Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

Critical Reviews™ in Biomedical Engineering, 38(1):1–20 (2010)

Models for Thermal Damage in Tissues:


Processes and Applications
John A. Pearce, PhD*
Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, TX

*Address all correspondence to John A. Pearce, Department of Electrical and Computer Engineering, The University of Texas at Austin, 1 University Station,
Austin, TX 78712; Tel.: 512-471-4984; Fax: 512-471-6964; jpearce@mail.utexas.edu.

Abstract: Irreversible thermal alterations in tissue function and structure are used in clinical applications to
achieve diverse goals, from lower-temperature tumor ablation to higher-temperature tissue fusion and surgical cut-
ting. The typical formulation in tumor hyperthermia studies, the thermal iso-effect dose, derives from cell-survival
studies but describes a single process only over a limited range of temperatures and is thus not suitable for multiple
higher-temperature events. Many other thermal damage processes have been described using the Arrhenius kinetic
rate of formation approach, which has the advantage that it is inherently quantitative in nature and can easily be
compared with quantitative markers of injury or histologic section. The vast majority of Arrhenius studies have been
directed toward measurable cellular effects at relatively low temperatures. Some emphasis in this paper has been
placed on what is known of higher-temperature processes to support the theme of this issue. This review compares
and contrasts the two thermal-damage formulations and reviews methods to convert between them.

Key words: ablation, thermal damage, thermal iso-effect dose, cumulative equivalent minutes, Arrhenius models

I. Introduction Diathermy, therapeutic heating at low temperatures


(between about 40°C and 45°C), is designed to achieve
Heat has been used therapeutically for millennia. analgesia, increase cellular metabolism, and stimulate
Major1 reports that Neolithic skulls unearthed in local perfusion below thermal-damage thresholds.
France show clear evidence of thermal cauterization. The increased perfusion facilitates healing processes
The Edwin Smith papyrus (ca. 3000 BCE)2 describes by increasing interstitial oxygen partial pressures, tis-
the use of thermal cautery for ulcers and tumors of the sue nutrients, antibodies, leukocytes, and phagocytes.
breast. According to Thevenet,3 French physician Guy Benefits result from the rapid clearing of metabolites
de Chaulliac (1300–1370) is one of the first surgeons and mechanical damage debris (as from a sprain).6 Cur-
to recommend “operating on hernias using golden rently, therapeutic heating from a multitude of sources
thread rather than cauterization.” According to Kelly is frequently used to promote healing in diathermy, to
and Ward,4 Nagelschmidt applied diathermy in the initiate apoptosis and necrosis in tumor hyperthermia
treatment of articular and circulatory disease in 1897. therapy; to kill tumors and ectopic foci in myocardium
Additionally, they report that Jules de Riviére demon- by ablation; to coagulate blood vessels, fuse tissues,
strated “white” coagulation (i.e., without sparking and and shrink collagen; and to achieve surgical cutting.
carbonization) before the French Surgical Congress Modeling these processes remains a challenging, but
in 1907—and that Doyen found temperatures in the important, avenue of research; the processes are kinetic
range of 65°C to 70°C for white coagulation. Doyen5 in nature, and models of them provide necessary deeper
also applied bipolar radiofrequency (RF) current to understanding than can be obtained from predictions
superficial tumors after the method of Pozzi. of temperature alone.

ABBREVIATIONS

CEM, cumulative equivalent minutes; CHO, Chinese hamster ovary; RF, radiofrequency; TID, thermal iso-effect dose.

11040-8401/10/$35.00  © 2010 by Begell House, Inc. 1


2 Pearce

I.A. Thermal Iso-effect Dose Concept where A is the “frequency factor” (s–1), Ea is an energy
barrier ( J mole–1), R is the gas constant (8.3143 J mole–1
In tumor hyperthermia therapy, the typically applied K–1), T is temperature (K), and τ is total experiment
assessment of clinical effectiveness is the “thermal iso- time (s). A and Ea characterize the thermal damage
effect dose” (TID), measured in units of cumulative
process and must be determined experimentally. An hour
equivalent minutes (CEM) at 43°C, as described by
of heating yields very different results from seconds at
Sapareto7 and adapted from the original work reported
the same temperature: temperatures that create thermal
in 1978 by Sapareto and Dewey.8 A successful hyper-
burns during long exposures may be well tolerated for
thermia treatment is generally held to require a thermal
short times. Consequently, it is not instructive to report
dose of 60 CEM, calculated from:
thermal-damage thresholds in terms of temperature
alone; the time of exposure is equally important and
(1) must be included whenever thermal damage is discussed.
Additionally, inspection of the functional nature of
where RCEM ≈ 0.5 for T > 43°C (the breakpoint Equation 2 suggests that damage accumulates at all
temperature) and 0.25 below 43°C; Ti is the constant temperatures above absolute zero. While this is implied
temperature (°C) for time ti (min); and N is the number
mathematically, in practice, no damage-process model
of epochs in the treatment. The method has been very
is valid below the lowest temperature at which the
widely applied since its introduction.9–11
process results have been observed.
Ultimately, the study of thermal damage reduces
I.B. Arrhenius Thermal Damage Models. to determining process kinetic parameters (activation
Arrhenius rate-of-reaction kinetic models describe the energy and frequency factor). This review describes the
evolution of thermal damage, and the rate of damage application of the two typically used forms of thermal-
accumulation is temperature dependent. Arrhenius damage models, the TID method and Arrhenius
models derive from standard physical-chemical ther- formulations, in models of particular processes and
modynamics typically used to predict reaction-product applications. These processes have been extensively
formation. The kinetic nature of thermal-damage devel- studied at moderate temperatures in cell culture, a very
opment is important to understanding observed results. important avenue of research. An elegant and thorough
For example, excessive longer-term heating, even in the survey of this work may be found in the comprehensive
relatively benign mid-40s Celsius, for greater than tens analysis by He and Bischof,13 which is filled with many
of minutes can result in edema as the gaps between pertinent and incisive observations of both heating and
endothelial cells widen to allow plasma migration into cryogenic modalities—one of many exemplary publica-
the interstitial space.6 Edema is not usually observed as tions from this group and its alumni. The emphasis in
a thermal-damage mechanism in short-term heating the current paper is on higher-temperature applications
(tens of seconds or less). Whether thermal damage for ablation, tissue fusion, and surgical cutting, about
will be observed depends on both the temperature which there have been considerably fewer studies and
and the time of exposure; the processes are kinetic in therefore much less is known.
nature, meaning that they are rate-limited. The kinetic
nature of thermal-damage development is often lost
II. THERMAL ISO-EFFECT DOSE:
in discussions of “threshold” temperatures but should
Cumulative Equivalent Minutes
not be overlooked.
The first application of Arrhenius rate-of-formation TID is derived from studies of the thermal sensitivity
kinetic models to thermal burns was by Henriques and of tumor cell lines in vitro, specifically from a standard
Moritz in 1947,12 who formulated the prediction in tumor cell line, Chinese hamster ovary (CHO) cells,
terms of a dimensionless damage parameter, Ω: but the method turns out to be equally applicable to
other cell lines as well.7 The basis for the measure is
(2) the ability of the cell line to continue to form colonies
as the temperature increases.

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 3

The thermal sensitivity of CHO cells was deter-


mined in vitro and is depicted in Figure 1. The slope of
the cell-survival curve (Fig. 1A) during the constant-rate
segment (i.e., where the process is first order) deter-
mines the cell-survival-time constant, D0, for the plot
in Figure 1B. Deviation from first-order behavior is
often observed when the cell count is very low, as can
be seen in the longer-duration segments in the figure.
Note that there is a prominent breakpoint (i.e., change
of slope) at 43°C in Figure 1B.

II.A. Foundations of the TID Method


The time constant of cell deactivation at constant
temperature, D0 (min), describes the cell-surviving
fraction, S/N0 , after exposure at constant temperature,
T, for (t – t0) min:

(3)

where N0 viable cells are counted at t0. From survival


curves, the “reaction rate,” k = dΩ/dt, is related to the
parameters of Equation 2 by:

(4)

Figure 1B is a plot of k = 1/D0 vs T–1 (K–1), with T


(°C) superposed (adapted from the original publica-
tion8). There is a prominent change in slope of the
Figure 1B plot (a breakpoint) at 43°C. For tem-
peratures above 43°C, the fit parameters are: ln{A} =
229.0 (i.e., A = 2.84 × 1099 s–1) and Ea = 6.18 × 105 ( J
mole–1), calculated from Equation 4 by least-squares
fit to the data in Figure 1B.
The model parameter, RCEM, signifies the rate at FIGURE 1. (A) Cell survival (colony formation rate) curves
as a function of temperature for asynchronous CHO cells
which the process accumulates. That is, RCEM ≈ 0.5
in culture. (B) Arrhenius plot of inactivation inverse time
means that an exposure of 1 min at 44°C is equivalent constants (1/D0) from Sapareto et al., 19788 for both
to an exposure of 2 min at 43°C. For time-varying asynchronous and G1-phase cells.
temperatures, the CEM43 criterion is calculated from:

(5) II.B. Limitations of the TID Method


and Tbreak is the breakpoint temperature. Different tis- Although this measure of thermal dose is in widespread
sues have different thermal sensitivities; for example, use in tumor-hyperthermia studies, it suffers from
tissues of the central nervous system are known to be several limitations. First, although derived from an
generally more thermally sensitive than those of the Arrhenius model, it constitutes a comparative rather
rest of the body.14 than predictive parameter: that is, it normalizes time-

