You are on page 1of 26

= 28 —

Hyperthermia

METHODS OF HEATING THERMAL DOSE


CELLULAR RESPONSE TO HEAT THE INTERACTION BETWEEN HEAT AND
SENSITIVITY TO HEAT AS A FUNCTION OF RADIATION
CELL AGE IN THE MITOTIC CYCLE THERMAL ENHANCEMENT RATIO
EFFECT OF pH AND NUTRIENT HEAT AND THE THERAPEUTIC GAIN
DEFICIENCY ON SENSITIVITY TO FACTOR
HEAT HEAT AND CHEMOTHERAPEUTIC AGENTS
HYPOXIA AND HYPERTHERMIA HUMAN APPLICATIONS
RESPONSE OF ORGANIZED TISSUES TO HYPERTHERMIA ALONE
HEAT HEAT PLUS RADIATION: CURRENT STATUS
THERMOTOLERANCE OF CLINICAL STUDIES
HEAT-SHOCK PROTEINS HYPERTHERMIA AND IMPLANTS
HEAT AND TUMOR VASCULATURE POSTSCRIPT ON HYPERTHERMIA
HYPERTHERMIA AND TUMOR MECHANISMS OF ACTION OF
REOXYGENATION HYPERTHERMIA
TEMPERATURE MEASUREMENT SUMMARY OF PERTINENT CONCLUSIONS

In Greek mythology the father of hyperther- ferum non sanat, ignis sanat. Quae vergo ig-
mia was Prometheus, a demigod who stole nis non sanat, insanobilia repotari oportet"
fire from Olympus and taught men to use it. ("Those who cannot be cured by medicine can
For this act he was chained to a rock by Zeus, be cured by surgery. Those who cannot be
the chief of the gods, and a vulture fed daily cured by surgery can be cured by fire [hyper-
on his liver. thermia]. Those who cannot be cured by hy-
The use of hyperthermia for the treatment perthermia, they are indeed incurable.")
of cancer is certainly not new. The very first The attempt in modern times to exploit el-
medical text known today contains a case evated temperatures to treat cancer has a his-
study describing a patient with a breast tumor tory longer than the use of ionizing radiations.
treated with hyperthermia. The case is found In 1866, 30 years before Rontgen discovered
in the Edwin Smith Surgical Papyrus, an "a new kind of ray," the German physician W.
Egyptian papyrus roll, which dates from more Busch described a patient with a sarcoma in
than 5,000 years ago. Later, heat was used in the face that disappeared after a prolonged in-
all cultures as one of the most prominent fection with erysipelas, an infectious disease
medical therapies for almost any disease, in- normally characterized by high fever. This
cluding cancer. Thus, Hippocrates (470-377 and similar cases led the New York surgeon
BC), in one of his aphorisms, states, "Quae William B. Coley to believe that the bacteria
medicamenta non sanat, fentm sanat. Quae causing erysipelas may be effective against

495
496 RADIOBIOLOGY FOR THE RADIOLOGIST

cancer. He extracted a toxin (Coley's toxin, or in this simplest of situations the tumor may
mixed bacterial toxin), with which he treated not be at the same temperature as the skin.
a number of patients. If localized hyperthermia is achieved by
Although it is difficult to evaluate the di- microwaves, radiofrequency-induced currents,
rect role of heat in this combined total-body or ultrasound, there are serious technical
hyperthermia and unspecific immunotherapy, problems and limitations. In the case of micro-
the work by Coley initiated a number of other waves, good localization can be achieved at
studies using local hyperthermia applied to shallow depths, but at greater tumor depths,
patients and tumors in experimental animals. even if the frequency is lowered to allow
In 1898, Westermark, a Swedish gynecolo- deeper penetration, the localization is much
gist, published a paper describing a marked poorer and surface heating limits therapy. If
regression of large carcinomas of the uterine ultrasound is used, the presence of bone or air
cervix after local hyperthermia, although the cavities causes distortions of the heating pat-
treatments involved were poorly controlled tern, but adequate penetration and good uni-
and the cases largely anecdotal. The use of hy- form temperature distributions can be
perthermia. either alone or in combination achieved in soft tissues, particularly with ul-
with radiation, has been attempted at irregular trasound in focused arrays. In practice, then,
intervals over the years but has never found a tumors such as recurrent chest-watl nodules
permanent place in the management of can- can be treated adequately with microwaves,
cer. Historical reviews of the early clinical use and it should theoretically be possible to heat
of hyperthermia can be found in several ex- deep-seated tumors below7 the diaphragm with
cellent articles (eg., by Dewey and col- focused ultrasound, regional microwave de-
leagues, 1977, and by Overgaard, 1984). vices, or interstitial techniques.
The interest in hyperthermia at the present In all cases, however, present methods of
time is based on documented clinical evi- heating pose a problem, though significant
dence of tumor regression as well as a bio- progress has been made as a result of the
logic rationale and encouraging results from clever application of focused arrays. For ex-
laboratory experiments. ample, the use of multiphased arrays allows
uniformity of temperature to the edge of the
field. The picture is complicated, and it is un-
METHODS OF HEATING likely that one simple answer to all the com-
The major problem in the development of plex problems will be found. This is the area,
clinical hyperthermia for cancer therapy has then, in which engineering developments are
been the design of equipment to heat desig- needed and a breakthrough in basic science
nated tumor volumes accurately and uni- would be welcome.
formly. Methods of local heating include (1) One method of producing localized hyper-
shortwave diathermy, (2) radiofrequency-in- thermia that suffers from fewer problems is
duced currents, (3) microwaves, and (4) ultra- the use of implanted microwave or radiofre-
sound. quency sources. Good temperature distribu-
Most of the work with cells cultured in tions can be achieved and maintained if ra-
vitro, as well as much of the early animal ex- diofrequency-induced currents or microwaves
perimentation, involves heating by hot water are applied to an array of wires actually im-
baths. The simplest and most reliable way to planted in the tumor and surrounding tissues.
heat a petn dish or a tumor transplanted into The "wires" used are frequently radioactive
the leg of a mouse is to immerse it totally in a sources, so that heat and radiation can be
thermostatically controlled bath of water. Wa- combined. Alternatively, microwave coaxial
ter temperature can be controlled within a antennae can be inserted into the catheters
fraction of a degree, and temperature mea- used to hold the radioactive iridium-92 wires,
surement involves no problem, although even and deep tumor volumes heated from the in-
HYPERTHERMIA 497

