Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

REVIEW

CURRENT
OPINION Neurotoxicity associated with cancer
immunotherapy: immune checkpoint inhibitors and
chimeric antigen receptor T-cell therapy
Claire Perrinjaquet a,, Nicolas Desbaillets b,, and Andreas F. Hottinger a,b

Purpose of review
Immune checkpoint inhibitors (ICPI) and chimeric antigen receptor T cells (CAR-T) represent novel therapies
recently approved to treat a number of human cancers. As both approaches modulate the immune system,
they can generate a number of immune-related adverse events (irAEs), including a large spectrum of novel
neurological toxicities. These are of special interest given their potential severity and risk of compromising
further oncologic treatment. We aim to provide a comprehensive review of the literature and discuss their
optimal management.
Downloaded from http://journals.lww.com/co-neurology by BhDMf5ePHKbH4TTImqenVLIlg0cBmue4nzEvPxp1Oh6WQ25vpQlwmRfP13/9PVPC on 12/15/2019

Recent findings
In contrast to irAEs involving other organs, neurological complications of ICPI are uncommon, may present
throughout the course of treatment and involve the peripheral and central nervous system, including
polyneuropathy, myositis, myasthenia gravis, demyelinating polyradiculopathy, myelitis, encephalitis and
others. If started early, ICPI-related neurologic irAEs are usually responsive to steroids. In contrast, as many
as 40% of patients undergoing CAR-T therapy will develop neurologic complications in the form of a
cytokine-release-associated encephalopathy. It includes delirium, aphasia, tremor/myoclonus, seizure and
seizure-like activity.
Summary
irAEs associated with CAR-T and ICPI therapy constitute new entities. Early identification and treatment are
essential to optimize the functional outcome and further oncologic management of the patient.
Keywords
chimeric antigen receptor T-cell therapy, chimeric antigen receptor T-cell-related encephalopathy, immune
checkpoint inhibitor, neurologic complication

INTRODUCTION by the immune system [2]. Antibodies against CTLA-4


In the last decade, intensive research has focused on (cytotoxic T-lymphocyte-associated antigen-4) and
the ability of cancer cells to evade the immune PD-1 or PD-L1 [programmed cell death-(ligand)-1]
system and how to harness this characteristic to have been the first approved ICPI given the impres-
fight cancer. Stimulating the immune system rather sive results in a growing number of cancer indications
than destroying cancer cells directly has been so [3] (Table 1). The CTLA-4 antibodies mainly
efficient that immunotherapy is now considered
the fifth pillar of cancer care after radiotherapy,
a
surgery, medical oncology and interventional Department of Oncology and bDepartment of Clinical Neurosciences,
oncology. To date, two main approaches have Centre Hospitalier Universitaire Vaudois & Lausanne University, Lau-
sanne, Switzerland
shown efficacy in the fight against cancer.
Correspondence to Andreas F. Hottinger, Department of Oncology;
Department of Clinical Neurosciences, Centre Hospitalier Universitaire
Vaudois & Lausanne University, BH 08-634, 1011 Lausanne,
IMMUNE CHECKPOINT INHIBITORS Switzerland. Tel: +41 21 314 1111; fax: +41 21 314 0737;
Immune checkpoint inhibitors (ICPI) inhibit specific e-mail: Andreas.hottinger@gmail.com
membrane receptors expressed by T cells which 
Claire Perrinjaquet and Nicolas Desbaillets contributed equally to the
dampen their activation to maintain self-tolerance article.
and prevent autoimmunity [1]. Cancer cells may Curr Opin Neurol 2019, 32:500–510
highjack this inhibitory pathway to evade destruction DOI:10.1097/WCO.0000000000000686

www.co-neurology.com Volume 32  Number 3  June 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Neurotoxicity associated with cancer immunotherapy Perrinjaquet et al.

isolated, activated and expanded ex vivo and then


KEY POINTS reinfused in patients after preparative lymphodeple-
 Patients treated with ICPI may develop a wide range of tion. This allows attacking tumors with much more
immune-related neurologic adverse events involving the favorable T-cell numbers. Moreover, preactivated
central or peripheral nervous system, including myositis, cells are less susceptible to immunosuppressive cues.
myasthenia gravis, demyelinating polyradiculopathy, However, this approach is sometimes unsuccess-
aseptic meningitis and encephalitis. ful, either because of a lack of intratumoral T cells or
 Moderate-to-severe neurologic immune-related because tumor cells don’t present immunogenic
neurologic adverse events occur in less than 1% of epitopes. One workaround consists in enabling T-
patients treated with ICPI. cell activation and tumor cell killing in a T-cell
receptor (TCR) and costimulation independent
 The first line of treatment consists in discontinuation of
manner. Such engineered T cells are called chimeric
ICPI and administration of corticosteroids, even in
diseases in which corticosteroids are usually not antigen receptor T cells (CAR-T) and express a chi-
considered, such as Guillain–Barré syndrome. In cases meric transmembrane-receptor harboring an extra-
that remain nonresponsive, second-line treatment must cellular scFv-fragment for tumor targeting linked via
be tailored to the specific situation. Options include a transmembrane domain to intracellular signaling
plasmapheresis, intravenous immunoglobulins and other domains. The signaling domains consist of CD3j,
immunosuppressive agents. the TCR Immune-receptor-Tyrosine-based-Activa-
 CAR-T therapy induces durable oncologic responses, tion-Motif-containing domain and costimulatory
but is associated with unique acute toxicities, which domains from CD28, 4-1BB or OX-40. This implies
can be severe or even fatal. The most common that a single target-antigen recognition event trig-
toxicities are cytokine-release syndrome and CAR-T- gers full activation of the T cell without the need for
related encephalopathy. major histocompatibility complex-epitope presen-
 Prompt and aggressive management of cytokine-release tation nor APC co-stimulation [4]. To date, two CAR-
syndrome and CAR-T-related encephalopathy are T therapies have been approved: Axicabtagene cil-
essential to minimize morbidity and mortality. oleucel (Yescarta, Gilead, Foster City, CA, USA) to
treat diffuse large B-cell lymphoma (DLBCL) [5] and
Tisagenlecleucel (Kymriah, Novartis, Basel,
act during T-cell priming when antigen pre- Switzerland) targeting CD19 against refractory B-cell
senting cells (APC) and the T cells interact in acute lymphoblastic leukemia and DLBCL [6,7].
the lymph node, whereas PD-1/PD-L1 anti- Currently, over 340 CAR-T trials with over 10 phase
bodies act directly at the tumor site when T 3 trials are ongoing [8]. Therefore, a growing number
cells and tumor cells interact. of patients will be eligible, and clinicians should be
prepared to deal with their adverse events.
ADOPTIVE T-CELL THERAPIES
Adoptive T-cell therapies (ACT) consist in adminis- COMPLICATIONS OF IMMUNE
trating a patient its own (autologous) or donor (allo- CHECKPOINT INHIBITORS
genic) antitumor lymphocytes. The approach mimics
successful T-cell immunization with activation and Systemic side effects of immune-related
expansion of antigen-specific T cells. In ACT this adverse events
expansion is however performed ex vivo to maximize Immune-related adverse events (IrAE) occur when T-
the response. Practically, intratumoral T cells are cell activation exceeds its normal range and induces