Volume 38, Number 1, 2010


4 Pearce

varying thermal histories so that different treatments


can be compared on a common basis. TID does this
very well, but it does not predict a particular tissue effect
and is therefore not very useful for the wide range of
thermal alterations observed in tissues, especially at that
elevated temperatures required for ablation, tissue fusion,
and surgical cutting (50°C to 150°C). Second, TID
provides no information directly applicable to ablation
studies; collagen, for example, can be exposed to 43°C FIGURE 2. Native-state molecules surmount an energy
for extremely long time intervals without showing any barrier, ΔH* (J mole–1), at overall reaction speed, k (s–1),
sign of thermal alteration. Third, the TID method can- to form denatured protein products.16
not be compared to quantitative means of assessment
such as a damage fraction in histologic section. Fourth, III.A. Foundations of the Arrhenius Method
TID is not in the form of a probabilistic model and
cannot be compared to ensembles of experiments. A The kinetic model of Equation 2 derives originally from
quantitative model that predicts the degree of damage, a uni-molecular reaction formulation typical in physical
or a probabilistic formulation, would be more generally chemistry texts; for example,15 when a concentration of
useful. The former can be directly compared to, for native-state molecules, C, surmount an energy barrier,
example, measured fluorescence intensity or to observed ∆H* ( J mole–1), to become “activated” (in some sense),
which is expressed as C*, and may either relax back
damage concentrations in histologic sections. The latter
to the inactivated original state or progress to, in our
can be compared to the frequency of observation in an
case, a thermally denatured state: ∆H* = Ea - RT ≈
ensemble of experiments in which the degree of dam-
Ea for first-order processes. The specific definition of
age is not measurable, but the frequency of occurrence
“concentration” is left to the particular investigator. ∆H*
is observably temperature dependent.
relates to the energy barrier, not the overall reaction
energy, ∆H ( J mole–1), as illustrated in Figure 2.
III. VOLUME FRACTION Arrhenius
Formulation 1. Physical Chemical Process Parameters
The original skin-burn studies of Henriques and In the uni-molecular description, the reaction speed, k
Moritz12 employed calculations of the non-dimensional (s–1), is the Arrhenius kernel:
parameter Ω (Equation 2), and classified relative burn
severity only in terms of Ω at the skin surface. For (6)
example, Ω = 0.53 was assigned to a "first-degree"
where C is the remaining unaltered concentration, and
burn (i.e., superficial irreversible erythema), Ω = 1.0
∆G* is the Gibbs free energy of activation. ∆G* includes
to a "second-degree" burn (i.e., partial thickness, the activation entropy, ∆S*, and enthalpy, ∆H*: ∆G* =
complete trans-epidermal necrosis), and Ω = 104 to a ∆H* – T ∆S*.
“third-degree” or full-thickness burn (i.e., completely In a few steps one can obtain the correspondences
involving the dermis as well). This calculation can be (for ∆S*/R >> 1):
used much more effectively. By looking at its origins in
reaction-product formation, it is obvious that a quan- (7)
titative and/or probabilistic model can be formulated
and is substantially more useful than the calculation of where N is Avogadro’s number (6.023 × 1023) and hP
Ω alone. It is much too limiting, and unnecessary, to is Planck’s constant (6.63 × 10–34 J s), from which, A
apply only one set of coefficients to describe multiple can be calculated by:
thermodynamically independent kinetic processes acting
in parallel, such as occur in complex tissue structures (8)
at substantially elevated temperatures.

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 5

At first glance, it looks as though A should be histology or a probabilistic prediction because the
treated as temperature dependent; however, T is in K damage process is plainly saturated.
here, and the temperature varies so little over a typical
thermal-damage experiment temperature range (com- III.B. Example Thermal-Damage Processes
pared with the activation enthalpy term) as to be of
little importance in the total calculation. Thermal-damage processes that are amenable to quan-
titative or probabilistic analysis generally fall into three
2. Comparative-Process Parameters categories: i) quantitatively measurable as the thermal
change develops, such as fluorescence intensity; ii)
Because the physical significance of the process param- quantitatively measurable, but only at the conclusion of
eters is not immediately obvious, we introduce three the exposure, such as by enzyme histochemistry or in
additional descriptors. The first of these is the “critical histologic section; or iii) not quantitatively measurable
temperature,” Tcrit, the temperature at which the damage necessarily, but identifiable by frequency of occurrence,
rate (reaction speed) dΩ/dt = k = 1 (as in Equations 2 such as the occurrence of pyknotic (shrunken and
and 7); the second is the (time-dependent) “threshold shriveled) nuclei.
temperature,” T TH, where Ω = 1 (or a specific reference
value, Ωref ); and the third is the time for the process to 1. Apoptosis
achieve Ω = 1 at a constant temperature of, say, 43°C,
τ43 (only useful at low temperature): Apoptosis, or programmed cell death, is a complex
biochemical cascade of protein alterations that even-
(9a) tually result in the death of cells in multicellular
organisms—a form of cellular suicide. It is presently
under intense investigation because of its far-reaching
implications in cancer (e.g., tumor necrosis factor and
the like),17 the treatment of disease,18 and the immune
(9b) response.19 Although not very well understood at this
point, programmed cell death is one hypothesis for the
effectiveness of tumor-hyperthermia therapy. Markers of
programmed cell death include morphological changes
such as “blebbing” (irregular buds in the cell membrane),
(9c) cell shrinkage, nuclear disruption (pyknosis), and chro-
matin condensation, among other changes.20
In Equation 9c: 2.629 × 103 = R × 316.2 (K), i.e. Apoptosis occurs naturally, for example, in tissue-
(8.3143)(273.16 + 43°C). With the Arrhenius formula- differentiation processes during embryonic develop-
tion, it is relatively straightforward to recast the damage ment,21 as an intrinsic response to extrinsic signals such
model in quantitative terms: as heat and radiation,22 and to intrinsic signals such as
viral infection.22 There are several applicable staining
techniques to identify apoptosis.23 Apoptosis is differ-
(10) entiated from necrosis in that necrosis is traumatic cell
death due to acute injury (e.g., membrane disruption
or disruption of mitochondria), not cell death due to
where P is either the damage fraction, 1 - C(τ)/C(0) triggering of internal biochemical mechanisms.
or the probability of observing tissue damage (%), Bhowmick et al.24 determined Arrhenius param-
depending on how the damage process has been eters for the combination of necrosis and apoptosis
measured. Consequently, Ω = 1 is well above the in vitro in tissue sections of human prostate: A =
"threshold": the damage process is 63.2% complete at 7.78 × 1022 (s–1), Ea = 1.61 × 105 ( J mole–1), Tcrit =
that point. Also, values of Ω above about 5 (i.e., P = 94.2°C, and τ43 = 5.11 × 103 s (85.3 min). The low
99.3%) are useless markers for either comparison with values of A and Ea coupled with the high Tcrit indicate

Volume 38, Number 1, 2010


6 Pearce

a relatively “slow” damage process, one that is more SN12 human renal carcinoma cells to heating rates up
likely to be observed in long-term heating at lower to 130°C min–1, with varying hold times between 0 and
temperatures, rather than in short-term, higher-tem- 10 min at hold temperatures between 45°C and 70°C,
perature heating. Borrelli et al.25 studied cell death in followed by a 65°C min–1 cooling sequence. The results
BhK cells and reported that A = 2.984 × 1080 (s–1), Ea for suspended cells differed significantly from those for
= 5.064 × 105 ( J mole–1), Tcrit = 55.5°C, and τ43 = 1.51 attached cells (Table 1) because of the effects of local
× 103 s (25.8 min). stress, suggesting that cellular attachment thermally
Of course, moderate heating also signals other stabilizes the cells.
cellular responses, complicating the analysis of tumor The dorsal skin flap is a standard construct that
responses. Some genes are up-regulated while others are allows continuous close observation of skin vasculature
down-regulated, and other heat-sensitive mechanisms in vivo without anesthesia (once implanted) in diverse
include the cell cycle and DNA repair.26 Moderate small animal species.41 The skin is opened, elevated, and
heating also induces cellular-protective mechanisms. clamped between plates supporting removable windows
Cells respond to hyperthermia by expressing heat-shock and allowed to heal. This fixture has been used success-
proteins, chaperone molecules that assist heat-denatured fully to study diverse vascular phenomena, including
proteins in refolding into their native morphology.27-33 thermal alterations.23,38,42–44 The thermal breakdown
Beckham et al.33 studied the role of the heat-shock of red blood cells, hemolysis, another primary skin-
protein Hsp70 in murine embryonic fibroblasts in vitro burn response, has also been studied with substantial
using bioluminescent imaging, and determined the dif- success.45
ference in thermotolerance between cells with intact Vital stains are used without fixation to identify
Hsp70 production and those in which Hsp70 produc- viable cells. There are many such stains; a partial list
tion was blocked. For Hsp-deficient cells pre-shocked includes: trypan blue, vital red, neutral red, Nile blue,
(i.e., heated again 4 h later), A = 6.9 × 10116 (s–1), Ea methylene blue, acrydine orange, Bismarck brown, and
= 7.3 × 105 ( J mole–1), Tcrit = 53.2°C, and τ43 = 5.87 × Janus green. Janus green B has been used to study mito-
103 s (97.9 min). Hsp70 increases thermotolerance in chondria in the cornea46 and elsewhere. Indocyanine
similarly reheated cells to A = 3.7 × 10157 (s–1), Ea = 9.8 green is another common vital stain (and laser chro-
× 105 ( J mole–1), Tcrit = 51.7°C, and τ43 = 22.1 × 103 s mophore), but its use in retinal surgery has recently come
(368 min). Thus, Hsp70 makes an effective marker of under scrutiny due to possible toxic effects.47 Another
thermal insult that can be imaged.34–36 interesting supravital stain is nitro blue tetrazolium,
which was used by Acosta and Wendel in 1975,48 by
2. Fluorescent and Vital Stain Markers Feldman et al.49 and Lucchesi et al. in 1976,50 and by
many others since, to study myocardial cell function.
Fluorescent markers have been used for several decades Nitro blue tetrazolium stains normal, metabolically
to indicate thermal processes as they develop. In an active tissue a dark purple/brown color.
early experiment series, Green and Diller37 measured In a follow-up to their earlier 2000 study,39 Bhow-
increased macromolecular leakage from capillary vessels mick et al.51 used a clonogenic dye regimen for the
in the hamster cheek pouch in vivo using fluorescein analysis of thermal damage in AT-1 cells: Hoechst 3342
isothiocyanate conjugated dextran. Heating increased is a fluorescent green stain for all cell nuclei (regardless
the capillary inter-endothelial cell gaps, resulting in of viability), and ethidium homodimer dye-1 stains red
increased migration of the dye-tagged dextran into the nuclei that have a compromised plasma membrane.
the interstitial space. Aggarwal et al.38 applied digital The total number of cells in an image are identified
image processing techniques to measure the diffusion by the green fluorescent dye, and the relative number
rates of the fluorescence-labeled dextran in a dorsal of killed cells with red nuclei are separately counted,
skin flap preparation. Bhowmick et al.39 measured with the ratio constituting a direct measure of P = [1 –
the loss of fluorescence in Dunning AT-1 prostate exp(–Ω)] for Arrhenius analysis. In this AT-1 cell study,
tumor cells in vitro heated at rates of 2°C and 5°C below the 50°C breakpoint, A = 1.66 × 1091 (s–1), Ea =
per minute to temperatures between 40°C and 70°C. 5.68 × 105 ( J mole–1), Tcrit = 52.1°C, and τ43 = 420 s (7
In an analogous preparation, He and Bischof40 exposed min); above the 50°C breakpoint, A = 173.5 (s–1), Ea =