side out. Some of the most promising results sequently. The similarity in the shape of the
have been obtained in this way for the small cell-survival curves for heat and x-rays is mis-
fraction of patients with cancer for whom an leading. It is important, therefore, not to draw
interstitial implant is practical. conclusions for heat based on the interpretation
of radiation dose-response curves, because the
amount of energy involved in cell inactivation
CELLULAR RESPONSE TO HEAT is a thousand times greater for heat than for x-
Heat kills cells in a predictable and repeat- rays. This reflects the different mechanisms in-
able way. Figure 28.1 shows a series of survival volved in cell killing by heat and x-rays.
curves for cells exposed for various periods of Families of survival curves similar to those
time to a range of temperatures from 41.5°C to in Figure 28.1 have been obtained for many
46.5°C. The cell-survival curves for heat are different cell types, and it is clear that cells
similar in shape to those obtained for x-rays differ widely in their sensitivity to hyperther-
{i.e., an initial shoulder followed by an expo- mia. As with ionizing radiations, there is no
nential region), except that the time of expo- consistent difference between normal and ma-
sure to the elevated temperature replaces the lignant cells, in spite of numerous individual
absorbed dose of x-rays. For lower tempera- reports claiming that tumor cells are inher-
tures the picture is complicated because the ently more sensitive to heat than their normal
survival curves flatten out after a protracted ex- tissue counterparts.
posure to hyperthermia, indicating the devel- Survival data for cells exposed to various
opment of a resistance or tolerance to the levels of hyperthermia (taken from Figure
elevated temperature. This is discussed sub- 28.1) are replotted in Figure 28.2, with 1/Do

0 100 200 300 400 500


EXPOSURE TO HYPERTHERMIA (MINUTES)
Figure 28.1. Survival curves for mammalian cells in culture (Chinese hamster ovary line) heated at
different temperatures for varying lengths of time. (Adapted from Dewey WC, Hopwood LE, Sapareto
LA, Gerweck LE: Cellular responses to combinations of hyperthermia and radiation. Radiology 123:
463-474, 1977, with permission.)
498 RADIOBIOLOGY FOR THE RADIOLOGIST

415 42.5 43.5 44.5 45.5 46.5


,0

Activation energy
1.0 148Kcal/mole

Activation energy
10- 2 365 Kcal/mole

10" JL J_ _L
3!8 317 316 315 314 313
1/T (x 105)

Figure 28.2. An. Arrhenius plot for heat inactivation of mammalian cells in culture. The reciprocals of
the Do values obtained for Fig 28.1 are plotted versus reciprocal of the absolute temperature.
(Adapted from Dewey WC, Hopwood LE, Sapareto LA, Gerweck LE: Cellular responses to combi-
nations of hyperthermia and radiation. Radiology 123:463-474, 1977, with permission.)

on the ordinate and 1/T on the abscissa. T is above 43°C, it cannot. On this basis the in-
the absolute temperature; Do is the reciprocal trinsic heat sensitivity is that observed above
of the slope of the exponential region of the 43°C; cell survival for continuous heating be-
survival curve (i.e., the time at a given tem- low this temperature is an expression of heat
perature that is necessary to reduce the frac- sensitivity modified by the induction, devel-
tion of surviving cells to 37% of their former opment, and decay of thermotolerance. The
value). This type of presentation is known as similarity of the activation energy for protein
an Arrhenius plot; its slope gives the activa- denaturation to the activation energy for heat
tion energy of the chemical process involved cytotoxicity, calculated from the Arrhenius
in the cell killing. The dramatic change of analysis, led to the hypothesis that the target
slope that occurs at a temperature of about for heat' cell killing may be a protein. The
'43°C. which means that the activation energy structural chromosomal proteins, nuclear ma-
is different above and below this temperature, trix and cytoskeleton repair enzymes, and
may reflect different mechanisms of cell membrane components all have been identi-
killing (i.e., different targets for cytotoxicity fied as possible targets that are denatured by
above and below 43°C). On the other hand, it hyperthermia.
may equally well be a manifestation of ther- The Arrhenius plot for a given cell line can
motolerance. Below 43°C, thermotolerance be modified by a number of things. For ex-
can develop gradually during the heating; ample, altering the pH of the cells raises the
HYPERTHERMIA 499

curves, and the break point occurs at a higher tively by hyperthermia compared with the
temperature. slowly turning over normal tissues responsi-
ble for late effects that are in a Gi or Go state.
SENSITIVITY TO HEAT AS A
FUNCTION OF CELL AGE IN THE EFFECT OF pH AND NUTRIENT
MITOTIC CYCLE DEFICIENCY ON SENSITIVITY TO
HEAT
The age-response function for heat com-
plements that for x-rays (Fig. 28.3). The phase Cells at acid pH appear to be more sensitive
of the cycle most resistant to x-rays, late in to killing by heat. This is certainly true of
the DNA synthetic phase (late S), is most sen- cells treated with heat soon after their pH is
sitive to hyperthermia treatment. On this ba- altered artificially by adjusting the buffer. The
sis, cycling tumor cells should be killed selec- pH dependence of cytotoxicity at elevated

1.0

i r
600-rad
x-rays
0.1
o
o

I
CD
\ -

2
5

-j 0.01
15 min
^-hyperthermia

- /

- /

0.001 W Gi

i i ~T r~~n r
0 2 4 6 8 10 12
HOURS AFTER PLATING MITOTIC CELLS
Figure 28.3. Comparison of the fraction of cells surviving heat or x-irradiation delivered at various
phases of the cell cycle. The heat treatment consisted of 15 minutes at 45.5°C, and the x-ray dose
was 600 rad (6 Gy). (Adapted from Westra A, Dewey WC: Variation in sensitivity to heat shock dur-
ing the cell cycle of Chinese hamster cells in vitro. Int J Radiat Biol 19:467-477, 1971, with permis-
sion.)
500 RAD1OBIOLOGYFOR THE RADIOLOGIST