Table 1. Approved immune checkpoint inhibitors and indications


CTLA-4 Ipilimumab (Yervoy) Melanoma
PD-L1 Pembrolizumab (Keytruda), NSCLC, small-cell lung cancer, head and neck carcinoma,
nivolumab (Optivo) RCC, Hodgkin lymphoma, cervical carcinoma, PMBCL,
urothelial carcinoma, hepatocellular carcinoma, gastric cancer,
MSI-H or dMMR solid tumor
PD1 Atezolizumab (Tecentriq), durvalumab Urothelial cancer, NLCLC, Merckel cell carcinoma
(Imfinzi), avelumab (Bavencio)
CTLA-4 þ PD-L1 Ipilimumab þ nivolumab Metastatic melanoma, RCC, colorectal cancer (MSI-H or dMMR)

CTLA-4, cytotoxic T-lymphocyte-associated antigen-4; dMMR, deficient mismatch repair; MSI-H, microsatellite instability high; NSCLC, nonsmall-cell lung cancer;
PD-L-1, programmed cell death-ligand 1; PMBCL, primary mediastinal large B cell lymphoma; RCC, renal-cell carcinoma.

1350-7540 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-neurology.com 501

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Widening spectrum of CNS inflammatory disorders of the CNS

inflammation. They have been described in various different causes. A multidisciplinary team should be
organs including colon, liver, kidney, lung, thyroid, involved in the management decision, as discontin-
pituitary gland, skin, muscle, joint and eye. The uation of the oncologic treatment could decrease its
physiopathological pathway remains only partially efficiency. A lumbar puncture should be realized
understood but immune checkpoints play an impor- with symptoms emerging from the central nervous
tant role in immune homeostasis, preventing auto- system. An MRI may be helpful to exclude other
&
immunity and promoting self-tolerance [9 ]. potential causes.
The overall reported incidence of irAEs varies
from 15 to 90% depending on study [10,11]. The rate
of severe irAEs requiring treatment discontinuation MANAGING NEUROLOGICAL
is however much lower, depending on the agents COMPLICATIONS OF IMMUNE
evaluated (0.5–13%) [12–18]. CHECKPOINT INHIBITORS
Overall, PD-1/PD-L1 inhibitors show a lower
There are no prospective trials to help determine the
incidence of irAEs compared with CTLA-4 inhibi-
optimal management of nAEs. Early intervention is
tors. Combined double-axis inhibition (CTLA-4 and
the key to reduce both severity and duration [38]. As
PD-1/PD-L1) results in higher irAEs with 30–50% of
nAEs are most likely caused by general immunologic
patients developing grades 3–4 side effects in con-
activation, temporary immunosuppression with cor-
trast to 10–20% with either monotherapy
ticosteroids is recommended as soon as symptoms are
[11,12,17,19–30]. Significantly, whereas anti-PD-
severe enough or in certain-specific situations
1/PD-L1 irAEs appear to be dose independent, risks &&
(encephalitis, myasthenia gravis) (Table 2) [31 ].
of severe adverse event increase from 7 to 25% when
Plasmapheresis and immunosuppressants like myco-
ipilimumab dosage increases from 3 to 10 mg/kg.
fenolate, methotrexate, cyclophosphamide and rit-
Overall, neurologic side effects linked to ICPI
uximab have been considered in refractory cases.
remain rare. The most commonly affected organs
Alternative immunosuppressive strategies including
are, in order, the skin and gastrointestinal tract
&& natalizumab, bortezomib, tacrolimus or IL-17 inhib-
followed by the lung, and thyroid [31 ]. Moreover,
itors have also been efficient [38,39]. Most nAEs
16–24% of patients report nonspecific symptoms
resolve with adequate management.
such as fatigue [32].

Neurologic side effects of immune Toxicities of the peripheral nervous system


checkpoint inhibitors
The overall incidence of reported neurological Inflammatory myopathies
adverse events (nAE) of any grade was 3.8% with Myopathy appears to be the most frequent nAE with
anti-CTLA-4, 6.1% for anti-PD-1/PD-L1 and 12% anti-PD-1/PD-L1 and occurs less commonly with
with the combination of anti-PD-1 and anti- anti-CTLA-4. The most frequent types are necrotizing
CTLA-4 therapy. The most frequent side effects autoimmune myositis, dermatomyositis and poly-
described were aspecific symptoms (headache, dys- &
myositis [42 ,43–50]. Of note, rare cases of orbital
geusia, dizziness, peripheral sensory neuropathy) myositis and eosinophilic fasciitis have also been
&
[33 ]. Severe (grades 3–4) nAE occurred in 1.9% of &&
reported [50,51 ,52]. The usual symptoms include
patients under anti-CTLA-4 (ipilimumab) [34], and muscle pain, proximal limb weakness, difficulties
0.2–0.4% under anti-PD-1 (nivolumab or pembro- speaking/swallowing, ptosis or oculomotor weak-
lizumab) [20,35] and 0.1–1% under anti-PD-L1 (ate- ness. Moreira et al. [53] described 19 cases of ICPI-
zolizumab, avelumab or durvalumab) [28,36,37]. induced myositis, in which 32% presented associated
myocarditis and 5% had concomitant myasthenia
Diagnosis of neurological adverse events gravis, all of them receiving at least anti-PD-1 treat-
When a patient under ICPI develops neurological ment. Some patients developed respiratory distress
symptoms, one should keep in mind that nAE linked to diaphragm involvement. Laboratory
&&
remains a diagnosis of exclusion. Therefore, investi- workup often shows elevated creatine kinase [51 ].
gating potential differential diagnoses, including vas- Electrophysiological findings show a myopathic pat-
cular, toxic, metabolic, epileptic, infectious causes or tern. Muscle biopsy may show necrotic myofibers
&&
the presence of progressive oncologic disease (brain and inflammatory changes [51 ]. Treatment with
metastasis, leptomeningeal carcinomatosis or spinal high-dose corticosteroids and ICPI discontinuation
cord compression) should be prioritized. usually allows symptom improvement. Most patients
Rapid diagnosis and treatment initiation are recover completely, although patients with sequelae
crucial as sequelae or death can occur with all these have been described.

