. trode are in place to target in prone position without collision
The use of intraoperative imaging in cordotomy is a recent of the electrode with the CT scanner. In my experience, a switch
technical addition. A report of O-arm−guided cordotomy was to general anesthesia, given the difficulty of maintaining the
published in 2013.10 Burchiel and associates reported use of intra- prone position for patients with sensitive facial pain, is preferred
operative CereTom (NeuroLogica, Danvers, MA) in CT-guided for this procedure. Other neurosurgeons perform the proce-
tractotomy and myelotomy, and with wider bore intraoperative dure in a lateral position to circumvent this problem (personal
1470 SECTION 6  Pain

Figure 183-4. Diagrammatic representation of the trigeminal tractotomy electrode in position between C0 and
C1 and on axial between the dorsal column tract and nuclei medially and the spinocerebellar tract laterally.

communication). When general anesthesia is used, muscle relax-


ants should be withheld to allow for motor stimulation and elec-
trophysiologic verification of target.

Preoperative Preparation
Patients are usually admitted the day of surgery. The use of anti-
coagulants and antiplatelets is stopped at least 1 week before the
procedure. Thirty minutes before the procedure, a lumbar injec-
tion of 12 mL of Omnipaque 300 mg/mL solution is performed,
and the patient is kept in the Trendelenburg position.

Surgical Positioning
TR-NC is performed in the prone or lateral position, and the
head is immobilized by taping to the CT gantry. It is important
to maintain the head in an orthogonal position to help the three-
dimensional orientation during needle placement. If an intraop-
erative CT scan is used, a three-pin head holder is often used.17
Figure 183-5. Computed tomographic myelography showing the
Adequate flexion of the head to allow opening the space between
needle in final position for trigeminal tractotomy.
C0 and C1 is critical.

Imaging and Data Acquisition posterolateral sulcus on the spinomedullary junction approxi-
Parallel, preferably zero gantry angle, 1- to 1.25-mm cuts are mately midway between the midline and the equator of the cord
scanned spanning the foramen magnum down to the top of C2, dorsally. Note that the medial displacement of the target will
with a wide field of view to allow of imaging the skin. The field lead to lesioning of the gracile or cuneate tracts, whereas ventral
of view and the gantry angle are adjusted to avoid dentures arti- displacement risks missing the tract. Because of the proximity
fact. The axial slice of choice to perform TR-NC is between C0 of the spinocerebellar tract to the trigeminal tract, ipsilateral
and C1. The laser beam of the scanner is used to guide the entry upper extremity ataxis is almost unavoidable after this procedure;
point into the skin. however, it is almost always mild or temporary, or both.
The skin dura thickness, anteroposterior diameter, and lateral
diameter of the cord are determined from the slice through which
skin entry will be done.
Target Verification
Radiologic.  The position of the electrode is confirmed by CT
checking of the position of the electrode in relation to the geom-
Needle Placement etry of the cord. Figures 183-4 and 183-5 show the final position
The technique is similar to cordotomy with a difference in the of the electrode and depict its relationship to surrounding tracts.
skin entry point and the target. The entry point is usually 2 to
3 cm off the midline ipsilateral to the side of lesion at the axial Clinical.  Usually, cord penetration produces pain in the area
level determined by the laser beam of the skin corresponding supposed to be ablated of pain sensation if the lesion is made in
to the level of the space between C0 and C1; the target is the that particular target; also, electric stimulation confirms this
CHAPTER 183  Percutaneous Cordotomy and Trigeminal Tractotomy-Nucleotomy 1471

phenomenon. However, this is only relevant in awake patients, tions and side effects include ipsilateral upper extremity ataxia,
and when general anesthesia is used, there is no clinical verifica- which is usually mild but common, cerebrospinal fluid leak, men- 183
tion of the target. ingitis, and persistence of pain.24