temperatures, however, is affected by pH his- not result from hypoxia per se but may be a
tory. Cells can adapt to pH changes and avoid consequence of the lowered pH and the nutri-
the heat sensitivity shown for low pH. tional deficiency that cells suffer as a result of
Cells deficient in nutrients are certainly prolonged hypoxia. Of utmost importance in a
heat sensitive. This can be demonstrated with practical situation is that hypoxic cells in tu-
cells in culture in which sensitivity to heat in- mors are often both nutrient-deficient and at a
creases progressively as cells have their en- lowered pH.
ergy supply compromised, either by depriving
them of glucose or by the use of a drug that
uncouples oxidative phosphorylation. RESPONSE OF ORGANIZED TISSUES
TO HEAT
These conclusions about pH and nutrients,
obtained under controlled conditions with Normal tissues respond to heat in a way
cells in culture, led to the speculation that that is substantially different from their more
cells in tumors that are nutritionally deprived familiar response to x-rays. The principal dif-
and at acid pH because of their location re- ference is that after irradiation, cells die only
mote from a blood capillary may be particu- in attempting the next or a subsequent mitosis
larly sensitive to heat. Because of their envi- (except in very unusual circumstances),
ronment, it is likely that these cells are out of whereas heated cells die by apoptosis, so that
cycle and possibly hypoxic, also. This conclu- heat damage is expressed early. In addition,
sion certainly correlates with the observation heat affects differentiating as well as dividing
that large necrotic tumors often shrink dra- cells. The familiar delay between exposure to
matically after a heat treatment. In this con- x-rays and the subsequent response of a cell-
text, also, heat and x-rays appear to be com- renewal tissue is because moderate doses of
plementary in their action, because the cells x-rays kill the dividing stem cells and leave
that are most resistant to x-rays (out of cycle the differentiated cells functional; the re-
and hypoxic because of their remoteness from sponse is delayed for a time that is related to
a capillary) show enhanced sensitivity to heat. the natural lifetime of the mature differenti-
A further complicating factor here is that re- ated cells and the time it takes for stem cells
gions of a tumor in which the vasculature is to progress through the process of differentia-
poorly developed tend to be at an elevated tion and become functional. This delay is ab-
temperature because the cooling effect of sent in the case of heat because all cells are af-
blood flow is reduced. fected, differentiated or dividing; the damage
to the tissue is expressed immediately.
As a result of these considerations, experi-
HYPOXIA AND HYPERTHERMIA
mental assay systems based on clonogenic
The response of hypoxic cells constitutes a survival may underestimate the effect of hy-
vital difference between x-rays and hyperther- perthermia seriously.
mia. Hypoxia protects cells from killing by x-
rays. By contrast, hypoxic cells are not more
THERMOTOLERANCE
resistant than aerobic cells to hyperthermia;
indeed, the evidence suggests that under some The development of a transient and nonher-
conditions they may be slightly more sensi- itable resistance to subsequent heating by an
tive. initial heat treatment has been described vari-
Cells made acutely hypoxic and then ously as induced thermal resistance, thermal
treated with heat have a sensitivity similar to tolerance, or, most commonly, thermotoler-
aerated cells. Cells subject to chronic hypoxia ance. In 1975, Henle and Leeper and Gerner
{i.e., deprived of oxygen for prolonged peri- and Schneider independently showed that the
ods) show a slightly enhanced sensitivity to resistance induced in cells by one heat expo-
heat. This increased sensitivity probably does sure exceeded anything that could be ex-
HYPERTHERMIA 501

pected from repair of sublethal damage or riod after an exposure of 2 or 3 hours. This
progression into a resistance phase of the cy- phenomenon is already apparent by the
cle. Figure 28.4 illustrates the phenomenon of change of slope in the survival curves of Fig-
thermotolerance. If heating at 44°C is inter- ure 28.1. Second, at temperatures above 43°C,
rupted after 1 hour and resumed some 2 hours thermotolerance cannot be produced during
later, the dose-response curve is much shal- the heating, and it takes some time to develop
lower (i.e., the cells have become resistant) after the heating has been stopped. It then de-
than if heating had been continued. Opera- cays slowly.
tionally, there are two ways in which heating Thermotolerance is a substantial effect; the
can induce thermotolerance. First, at lower slope of a survival curve may be altered by a
temperatures of around 39°C to 42°C, ther- factor of 4 to 10, which translates into a dif-
motolerance is induced during the heating pe- ference in cell killing of several orders of
magnitude. It is a factor to be reckoned with
in fractionated hyperthermia. For cells in cul-
ture the time taken for cells that have become
100-k
thermotolerant to revert to their normal sensi-
tivity (i.e., the decay of thermotolerance) may
be as long as 160 hours. Groups led by Over-
gaard and by Urano have shown that thermo-
tolerance can be induced in transplantable
mouse tumors and that this thermotolerance
10- also decays very slowly, requiring something
on the order of 120 hours before the decay is
complete.
There is evidence from the work of Field
and Law and their colleagues at Hammer-
(0 smith that in normal tissue systems, such as
gut, skin, and cartilage, the appearance of
3
CO thermotolerance may not reach a maximum
until 1 or 2 days after heating, depending on
the initiating treatment. It may take as long as
1 or 2 weeks to decay completely.
Thermotolerance is a serious problem in the
0.1
clinical use of hyperthermia. Figure 28.5 illus-
trates why by contrasting heat and radiation.
The top graph shows the familiar pattern for a
multifraction regimen of x-rays given in daily
doses, in which the shoulder must be repeated
0.01 each time and each dose produces about the
1 2 3 same amount of cell killing. The bottom graph
Total time at 44° hr) shows a strikingly different pattern for hyper-
Figure 28.4. Development of thermotolerance
thermia. The first heat dose kills a substantial
in HeLa cells. Closed circles indicate cell survival fraction of cells, but subsequent daily treat-
to single heat exposures; open circles indicate ments are comparatively ineffective because of
response of cells treated at 44°C for 1 hour, re- the development of thermotolerance, which oc-
turned to 37°C for 2 hours, and given second curs a few hours after the first treatment and
graded doses of 44°C for graded times. (From may take as much as a week to decay. Because
Gerner EW, Schneider MJ: Induced thermal re-
sistance in HeLa cells. Nature 256:500-502, of the problems and uncertainties involved,
1975, with permission.) Field advises, "The best way to deal with ther-
502 RADIOBIOLOGY FOR THE RADIOLOGIST