502 www.co-neurology.com Volume 32  Number 3  June 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Table 2. Differential diagnosis and work up of common immune checkpoint inhibitor-related neurological adverse events

Work up and management of common neurological complications of immune checkpoint inhibitors


Diagnosis Differential diagnosis Recommended work-up Grade Management

Peri pheral nervous system


Polyneuropathy
Metabolic Grade 1 (mild symptoms, without Low threshold to withhold immune
interference in daily life checkpoint inhibitors. Close monitoring
activities) for any progression
Toxic (previous chemotherapies, CAUTION: any cranial nerve
vitamin deficits) involvement must be considered
as grade 2
Exclude leptomeningeal spread in Grade 2 (moderate symptoms Withhold immune checkpoint inhibitor
case of cranial nerve involvement with interference in activities of treatment/close monitoring or steroids
daily life) (prednisolone/0.5–1 mg/kg).
Pregabalin or duloxetine for pain
Grade 3 (severe limitation in Hospitalize patient/withhold ICPI, high-
activities of daily life or life dose steroids (prednisolone 2 mg/kg)
threatening problems
(respiratory, other)
Guillain–Barré syndrome (progressive symmetric ascending muscle weakness with decreased reflexes  facial, respiratory or oculomotor/bulbar muscle involvement  autonomic nerve
dysregulation)
Spinal cord compression, myelitis ENMG (acute demyelinating First line: high-dose corticosteroids.

1350-7540 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved.
polyneuropathy) CAUTION: possible initial
deterioration
Infection (Lyme, HIV, HSV, HZV) CSF: normal WBC, elevated proteins In case of lack of improvement:
plasmapheresis or IVIG
Metabolic (acute intermittent Pulmonary function test with measure Autonomic dysfunction or pulmonary
porphyria, hypokalemia, of vital capacity and max function deterioration determines
hyperkalemia) inspiratory/expiratory pressures requirement for management in
intensive care
Drug induced (amiodarone, Anti-GQ1b (positive in Miller Fisher
disulfiram, organophosphates) variant)
Critical care polyneuropathy
CIDP, vasculitis
Myasthenia gravis (fluctuating muscle weakness, including cranial nerves with fatiguability, possible respiratory nerve involvement)
Metabolic myopathy Serum: acetylcholinereceptor and anti High-dose corticosteroids CAVE: risks of

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


musk antibodies deterioration
Toxic-induced myasthenic syndrome Tensilon or ice pack test Pyridostigmine

www.co-neurology.com
Polymyositis Repetitive nerve stimulation and If no improvement: plasmapheresis, IVIG,
single fiber EMG azathioprine cyclosporine or
mycophenolate to be considered
Permanently discontinue ICPI
Neurotoxicity associated with cancer immunotherapy Perrinjaquet et al.

503
504
Table 2 (Continued)

Work up and management of common neurological complications of immune checkpoint inhibitors


Diagnosis Differential diagnosis Recommended work-up Grade Management

Myositis (bilateral proximal limb weakness, possible muscle pain, fever, possible bulbar weakness, oculoparesis, bilateral ptosis)
Steroid myopathy Serum: elevated creatine kinase High-dose corticosteroids
Drugs (statins) ENMG: fibrillations Monitor patient closely if respiratory
muscle weakness
Consider muscle biopsy: necrotizing CAVE: possible concomitant myocarditis
myopathy with inflammatory infiltrate
Central nervous system

www.co-neurology.com
Encephalitis (confusion or altered behavior, headaches, motor or sensory deficits, dysphasia, fever)
Infection Brain  spine MRI Grade 1 (mild symptoms, no limits Hold ICPI/Initiate diagnostic work-up,
on activities of daily life) consider permanent discontinuation in
case of lack of improvement or if
worsening
Metabolic disturbances Serum: electrolytes, glucose, total Grade 2 Exclude viral and bacterial infection
proteins, serum electrophoresis, (consider concurrent antiviral and
consider viral serologies antibacterial treatment until infection
ruled out)
Brain metastasis CSF: WBC (usually <250/ml with High-dose corticosteroids (0.5–1 mg/kg/
lymphocytic predominance), day) methylprednisolone once
Widening spectrum of CNS inflammatory disorders of the CNS

normal gram stain, protein, infection excluded


glucose, PCR for HSV and other
viruses, cytology
Leptomeningeal dissemination
Cerebrovascular infarct
Intracerebral hemorrhage
Meningitis (headache, photophobia, neck stiffness, possible fever, vomiting, normal cognition)
Infection Brain and spine MRI Exclude bacterial and viral infection
Metabolic disturbances CSF: WBC (typically <500 ml), PCR High-dose corticosteroids
for viral infection, cytology
Leptomeningeal dissemination
Metastasis
Transverse myelitis (acute or subacute, often bilateral motor or sensory symptoms  autonomic deficits, sensory level)

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Spinal metastasis Brain and spine MRI High-dose cortcosteroids
Spinal cord compression CSF: usually normal Plasmapheresis or IVIG if non responsive
to steroids
Infection Serum: B12, TSH, HIV, syphilis,
ANA, anti-RO and La antibodies,
anti aquaporin-4 IgGs

Volume 32  Number 3  June 2019


CIDP, chronic inflammatory demyelinating polyneuropathy; CSF, cerebrospinal fluid; ENMG, electroneuromyography; HSV, herpes simplex virus; HZV, herpes zoster virus; ICPI, immune checkpoint inhibitor; IVIG,
&&
intravenous immunoglobulins; TSH, thyroid stimulating hormone; WBC, white blood cells. Adapted from [31 ,40,41].
Neurotoxicity associated with cancer immunotherapy Perrinjaquet et al.