Electrophysiologic
Impedance Monitoring.  Impedance monitoring is mainly
CONCLUSION
beneficial in identifying cord penetration; however, it was of very Neuroablation of the spinothalamic tract or the trigeminal tract
limited significance in regard to verification of the target inside is an effective method for control of intractable pain. These
the cord. procedures should not be considered first-line management but
Macrostimulation.  The electrodes used for lesioning allow can be an effective strategy in the right context for severe, refrac-
one to stimulate the spinothalamic tract (the target) in both tory pain. There is a renewed interest in these procedures, owing
sensory and motor frequencies to verify the target. to technologic advances and the emergence of new indications
All patients are stimulated with 100 Hz, with a pulse duration for patients who fail neuromodulation therapy.
0.1 msec as a sensory frequency, and no lesion is made except
after obtaining a sense of temperature change or painful sensation SUGGESTED READINGS
in the face. That verification is only possible in awake patients, Collins KL, Patil PG. Flat-panel fluoroscopy O-arm-guided percutane-
and it should be noted that stimulation of the trigeminal tract of ous radiofrequency cordotomy: a new technique for the treatment of
the nucleus is extremely painful. Motor stimulation at 2 Hz, with unilateral cancer pain. Neurosurgery. 2013;72(1 suppl Operative):27-34,
a pulse duration 0.1 msec, is then performed up to 1 V. There discussion 34.
should be no motor response in the extremities to stimulation, Kanpolat Y, Deda H, Akyar S, et al. CT-guided trigeminal tractotomy.
this part done irrespective of anesthesia used to rule out proxim- Acta Neurochir (Wien). 1989;100(3–4):112-114.
Kanpolat Y, Ugur HC, Ayten M, et al. Computed tomography-guided
ity to motor tracts. percutaneous cordotomy for intractable pain in malignancy. Neurosur-
gery. 2009;64(3 suppl):ons187-193, discussion ons193-194.
Lesion Making Raslan AM. Percutaneous computed tomography-guided radiofrequency
ablation of upper spinal cord pain pathways for cancer-related pain.
If patients are awake, creation of the lesion is painful, and lower Neurosurgery. 2008;62(3 suppl 1):226-233, discussion 233-224.
temperatures over longer periods have been advocated by Kan- Raslan AM, Cetas JS, McCartney S, et al. Destructive procedures for
polat. The author is performing the procedure with patients control of cancer pain: the case for cordotomy. J Neurosurg. 2011;
asleep, and TR-NC lesions are created using 75°C for 90 seconds, 114(1):155-170.
two lesions usually suffice. Thompson EM, Burchiel KJ, Raslan AM. Percutaneous trigeminal
tractotomy-nucleotomy with use of intraoperative computed tomog-
Postoperative care is very similar to cordotomy. raphy and general anesthesia: report of 2 cases. Neurosurg Focus. 2013;
35(3):E5.
Results and Complications
When patients are adequately selected, adequate pain control
rates exceed 90% for follow-up reported up to 4 years. Complica- See a full reference list on ExpertConsult.com
CHAPTER 183  Percutaneous Cordotomy and Trigeminal Tractotomy-Nucleotomy 1471.e1