Cell Survival after Multiple Doses

D RADIATION

CO

A given
reaction
on

Tota.1 Radiation Dose


1 .Oh
HYPERTHERMIA
en

D2
$

A given
reaction I

Total Treatment Time

Figure 28.5. Why the development of thermotolerance is such a problem in a daily fractionated reg-
imen. Top: X-rays. Each dose in a fractionated regimen has about the same effect (i.e., kills the same
proportion of cells). The shoulder of the curve must be reexpressed for each dose fraction. Bottom:
Hyperthermia. The first heat treatment results in a substantial biologic effect but also triggers the de-
velopment of thermotolerance, which may take as long as a week to decay. Subsequent daily heat
treatments would be relatively ineffective because of the acquired thermoresistance of the cells.
(From Urano M: Kinetics of thermotolerance in normal and tumor tissues: A review. Cancer Res
46:474-482, 1986, with permission.)

motolerance is to avoid it." This advice gener- kd) are produced. The appearance of these
ally has been followed in the clinical use of hy- heat-shock proteins tends to coincide with the
perthermia. which imposes a limit of one or at development of thermotolerance and their
most two heat treatments per week. disappearance with the decay of thermotoler-
ance. Although the correlation is clear, it is
not known if the heat-shock proteins are in-
HEAT-SHOCK PROTEINS
volved in the mechanism of the production of
If cells are exposed to heat, proteins of a thermotolerance or an independent manifesta-
defined molecular weight (mainly 70 or 90 tion of the heat insult. In fact, although they
HYPERTHERMIA 503

have been given the name heat-shock pro- hours later. Comparison with the control indi-
teins, they are produced after treatment with cates a molecular weight of about 70 to 110
other agents, including arsenite and ethanol. kd. The time of appearance of these proteins
Proteins of roughly the., same molecular' coincides with the development of thermotol-
weight are found in cells of many species and erance.
first were discovered, in fact, in the fruit fly
Drosophila. They appear to be a ubiquitous
HEAT AND TUMOR VASCULATURE
cellular response to stress.
Heat-shock proteins are identified by gel Tumors in general have a less organized
electrophoresis, in which they show up as and less efficient vasculature than most nor-
clearly defined bands of specific molecular mal tissues. All functional capillaries in tu-
weights. Methionine labeled with sulfur-75 is mors are open and used to capacity, even un-
used to label the protein in the treated cells, der ordinary conditions; in normal tissues
which then are run on a polyacrylamide gel in many capillaries are closed under ambient
an electric field. The proteins move a distance conditions. Consequently, although blood
that depends on their molecular weight, with flow may be greater in tumors (particularly
smaller proteins going farther. This separates those that are small) compared with normal
out the various proteins in the cell (Fig. 28.6). tissues at physiologic temperatures, the ca-
After a treatment of 45°C for 20 minutes, pro- pacity of tumor blood flow to increase during
teins that are present only in small quantities heating appears to be rather limited in com-
before heat treatment are synthesized some parison with normal tissues. It is well docu-

Colon Carcinoma
Molecular wt
kitodaltons

Figure 28.6. Autoradiograph of cells


labeled with methionine labeled with
sulfur-35 in a polyacrylamide gel, to il-
lustrate the synthesis of heat-shock
proteins in cells derived from human
colon carcinoma. The left column
shows the normal pattern of protein
synthesis in untreated cells. The other
columns show patterns of protein syn-
thesis at various times (0, 2, 6, and 24
hours) after a heat treatment of 20 min-
utes at 45°C. Immediately after treat-
ment (0 hours), protein synthesis is
Actin completely shut down. At 2 and 6 hours
after heat treatment, proteins of molec-
ular weight 70, 90, and 110 kd are
overexpressed, as evidenced by the
dark bands. These are the heat-shock
proteins whose appearance coincides
Con 0' 2 6 with the development of thermotoler-
ance. (Courtesy of Dr. Laurie Roizin-
Time after heating (hrs)
Towle.)
504 RADIOBIOLOGY FOR THE RADIOLOGIST

shunts open
Normal Tissue

damaged
neo-v/ascu!ature
Blood flow
decreases

Heat
retained

Temperature
Tumor rises
\

Figure 28.7. Possible mechanism explaining why tumors get hotter than surrounding normal tis-
sues. Normal tissues have a relatively high ambient blood flow, which increases in response to ther-
mal stress, thereby dissipating heat. Tumors, with relatively poor blood flow and unresponsive neo-
vasculature, are incapable of augmenting flow (i.e., shunting blood) and act as a heat reservoir. (Idea
courtesy of Dr. F. K. Storm.)

11 -

10-

T3
O
_O
CD

CD
O>
c
CD
sz
O
o Figure 28.8. Relative changes in blood flow
in the skin and muscle of a rat and in vari-
S ous animal tumors at different tempera-
cc tures. In general, blood flow increases in
normal tissues during hyperthermia and de-
creases in tumors, often leading to differen-
tial tumor heating. (From Song CW: Effect of
i
38 39 40 41 42 43 44 45 46 47 48 local hyperthermia on blood flow and mi-
croenvironment: A review. Cancer Res
Temperature °C 44(suppl):4721S-4730S, 1984, with per-
(Heating for 30-40 Mm.] mission.)
HYPERTHERMIA 505

mented that in normal tissues heat induces a higher than in the surrounding normal tissues.
prompt increase in blood flow accompanied In a practical situation, therefore, the differ-
by dilation of vessels and an increase in per- ence in intrinsic sensitivity between normal
meability of the vascular wall. As a result, and malignant tissues becomes a moot point,
heat dissipation by blood flow is slower in tu- because the tumor is often hotter anyway. A
mor than in normal tissues, and so it often is postulated mechanism for the selective solid
found that the temperature within a tumor is tumor heating is shown in Figure 28.7. Lest it