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 7

Table 1. Collected Representative Arrhenius Kinetic Coefficients

Process Parameters
A Ea Tcrit τ43
Process (s–1) (J mol–1) (°C) (s) Notes
Cell Death
Sapareto8 2.84 × 1099 6.18 × 105 51.4 447 CHO cells, T > 43°C
Beckham33 6.9 × 10116 7.3 × 105 53.2 5.87 × 103 Murine w/o Hsp
3.7 × 10157 9.8 × 105 51.7 22.1 × 103 Murine w/ Hsp
Bhowmick24 7.78 × 1022 1.61 × 105 94.2 5.11 × 103 H. Prostate
Bhowmick 51 1.66 × 10 91 5.68 × 10 5 52.1 420 AT-1 Cells < 50°C
173.5 1.97 × 104 186 AT-1 Cells > 50°C
Borrelli25 2.984 × 1080 5.064 × 105 55.5 1.51 × 103 BhK Cells
He40 4.362 × 1043 2.875 × 105 71.0 SN12 cells, suspended
3.153 × 1047 3.149 × 105 73.1 SN12 cells, attached
Erythrocytes
Lepock45 7.6 × 1066 4.55 × 105 82.2 Hemolysis
Skin Burns
Henriques12 3.1 × 1098 6.28 × 105 59.9 Not recommended
Diller 60 8.82 × 10 94 6.03 × 105 58.6 T ≤ 53°C (same data)
1.297 × 1031 2.04 × 105 69.3 T > 53°C
Weaver61 2.19 × 10124 7.82 × 105 55.4 T ≤ 50°C
1.82 × 1051 3.27 × 105 60.1 T > 50°C
Brown62 1.98 × 10106 6.67 × 105 54.6 8.04 × 103 Microvessels
Retinal Damage
Welch134 3.1 × 1099 6.28 × 105 56.6 Whitening
Collagen Changes
Maitland78 1.77 × 1056 3.676 × 105 68.2 Rat tail birefringence
Pearce74 1.61 × 1045 3.06 × 105 80.4 Rat skin birefringence
Aksan102 1.136 × 1086 5.623 × 105 68.1 Rabbit patellar tendon
Miles103 Rat tail tendon, differential scanning
6.658 × 1079 5.21 × 105 67.8
calorimetry
Chen99–101 1.46 × 1066 4.428 × 105 76.4 Shrinkage, rat tail tendon
Muscle
Jacques68 2.94 × 1039 2.596 × 105 70.4 Myocardium whitening
Liver
Jacques135 5.51 × 1041 2.769 × 105 73.4 Whitening, pig liver

1.97 × 104 ( J mole–1), and Tcrit = 186°C—the latter was 3. Skin Burns and Vascular Disruption
a somewhat surprising result relative to other studies
of thermal damage, which nevertheless plots well on Most of the damage mechanisms discussed to this point
Wright’s line in Figure 3. have typically been studied in colonies of single-cell
The above remain active and important areas preparations. Tissues and organs are composed of diverse
of research, but in this issue we are more interested populations of cells with varying degrees of sensitivity
in thermal-damage processes and mechanisms that and are surrounded by supporting structures, which
completely overwhelm all subtle thermal changes often makes a difference in their response to thermal
and protective mechanisms and result in substantial insult. In contrast to single cells, reversible thermal
structural and functional alterations to tissue. injury in tissues and organs usually involves some level

Volume 38, Number 1, 2010


8 Pearce

(Table 1). Breakpoints in Arrhenius plots are indications


that a different thermal process has taken over.
The primary mechanism of lower-temperature-
range thermal damage in skin burns is the disruption
of vasculature in the dermis and in the subpapillary and
subdermal vascular plexuses. In a related process, Brown
et al.62 reported coefficients for the disruption of tumor
microvasculature of A = 1.98 × 10106 (s–1), Ea = 6.67 ×
105 ( J mole–1), Tcrit = 54.6°C, and τ43 = 134 min.

4. Ablation
The term “ablation,” from the Latin “ablatus,” the
FIGURE 3. Wright’s line126 plot of the Arrhenius coef- past participle of “auffere” (to carry away),63 is used
ficients from Table 1 (Equation 23). in several different contexts. Surgical ablation means
excision or extirpation. Similarly, in general engineering
terminology, ablation usually implies the creation of a
of cell death; however, the overall function may not be mass defect, as in ablative heat transfer (aerospace) or
impaired significantly.52 Minutes to hours after heating, glacial erosion (earth science). In clinical ablation—
edema and hyperemia appear due to the release of vaso- cardiac ablation, tumor ablation, endometrial ablation,
active agents, increasing the gaps between endothelial and like processes—the goal is frequently to inactivate
cells, and repair begins a few days later.53 More severe the tissue in situ, leaving it in place to form scar tissue
heating results in irreversible vascular disruption and (i.e., to remove it from the inventory of physiologically
consequent progression of tissue death “downstream” active tissue). The US Food and Drug Administration
due to ischemia, hypoxia, and other conditions. Novel indexes approximately 11,000 articles related to clinical-
methods for assessing skin-burn depth using optical ablation devices,64 a testament to the importance of
these clinical applications.
coherence tomography imaging were described by
Collectively, thermal damage from cryogenic, RF,
Srinivas et al.54 and Pierce et al.55
microwave, ultrasound, or laser sources is applied to
As mentioned in the introduction, skin burns were
specific sites to achieve ablation. Interstitial applicators
the first kinetic thermal damage mechanism studied in
are typically used, and the usual goal is an ablative lesion
detail.12,56–58 The kinetic coefficients reported by Hen-
larger in dimension than the applicator. He and Bischof13
riques and Moritz,12 A = 3.1 × 1098 (s–1), Ea = 6.28 ×
provide a thorough description of cryogenic ablation.
105 ( J mole–1), Tcrit = 59.9°C, and τ43 = 1.81 × 105 s (> Here, we look at elevated temperature ranges.
3 × 103 min, 2.09 d), have been widely used since but One or more of the previously described moderate-
actually do not fit their original data set very well and temperature mechanisms are only likely to be observed
are not recommended. In 1993, Diller and Klutke59 took at the edges of the lesion. The essential feature of an
another look at the original data (up to the 50°C point), ablation lesion is that cellular protective and repair
and if the datum at 52°C is included, as in the followup mechanisms, such as the heat-shock proteins, are
publication,60 one obtains: A = 8.82 × 1094 (s–1), Ea = essentially irrelevant due to the level and rate of heat-
6.03 × 105 ( J mole–1), Tcrit = 58.6°C, and τ43 = 4.78 × ing applied for the bulk of the lesion volume (although
104 s (13.3 h). Above 53°C, an obvious breakpoint, they may effectively mark the lesion’s outer boundary).
the Henriques and Moritz data are fit by: A =1.297 × A typical severe acute lesion consists of a hemorrhagic
1031 (s–1), Ea = 2.04 × 105 ( J mole–1), and Tcrit = 69.3°C outer ring due to vascular disruption, a coagulative
(τ43 = 406 s, but this is irrelevant at these temperatures). heat-fixed “white” necrotic annulus, and often an inner
The improved coefficients are relatively close to the val- carbonized disk that may include steam vacuoles or
ues obtained by Weaver and Stoll in thermal-radiation craters.52,65,66 The following higher-temperature thermal
skin burns,61 who also observed a breakpoint, but at 50°C damage mechanisms may all be at work in the central

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 9

region of ablation lesions, while the cellular damage anatomy is significantly different from that of human
processes may be observed at their periphery. and pig skin.79

5. Birefringence Loss in Muscle and Collagen 6. Collagen Denaturation and Shrinkage


Native-state muscle, cardiac, skeletal, and smooth, is Extensive thermal damage in collagen results in
“birefringent,” or able to rotate the polarization of an amorphous glassified or gelled state, “hyaliniza-
visible light. This is the source of the histologically tion.”52,66,76 Achieving a hyalinized, gelled state is
identifiable “A-band” (anisotropic band) in the sarcom- essential in tissue-fusion processes.80-83
ere of skeletal and cardiac muscle. Thermally damaged Less-severe thermal alteration shrinks collagen
muscle loses this property due to disruption in the fibers in length by a helix-to-coil transformation.86
actin-myosin array, a marker of substantial structural The original studies of collagen shrinkage were in the
and functional damage in the sarcomere.67 As a result, context of leather tanning processes,87 and have been
easily identifiable boundaries of thermal damage and intensively studied since. As the fibers shrink in length,
gradation in relative intensity directly comparable to they swell, maintaining an approximately constant
C(τ)/C(0) can be identified in histologic section through volume in a visco-elastic creep process.88 Targeted col-
transmission polarizing microscopy (Fig. 4a). In a lagen shrinkage has many practical clinical applications,
good section, one can measure the relative intensity from corneal reshaping83–85,89-91 to cosmetic treatment
of the birefringence signal, which makes a quantitative for wrinkles.92-98
thermal damage assay amenable to damage-fraction Chen et al.99–101 have developed a kinetic model
calculation. The zone of birefringence loss corresponds that predicts collagen shrinkage based on a series
approximately to the grossly observable whitened zone of experiments in rat tail tendons. The correlations
in an acute myocardial lesions, for which Jacques and normalize all of their data to include the influence
Gaeeni provide an estimate of kinetic parameters of applied stress during shrinkage, indicated by the
(Table 1).68 normalized shrinkage time, τ2:
Cardiac ablation is used as a palliative treatment
(11)
for several types of atrial and ventricular fibrillation
induced by ectopic foci, although it is most practical where α is –ln{A} = –152.35, β = 0.0109 (kPa ), P is
–1

for procedures in the atria.69,70 The most common applied stress (kPa), M = Ea/R = 5.3256 × 104 (K);
heating method is by RF current,69 although micro- that is, A = 1.46 × 1066 (s–1) and Ea = 4.428 × 105 ( J
wave and ultrasound are also feasible. Certainly, by mole–1). The signs are reversed in Equation (11) because
the time birefringence is lost in cardiac muscle, it has τ2 effectively appears in the denominator of a shrinkage
been damaged to the point of complete dysfunction. calculation. In use, the effective τ2 is integrated (i.e.,
However, cardiac muscle may be irreversibly damaged ∆t/τ2), and at the conclusion of heating the normal-
at less-severe heating levels. Cardiac muscle has a sub- ized time, ν = ln{t/τ2}, is fit to the data to determine
stantial electrical permittivity due to the transmembrane the shrinkage:
charge distribution that is substantially reduced when
it collapses.71,72 Changes in permittivity may also prove
(12)
to be a useful measure of cardiac ablation success, but
this has not been studied to date.
Collagen is similar in that the regular structure where ξ is the collagen shrinkage (%), νm = –0.77 ±
of its rope-like fibers (Fig. 4b) is also birefringent, 0.26, a normalized reference time, a = 2.48 ± 0.438,
and this birefringence is lost when the collagen is a0 = 1.80 ± 2.25, a1 = 0.983 ± 0.937, b0 = 42.4 ± 2.94,
thermally damaged.52,67,74–77 Arrhenius coefficients for and b1 = 3.17 ± 0.47 (all in %). A predicted shrink-
birefringence loss in collagen may be found in Pearce age in excess of about 60% signifies gelled amorphous
et al.74 and Maitland et al.75,78 Collagen damage is (hyalinized) collagen, so the model can be used for
another central feature of skin burns; most animal skin multiple purposes. After shrinkage (in the absence of