Myasthenia gravis fatigue and weakness. Up to 10% of patients under


Makarious et al. [54] reported 23 cases of ICPI-asso- ipilimimab develop hypophysitis. Anti-PD-1/PD-L1
ciated myasthenia gravis with 70% of de novo cases is rarely implicated. Typically, hypophysitis appears
(13 receiving anti-PD-1, four anti-CTLA-4 and three 2–3 months after treatment initiation [64]. The
a combination). The incidence was 0.12% in a large diagnostic workup includes blood tests to analyze
set of patients treated with nivolumab [55]. Clinical all endocrine axes (adrenocorticotrophic hormone,
manifestations included ptosis, diplopia, muscle cortisol, thyroid stimulating hormone, f-T4, lutei-
weakness, dyspnea and dysphagia. Acetylcholine nizing hormone, follicle stimulating hormone, tes-
receptor antibodies were positive in only 59% of tosterone/estrogen, electrolytes). MRI may show an
patients. Concomitant myositis was present in nine enlargement of the gland and exclude other differ-
cases. Symptoms began 7–11 weeks after treatment ential diagnoses (metastasis) in the presence of
initiation. One-third of patients passed away despite endocrine modification.
aggressive treatments including pyridostigmine, ste- High-dose steroids are not prescribed anymore
roids, intravenous immunoglobulins (IVIG), plas- (unless in a clearly symptomatic patient), as this
mapheresis or plasma exchange. treatment doesn’t improve the outcome. Most of
the time, the pituitary gland is definitely destroyed,
Guillain–Barré syndrome and other requiring long-term substitution [65]. Once the
peripheral neuropathies patient is substituted, ICPI can be resumed.
Guillain–Barré syndrom (GBS) is a rare side effect.
Supakornnumporn et al. described five patients. All Encephalitis
but one developed symptoms around the third treat- Autoimmune encephalitis occurs in 0.1–0.25% of
ment-cycle, with sensory loss, paresis, areflexia, ICPI-treated patients, especially when combined
weakness, paresthesia, numbness, dysphagia as [66–69]. Larkin et al. reported six patients who devel-
main symptoms. Diagnosis is established by the oped encephalitis under nivolumab or nivolumab/
cerebrospinal fluid (CSF) showing an albuminocy- ipilimumab combination for melanoma 18–297 days
tologic dissociation and a demyelinating polyneur- after treatment introduction. Clinical presentation
opathy on electroneuromyography (ENMG). The was varied and included altered mental status with
treatment consisted in immunoglobulins alone or confusion, aphasia, agitation, memory problems,
with added corticoids for two patients. One was seizures, fever, fatigue, weakness, headache, stiff neck
treated with corticosteroids alone. The last patient [62]. Some patients may present with positive para-
received tacrolimus, corticoids followed by plasma neoplastic antibodies including anti-Hu and anti-N-
exchanges. Regarding outcome, two patients died, methyl-D-aspartate (NMDA) [70]. One case of cere-
two improved and one remained debilitated [56]. bellitis developing after three cycles of nivolumab has
ICPI-induced neuropathies occur in less than 1% been described [71]. The differential diagnosis
of patients and show great variability in severity. ICPI includes brain metastasis, leptomeningeal dissemi-
may affect small sensory-type fibers or show more nation, metabolic encephalopathy and infectious
typical presentations of immune-mediated phenom- origin, requiring a large diagnostic workup including
ena such as chronic inflammatory demyelinating pol- lumbar puncture (with viral, bacterial, paraneoplastic
&
yneuropathy [33 ,45,57–60]. For instance, Gu et al. panel, cytology analyses), MRI, EEG, pituitary hor-
reported 14 cases of acute neuropathy, mainly elicit- monal axes, toxic screening, complete blood tests.
ing sensorimotor neuropathy and hyporeflexia. Half Rapid management of encephalitis with treatment
of the patients showed lymphocytosis in the CSF and discontinuation and high-dose corticosteroids seem
an axonal pattern on ENMG allowing the distinction to be crucial for better outcome. Half of patients also
with GBS [61]. received IVIG. Empirically antiviral and broad-spec-
Polyradiculopathies and meningoradiculoneur- trum antibiotics should be started until an infectious
itis with facial diplegia, muscle weakness and con- cause is excluded.
trast enhancement of the caudal nerve fibers have
been described [46,57,58], as have cases of isolated Vasculitis
cranial mononeuropathies [62,63]. Steroids and Cases of giant cell arteritis and polymyalgia rheu-
IVIG are typically used. matica have been described in patients treated with
ipilimumab [45,46,72].
Toxicities of the central nervous system
Aseptic meningitis
Hypophysitis Aseptic meningitis is a rare side-effect, described
Clinical manifestations of a hypophysitis can evoke mostly with ipilimumab, which typically develops
neurological symptoms, in particular headache, between weeks 1 and 7 after initiation of ICPI. Main

1350-7540 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-neurology.com 505

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Widening spectrum of CNS inflammatory disorders of the CNS

symptoms include neck stiffness (but not always), is a frequent cause of death in CAR-T-treated
fever and headache [57]. CSF shows sterile liquid patients [80].
with lymphocytic predominance. Meningeal CRS is triggered by the activation of CAR-T when
enhancement is usually present on MRI. Steroid they encounter their target cells. This leads to a
treatment is generally effective [38,62]. massive release of cytokines such as IL-2, sIL-2Ra,
IFNg, IL-6, s-IL-6R and granulocyte colony stimulat-
Multiple sclerosis ing factor by the T cells and neighboring immune
De novo or relapsed multiple sclerosis, including cells, producing what is sometimes called a cytokine
optic neuritis, transverse myelitis and acute tumefac- storm. Serum IL-6 levels correlate with CRS severity,
tive demyelinating lesions have been described under and thus, IL-6R blockade with tocilizumab has been
ipilimumab treatment [73–76]. Significantly, even in approved as standard of care to treat CRS following
&&

the absence of ICPI therapy, expression of CTLA-4, tisagenlecleucel infusion [81 ]. Similarly, blocking
PD-1 and T cell immunoglobulin and mucin domain IL-6 directly with siltuximab is also used. Cortico-
3 is downregulated in multiple sclerosis patient when steroids can be used, but as they inhibit T-cell func-
compared with controls, strengthening the crucial tion and induce T-cell apoptosis, thereby reducing
role of inhibitory receptors in the maintenance of the efficacy of the oncologic treatment, they should
immune homeostasis [77]. be avoided whenever possible.

Neurosarcoidosis
Chimeric antigen receptor T-cell-related
A 68-year-old patient treated with combined ICPI encephalopathy
(nivolumab/ipilimumab) developed several systemic
irAEs including colitis, rash and transaminitis fol- CAR-T-related encephalopathy (CRES) is a central
lowed by pulmonary sarcoidosis 4 months after dis- nervous system toxicity mediated by CAR-T which
continuation of ICPI treatment that further evolved represents a significant cause of morbidity and mor-
to neurosarcoidosis with hemianopsia, agnosia, acal- tality potentially hindering the expansion of CAR-T
culia, disorientation and alexia. MRI revealed subtle treatments. Such nAEs are not rare, with up to 40%
leptomeningeal enhancement and T2-weighted sig- of patients developing severe and sometimes even
nal abnormalities. The patient initially improved fatal neurologic symptoms, [79,80,82,83]. Adverse
with dexamethasone but relapsed during dose taper- events have been observed in all CD19 CAR studies
ing. Methotrexate allowed an almost complete recov- and seem to occur with both CD28 and 4-1BB cos-
ery and stable oncological status [78]. timulatory domains although their incidence may
&&
be different [81 ]. The mechanisms underlying
these toxicities are only starting to emerge but dis-
&&
TOXICITY OF CHIMERIC ANTIGEN ruption of the brain–blood barrier [81 ,82,83] and
RECEPTOR T-CELL THERAPY cerebellar edema [5] via cytokine release by CAR-T
&&

In contrast to ICPI adverse events, CAR-T compli- are key features of CRES [81 ]. This leakage results in
cations are typically linked to acute complications cerebral edema, hemorrhage, infarction and necro-
during T-cell reinfusion, that is, cytokine-release sis due to intravascular coagulation [83]. The later
syndrome (CRS). It must however, be recognized can be measured in the patients’ blood with severe
that nAEs may arise both in the frame of the CRS neurotoxicity as they have significantly elevated
&&

and with distinct timing. DIC markers [81 ].