J9EHMK

B m&m
Figure 28.9. Hyperthermia-induced compression and occlusion of tumor vessels (x 10). The pho-
tographs are of a squamous cell carcinoma grown in a transparent cheek pouch chamber of a Syr-
ian hamster. A: Vascular pattern at 34°C before heating. Note the prominent feeding arteriole. B: Oc-
cluded vascular network after 30 minutes of heating at 45°C. Blood flow to the area stopped because
of marked constriction of the arteriole. Interstitial pressure then exceeded hydrostatic pressure to
compress tumor vessels. (From Eddy HA: Radiology 137:515-521, 1980, with permission.)
506 RADIOBIOLOGY FOR THE RADIOLOGIST

should be thought that this is a function of the Temperature|


artificial "encapsulated" nature of most trans-
planted tumors in experimental animals, it
should be noted here that differential heating Biood Flow ||
of tumors relative to normal tissues frequently
has been observed in humans in clinical trials
PO, pH Nutrients]
of hyperthermia. It may be one of the reasons
why excessive normal tissue damage has sel-
\ \
dom been observed m clinical studies with
heat. Cell death
There is, however, more to the story of Figure 28.10. The complex interplay between
heat and tumor vasculature. For reasons that hyperthermia and blood flow. An elevated tem-
are not fully understood, heat appears to perature leads to direct cell killing, but it also
causes a reduction in blood flow in tumors; this,
preferentially damage the fragile vasculature in turn, causes changes in pH, pC>2, and nutri-
of tumors; as a consequence, the -heat-in- ents, leading to enhanced cell killing. The reduc-
duced change in blood flow in at least trans- tion in blood flow also further elevates the tem-
plantable tumors in animals is quite different perature in the tumor, because heat is not being
from that in normal tissues. The relative carried away. (Based on the ideas of Dr. Chang
Song.)
change in blood flow in the skin and muscle
of rats, as well as in various transplanted tu-
mors, is summarized graphically in Figure
28.8. After hyperthermia, tumor blood flow cells are affected, leading to enhanced cell
in most cases goes down: blood flow in rep- killing.
resentative normal tissues goes up by a fac-
tor of 8 to 10. This further exacerbates the
difference in blood flow and further in- HYPERTHERMIA AND TUMOR
creases the temperature of the tumor com- OXYGENATION
pared with the surrounding normal tissues. Early studies with transplanted tumors in
Eddy has studied the microcirculation of tu- rodents showed that heat caused vascular
mors after hyperthermia at 41 °C to 45°C. At damage. It was concluded therefore that care
the higher temperatures, particularly, com- was needed in combining heat and radiation,
pression, occlusion, hemorrhage, and stasis because the oxygen status of the tumor would
thrombosis were observed (Fig. 28.9). He be compromised by heating. Flowever, it turns
concluded that pathophysiologic changes in out that tumor vasculature in spontaneous hu-
the tumor microvasculature during hyper- man tumors is considerably more resistant to
thermia can play an important role in tumor thermal damage than in transplanted tumors
response and may account for cure rates that in mice, and that in any case, much of the
are better than would be predicted from di- early work with mice utilized thermal doses
rect cell killing. far in excess of those that can be achieved
There is a complex interplay in tumors be- clinically. It now is recognized from animal
tween hyperthermia, blood flow, and cell studies that mild hyperthermia actually can
killing; this is illustrated in Figure 28.10. If a promote tumor reoxygenation. with the de-
tumor is heated to an elevated temperature, gree of reoxygenation correlating with the
there is direct cell killing, but there is also a level of cytotoxicity. This also has been con-
reduction in blood flow. This causes the tumor firmed in a clinical study of patients with
to get even hotter, because the reduced blood soft-tissue sarcomas. Brizel and colleagues
flow carries less heat away and at the same showed that one heat treatment led to reoxy-
time the pH, pCb, and nutrient status of the genation within 24 to 48 hours, whereas there
HYPERTHERMIA 507

was no measurable reoxygenation during a is exceeded by 90% of the measurements


week of standard radiation therapy. within the tumor.
As demonstrated by both in vitro and in
vivo experiments, hyperthermia cytotoxicity
TEMPERATURE MEASUREMENT
is dependent on both temperature and time.
The control and measurement of tempera- The time-integrated temperature descriptor,
ture in tissues, if heating is achieved by an namely cumulative equivalent minutes at
external source, is not a trivial problem. If 43°C, is perhaps the best concept of thermal
microwaves, short-wave diathermy, or radio- dose.
frequency-induced currents are used, accu- A formal definition of thermal dose might
rate temperature measurements cannot be be: the time in minutes for which the tissue
made with metal thermometers, such as ther- would have to be held at 43°C to suffer the
mocouples or thermistors, because of direct same biologic damage as produced by the ac-
heating of the electrically conducting compo- tual temperature, which may vary with time
nents and the perturbations of the electro- dining a long exposure.
magnetic field. These initial problems were Because in practice the tumor temperature
solved largely by the development of non- may vary up and down during a typical treat-
metallic, temperature-sensitive crystal and ment, some method is needed to convert the
fiberoptic probes. varying times at various temperatures into
It is clearly desirable to replace invasive equivalent minutes at 43°C. Probably the best
methods that measure temperature at a lim- method proposed is to use the relationship be-
ited number of points with a nonmvasive tween treatment time and temperature for a
technique that gives an accurate picture of the biologic isoeffect (the Arrhenius plot, shown
temperature distribution. The most developed for cells in culture in Figure 28.2). This has
method of noninvasive thermometry is based been confirmed in principal for a variety of
on magnetic resonance imaging. A conven- normal tissues and tumors. A marked change
tional magnetic resonance scanner, measur- of slope occurs somewhere between 42°C and
ing the chemical shift of water, can yield a 43°C. Above this transition temperature, the
0.5°C resolution m 0.02-cm3 voxels in both slope is consistent for a variety of cells and
normal and malignant tissues. This and simi- tissues. It generally is agreed that a 1°C rise of
lar ideas are likely to receive a lot of attention temperature is equivalent to a reduction of
in the future. time by a factor of 2. Consequently, above this
transition temperature,

THERMAL DOSE 1± _ 9TI-T2


ti
The temperature distribution m a tumor
in which tj and tj are the heating times at tem-
during hyperthermia treatment is almost al-
peratures Tj and T2 to produce equal biologic
ways nonuniform. This nonuniformity stems
effect.
from two spatially varying sources: power de-
For temperatures below the transition tem-
position and tumor blood perfusion, which
perature, an increase in temperature by PC-
carries the heat away.
requires that time be decreased by a factor of
The most common descriptors of the mea-
4 to 6:
sured temperature distribution that are be-
lieved to correlate with biologic outcome are — = (4 to 6)T1~T2
the minimum tumor temperature, because ti
clonogens surviving in any region of lower The equivalent heating time at 43°C—the
temperature may constitute a focus for the re- thermal dose—may be calculated from one or
growth of the tumor, and the temperature that the other of these expressions or a combina-
508 RADIOBIOLOGY FOR THE RADIOLOGIST