Volume 38, Number 1, 2010


10 Pearce

FIGURE 4. a) Birefringence loss in rab-


bit cardiac muscle due to argon laser
irradiation; Mallory’s trichrome stain,
original magnification ×40. b) Sketch of
collagen fibril morphology from Pearce
200673 (used with permission).

gellification) and during cooling, the collagen fibers ily absorbed by tissue water, such as for Tm:YAG (λ
relax to slightly longer lengths.86, 99–101 = 2.01 µm), Ho:YAG (2.09 µm), Er:YAG (2.94 µm),
Other models for collagen denaturation include and CO2 (10.6 µm) lasers.
those applied by Aksan et al.86,102 and Miles et al.103,104 At higher temperatures and heating rates, such as
based on coefficients derived from differential scanning for surgical cutting, steam formation is almost always
calorimetry, a very powerful experimental method.13 a dominant thermodynamic mechanism; tissue water is
Figure 5 illustrates collagen shrinkage in the cornea. by far the most thermodynamically active constituent
in tissue. Laser and RF electrosurgical cutting processes
7. Vaporization and Steam Formation depend on rapid steam evolution to separate tissue
layers with minimal radiating thermal damage.4,108–110
Evaporation of tissue water from the surface makes The thermodynamics are intricate: at only a few degrees
a measurable contribution to the overall surface heat of superheat (above the saturation boiling temperature
transfer above approximately 60°C.105 Torres et al. suc- at local pressure), the steam evolution rate increases
cessfully applied experimentally derived evaporation extremely rapidly, and there are numerous steam-bubble
rate correlations,106 Stelling’s formula, to predict the nucleation centers in all tissue structures. Consequently,
temperature history of continuous-wave argon laser as a practical matter in relatively “open” tissue structures
irradiation (514 nm) in air on aorta, beef myocardium, (i.e., not highly compartmentalized), one may often
and polyacrylamide gel. Stelling’s formula is: treat the boiling process as essentially isothermal at the
saturation temperature.52 In compartmentalized tissues
(13) such as large arteries, as steam evolves the local pressure
where ζ is surface water loss rate (m s ), As = 7.31 ×
–1
rises: at 100°C, the specific volume of saturated steam is
10–11 (m Pa–1 s–1), Bs = 1.2 × 10–11 (Pa–1), u is free more than 1.3 × 103 times that of the saturated-liquid
stream velocity above the surface (m s–1), Ps is saturation phase.111 Below the surface, evolved steam raises the
pressure (Pa) at the surface temperature, T, and at the local pressure, which in turn raises the boiling-point
environmental temperature and relative humidity, Tenv temperature.52,112 Figure 6A illustrates such an event, in
and RH, respectively. Stelling’s formula was originally which a temperature of 175°C corresponds to a satura-
derived from solar-pond data, but provides a useful and tion pressure of 10 atmospheres, which is the energy
easily implemented construct for numerical model work. source that drives the explosion plume.
Surface-water evaporation is even more important to Including steam evolution in the energy balance
the calculations when the laser wavelength is primar- reveals some of the intricacy. The development summa-

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 11

( J kg–1). This formulation includes phase change in


both the left and right sides; it applies to the liquid-
phase water-tissue composite and can be inserted into
numerical models by employing the following correla-
tions for saturation pressure and density, derived from
steam tables:111

(15)

where ∆hfg is in J kg–1 and T is in °C. The liquid-phase


saturation pressure is fit by (r2 = 1.000):

(16)

where Psat is in kPa (kN m–2) and T is in °C. Similarly,


the liquid-phase density is (r2 = 0.998):

(17)

where ρsat is in kg m–3 and, T is in °C. From these cor-


relations, it is a simple matter to derive temperature-
dependent expressions for ∂P/∂T and ∂ρ/∂T. Ordinarily,
including the expansion term ∂ρ/∂T would not be
FIGURE 5. Application of collagen shrinkage. a) Corneal important in free surface water, but it is the source of
collagen heated by a RF needle electrode; original mag- energy driving a debris plume and so is important in
nification ×25; bar = 0.1 mm, H&E stain. b) Transmission
confined spaces such as cells and tissue structures such
polarizing microscopy view of the same section; birefrin-
gence loss near needle electrode is evident, as is “straight- as vessel walls, as shown in Figure 6A. An additional
ening” of collagen fibers just outside of the birefringence energy balance similar to Equation 14 is required for
loss zone, indicating shrinkage. From Pearce 200184 and the generated gas phase as well (coupled with a suitable
Pearce and Ikei 200785 (used with permission).
state equation because of its compressibility); however,
rized here is more completely described in the chapters none has been published to date because of a shortage
by Pearce and Thomsen.52,113 Briefly, the left side of of suitable biomechanical parameters for the migration
the energy balance (net rate of energy deposition) is of vapor phase through tissues.
modified to include expansion and phase-change effects The embedded lesson here is important. Bipolar
(metabolic and perfusion heat are truly negligible in electrosurgical sealing of large vessels was not practical
these processes): for many years because evolved steam disrupted the
fusion process before reliable fusion of the collagen/
elastin matrix could be achieved (Fig. 6B). The Ligasure®
(14) system (Covidien, Mansfield, MA) employs substan-
tially elevated tissue pressures to raise the equilibrium
boiling point such that vessel fusion temperatures can
where ρ is density (kg m–3), h is specific enthalpy readily exceed 100°C, as required for a successful and
( J kg–1), P is liquid pressure (Pa), Qgen is the volume repeatable result.114
power deposition (W m–3), k is thermal conductivity Recently, Yang et al.115 suggested using a modified
(W m–1 K–1), ∂m/∂t is the rate of mass vaporization specific heat, C’, on the left side and adding the den-
of water per unit volume (kg s–1 m–3), and ∆hfg(T) is sity dependence to the perfusion term in the bioheat
the temperature-dependent phase-change enthalpy equation:

Volume 38, Number 1, 2010


12 Pearce

The right side is certainly not zero—imagine tissue


with zero blood flow at uniform temperature (so that
both the Fourier conduction term and the blood flow
term are zero); Qgen is not zero. The energy-balancing
effect of the mass-loss term (∂m/∂t) on the right side of
Equation 14 has been eliminated, leading to an invalid
conclusion from Equation 18.
The biomechanics of water-vapor diffusion through
tissue have been little studied to date, although Majaron
et al.116 have included some estimates. We lack data for
a complete model, and consequently the relationship
between evolved steam and local pressure elevation is
essentially indeterminate at this point. Steam evolution
is plainly the driving force behind the ejection plumes
that attend pulsed laser110 and similar RF heating appli-
cations. A subsurface steam vacuole is readily observed
in Figure 4a, which was created by a continuous-wave
argon laser (514 nm) at 1.8 W, 3.3-mm beam diameter,
for 2 s in rabbit myocardium.117 Subsurface dissection
FIGURE 6. Boiling processes. (A) Cover illustration due to steam formation is characteristic of high-power
thermogram, argon laser on excised aorta immediately lesions from highly scattered laser sources such as argon
prior to explosive boiling; each color is 5°C from 80°C to
and Nd:YAG lasers. In one such experiment on excised
180°C107; the center temperature in the image is 175°C,
resulting in a saturation pressure of 10 atmospheres. human aorta in vitro, we measured a surface temperature
(B) Cross-section of sealed canine femoral artery by of 175°C immediately prior to a steam-driven explo-
bipolar RF plate electrodes; Mallory’s trichrome stain, sion (Fig. 6A).107 Aorta and other arteries are highly
original magnification ×40; central hole is location of compartmentalized, in a layered sense, consisting of
intra-luminal thermocouple (250 mm diameter); steam
multiple elastin and collagen layers interspersed with
vacuoles (arrows) resulted from low-pressure apposition
of the vessel luminal surfaces. smooth muscle.118 The tissue layers create compart-
ments of evolved steam when heated that burst when
the yield stresses are exceeded.
Excimer (excited dimer) lasers, in particular the
(18) ArF laser at 193 nm (UV), have found widespread
use in radial keratotomy procedures.119–124 Ejection
plumes observed in pulsed excimer laser applications
where c is the composite specific heat of the tissue ( J are steam driven as well, even though excimer lasers
kg–1 K–1), and W includes tissue water density. This are frequently incorrectly described as “cold” lasers.122
formulation includes the latent heat of vaporization, In fact, the UV-absorbing bodies (cell organelles such
but not the changes in tissue density or pressure. as the mitochondria) transfer heat rapidly to the cell
The formulation of Equation 18 is convenient in water during and for a very short time after the femto-
that it has the form of the bioheat equation, but it or nanosecond pulse, thus generating the high pressure
has to be used with considerable caution. First, “W” required to drive the plume.
includes much more than the density of water; in fact,
W includes the mass fraction of water in the tissue, III.C. Determining Arrhenius Coefficients
so W = m in Equation 14. Second, applying the rela-
From End Point Data
tion directly leads to an aphysical result in the case of
equilibrium vaporization at the saturation temperature, The methods for determining Arrhenius coefficients
where dT/dt = 0. In that event, the left side of Equa- from differential scanning calorimetry data described
tion 18 is zero for equilibrium boiling at saturation. by Miles103,104 and employed to great advantage by

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 13

many investigators13,24,40,86,102,125 are powerful. When Briefly, given an ensemble of experiments, a sample
the damage marker can be measured as the damage function, transient heating history, Ti(t), is integrated by
process develops, one can determine dΩ/dt directly, and Equation 2 using assumed values for A and Ea. A suitable
the parameters can be obtained from a straightforward segment of the ln{A} – Ea plane is scanned (reasonable
curve fit to an ensemble of measured slopes, as, for estimates can be quickly assembled from Table 1 data,
example, in cell-survival curves.8,39 or from Wright’s line, Equation 23) to find points that
yield the experimentally observed Ωi(τi). The locus of
1. Constant Temperature Experiments possible solutions lies along a straight line:

When the heating method is isothermal for a very long (22)


time compared with the warm-up time, the resulting
damage can reasonably be assumed to derive from a for which the slope is the temperature and the intercept
constant-temperature experiment. As long as the dam- the exposure time of an equivalent constant-temperature
age marker is quantifiable, Arrhenius parameters can experiment, τeq Teq, for which Ω = 1. Note that the
be obtained. That was the method used to determine measured Ωi values need not be equal in this method
A and Ea for birefringence loss in rat skin collagen.81 as long as they are known for each experiment in
Given an ensemble of such experiments for which Ω the ensemble. The ensemble of equivalent constant-
= 1, one plots the points on Arrhenius axes (ln{τ} vs temperature experiment conditions are then plotted
1/T) and determines the parameters from the slope on standard Arrhenius axes, ln{τeq} vs 1/Teq, to estimate
and intercept: the parameters for the underlying damage process, as
in Equation 19.
(19) Scanning the ln{A} – Ea plane is not as random a
A constant-temperature experiment for which search process as it might first appear. In 2001, Wright126
Ωi is not 1 can be converted to an equivalent Ω = 1 observed that all of the published A and Ea pairs plot
exposure time, τeq, from: very closely to a single line:

(20) (23)
That is, τeq = τ/Ωi. The inspiration for Wright’s plot derives from Miles
If Ω is not 1 for an experiment ensemble with and Ghelashvili’s127 use of a “polymer in a box” construct
known kinetic parameters, but S0 = C(τ)/C(0), the sur- for the process activation entropy to demonstrate that
vival fraction, is known and uniform, then the resulting ∆S* and ∆H* (Equation 7) are proportional. Wright’s
AS0 value can be converted to the Ω = 1 (i.e., general)
incisive technical brief126 is a necessary reference for
case, A36.8% by13,125:
anyone even slightly interested in this area of investiga-
tion; it has to be described as a seminal contribution,
(21)
and provides many illuminating insights and so it seems
No adjustment in Ea need be made for S0 not equal fitting to label Equation 23 “Wright’s line.” Equation
to 36.8% because the slope is not affected.13,125 23 makes an excellent sanity check for any proposed
set of Arrhenius coefficients. A Wright’s line plot of
the Arrhenius coefficients listed in Table 1 has been
2. Transient Temperature Histories, T(t)
included (Fig. 3).
There remains the difficult case of a quantifiable dam-
age marker that can only be measured as an end point 3. Experiment Design Considerations
(i.e., not as it develops), Ω(τ), and results from plainly
transient heating histories that cannot be assumed to In view of the extreme sensitivity of the Arrhenius
be constant temperature. We have developed a method formulation to the temperature, it is absolutely essential
to estimate Arrhenius parameters in that difficult case, that all effort be expended to maximally reduce the
more completely described in the book chapter.113 uncertainty in T. Experiments in which substantial

Volume 38, Number 1, 2010


14 Pearce

thermal gradients appear, such as under laser beams,


lead to high uncertainty in the appropriateness and
general usefulness of the model coefficients because (25)
small uncertainties in position lead to large uncertain-
ties in T(t). Badly estimated temperature histories often
manifest in plainly unrealistic predictions of damage that is, Tj = 50°C means that tj = 3.7420 s. Applying
when the timescale of the experiment changes by a Equation 19:
few orders of magnitude. Another hallmark of good
experimental technique is to ensure that several orders (26)
of magnitude in time are included in the experiment
From which, ln{A} = 232.7 (i.e. A = 1.176 × 10101)
set. An Arrhenius data set comprises holding the short
and Ea = 6.29 × 105 ( J mole–1), which is similar to, but
end of a very long stick indeed, and the uncertainty in observably different from, the values obtained from
the offset, ln{A}, is enormously enhanced when ln{τ} linear regression on Figure 1b data (compare to ln{A}
covers only a narrow range. = 229.0 and Ea = 6.18 × 105). This illustrative calcula-
tion serves to demonstrate the extreme sensitivity of
IV. Conversion Between the Two the results to round-off errors. The sensitivity arises
from the subtraction of large numbers; for example,
Methods
–6.5013 = ln{A} – 239.225 and –1.3196 = ln{A} –
A thorough analysis of the relationship between the 234.10 in Equation 26. In the illustrative calculations,
TID and Arrhenius methods, and methods for conver- an inordinate number of significant digits have been
used, far in excess of those warranted by the accuracy
sion between them, may be found in recent papers by
of the interpolation of the plotted data in Figure 1b,
He et al.125 and Pearce.16 They are briefly summarized
to illustrate this limitation.
here to conclude the discussion.
RCEM is the ratio of the time (in minutes) required to
V. Summary
achieve an iso-effect dose for a 1°C rise in temperature.
It is related to the Arrhenius coefficients by125: Thermal alterations in tissues are not phase-change
processes, although they have, very unfortunately, been
(24) described that way.128 Miles et al.104,129,130 were criticized
This relationship can be quickly derived from two by Engel and Bachinger131 for making this observa-
tion based on their differential scanning calorimetry
applications of Equation 2 by setting Ω1 = Ω2, T2 = T1
experiments. Miles and Bailey successfully defended
+ 1 and RCEM = the ratio τ2/τ1. So, for the CHO cell their newly discovered point of view in a subsequent
data (Table 1) at temperatures of 43°C and 44°C, RCEM letter132 pointing out that they also originally thought
= exp{–6.18 × 105/8.337 × 105} = 0.477. collagen denaturation was a phase-change process (as
A value for RCEM alone is not sufficient information was the prevailing notion among collagen chemists), but
to derive Arrhenius coefficients from the TID param- were convinced by their experiments that it was actually
eter; note that RCEM contains no information about A. a rate-limited, irreversible kinetic change. Thermal-
One also requires a value for the time constant, D0, damage processes in general are not phase changes
when they are used to describe major alterations in
for at least one convenient temperature, at minimum.
tissue structure or function, primarily because they are
Given RCEM and D0(T), the values for A and Ea can
just not reversible. I first encountered this simple but
be calculated, again from two applications of Equa- instantly crystallizing observation in a brilliant seminar
tion 2. For example, at 43°C D0(43) ≈ 666 s (Fig. 1b), presented by Neil Wright in 2000.133 Ice melts and re-
and with RCEM = 0.477, a temperature of 50°C would freezes, water boils and condenses: all true phase-change
require: processes reverse when the temperature changes simply

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 15

by absorbing or releasing the phase-change enthalpy by it a useful way to analyze such an experiment series
heat transfer or related means. In contra-distinction, an near the threshold of the particular damage process
egg cannot be “un-fried”; there is no verb for that in our under study.
language for a reason. Resolving the thermal damage In surveying the array of process parameters shown
site entails replacing the denatured proteinaceous and in Table 1, it might be shocking to observe the range of
membrane structures with scar tissue, followed by the A values reported: a spread of approximately 10100 can
healing process, a completely separate and thoroughly be seen. Indeed, these are certainly impressive, if not
independent thermodynamic event. frightening, numbers that often require some manipula-
Irreversible thermal alterations in tissues are tion to be applied; most calculators only accept up to
routinely used in surgery to simultaneously cut and 1099 as a floating-point number, for example. Practically,
coagulate tissues, to ablate ectopic foci in myocar- A—or, rather, its logarithm, ln{A}, the process activa-
dium, to ablate tumors by severe heating, and in more tion entropy—represents the offset for the process; in
subtle hyperthermia tumor treatments. Surgical cutting concert with Ea, it establishes the temperature at which
requires rapid boiling of tissue water, an extremely the process becomes active. The rate of damage (the
thermodynamically active tissue constituent, whether slope) is determined by the activation energy, Ea, and
by RF current or laser energy. The fusion of tissues small differences in Ea can make huge differences in A.
and sealing of blood vessels depend on gellification of
The logarithm is much less sensitive for obvious reasons,
collagens and elastin plates at high temperature and
and in numerical model work the numbers are often
high apposition pressure. Collagenous tissues can also
too large to be handled in terms of A, so the formula-
be shrunk to achieve a desired clinical result. Cellular
tion is routinely set up to be in the form of dΩ/dt =
necrosis and apoptosis may both be observed in tumor
exp(ln{A} – Ea/RT). Therefore, in view of the functional
treatment and like applications, and near the edges of
relationship involved and the range of temperatures for
higher-temperature acute lesions.
Using Arrhenius models to predict the probabil- the particular processes, the wide range in A is not really
ity of observation of a discrete (i.e., non-quantitative, too surprising; it is characteristic of this type of model.
or 0–1) thermal damage process by calculating P, as Wright’s cogent observations126 provide an important
introduced earlier, might at first appear to inappropri- additional illuminating perspective.
ately stretch the imagination. An “ergodic” stochastic Despite the temptation to discuss thermal-damage
process is one in which calculation of any statistical processes in terms of threshold temperatures alone,
moment with respect to time in one experiment (i.e., it must always be kept in mind that the underlying
sample function) gives the same result as calculating processes are kinetic in nature: the time of exposure
the same statistic across the sample functions at one is equally important as the temperature. In fact, if a
fixed time in an ensemble of experiments. Therefore, “threshold” temperature must be discussed, the kinetic
if the stochastic nature of the damage process under nature can easily be included, as was described in
study is (at least) ergodic with respect to the mean, then Equation 9b:
we can reasonably expect the first statistical moment
to be the same whether computed across an ensemble
of experiments or with time for a single experiment. (9b)
Consequently, as mentioned earlier, it may be useful
to apply the Arrhenius model in this way to a damage
process that is not quantifiable in an analog sense, but If the CHO cells are used as an illustrative example,
is discretely observable (i.e., present or not present) at T TH for a 30-min exposure would be 41.1°C, while if
temperatures near the threshold, T TH, for the process. the exposure time were reduced to 1 min, T TH would
In fact, I was surprised to find that this approach suc- be 45.7°C. This is a substantial difference if one is
cessfully modeled an unpublished series of just such concerned about either staying just below or ensuring
discrete experiments: the Diller and Klutke Arrhenius that one exceeds the damage threshold. Of course, this
coefficients were predictive of the frequency of observa- particular damage process is only valid for temperatures
tion in a large ensemble. Future investigators may find above 43°C (the calculation grossly overestimates the