Although CRES can occur without a CRS, patients
usually present both (91%) simultaneously. The
Cytokine-release syndrome severity of CRES is proportional to the CRS, thus
The most common complication faced by patients being tightly linked to in-vivo T-cell expansion with
receiving CAR-T is a CRS. Clinically, it resembles CRES occurring when CAR-T reach high levels
&&
sepsis, with high fever, hypotension, tachycardia, [81 ,83]. Neurotoxic symptoms usually follow a ste-
respiratory insufficiency, myalgias, vascular leak, reotypical progression beginning with somnolence,
coagulopathy (DIC) and oliguria leading to multiple disorientation, confusion, diminished attention,
organ failure in severe cases. CRS can affect any mild aphasia, progressing to delirium, hallucina-
organ, including the nervous system. Symptoms tions, global aphasia, myoclonus or tremor. Finally,
usually start within the 1st days following T-cell severe cases evolve to generalized seizures and
infusion and peak between 1 and 2 weeks postin- encephalopathy potentially leading to coma and
&&
fusion. All CAR-T cell therapies elicit some degree of death [81 ]. EEG findings are nonspecific with dif-
CRS; 27% present severe CRS requiring intensive fuse generalized slowing  triphasic waves. These
care support [79]. Even when well managed, CRS patients are at risk of developing posterior reversible

506 www.co-neurology.com Volume 32  Number 3  June 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Neurotoxicity associated with cancer immunotherapy Perrinjaquet et al.

Table 3. Diagnosis and management of cytokine release syndrome and chimeric antigen receptor T-cell-related
encephalopathy syndrome
Grading and management of CRS and CRES
1. Grading of CRS
CRS grade 1 CRS grade 2 CRS grade 3 CRS grade 4

Symptoms or signs of CRS


(A) Temperature >38 8C Yes Yes Yes Yes
(B) SBP < 90 mmHg No Yes, responds to IV fluids/does Requires vasopressor with or Requires multiple vasopressors (excluding
not require vasopressors without vasopressin vasopressin)
(C) AND/OR hypoxia No Requiring low-flow nasal Requiring high-flow cannula Requies positive pressure (CPAP, BiPAP,
canula (6 l/min) or blow- (>6 l/min), facemask, non intubation and mechanical ventilation)
by rebreather mask or Venturi
mask

2. Grading of CRES
CRES grade 1 CRES grade 2 CRES grade 3 CRES grade 4

Neurological assessment score (by ICE 7–9 (Mild 3–6 (Moderate 0–2 (Severe impairment) 0 Patient in critical condition and/
score) impairment) impairment) or unable to perform assessment
Depressed level of consciousness (not Spontaneous Awakens to voice Awakens only to tactical Patient is unarousable or need for
attributable to other factors (sedation, awakening stimulus vigourous/repetitive tactical
. . .) stimulus. Stupor or coma
Raised intracranial pressure NA NA Focal/local edema on Diffuse cerebral edema on
neuroimaging neuroimaging; decerebrate or
decorticate posturing or cranial
nerve VI palsy; or papilledema
or Cushing’s triad
Seizure NA NA Any clinical seizure that Life-threatening generalized seizure
resolves rapidly or non (>5 min) or repetitive clinical or
convulsive seizure on electrical seizures without return
EEG that resolves with to baseline in between
intervention
Motor findings NA NA NA Deep focal motor weakness such
as hemi or paraparesis

3. Management recommendations for CRS

Grade 1
Fever or organ toxicity Acetaminophen and hypothermia blanket to treat fever/tocilizuma 8 mg/kg IV or siltuximab 11 mg/kg
IV for fever lasting >3 days Ibuprofen as 2nd line option/assess for infection/hydration with
IV fluids
Grade 2
Hypotension 500–1000 ml normal saline IV bolus (may be repeated)/tocilizumab or siltuximab if refractory to fluid
boluses/if refractory: start vasopressors, transfer to ICU/consider starting corticosteroids
Hypoxia O2 substitution (high flow  noninvasive positive pressure ventilation)/tocilizumab or siltuximab and
high-dose corticosteroids
Organ toxicity Symptomatic management/Tocilizumab or siltuximab and high-dose corticosteroids
Grade 3
Hypotension 500–1000 ml normal saline IV bolus (may be repeated)/tocilizumab or siltuximab if refractory to fluid
boluses/vasopressors as needed/transfer to ICU, echocardiogram and hemodynamic monitoring/
corticosteroids (dexamethasone 10 mg IV q6h, if refractory increase to 20 mg)
Hypoxia O2 substitution (high flow  non invasive positive pressure ventilation)/tocilizumab or siltuximab and
high-dose corticosteroids
Organ toxicity Symptomatic management/tocilizumab or siltuximab and high-dose corticosteroids
Grade 4
Hypotension ICU management: IV fluids, tocilizumab or siltuximab and methylprednisolone 1 g/day IV, vasopressors
and hemodynamic monitoring
Hypoxia Mechanical ventilation/tocilizumab or siltuximab and high-dose corticosteroids and supportive care as
described for grade 3 CRS
Organ toxicity Symptomatic management of organ toxicity/tocilizumab or siltuximab and high-dose corticosteroids and
supportive care as described for grade 3 CRS

1350-7540 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-neurology.com 507

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Widening spectrum of CNS inflammatory disorders of the CNS

4. Management recommendations for CRES

Grade 1
IV hydration, vigilant supportive care, aspiration precautions
Withhold oral intake of food, fluids and medicines, assess swallowing
Avoid all medications that may cause CNS depression
Agitated patients may be controlled with low-dose lorazepam (0.25–0.5 mg IV q8h) or haldoperidol (0.5 mg IV q6h)
Search for and follow a possible papilledema
Brain MRI
Lumbar puncture with measurement of opening pressure
MRI of spine if focal peripheral neurological deficits
Daily 30 min EEG until toxicity symptoms resolve, in absence of seizure prophylactic levetiracetam q12h
Consider tocilizumab 8 mg/kg IV or siltuximab 11 mg/kg IV if concomitant CRS
Grade 2
Consider transferring patient to ICU if CRES associated with grade 2 CRES
Supportive care and neurological work-up as indicated for grade 1 CRES
Toxilizumab 8 mg/kg IV or siltuximab 11 mg/kg IV if concurrent CRS
Dexamethasone 10 mg IV q6h if refractory to anti-IL-6 or for CRES without CRS
Grade 3
Transfer to ICU
Supportive care and neurological work-up as indicated for grade 1 CRES
Anti IL-6 therapy if associated with concurrent CRS
Dexamethasone 10 mg IV q6h if CRES symptoms worsen despite anti IL-6 therapy or for CRES without CRS. Taper once CRES is resolved to grade 1
Follow papilledema
Consider repeat MRI every 2–3 days if persistent CRES grade 3
Grade 4
ICU monitoring, consider mechanical ventilation for airway protection
Supportive care and neurological work-up as indicated for grade 1 CRES
Anti IL-6 therapy and repeat neuroimaging as described for grades 2 and 3 CRES
High-dose corticosteroids continued until improvement to CRES grade 1 (methylprednisolone IV 1 g/day, followed by rapid taper at 250 mg q12h for 2 days,
125 mg q12h for 2 days, 60 mg q12h for 2 days)
Stage 3 papilledema with CSF opening pressure 20 mmHg: high-dose corticosteroids (methylprednisolone IV 1 g/day)/elevate head end of patient’s bed to
308/hyperventilation to achieve a partial pressure of arterial carbon dioxide (PaCO2) of 28–30 mmHg, but maintained for no longer than 24 h/
hyperosmolar therapy with mannitol or hypertonic saline/consider neurosurgery consultation for Ommaya reservoir/drain/consider anesthetics for burst-
suppression pattern on EEG/metabolic profiling and daily imaging of brain to prevent rebound cerebral edema, renal failure, electrolyte abnormalities,
hypovolemia and hypotension