REFERENCES 13. Guyatt GH, Oxman AD, Vist GE, et al. GRADE: an emerging con-
1. Sjöqvist O. Studies on pain conduction in the trigeminal nerve: sensus on rating quality of evidence and strength of recommenda- 183
a contribution to the surgical treatment of facial pain. JAMA. 1940; tions. BMJ. 2008;336(7650):924-926.
115(5):408. 14. Raslan AM, Cetas JS, McCartney S, et al. Destructive procedures for
2. Spiller WG, Martin E. THe treatment of persistent pain of organic control of cancer pain: the case for cordotomy. J Neurosurg.
origin in the lower part of the body by division of the anterolateral 2011;114(1):155-170.
column of the spinal cord. JAMA. 1912;LVIII(20):1489-1490. 15. Tasker R. Percutaneous cordotomy. In: Tasker R, Gildenberg P, eds.
3. Mullan S, Harper PV, Hekmatpanah J, et al. Percutaneous inter- Textbook of Stereotactic and Functional Neurosurgery. Berlin: Springer-
ruption of spinal-pain tracts by means of a strontium90 needle. Verlag; 2009.
J Neurosurg. 1963;20:931-939. 16. Kanpolat Y. CT guided percutaneous cervical cordotomy for cancer
4. Rosomoff HL, Brown CJ, Sheptak P. Percutaneous radiofrequency pain. In: Tasker R, Gildenberg P, eds. Textbook of Stereotactic and
cervical cordotomy: technique. J Neurosurg. 1965;23(6):639-644. Functional Neurosurgery. Berlin: Springer-Verlag; 2009.
5. Hitchcock E. Stereotactic trigeminal tractotomy. Ann Clin Res. 17. Thompson EM, Burchiel KJ, Raslan AM. Percutaneous trigeminal
1970;2(2):131-135. tractotomy-nucleotomy with use of intraoperative computed tomog-
6. Tasker RR. Percutaneous cervical cordotomy. Appl Neurophysiol. raphy and general anesthesia: report of 2 cases. Neurosurg Focus.
1976;39(2):114-121. 2013;35(3):E5.
7. Jackson MB, Pounder D, Price C, et al. Percutaneous cervical cor- 18. Fonoff ET, de Oliveira YS, Lopez WO, et al. Endoscopic-guided
dotomy for the control of pain in patients with pleural mesothelioma. percutaneous radiofrequency cordotomy. J Neurosurg. 2010;113(3):
Thorax. 1999;54(3):238-241. 524-527.
8. Raslan AM. Percutaneous computed tomography-guided transdiscal 19. Falconer MA. First and second division trigeminal neuralgia treated
low cervical cordotomy for cancer pain as a method to avoid sleep by intramedullary trigeminal tractotomy. Proc R Soc Med. 1954;47(4):
apnea. Stereotact Funct Neurosurg. 2005;83(4):159-164. 299-300.
9. Cetas JS, Saedi T, Burchiel KJ. Destructive procedures for the treat- 20. Fox JL. Intractable facial pain relieved by percutaneous trigeminal
ment of nonmalignant pain: a structured literature review. J Neuro- tractotomy. JAMA. 1971;218(13):1940-1941.
surg. 2008;109(3):389-404. 21. Crue BL, Carregal EJA, Felsoory A. Percutaneous stereotactic radio-
10. Collins KL, Patil PG. Flat-panel fluoroscopy O-arm-guided percu- frequency trigeminal tractotomy with neurophysiological recordings.
taneous radiofrequency cordotomy: a new technique for the treat- Stereotact Funct Neurosurg. 1972;34(6):389-397.
ment of unilateral cancer pain. Neurosurgery. 2013;72(1 suppl 22. Kanpolat Y, Deda H, Akyar S, et al. CT-guided trigeminal tractot-
Operative):27-34, discussion 34. omy. Acta Neurochir (Wien). 1989;100(3–4):112-114.
11. Raslan AM. Percutaneous computed tomography-guided radiofre- 23. Bernard EJ Jr, Nashold BS Jr, Caputi F, et al. Nucleus caudalis DREZ
quency ablation of upper spinal cord pain pathways for cancer- lesions for facial pain. Br J Neurosurg. 1987;1(1):81-91.
related pain. Neurosurgery. 2008;62(3 suppl 1):226-233, discussion 24. Kanpolat Y, Kahilogullari G, Ugur HC, et al. Computed tomography-
233-224. guided percutaneous trigeminal tractotomy-nucleotomy. Neurosur-
12. Kanpolat Y, Ugur HC, Ayten M, et al. Computed tomography- gery. 2008;63(1 suppl 1):ons147-153, discussion ons153-155.
guided percutaneous cordotomy for intractable pain in malignancy.
Neurosurgery. 2009;64(3 suppl):ons187-193, discussion ons193-194.

You might also like