Volume 38, Number 1, 2010


16 Pearce

damage rate below 43°C), so the result is for illustra- REFERENCES


tive purposes only. 1. Major RH. A history of medicine. Springfield, IL: Charles
Kinetic descriptions of the propagation of thermal C. Thomas; 1954.
damage may be easily incorporated into numerical 2. Breasted JH. The Edwin Smith surgical papyrus. Chicago:
The University of Chicago Press; 1930.
model predictions to extend the significance of the 3. Thevenet A. Guy de Chauliac (1300–1370): The “father of
model results to observable tissue effects, rather than surgery.” Ann Vasc Surg. 1993 Mar;7(2):208–12.
being limited to predictions of temperature fields 4. Kelly HA, Ward GE. Electrosurgery. Philadelphia: W.B.
Saunders; 1932.
alone. Multiple processes can be included in parallel 5. Doyen E. Sur la destruction des tumeurs cancereuses acces-
and actually compared with clinical assessments such sibles; par la methode de la voltaisation bipolaire et de l’electro-
coagulation thermique. Archives d’Electicitie Medicale et de
as histologic sections or vital stain images. The signifi- Physiotherapie du Cancer. 1909;17:791–5.
cance and usefulness of the results are thus magnified 6. Stilwell GK. General principles of thermotherapy. In: Licht S,
many times over. As a consequence, the number of editor. Therapeutic heat and cold. New Haven, CT: Elizabeth
Licht; 1965.
experiments required to complete a device design or 7. Sapareto SA. The biology of hyperthermia in vitro. In:
to establish the efficacy of a method or device may be Nussbaum GH, editor. Physical aspects of hyperthermia.
reduced. Dosimetry and treatment planning may also New York: American Institute of Physics; 1982.
8. Sapareto SA, Hopwood LE, Dewey WC, Raju MR, Gray JW.
be placed on a more quantitative basis. Effects of hyperthermia on survival and progression of Chinese
Many important thermal-damage processes at hamster ovary cells. Cancer Res. 1978;38(2):393–400.
9. Dewey WC. Arrhenius relationships from the molecule and
moderate and low temperatures have been quantitatively cell to the clinic. Int J Hyperthermia. 1994;25(1):3–20.
studied in cell preparations, but many others remain. A 10. Dewey WC. Thermal tolerance during S phase for cell
smaller number of processes have been quantitatively killing and chromosomal aberrations. Radiation Res.
1990;122(2):193–6.
studied at higher temperatures in intact tissues, a difficult 11. Dewhirst MW, Viglianti BL, Lora-Michiels M, Hanson
arena. A key element in higher-temperature thermal- M, Hoopes PJ. Basic principles of thermal dosimetry and
damage studies is the identification of a quantitative thermal thresholds for tissue damage from hyperthermia. Int
J Hyperthermia. 2003;19(3):267–94.
marker. At this time, we have only a few of them, and 12. Henriques FC. Studies of thermal injury, V. The predict-
those that we do have appear acutely. It is well known ability and significance of thermally induced rate processes
leading to irreversible epidermal injury. Arch Pathol. 1947;43:
that thermal damage accumulates post-exposure as tissue 489–502.
vasculature collapses and cells and tissues that survived 13. He X, Bischof JC. Quantification of temperature and injury
the initial thermal insult die from ischemia, hypoxia, response in thermal therapy and cryosurgery. Crit Rev Biomed
Eng. 2003;31(5 & 6):355–421.
or a related cause. The post-injury healing response is 14. Haveman J, Sminia P, Wondergem J, van der Zee J, Hulshof
another matter entirely, and virtually no quantitative MCCM. Effects of hyperthermia on the central nervous sys-
tem: what was learnt from animal studies? Int J Hyperthermia.
descriptions are presently available.
2005;21(5):473–87.
Higher-temperature exposures become substantially 15. Maron SH, Lando JB. Fundamentals of physical chemistry.
more complicated when tissue water boils. A number New York: Macmillan Publishing Co.; 1974.
16. Pearce JA. Relationship between Arrhenius models of thermal
of biomechanical parameters are required before more damage and the CEM 43 thermal dose. In: Energy-based
complete analyses are feasible, analyses that include treatment of tissue and assessment V, (Proceedings of the
mechanical disruption due to induced stresses such as Society of Photographic Instrumentation Engineers, vol 7181).
Bellingham, WA: SPIE; 2009. p. 718104.
surgical cutting. Obviously, much more work remains 17. Chen G, Goeddel DV. TNF-R1 signaling: a beautiful pathway.
to be done on higher-temperature measures and models Science. 2002;296(5573):1634–5.
18. Thompson CB. Apoptosis in the pathogenesis and treatment
of thermal damage and repair.
of disease. Science. 1995;267(5203):1456–62.
19. Everett H, McFadden G. Apoptosis: an innate immune
response to virus infection. Trends Microbiol. 1999;7(4):
ACKNOWLEDGMENT
160–5.
20. Kerr JF. A histochemical study of hypertrophy and ischaemic
Partial support for this work was provided by the T.L.L. injury of rat liver with special reference to changes in lysosomes.
Temple Foundation. J Pathol Bacteriol. 1965;90:419–35.

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 17

21. Kerr JF, Wylie AH, Currie AR. A histochemical study 36. Wilmink GJ, Opalenik SR, Beckham JT, Abraham AA,
of hypertrophy and ischaemic injury of rat liver with Nanney LB, Mahadevan-Jansen A, Davidson JM, Jansen ED.
special reference to changes in lysosomes. Brit J Cancer. Molecular imaging-assisted optimization of hsp70 expression
1972;26(4):239–57. during laser-induced thermal preconditioning for wound repair
22. Cotran RS, Kumar V, Collins T, Robbins SL. Robbins patho- enhancement. J Invest Dermatol. 2009 Jan;129(1):205–16.
logic basis of disease. Philadelphia: W.B. Saunders; 1998. 37. Green DM, Diller KR. Measurement of burn-induced
23. Barton J, Rollins A, Yazdanfar S, Pfefer TJ, Westphal V, Izatt leakage of macromolecules in living tissue. J Biomech Eng.
JA. Photothermal coagulation of blood vessels: A comparison 1978;100(3):153–8.
of high-speed optical coherence tomography and numerical 38. Aggarwal SJ, Shah SJ, Diller KR, Baxter CR. Fluorescence
modelling. Phys Med Biol. 2001;46(6):1665–78. digital microscopy of interstitial macromolecular diffusion in
24. Bhowmick P, Coad JE, Bhowmick S, Pryor JL, Larson T, De burn injury. Comput Biol Med. 1989;19(4):245–61.
La Rosette J, Bischof JC. In vitro assessment of the efficacy 39. Bhowmick S, Swanlund DJ, Bischof JC. Supra-physiological
of thermal therapy in human benign prostatic hyperplasia. thermal injury in Dunning AT-1 prostate tumor cells. J
Int J Hyperthermia. 2004 Jun;20(4):421–39. Biomech Eng. 2000;122(1):51–9.
25. Borrelli MJ, Thompson LL, Cain CA, Dewey WC. Time- 40. He X, Bischof JC. The kinetics of thermal injury in human renal
temperature analysis of cell killing of BhK cells heated at carcinoma cells. Ann Biomed Eng. 2005;33(4):502–10.
temperatures in the range of 43.5°C to 57.0°C. International 41. Taormina M, Diller KR, Baxter CR. Burn induced altera-
Journal of Radiation Oncology and Biological Physics. tion of vasoactivity in the cutaneous microcirculation. In: In:
1990;19:389–99. Diller K, Roemer RB, editors. Heat and mass transfer in the
26. Wust P, Hildebrandt B, Sreenivasa G, Rau B, Gellermann microcirculation of thermally significant vessels. Anaheim,
J, Riess H, Felix R, Schlag PM. Hyperthermia in combined CA: American Society of Mechanical Engineers; 1986. p.
treatment of cancer. Lancet Oncol. 2002 Aug;3(8):487–97. 81–5.
27. Bowman PD, Schuschereba ST, Lawlor DF, Gilligan GR, 42. Gourgouliatos ZF, Welch AJ, Diller KR. Microscopic instru-
Mata JR, DeBaere DR. Survival of human epidermal mentation and analysis of laser-tissue interaction in a skin
keratinocytes after short-duration high temperature: syn- flap model. J Biomech Eng. 1991;113(3):301–7.
thesis of HSP70 and IL-8. Am J Physiol. 1997;272(6 pt 43. Hoffmann NE, Bischof JC. Cryosurgery of normal and
1):C1988–94. tumor tissue in the dorsal skin flap chamber: Part I--thermal
28. Zhang XP, Elofsson A, Andreu D, Glaser E. Interaction of response. J Biomech Eng. 2001;123(4):301–9.
mitochondrial presequences with DnaK and mitochondrial 44. Hoffmann NE, Bischof JC. Cryosurgery of normal and
hsp70. J Mol Biol. 1999;288(1):177–90. tumor tissue in the dorsal skin flap chamber: Part II--Injury
29. Grimshaw JP, Jelesarov I, Schönfeld HJ, Christen P. Response. J Biomech Eng. 2001;123(4):310–6.
Reversible thermal transition in GrpE, the nucleotide 45. Lepock JR, Frey HE, Bayne H, Markus J. Relationship of
exchange factor of the DnaK heat-shock system. J Biol hyperthermia-induced hemolysis of human erythrocytes to
Chem. 2001;276(9):6098–104. the thermal denaturation of membrane proteins. Biochim
30. Beckham JT, Mackanos MA, Crooke C, Takahashi T, Biophys Acta. 1989;980:191–201.
O’Connell-Rodwell C, Contag CH, Jansen ED. Assessment 46. Van Delft JL, Oosterhuis JA, Barthen ER, Eijkelenboom AM.
of cellular response to thermal laser injury through biolu- Janus green vital staining of the cornea. Doc Ophthalmol.
minescence imaging of heat shock protein 70. Photochem 1983;55(1-2):47–50.
Photobiol. 2004 Jan;79(1):76–85. 47. Bhisitkul RB. Second generation vital stains in retinal surgery.
31. Rylander MN, Diller KR, Wang S, Aggarwal SJ. Correlation Br J Ophthalmol. 2003 Jun;87(6):664–5.
of HSP70 expression and cell viability following thermal 48. Acosta D, Wendel DG. A permeability test for the study of
stimulation of bovine aortic endothelial cells. J Biomech Eng. mitochondrial injury in in vitro cultured heart muscle and
2005;127(5):751–7. endothelioid cells. Histochem J. 1975;7(1):45–56.
32. Rylander MN, Feng Y, Zhang Y, Bass J, Jason Stafford R, 49. Feldman S, Glagov S, Wissler RW, Hughes RH. Postmortem
Volgin A, Hazle JD, Diller KR. Optimizing heat shock protein delineation of infarcted myocardium. Coronary perfusion
expression induced by prostate cancer laser therapy through with nitro blue tetrazolium. Arch Pathol Lab Med. 1976
predictive computational models. J Biomed Opt. 2006 Jul- Jan;100(1):55–8.
Aug;11(4):041113. 50. Lucchesi BR, Burmeister WE, Lomas TE, Abrams GD.
33. Beckham JT, Wilmink GJ, Mackanos MA, Takahashi K, Ischemic changes in the canine heart as affected by the
Contag CH, Takahashi T, Jansen ED. Role of HSP70 dimethyl quaternary analog of propranolol, UM-272 (SC-
in Cellular Thermotolerance. Lasers Surg Med. 2008 27761). J Pharmacol Exp Ther. 1976 Nov;199(2):310–28.
Dec;40(10):704–15. 51. Bhowmick S, Swanlund DJ, Bischof JC. In vitro thermal ther-
34. Wilmink GJ, Opalenik SR, Beckham JT, Davidson JM, Jansen apy of AT-1 Dunning prostate tumours. Int J Hyperthermia.
ED. Assessing laser-tissue damage with bio-luminescent 2004;20(1):73–92.
imaging. J Biomed Opt. 2006;11(4). 52. Pearce JA, Thomsen S. Rate process analysis of thermal dam-
35. Wilmink GJ, Opalenik SR, Beckham JT, Mackanos MA, age. In: Welch AJ, vanGemert MJC, editors. Optical-thermal
Nanney LB, Contag CH, Davidson JM, Jansen ED. In-vivo response of laser-irradiated tissue. New York: Plenum Press;
optical imaging of hsp70 expression to assess collateral tis- 1995. p. 561–606.
sue damage associated with infrared laser ablation of skin. J 53. Robbins SL, Kumar V. Basic pathology. 4 ed. Philadelphia:
Biomed Opt. 2008 Sep-Oct;13(5):054066. W.B. Saunders; 1987.