Comments: ICE score: 1 point for each of the following points: orientation to year, month, city, hospital (4 pts), name 3 objects (3 pts), ability to follow simple
command (show me 2 fingers: 1 pt), write a standard sentence (1 pt), count backwards from 100 in tens (1 pt). BiPAP, Bilevel Positive Airway Pressure; CNS,
central nervous system; CPAP, continuous positive airway pressure; CSF, cerebrospinal fluid; CRES, chimeric antigen receptor-T-related encephalopathy; CRS,
cytokine-release syndrome; EEG, electroencephalogram; ICE score, immune effector cell-associated encephalopathy score; IV, intravenous. Adapted from
&& &&
[80,81 ,85 ].

encephalopathy syndrome and acute necrotizing CONCLUSION


encephalopathy [83]. Immunotoxicity and autoimmunity are important
On the contrary, CRES remains unresponsive to issues to consider in patients treated with ICPI or
IL-6R blockade (tocilizumab) which is used to treat CAR-T therapies. Following ICPI therapy, neurolog-
CRS. Dexamethasone intravenous and anticonvul- ical irAEs are rare but may become severe or even
sive drugs (levetiracetam) are the only available life-threatening if not managed adequately. More-
&&
treatments [80,81 ]. Plasmapheresis has been pro- over, they may occur at any time during or even
posed, as it works for thrombotic thrombocytopenic after ICPI treatment. In contrast, neurologic toxic-
purpura, which shares similar endothelial cell acti- ity of CAR-T therapy is often associated closely
vation mechanisms but this approach still requires (within days or weeks) of the treatment infusion.
validation [84]. Consensus guidelines have been Preventing or reducing the severity and duration of
published by the American Society of Bone Marrow neurologic symptoms remains a key challenge. Cur-
&&
Transplant (Table 3) [85 ]. rent consensus guidelines remain however based

508 www.co-neurology.com Volume 32  Number 3  June 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Neurotoxicity associated with cancer immunotherapy Perrinjaquet et al.