tion of both. In principle, at least, the heat ied with cells in culture and with experimen-
dose associated with a changing temperature tal animal systems.
may be calculated as the sum of equivalent In the case of cells cultured in vitro, the
heating times at 43°C for each temperature.- shape of the x-ray survival curve is changed
Although the concept of thermal dose is at- by the addition of heat.
tractive, there are problems in its implementa- For acute hyperthermia (i.e., brief expo-
tion: sures to temperatures of about 45°C, which
lead to a substantial amount of cell killing)
• Nonuniformity of temperature occurs
the principal effect of hyperthermia is a steep-
throughout the tumor.
ening (i.e., reduced Do) of the x-ray survival
• The concept relates only to cell killing by
curve. The change in the shoulder of the curve
heat and does not include radiosensitiza-
is minimal. On the other hand, if a more mod-
tion. est level of hyperthermia is used (4CM3°C),
• It relates to one heat treatment, so it is not which involves little or no cell killing, the
possible to add one treatment to the next principal effect observed is a removal of the
given a few days later, because of the prob- shoulder from the x-ray survival curve, and
lem of thermotolerance. then heat treatment after irradiation is the
more effective sequence. These differences
between higher and lower temperatures, asso-
THE INTERACTION BETWEEN HEAT ciated with the break in the Arrhenius plot at
AND RADIATION around 43 °C, may reflect different critical tar-
The biologic effect of a combination of gets or simply be a consequence of the fact
heat and radiation may be a consequence of: that thermotolerance can develop during the
treatment at lower temperatures.
1. The independent but additive cytotoxic
Heat inhibits the repair of radiation-in-
effects of the heat and radiation, with
duced single-strand breaks and radiation-in-
their complementary patterns of sensitiv-
duced chromosome aberrations. This inability
ity through the cell cycle and the greater
to repair molecular damage translates into the
sensitivity to heat of nutritionally de-
inability to repair both sublethal damage and
prived cells at low pH
potentially lethal damage produced by radia-
2. The interaction between heat and radia- tion. Repair of sublethal damage does not oc-
tion, in the form of sensitization of the ra- cur if hyperthermia is applied during the in-
diation cytotoxicity by heat, resulting terval between the two doses of x-rays.
from the inhibition of repair of radiation-
If heat and radiation are combined, an im-
induced damage
portant consideration is sequencing. Sequenc-
In a practical situation in the clinic, the in- ing in vitro is discussed first. Figure 28.11
teraction is most likely to be the independent shows the fraction of cells that survived a
and additive cytotoxic effects as described in combination of 5 Gy (500 rad) of x-rays and
the first point listed, because of the modest 40 minutes of heating at 42.5°C when the se-
heat levels that can be achieved, and the time quence of the two treatments and the time in-
interval usually used between hyperthermia terval between them were varied. The heat
and radiation treatment. Of course, both may treatment itself did not kill any cells, but it po-
occur simultaneously, in which case the com- tentiated the effect of the x-rays, even if given
bination of hyperthermia and x-rays results in an hour or more before or after the radiation.
a greater cytotoxicity than can be accounted The greatest reduction in cell survival was
for by the addition of the cytotoxic effects of caused by irradiation during heating. Compa-
the agents alone; that is, the interaction be- rable data for mouse skin are shown in Figure
tween the two modalities is synergistic, or 28.12. Heating at 43°C for 1 hour was done
supra-additive. This interaction has been stud- before or after a single 20 Gy (2,000-rad)
l.Op
ALONE

O
10"
§ - 500-rad X-RAYS
^~~ ALONE

120 100 80 60 40 20 0 20 40 60 80 mm
X-RAYS BEFORE HEAT X-RAYS AFTER HEAT

Figure 28.11. Survival of Chinese hamster cells irradiated with 5 Gy (500 rad) of x-rays before, dur-
ing, or after a heat treatment of 40 minutes at 42.5°C. No killing was observed with this heat treat-
ment alone (open triangle). The effect of 5 Gy (500 rad) of x-rays alone is shown by the arrow. There
is a clear interaction between heat and x-rays, with the maximum effect being produced if the x-rays
are delivered midway through the heat treatment. (Adapted from Sapareto SA, Hopwood LE, Dewey
WC: Combined effects of x-irradiation and hyperthermia on CHO cells for various temperatures and
orders of application. Radiat Res 43:221-233, 1978, with permission.)

9r-

Response due
- - t o 2000-rad
x-rays alone

Response due
to 43°C for
0 hour alone
T
6 4 2 0 2 4 6 8 HOURS
HEAT BEFORE X-RAYS t HEAT AFTER X-RAYS

X-RAYS
Figure 28.12. Response of mouse skin to the effect of combined heat and x-rays. The heat treat-
ment was 43°C for 1 hour, and the x-ray dose was 20 Gy (2,000 rad). Heat was given before or af-
ter irradiation, and the time interval between the two was varied. (Adapted from Field SB, Hume S,
Law MP, Morris C, Myers R: Presented at the International Symposium on Radiological Research
Needed for the Improvement of Radiotherapy, Vienna, November 22-26, 1976, with permission.)
510 RADIOBIOLOGYFOR THE RADIOLOGIST