Volume 38, Number 1, 2010


18 Pearce

54. Srinivas SM, de Boer JF, Park H, Keikhanzadeh K, Huang 71. Cinca J, Warren M, Carreño A, Tresànchez M, Armadans
HE, Zhang J, Jung WQ, Chen Z, Nelson JS. Determination L, Gómez P, Soler-Soler J. Changes in myocardial electri-
of burn depth by polarization-sensitive optical coherence cal impedance induced by coronary artery occlusion in pigs
tomography. J Biomed Opt. 2004;9(1):207–12. with and without preconditioning. Circulation. 1997;96:
55. Pierce MC, Sheridan RL, Park BH, Cense B, de Boer JF. 3079–86.
Burn depth determination in human skin using polarization- 72. Raghavan K, Porterfield JE, Kottam AT, Feldman MD,
sensitive optical coherence tomography. In: Progress in Escobedo D, Valvano JW, Pearce JA. Electrical conductivity
Biomedical Optics and Imaging: Coherence Domain Optical and permittivity of murine myocardium. IEEE Trans Biomed
Methods and Optical Coherence Tomography in Biomedicine Eng. 2009 Aug;56(8):2044–53.
VII; 27–29 Jan 2003; San Jose, CA. Bellingham, WA: Society 73. Pearce JA. Electrosurgical unit. In: Webster JG, editor.
of Photographic Instrumentation Engineers. p. 263–70. Encyclopedia of medical devices and instrumentation. New
56. Henriques FC, Moritz AR. Studies of thermal injury in the York: John Wiley and Sons; 2006. p. 156–77.
conduction of heat to and through skin and the temperatures 74. Pearce JA, Thomsen SL, Vijverberg H, McMurray TJ. Kinetics
attained therein: a theoretical and experimental investigation. for birefringence changes in thermally coagulated rat skin col-
Am J Pathol. 1947 Jul;23(4):530–49. lagen. In: Laser Isotope Separation, Society of Photographic
57. Moritz AR, Henriques FC. Studies in thermal injury II: the Instrumentation Engineers; 19–20 Jan 1993; Los Angeles,
relative importance of time and surface temperature in the cau- CA. Bellingham, WA: SPIE; 1993. p. 180­–6.
sation of cutaneous burns. Am J Pathol. 1947;23:695–720. 75. Maitland DJ, Walsh JT Jr. Intensity-based feedback of the
58. Moritz AR. Studies of thermal injury III. The pathology and thermally induced denaturation of collagen. In: Proceedings
pathogenesis of cutaneous burns: An experimental study. Am of 17th Annual International Conference of the IEEE
J Pathol. 1947;23:915–34. Engineering in Medicine and Biology Society, Montreal,
59. Diller KR, Klutke GA. Accuracy analysis of the Henriques Quebec, Canada, 20–24 Sep 1995. Los Alamitos, CA: IEEE;
model for predicting thermal burn injury. In: Advances 1995. p. 1735–6.
in Heat and Mass Transfer in Biotechnology. American 76. Thomsen S, Cheong WF, Pearce JA. Changes in collagen
Society of Mechanical Engineers International Mechanical birefringence: a quantitative histologic marker of thermal
Engineering Congress & Exposition; 15–18 Nov 1999; damage in skin. In: Proceedings of Lasers in Dermatology
Nashville, Tennessee. New York: ASME. p. 117–23. and Tissue Welding, Society of Photographic Instrumentation
60. Diller KR, Valvano JW, Pearce JA. Bioheat transfer. In: Kreith Engineers; 21–22 Jan 1991; Los Angeles, CA. Bellingham,
F, editor. CRC handbook of thermal engineering. Boca Raton: WA: SPIE; 1991. p. 34–42.
CRC Press; 2000. p. 114–215. 77. Thomsen S. Practical and theoretical considerations on the
61. Weaver JA, Stoll AM. Mathematical model of skin exposed use of quantitative histology to measure thermal damage
to thermal radiation. Aerosp Med. 1969;40(1):24–30. of collagen in cardiovascular tissues. In: Proceedings of
62. Brown SL, Hunt JW, Hill RP. Differential thermal sensitivity Diagnostic and Therapeutic Cardiovascular Interventions IV,
of tumour and normal tissue microvascular response during Society of Photographic Instrumentation Engineers; 22–23
hyperthermia. Int J Hyperthermia. 1992;8:501–4. Jan 1991; Los Angeles, CA. Bellingham, WA: SPIE; 1991.
63. Webster’s new world dictionary. 2 ed. New York: Simon and p. 110–3.
Schuster; 1982. 78. Maitland DJ, Walsh JT Jr. Quantitative measurements of
64. US Department of Health & Human Services Food and linear birefringence during heating of native collagen. Lasers
Drug Administration website. Available at: http://www Surg Med. 1997;20(3):310–8.
.fda.gov/. 79. Chen WR, Bartels KE, Liu H, Nordquist RE. Laser-
65. Thomsen S. Targeted thermal injury: mechanisms of cell photothermal effect on skin tissue--damage and recovery. J
and tissue death. In: Energy-based treatment of tissue and X-Ray Sci Technol. 2006;14(3):207–15.
assessment V (Proceedings of the Society of Photographic 80. Pearce JA, Cilesiz I, Welch AJ, Chan EK, McMurray TJ,
Instrumentation Engineers, vol 7181). Bellingham, WA: SPIE; Thomsen SLMD. Comparison of Ho:YAG, Tm:YAG, and
2009. p. 718102. argon lasers for fusion of intestinal tissues. In: International
66. Godwin BL, Coad JE. Healing responses following cryo- Symposium on Optics, Imaging, and Instrumentation, Society
thermic and hyperthermic tissue ablation. In: Energy-based of Photographic Instrumentation Engineers; 24-29 July 1994;
treatment of tissue and assessment V (Proceedings of the San Diego, CA. Bellingham, WA: SPIE; 1994. p. 517–26.
Society of Photographic Instrumentation Engineers, vol 81. Pearce JA, Thomsen S. Kinetic models of laser-tissue fusion
7181). Bellingham, WA: SPIE; 2009. p. 718103. processes. In: Proceedings of the 30th Annual Rocky Mountain
67. Thomsen S, Pearce JA, Cheong WF. Changes in birefringence Bioengineering Symposium and the 30th International ISA
as markers of thermal damage in tissues. IEEE Trans Biomed Biomedical Sciences Instrumentation Symposium; 2–3 Apr
Eng. 1989 Dec;36(12):1174–9. 1993; San Antonio, TX. Research Triangle Park, NC: ISA
68. Jacques SL, Gaeeni MO. Thermally induced changes in Services Inc.; 1993. p. 355–60.
optical properties of heart. IEEE Eng Med Biol Mag. 82. Bass LS, Maozami N, Pocsidio J, Oz MC, LoGerfo P, Treat
1989(11):1199–200. MR. Electrophoretic mobility patterns of collagen following
69. McRury ID, Haines DE. Ablation for the treatment of laser welding. In: Proceedings of Lasers in Dermatology and
arrythmias. Proc IEEE 1996;84:404–16. Tissue Welding, Society of Photographic Instrumentation
70. McDaniel GM, Van Hare GF. Catheter ablation in children Engineers; 21–22 Jan 1991; Los Angeles, CA. Bellingham,
and adolescents. Heart Rhythm. 2006 Jan;3(1):95–101. WA: SPIE; 1991. p. 123–7.