21. Brahmer J, Reckamp KL, Baas P, et al. Nivolumab versus docetaxel in


on empirical data. Prospective clinical trials remain advanced squamous-cell non-small-cell lung cancer. N Engl J Med 2015;
a key priority. 373:123–135.
22. Borghaei H, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in
advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015;
Acknowledgements 373:1627–1639.
23. Carbone DP, Reck M, Paz-Ares L, et al. First-line nivolumab in stage IV or
None. recurrent non-small-cell lung cancer. N Engl J Med 2017; 376:2415–2426.
24. Reck M, Rodrı́guez-Abreu D, Robinson AG, et al. Pembrolizumab versus
chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med
Financial support and sponsorship 2016; 375:1823–1833.
25. Rittmeyer A, Barlesi F, Waterkamp D, et al. Atezolizumab versus docetaxel in
None. patients with previously treated nonsmall-cell lung cancer (OAK): a phase 3,
open-label, multicentre randomised controlled trial. Lancet 2017;
389:255–265.
Conflicts of interest 26. Motzer RJ, Escudier B, McDermott DF, et al. Nivolumab versus everolimus in
There are no conflicts of interest. advanced renal-cell carcinoma. N Engl J Med 2015; 373:1803–1813.
27. Younes A, Santoro A, Shipp M, et al. Nivolumab for classical Hodgkin’s
lymphoma after failure of both autologous stem-cell transplantation and
brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial.
Lancet Oncol 2016; 17:1283–1294.
REFERENCES AND RECOMMENDED 28. Rosenberg JE, Hoffman-Censits J, Powles T, et al. Atezolizumab in patients
with locally advanced and metastatic urothelial carcinoma who have pro-
READING gressed following treatment with platinum-based chemotherapy: a single-arm,
Papers of particular interest, published within the annual period of review, have multicentre, phase 2 trial. Lancet 2016; 387:1909–1920.
been highlighted as: 29. Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric
& of special interest or gastro-oesophageal junction cancer refractory to, or intolerant of, at least
&& of outstanding interest
two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a
randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;
1. Swaika A, Hammond WA, Joseph RW. Current state of anti-PD-L1 and anti- 390:2461–2471.
PD-1 agents in cancer therapy. Mol Immunol 2015; 67:4–17. 30. Soldatos TG, Dimitrakopoulou-Strauss A, Larribere L, et al. Retrospective
2. Sharma P, Allison JP. The future of immune checkpoint therapy. Science side effect profiling of the metastatic melanoma combination therapy ipilimu-
2015; 348:56–61. mab-nivolumab using adverse event data. Diagnostics (Basel) 2018; 8:1–13.
3. Wei SC, Duffy CR, Allison JP. Fundamental mechanisms of immune check- 31. Haanen JBAG, Carbonnel F, Robert C, et al. Management of toxicities from
point blockade therapy. Cancer Discov 2018; 8:1069–1086. && immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment
4. Wilkins O, Keeler AM, Flotte TR. CAR T-cell therapy: progress and prospects. and follow-up. Ann Oncol 2017; 28:iv119–iv142.
Hum Gene Ther Methods 2017; 28:61–66. The results of the European Society of Medical Oncology working group on the
5. Neelapu SS, Locke FL, Bartlett NL, et al. Axicabtagene ciloleucel CAR T-cell management of irAEs.
therapy in refractory large B-cell lymphoma. N Engl J Med 2017; 32. Johnson DB, Chandra S, Sosman JA. Immune checkpoint inhibitor toxicity in.
377:2531–2544. JAMA 2018; 320:1702–1703.
6. Schuster SJ, Bishop MR, Tam CS, et al. Tisagenlecleucel in adult relapsed 33. Cuzzubbo S, Javeri F, Tissier M, et al. Neurological adverse events associated
or refractory diffuse large B-cell lymphoma. N Engl J Med 2019; 380:45– & with immune checkpoint inhibitors: review of the literature. Eur J Cancer 2017;
56. 73:1–8.
7. Prasad V. Immunotherapy: tisagenlecleucel – the first approved CAR-T-cell A comprehensive review of neurological irAEs.
therapy: implications for payers and policy makers. Nat Rev Clin Oncol 2018; 34. Eggermont AM, Chiarion-Sileni V, Grob JJ, et al. Adjuvant ipilimumab versus
15:11–12. placebo after complete resection of high-risk stage III melanoma (EORTC
8. Köhl U, Arsenieva S, Holzinger A, Abken H. CAR T cells in trials: recent 18071): a randomised, double-blind, phase 3 trial. Lancet Oncol 2015;
achievements and challenges that remain in the production of modified T cells 16:522–530.
for clinical applications. Hum Gene Ther 2018; 29:559–568. 35. Weber JS, D’Angelo SP, Minor D, et al. Nivolumab versus chemotherapy in
9. Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events asso- patients with advanced melanoma who progressed after anti-CTLA-4 treat-
& ciated with immune checkpoint blockade. N Engl J Med 2018; 378: ment (CheckMate 037): a randomised, controlled, open-label, phase 3 trial.
158–168. Lancet Oncol 2015; 16:375–384.
A comprehensive summary of general systemic immune-related adverse events 36. Kaufman HL, Russell J, Hamid O, et al. Avelumab in patients with chemother-
(irAEs). apy-refractory metastatic Merkel cell carcinoma: a multicentre, single-group,
10. Eggermont AM, Chiarion-Sileni V, Grob JJ, et al. Prolonged survival in stage III open-label, phase 2 trial. Lancet Oncol 2016; 17:1374–1385.
melanoma with ipilimumab adjuvant therapy. N Engl J Med 2016; 37. Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after chemoradiotherapy in
375:1845–1855. stage III non-small-cell lung cancer. N Engl J Med 2017; 377:1919–1929.
11. Ferris RL, Blumenschein G, Fayette J, et al. Nivolumab for recurrent squa- 38. Hottinger AF. Neurologic complications of immune checkpoint inhibitors. Curr
mous-cell carcinoma of the head and neck. N Engl J Med 2016; 375: Opin Neurol 2016; 29:806–812.
1856–1867. 39. Touat M, Talmasov D, Ricard D, Psimaras D. Neurological toxicities asso-
12. Hodi FS, O’Day SJ, McDermott DF, et al. Improved survival with ipilimumab in ciated with immune-checkpoint inhibitors. Curr Opin Neurol 2017;
patients with metastatic melanoma. N Engl J Med 2010; 363:711–723. 30:659–668.
13. Wolchok JD, Neyns B, Linette G, et al. Ipilimumab monotherapy in patients 40. Astaras C, de Micheli R, Moura B, et al. Neurological adverse events
with pretreated advanced melanoma: a randomised, double-blind, multicen- associated with immune checkpoint inhibitors: diagnosis and management.
tre, phase 2, dose-ranging study. Lancet Oncol 2010; 11:155–164. Curr Neurol Neurosci Rep 2018; 18:3.
14. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor remission, 41. Puzanov I, Diab A, Abdallah K, et al. Managing toxicities associated with
and long-term safety in patients with advanced melanoma receiving nivolu- immune checkpoint inhibitors: consensus recommendations from the Society
mab. J Clin Oncol 2014; 32:1020–1030. for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J
15. Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune Immunother Cancer 2017; 5:95.
correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366: 42. Kao JC, Liao B, Markovic SN, et al. Neurological complications associated
2443–2454. & with anti-programmed death 1 (PD-1) antibodies. JAMA Neurol 2017;
16. Ribas A, Kefford R, Marshall MA, et al. Phase III randomized clinical trial 74:1216–1222.
comparing tremelimumab with standard-of-care chemotherapy in patients A comprehensive review of neurological irAEs.
with advanced melanoma. J Clin Oncol 2013; 31:616–622. 43. Johnson DB, Sullivan RJ, Ott PA, et al. Ipilimumab therapy in patients with
17. Larkin J, Chiarion-Sileni V, Gonzalez R, et al. Combined nivolumab and advanced melanoma and preexisting autoimmune disorders. JAMA Oncol
ipilimumab or monotherapy in untreated melanoma. N Engl J Med 2015; 2016; 2:234–240.
373:23–34. 44. Spain L, Walls G, Julve M, et al. Neurotoxicity from immune-checkpoint
18. Postow MA, Chesney J, Pavlick AC, et al. Nivolumab and ipilimumab versus inhibition in the treatment of melanoma: a single centre experience and review
ipilimumab in untreated melanoma. N Engl J Med 2015; 372:2006–2017. of the literature. Ann Oncol 2017; 28:377–385.
19. Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as second-line therapy 45. Cappelli LC, Gutierrez AK, Bingham CO, Shah AA. Rheumatic and muscu-
for advanced urothelial carcinoma. N Engl J Med 2017; 376:1015–1026. loskeletal immune-related adverse events due to immune checkpoint inhibi-
20. Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated tors: a systematic review of the literature. Arthritis Care Res (Hoboken) 2017;
melanoma without BRAF mutation. N Engl J Med 2015; 372:320–330. 69:1751–1763.

1350-7540 Copyright ß 2019 Wolters Kluwer Health, Inc. All rights reserved. www.co-neurology.com 509

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.