dose of x-rays. There was a marked variation zation has been studied, typical TER values
in the normal tissue response after a given are 1.4 at 41°C, 2.7 at 42.5°C, and 4.3 at
dose of x-rays, depending on the time interval 43°C, with heat applied for 1 hour.
between heat and irradiation and on the order
in which the two treatments were given. Heat-
HEAT AND THE THERAPEUTIC GAIN
ing immediately before or immediately after
FACTOR
irradiation produced a similar effect.
Although many experiments have been per- The therapeutic gain factor can be de-
formed with cultured cells and with labora- fined as the ratio of the TER in the tumor to
tory animals to investigate the interaction of the TER in normal tissues. There is no advan-
heat and radiation, if hyperthermia is used in tage to using heat plus lower doses of x-rays if
the clinic as an adjunct to radiotherapy, their there is no therapeutic gain compared with the
effects are probably not interactive at all. Be- use of higher doses of x-rays alone.
cause the daily doses used in conventional The question of a therapeutic gain factor is
fractionated radiotherapy are small (2 Gy) complicated in the case of heat, because the
and the levels of heating achieved are modest, tumor and normal tissues are not necessarily
the cytotoxicity of the heat and radiation are at the same temperature. If the statement is
more probably independent but additive. made that heat preferentially damages tumor
cells compared with normal tissue, it is im-
plied that they both are at the same tempera-
THERMAL ENHANCEMENT RATIO ture. In a practical situation, however, this is
In the case of either normal tissues or trans- not always the case. For example, if a poorly
plantable tumors in experimental animals, the vascularized tumor is treated with mi-
extent of the interaction of heat and radiation crowaves, it may reach a higher temperature
is expressed in terms of the thermal en- than the surrounding normal tissue, because
hancement ratio (TER), defined as the ratio less heat is carried away by the flow of blood.
of doses of x-rays required to produce a given In addition the overlying skin can be cooled
level of biological damage with and without actively by a draft of air or even a cold water
the application of heat. pack. In these circumstances the normal tis-
The TER has been measured for a variety sues may be at a significantly lower tempera-
of normal tissues, including skin, cartilage, ture than the tumor, which therefore exagger-
and intestinal epithelium. The data form a ates the differential response m a favorable
consistent pattern of increasing TER with in- direction.
creasing temperature, up to a value of about 2 With these reservations in mind, there are
for a 1-hour heat treatment at 43°C. The TER still good reasons for believing that the effects
is more difficult to measure in transplanted of heat, alone or in combination with x-rays,
tumors m laboratory animals, because the di- may be greater on tumors than on normal tis-
rect cytotoxic effect of the heat tends to dom- sues, for reasons discussed previously.
inate. Heat often can control experimental tu-
mors with acceptable damage to normal
tissues, because cell killing by heat is strongly HEAT AND CHEMOTHERAPEU TIC
enhanced by nutritional deprivation and in- AGENTS
creased acidity, conditions that are typical of The cell-killing potential of some but not
the poorly vascularized parts of solid tumors. all chemotherapeutic agents is enhanced sub-
Thus a moderate heat treatment, which can be stantially by a temperature elevation of even a
tolerated by well-vascularized normal tissues, few degrees. This is illustrated for cisplatin in
destroys a large proportion of the cells of Figure 28.13. There is also a striking syner-
many solid tumors in experimental animals. gism in cytotoxicity if hyperthermia (42-
In those cases in which thermal radiosensiti- 43°C) is combined with either bleomycm or
HYPERTHERMIA 511

CIS-PLATINUM
1 Hour Treatmant

Figure 28.13. Effect of elevated tem-


peratures on the cytotoxicity of cisplatin
in V79 hamster cells in vitro. Cells were
heated for 1 hour at the temperatures
indicated. (From Roizin-Towle L, Hall
EJ, Capuano L: Interaction of hyper-
10
300
thermia and cytotoxic agents. Natl Can-
100 200
cer Inst Monogr 61:149-151, 1982, with
Dose (AJM) permission.)

doxorubicin. It is probable that at least part of not been done in this area in view of the sub-
the cell's sensitization to bleomycin at 43°C is stantial potential benefits. Table 28.1 is a list-
related to the inhibition of a repair mecha- ing of drugs that are potentiated by heat and
nism. No such explanation can account for the those that are not. There are several different
sensitization to doxorubicin, but it has been mechanisms that may be involved, some of
shown clearly that the drug gets into the cells which are listed in Table 28.2.
more easily at 43 than at 37°C, possibly be-
cause of a change in the properties of the
TABLE 28.1. Interaction of Heat and
plasma membrane at the higher temperature. Chemotherapeutic Agents
Once thermotolerance develops, however, less
Effect Drug
doxorubicin gets into the cell.
Whatever the mechanisms involved, the Potentiated by heat Melphalan
Cyclophosphamide
synergism between heat and drugs may prove BCNU
very useful in the chemotherapy of solid tu- Cis DDP
mors. The addition of local hyperthermia to a Mitomycin C
Bleomycin
chemotherapy schedule would have the obvi- Vincristine
ous advantage of "'targeting" and localizing Unaffected by heat Hydroxyurea
the principal effect of the drug, allowing Methotrexate
Vinblastine
greater tumor cell killing for a given systemic Complex interaction Doxorubicin
toxicity. This would help to overcome one of
From Kano E: Hyperthermia and drugs. In Over-
the principal problems and limitations of gaard J (ed): Hyperthermic Oncology, pp 277-282.
chemotherapy. It is surprising that more has London, Taylor & Francis, 1985, with permission.
512 RADIOBIOLOGY FOR THE RADIOLOGIST

TABLE 28.2. Mechanisms of Interaction of however, perhaps because the fever rather than
Hyperthermia and Drugs the toxin was the tumoricidal agent. Because
Mechanism Drug the immune system undoubtedly was influ-
Increased rate of alkylation Thiotepa .. enced by Coley's toxins, one can speculate that
Nitrosoureas in those patients in whom permanent cures
Mitomycin C were obtained the cures could be the result of
Single-strand DNA breaks Bleomycin
Inhibition of repair Methylmethane immune stimulation rather than direct thermal
sulphonate effect on the tumor alone.
Lonidamine
Bleomycin
Altered drug uptakea Doxorubicin +/- HYPERTHERMIA ALONE
Bleomycin -
Melphalan +/-
Thiotepa -
It generally is agreed based on a consider-
Cyclo- able body of data, that local hyperthermia has
phosphamide - no role in the curative treatment of tumors. Re-
Methyl-CCNU - gardless of treatment schedules, the complete
Fluorouracil +
Methotrexate +/- response rate to heat alone does not exceed
Cisplatin +/- about 10% and seems to be of short duration.
Dactinomycin +/- Local tumors can be sterilized only at high tem-
Common membrane target Polyene antibiotics
Local anesthetics peratures, which cause significant damage to
Alcohols surrounding normal tissues. This is in agree-
Energy metabolism Several ment with experimental studies in the labora-
Production of oxygen radicals Several
tory, which suggest that rumor cells in a well-
Other mechanisms for the interaction of drugs and vascularized area under normal conditions have
heat have been suggested, including increased ability
of an agent to penetrate membranes and prolongation
about the same sensitivity as normal tissues.
by drugs of the heat-sensitive cell-cycle phases.
a
Nevertheless, local hyperthermia has been
+, A reported increase from hyperthermia: -, a re- shown to be useful in palliation, in the relief
ported decrease.
Based on Dahl O: Hyperthermia and drugs. In Wat- of pain. Treatment with heat alone, therefore,
mougl DJ, Ross WM [eds]: Hyperthermia, Blackie, plays a limited role in cancer, and it is now
Glasgow, 1986, with permission. usually recommended that it be performed
only m patients who have limited life ex-
pectancies or if conventional therapy is con-
HUMAN APPLICATIONS traindicated.
Hyperthermia has been widely used as a
form of cancer therapy. Its use in the treatment
HEAT PLUS RADIATION: CURRENT
of human tumors dates from early translations
STATUS OE CLINICAL STUDIES
of Ramajama (2000 BC). In 1891, Coley noted
that the regression of an inoperable "round cell Hyperthermia has been used extensively as
sarcoma of the neck" was associated with a an adjuvant to radiation in the treatment of lo-
febrile bout of erysipelas. At this stage it was cal and regional cancer, and this is the area in
not clear whether the shrinkage of the tumor which it appears to offer the biggest advan-
was caused by fever or a direct effect of the tage. The protocol that has emerged from the
bacterial toxins. In 1953. Nauts, Fowler, and clinic is of the sequential application of radi-
Bogatko repeated Coley's work, and 25 of their ation and heat, with one or two applications of
30 selected patients with soft-tissue sarcoma, heat interspersed with a normal multifraction
lymphosarcoma, or carcinoma of the cervix course of radiotherapy.
and breast were alive and disease-free at 10 The radiosensitizing effect of heat is most
years. The earlier results of Coley with the evident if the heat and radiation are applied si-
highly pyrogenic agents never were equaled multaneously, but the effect is generally of the
with lesser agents or systemic hyperthermia, same magnitude in both tumors and normal
HYPERTHERMIA 513