Critical Reviews™ in Biomedical Engineering


Models for Thermal Damage in Tissues 19

83. Lemole GM, Anderson RR, DeCoste S. Preliminary evalu- and Cells XVIII Society of Photographic Instrumentation
ation of collagen as a component in the thermally-induced Engineers; 20–25 Jan 2007; San Jose, CA; Bellingham, WA:
‘weld.’ In: Proceedings of Lasers in Dermatology and Tissue SPIE; 2007. Article no. 64350Z.
Welding, Society of Photographic Instrumentation Engineers; 96. Longo L, Piccinetti AL, Dalle Monarche G, Botta G, Mancini
21–22 Jan 1991; Los Angeles, CA. Bellingham, WA: SPIE; S. Laser treatment of stretch marks: preliminary results. In:
1991. p. 116–22. Laser Florence ‘99: A Window on the Laser Medicine World,
84. Pearce JA. Corneal reshaping by radio frequency current: Society of Photographic Instrumentation Engineers; 28–31
numerical model studies in thermal treatment of tissue. In: Oct 1999. Florence, Italy. Bellingham, WA: SPIE; 1999. p.
Energy Delivery and Assessment, Society of Photographic 164–8.
Instrumentation Engineers; 21–22 Jan 2001; San Jose, CA, 97. Dang Y, Ren Q, Li W, Liu H, Liu Y, Zhang J. In vivo
United States. Bellingham, WA: SPIE; 2001. p. 09–18. experiment study of non-ablative photo-rejuvenation by using
85. Pearce JA, Ikei C. Increasing corneal curvature by RF current: a 595-nm pulsed dye laser. In: Optics in Health Care and
Numerical model studies of governing physical processes. In: Biomedical Optics: Diagnostics and Treatment II, Society of
Thermal Treatment of Tissue: Energy Delivery and Assessment Photographic Instrumentation Engineers. 9–12 Nov 2004;
IV, Society of Photographic Instrumentation Engineers; Beijing, China: Bellingham, WA: SPIE; 2004. p. 772–9.
25–27 Jan 2007; San Jose, CA, United States. Bellingham 98. Yetkinier D, Bessette A, Woloszko J. A novel radiofrequency
WA: SPIE; 2007. Article no. 64400L. technology (Coblation®) for dermatologic surgery applica-
86. Aksan A, McGrath JJ. Thermomechanical analysis of tions. In: Lasers in Surgery: Advanced Characterization,
soft-tissue thermotherapy. J Biomech Eng. 2001;125(5): Therapeutics, and Systems XII, Society of Photographic
700–8. Instrumentation Engineers; 19–22 Jan 2002; San Jose, CA.
87. Weir CE. Rate of shrinkage of tendon collagen: heat, entropy, Bellingham, WA: SPIE; 2002. p. 276–83.
and free energy of activation of shrinkage of untreated ten- 99. Chen SS, Wright NT, Humphrey JD. Heat-induced changes
don: effect of acid, salt, pickle, and tannage on activation of in the mechanics of a collagenous tissue: isothermal, isotonic
tendon collagen. Journal of the American Leather Chemists shrinkage. J Biomech Eng. 1998;120:382–8.
Association. 1949;44(3):108–38. 100. Chen SS, Wright NT, Humphrey JD. Phenomenological
88. Cohen RE, Hooley CJ, McCrum NG. Viscoelastic creep of evolution equations for heat-induced shrinkage of a col-
collagenous tissue. J Biomech. 1976;9(4):175–84. lagenous tissue. IEEE Trans Biomed Eng. 1998 Oct;45(10):
89. Trembly BS, Keates RH. Combined microwave heating and 1234–40.
surface cooling of the cornea. IEEE Trans Biomed Eng. 1991 101. Chen SS, Wright NT, Humphrey JD. Heat-induced changes
Jan;38(1):85–91. in the mechanics of a collagenous tissue: isothermal free
90. Brinkman R, Koop N, Kamm K, Geerling G, Kampmeier J, shrinkage. J Biomech Eng. 1997;119(4):372–8.
Birngruber R. Laser thermokeratoplasty by means of a con- 102. Aksan A, McGrath JJ, Nielubowicz DS Jr. Thermal damage
tinuously emitting laser diode in the mid-IR. In: Lasers in prediction for collagenous tissues part I: a clinically relevant
Ophthalmology IV, Society of Photographic Instrumentation numerical simulation incorporating heating rate dependent
Engineers; 9–10 Sep 1996; Vienna, Austria. Bellingham, WA: denaturation. J Biomech Eng. 2005;127(1):85–97.
SPIE; 1996. Vol. 2930. p. 66–74. 103. Miles CA. Kinetics of collagen denaturation in mammalian
91. Timberlake GT, Reinke MH, Miller A. Changes in cor- lens capsules studied by differential scanning calorimetry. Int
neal collagen induced by holmium:YAG laser irradiation. J Biol Macromol. 1993 Oct;15(5):265–71.
In: Laser-Tissue Interaction VII, Society of Photographic 104. Miles CA, Burdjanadze TV, Bailey AJ. The kinetics of the
Instrumentation Engineers; 28 Jan–2 Feb 1996; San Jose, thermal denaturation of collagen in unrestrained rat tail tendon
CA. Bellingham, WA: SPIE; 1996. p. 44–51. determined by differential scanning calorimetry. J Mol Biol.
92. Majaron B, Srinivas SM, Huang HL, Nelson JS. Deep 1995;245:437–46.
coagulation of dermal collagen with repetitive Er:YAG laser 105. Torres JH, Motamedi M, Pearce JA, Welch AJ. Experimental
irradiation. Lasers Surg Med. 2000;26:215–22. evaluation of mathematical models for predicting the ther-
93. Mordon S, Capon A, Creusy C, Fleurisse L, Buys B, Faucheaux mal response of tissue to laser irradiation. Applied Optics.
M, et al. In vivo experimental evaluation of non-ablative skin 1993;32(4):597–606.
remodeling using a 1.54 μm laser with surface cooling. In: 106. Brutsaert W. Evaporation into the atmosphere: theory, his-
Lasers in Surgery: Advanced Characterization, Therapeutics, tory and applications. Dordrecht, The Netherlands: D. Reidel;
and Systems X, Society of Photographic Instrumentation 1982.
Engineers; 22–23, 25 Jan 2000; San Jose, CA. Bellingham, 107. Pearce JA. [Cover illustration] J Assoc Adv Med Instrum.
WA: SPIE; 2000. p. 12–22. 1984;18(4):248.
94. Paithankar D, Hsia J, Ross EV. Sub-surface wrinkle removal 108. Pearce JA. Electrosurgery. In: Meldrum SJ, Watson BW,
by laser treatment in combination with dynamic cooling. In: Geddes LA, editors. London: Chapman & Hall; 1986.
Lasers in Surgery: Advanced Characterization, Therapeutics, 109. Oringer MJ. Electrosurgery in dentistry. Philadelphia: W.B.
and Systems X, Society of Photographic Instrumentation Saunders; 1975.
Engineers; 22–23, 25 Jan 2000; San Jose, CA. Bellingham, 110. Hartman RA, Whitaker P. The physics of transmyocardial
WA: SPIE; 2000. p. 4–11. laser revascularization. Journal of Clinical Laser Medicine
95. Liu H, Dang Y, Zhan W, Ren Q. Non-ablative collagen and Surgery. 1997;15(6):255–9.
remodeling initiated by different laser effects: A comparative 111. Keenan JH, Keyes FG. Thermodymanic properties of steam.
study on mouse model. In: Optical Interactions with Tissue New York: John Wiley and Sons; 1936.

Volume 38, Number 1, 2010


20 Pearce

112. Ravid A, Katzir A. Theoretical model simulating CO2 laser 123. Wallau AD, Campos M. One-year outcomes of a bilat-
ablation of biological tissue due to steam pressure generation. eral randomised prospective clinical trial comparing PRK
In: Proceedings of Lasers-Tissue Interaction, Tissue Optics and with mitomycin C and LASIK. Br J Ophthalmol. 2009
Laser Welding III, Society of Photographic Instrumentation Dec;93(12):1634–8.
Engineers; 4–6 Sep 1997; San Remo, Italy. Bellingham, WA: 124. Roundy CB. Electronic beam analysis of excimer lasers used
SPIE; 1997. p. 287–95. for photorefractive keratotomy. In: Fifth Conference on Optics:
113. Thomsen S, Pearce JA. Thermal damage and rate processes ROMOPTO ‘97, Society of Photographic Instrumentation
in biologic tissue. In: Welch AJ, van Gemert MJC, editors. Engineers; 9–12 Sep 1998; Bucharest, Romania. Bellingham
Optical-thermal response of laser-irradiated tissue. 2nd ed. WA: SPIE; 1998. p. 673–7.
Dordrecht, The Netherlands: Springer-Verlag; In Press. 125. He X, Bhowmick S, Bischof JC. Thermal therapy in urologic
114. Covidien. Ligasure® tissue fusion devices product informa- systems: a comparison of arrhenius and thermal isoeffective
tion. Available on the Covidien website at: http://www dose models in predicting hyperthermic injury. J Biomech
.ligasure.com/ligasure/pages.aspx. Eng. 2009 Jul;131(7):074507.
115. Yang D, Converse MC, Mahvi DM, Webster JG. Expanding 126. Wright NT. On a relationship between the Arrhenius
the bioheat equation to include internal water evaporation dur- parameters from thermal damage studies. J Biomech Eng.
ing heating. IEEE Trans Biomed Eng. 2007;54(8):1382–8. 2001;125(2):300–4.
116. Majaron B, Plestenjak P, Lukac M. Quantitative investiga- 127. Miles CA, Ghelashvili M. Polymer-in-a-box mechanism
tion of thermal damage in Er:YAG laser skin resurfacing. In: for the thermal stabilization of collagen molecules in fibers.
Lasers in Surgery: Advanced Characterization, Therapeutics, Biophys J. 1999 Jun;76(6):3243–52.
and Systems VIII, Society of Photographic Instrumentation 128. Flory PJ, Garrett RR. Phase transition in collagen and gelatin
Engineers; 8–9 Feb 1997; San Jose, CA; Bellingham, WA:
systems. J Am Chem Soc. 1958;80:4836–45.
SPIE; 1997. p. 366–73.
129. Miles CA, Bailey AJ. Thermal denaturation of collagen
117. Pearce JA, Liao WH, Thomsen S. The kinetics of thermal
revisited. Journal of Chemical Sciences/Indian Academy of
damage: estimation and evaluation of model coefficients. In:
Sciences. 1999;111(1):71–80.
Proceedings of the 1998 American Society of Mechanical
130. Miles CA, Knott L, Sumner IG, Bailey AJ. Differences
Engineers International Mechanical Engineering Congress
between the thermal stabilities of the three triple-helical
and Exposition; 15–20 Nov 1998; Anaheim, CA. Anaheim,
domains of type IX collagen. J Mol Biol. 1998;277:
CA: ASME; 1998. p. 71-5.
118. Pearce JA, Thomsen S. The effect of vessel architecture on 135–44.
fusion by radio frequency current. In: Surgical Applications of 131. Engel J, Bachinger. Cooperative equilibrium transitions
Energy, Society of Photographic Instrumentation Engineers; coupled with a slow annealing step explain the sharp-
25–26 Jan 1998; San Jose, CA. Bellingham, WA: SPIE; 1998. ness and hysteresis of collagen folding. Matrix Biol. 2000
p. 217–28. Jul;19(3):235–44.
119. Beckman H, Fuller TA, Boyman R, Mandell G, Nathan 132. Miles CA, Bailey AJ. Response to Engel and Bachinger’s
LE Jr. Carbon dioxide laser surgery of the eye and adnexa. critique of collagen denaturation as a rate process [letter to
Ophthalmology. 1980;87(10):990–1000. the editor]. Matrix Biol. 2001;20:263–5.
120. Puliafito CA, Steinert RF, Deutsch TF, Hillenkamp F, Dehm 133. Wright NT. Thermal damage processes in tissues. In: Seminar
EJ, Adler CM. Excimer laser ablation of the cornea and lens: in Biomedical Engineering, The University of Texas at Austin;
Experimental studies. Ophthalmology. 1985;92(6):741–8. April 2000.
121. Loree TR, Thomas SJ, Johnson TM, Birmingham BS, Rowsey 134. Welch AJ, Polhamus GD. Measurement and prediction of
JJ. Incising the cornea with ArF laser beams. In: Conference thermal injury in the retina of Rhesus monkey. IEEE Trans
on Lasers and Electro-Optics: Digest of Technical Papers; 26 Biomed Eng. 1984;31:633–44.
Apr–1 May 1987; Baltimore, MD. Washington, DC: Optical 135. Jacques SL, Newman C, He XY. Thermal coagulation of tis-
Society of America; 1987. p. 74. sues: liver studies indicate a distribution of rate parameters not
122. Yuan C, Yaokang L, Jiang B, Songging X, Wang S, Humei a single rate parameter describes the coagulation process. In:
J, Zheng Y. Zhang Z. Experimental study of radial kerato- Proceedings of the Winter Annual Meeting of the American
tomy using excimer laser. Yingyong Jiguang/Applied Laser Society of Mechanical Engineers; 1–6 Dec 1991; Atlanta,
Technology. 1991;11(6):272–4. GA. New York: ASME; 1991. p. 71–3.

Critical Reviews™ in Biomedical Engineering

You might also like