Widening spectrum of CNS inflammatory disorders of the CNS

46. Zimmer L, Goldinger SM, Hofmann L, et al. Neurological, respiratory, muscu- 67. Burke M, Hardesty M, Downs W. A case of severe encephalitis while on PD-1
loskeletal, cardiac and ocular side-effects of anti-PD-1 therapy. Eur J Cancer immunotherapy for recurrent clear cell ovarian cancer. Gynecol Oncol Rep
2016; 60:210–225. 2018; 24:51–53.
47. Sheik Ali S, Goddard AL, Luke JJ, et al. Drug-associated dermatomyositis 68. Kopecký J, Kubeček O, Geryk T, et al. Nivolumab induced encephalopathy in a
following ipilimumab therapy: a novel immune-mediated adverse event asso- man with metastatic renal cell cancer: a case report. J Med Case Rep 2018;
ciated with cytotoxic T-lymphocyte antigen 4 blockade. JAMA Dermatol 2015; 12:262.
151:195–199. 69. Shah S, Dunn-Pirio A, Luedke M, et al. Nivolumab-induced autoimmune
48. Hunter G, Voll C, Robinson CA. Autoimmune inflammatory myopathy after encephalitis in two patients with lung adenocarcinoma. Case Rep Neurol
treatment with ipilimumab. Can J Neurol Sci 2009; 36:518–520. Med 2018; 2018:2548528.
49. Yoshioka M, Kambe N, Yamamoto Y, et al. Case of respiratory discomfort due 70. Papadopoulos KP, Romero RS, Gonzalez G, et al. Anti-Hu-associated auto-
to myositis after administration of nivolumab. J Dermatol 2015; immune limbic encephalitis in a patient with PD-1 inhibitor-responsive myxoid
42:1008–1009. chondrosarcoma. Oncologist 2018; 23:118–120.
50. Khoja L, Maurice C, Chappell M, et al. Eosinophilic fasciitis and acute 71. Zurko J, Mehta A. Association of immune-mediated cerebellitis with immune
encephalopathy toxicity from pembrolizumab treatment of a patient with checkpoint inhibitor therapy. Mayo Clin Proc Innov Qual Outcomes 2018;
metastatic melanoma. Cancer Immunol Res 2016; 4:175–178. 2:74–77.
51. Touat M, Maisonobe T, Knauss S, et al. Immune checkpoint inhibitor-related 72. Läubli H, Hench J, Stanczak M, et al. Cerebral vasculitis mimicking intracranial
&& myositis and myocarditis in patients with cancer. Neurology 2018; metastatic progression of lung cancer during PD-1 blockade. J Immunother
91:e985–e994. Cancer 2017; 5:46.
The work highlights the importance of myositis as a complication of immune 73. Yshii LM, Hohlfeld R, Liblau RS. Inflammatory CNS disease caused by
checkpoint inhibitors. immune checkpoint inhibitors: status and perspectives. Nat Rev Neurol
52. Henderson AD, Thomas DA. A case report of orbital inflammatory syndrome 2017; 13:755–763.
secondary to ipilimumab. Ophthalmic Plast Reconstr Surg 2015; 74. Gerdes LA, Held K, Beltrán E, et al. CTLA4 as immunological checkpoint in
31:e68–e70. the development of multiple sclerosis. Ann Neurol 2016; 80:294–300.
53. Moreira A, Loquai C, Pföhler C, et al. Myositis and neuromuscular side-effects 75. Dalakas MC. Neurological complications of immune checkpoint inhibitors:
induced by immune checkpoint inhibitors. Eur J Cancer 2019; 106:12–23. what happens when you ‘take the brakes off’ the immune system. Ther Adv
54. Makarious D, Horwood K, Coward JIG. Myasthenia gravis: an emerging Neurol Disord 2018; 11:1756286418799864.
toxicity of immune checkpoint inhibitors. Eur J Cancer 2017; 82:128–136. 76. Gettings EJ, Hackett CT, Scott TF. Severe relapse in a multiple sclerosis
55. Suzuki S, Ishikawa N, Konoeda F, et al. Nivolumab-related myasthenia gravis patient associated with ipilimumab treatment of melanoma. Mult Scler 2015;
with myositis and myocarditis in Japan. Neurology 2017; 89:1127–1134. 21:670.
56. Supakornnumporn S, Katirji B. Guillain–Barré syndrome triggered by immune 77. Mohammadzadeh A, Rad IA, Ahmadi-Salmasi B. CTLA-4, PD-1 and TIM-3
checkpoint inhibitors: a case report and literature review. J Clin Neuromuscul expression predominantly downregulated in MS patients. J Neuroimmunol
Dis 2017; 19:80–83. 2018; 323:105–108.
57. Liao B, Shroff S, Kamiya-Matsuoka C, Tummala S. Atypical neurological 78. Dunn-Pirio AM, Shah S, Eckstein C. Neurosarcoidosis following immune
complications of ipilimumab therapy in patients with metastatic melanoma. checkpoint inhibition. Case Rep Oncol 2018; 11:521–526.
Neuro Oncol 2014; 16:589–593. 79. Weber JS, Yang JC, Atkins MB, Disis ML. Toxicities of immunotherapy for the
58. Bompaire F, Mateus C, Taillia H, et al. Severe meningo-radiculo-neuritis practitioner. J Clin Oncol 2015; 33:2092–2099.
associated with ipilimumab. Invest New Drugs 2012; 30:2407–2410. 80. Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell
59. Wilgenhof S, Neyns B. Anti-CTLA-4 antibody-induced Guillain–Barre syn- therapy – assessment and management of toxicities. Nat Rev Clin Oncol
drome in a melanoma patient. Ann Oncol 2011; 22:991–993. 2018; 15:47–62.
60. Thaipisuttikul I, Chapman P, Avila EK. Peripheral neuropathy associated with 81. Santomasso BD, Park JH, Salloum D, et al. Clinical and biological correlates
ipilimumab: a report of 2 cases. J Immunother 2015; 38:77–79. && of neurotoxicity associated with CAR T-cell therapy in patients with B-cell
61. Gu Y, Menzies AM, Long GV, et al. Immune mediated neuropathy following acute lymphoblastic leukemia. Cancer Discov 2018; 8:958–971.
checkpoint immunotherapy. J Clin Neurosci 2017; 45:14–17. The work highlights the neurological complications of chimeric antigen receptor T-
62. Larkin J, Chmielowski B, Lao CD, et al. Neurologic serious adverse events cell (CAR-T) therapies and correlations with biological markers.
associated with nivolumab plus ipilimumab or nivolumab alone in advanced 82. Torre M, Solomon IH, Sutherland CL, et al. Neuropathology of a case with fatal
melanoma, including a case series of encephalitis. Oncologist 2017; CAR T-cell-associated cerebral edema. J Neuropathol Exp Neurol 2018;
22:709–718. 77:877–882.
63. Altman AL, Golub JS, Pensak ML, Samy RN. Bilateral facial palsy following 83. Gust J, Hay KA, Hanafi LA, et al. Endothelial activation and blood–brain
ipilimumab infusion for melanoma. Otolaryngol Head Neck Surg 2015; barrier disruption in neurotoxicity after adoptive immunotherapy with CD19
153:894–895. CAR-T cells. Cancer Discov 2017; 7:1404–1419.
64. Faje A. Immunotherapy and hypophysitis: clinical presentation, treatment, and 84. Mackall CL, Miklos DB. CNS endothelial cell activation emerges as a driver
biologic insights. Pituitary 2016; 19:82–92. of CAR T cell-associated neurotoxicity. Cancer Discov 2017; 7:
65. Min L, Hodi FS, Giobbie-Hurder A, et al. Systemic high-dose corticosteroid 1371–1373.
treatment does not improve the outcome of ipilimumab-related hypophysitis: a 85. Lee DW, Santomasso BD, Locke FL, et al. ASBMT consensus grading for
retrospective cohort study. Clin Cancer Res 2015; 21: 749–755. && cytokine release syndrome and neurologic toxicity associated with immune
66. Reynolds KL, Guidon AC. Diagnosis and management of immune checkpoint effector cells. Biol Blood Marrow Transplant 2018. [Epub ahead of print]
inhibitor-associated neurologic toxicity: illustrative case and review of the The results of the American Society for Bone Marrow Transplant working group on
literature. Oncologist 2018. [Epub ahead of print] the management of irAEs of CAR-T therapy.

510 www.co-neurology.com Volume 32  Number 3  June 2019

Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.

You might also like