tissues and does not improve the therapeutic and neck nodes with full-dose radiation therapy
ratio unless the tumor is heated to a higher do not show any benefit from the use of hyper-
temperature than the normal tissue. On the thermia. It may be that heating techniques were
other hand, the hyperthermic cytotoxic mech- inadequate in these sites. Three trials show a
anism predominates if heat follows radiation clear and significant advantage for the addition
in a sequential procedure. The heat-sensitive of hyperthermia to standard radiation therapy.
cells are those found in a nutritionally de- These may be summarized as follows.
prived, chronically hypoxic and acidic envi-
ronment, conditions that are found in tumors
Superficial Localized Breast Cancer
but not generally seen in normal tissues. In
addition, as a result of heat killing of the hy- An international collaborative hyperthermia
poxic radioresistant tumor cells, the actual ra- group combined the results of five randomized
diation dose necessary to effectively control controlled clinical trials conducted in the
the tumor should be reduced. Thus, not only United Kingdom, Europe, and Canada, in
do we see a preferential enhancement occur- which external-beam radiation therapy alone
ring in tumors, but the overall radiation dam- was compared with radiation therapy plus hy-
age to normal tissues also is decreased, be- perthermia. Hyperthermia was induced using
cause the radiation dose is decreased. various externally applied electromagnetic ap-
Consequently, a therapeutic gam should re- plicators, aiming at a tumor temperature of
sult. 42.5 or 43°C. In fact, the minimum tempera-
A sequential administration of radiation and ture rarely exceeded 41°C, because of difficul-
heat has several other advantages over a simul- ties in heating. Not all trials demonstrated an
taneous treatment. The aim of giving heat is to advantage for the combined treatment, but the
improve the already existing radiotherapy treat- pooled data showed a significant improvement
ment. If heat is combined with radiation in a si- in complete tumor response. The greatest effect
multaneous protocol, it has to be given with was seen in patients with recurrent lesions in
each radiation fraction. Thermotolerance then previously treated areas, in whom further irra-
becomes a problem. This can be overcome by diation was limited to low doses.
increasing the fraction interval, but that would
be expected to reduce the effectiveness of the
radiation alone. With sequential heat and radia- Recurrent or Metastatic Malignant
tion treatment the heat can be administered Melanoma
once or twice, without interfering with the nor- Within the framework of the European So-
mal radiotherapy protocol. Last but not least, ciety for Hyperthermic Oncology, a multicen-
the sequential application of the two modalities ter randomized trial show improved local con-
is much more practical; to heat and irradiate si- trol at 2 years and survival at 5 years for
multaneously would be very difficult indeed. patients with recurrent malignant melanoma.
There is abundant evidence from phase I and The patients received three fractions of 8 or 9
II trials that heat may enhance radiotherapy to a Gy (800 or 900 rad) within a period of 8 days,
significant degree; these data were chronicled either alone or combined with hyperthermia
in some detail in previous editions of this book. aiming at 43°C for 60 minutes.
In more recent years, a number of randomized
phase III multicenter trials have been conducted
HYPERTHERMIA AND IMPLANTS
that must replace the more extensive, but less
convincing, data from the past. The results of The dose-rate effect was described in detail
these trials are summarized in Table 28.3. Some in Chapter 5. In short, if x- or y-rays are deliv-
show a benefit for the addition of hyperthermia ered at low dose rates, the biologic effective-
as an adjuvant to radiation therapy, and some do ness of a given dose is reduced because of the
not. The trials involving advanced breast cancer repair of sublethal damage that takes place dur-
514 RADIOBIOLOGYFOR THE RADIOLOGIST

TABLE 28.3. Results of Six Randomized Clinical Trials Comparing the Response of
Thermoradiotherapy Versus Radiotherapy Alone
Response
Complete Response, % Local Control, 2 y (%) Survival, 5 y (actuarial), %
Number
of
Site Lesions HT+RT RT HT+RT RT HT+RT RT
Head and Necka 65 46 12 25 8
(Stage III & IV)
Head and Neckfa 44 83 41 69 24 53 0
(Stage IV nodes)
Melanomac 134 62 32 46 28
Breast" (Primary 396 59 41 45 29
and recurrent)
Breast" (Recurrent, 210 57 31
previously irradiated)
Glioma e (Interstitial 64 Survival = Survival =
HT+RT and RT) 91 wk 76 wk;
P = 0.014
3 yr Survival
Pelvic Tumors'' P= P=0.014
0.005
Overall 360 56 39 32 24
p- P=0.01
0.013
Cervix1' (68 Gy) 115 83 57 52 27
Bladder'(66 Gy) 102 74 51 NS
Rectum' (56 Gy) 143 21 15 NS
All differences significant to P < 0.05.
HT + RT, thermoradiotherapy; RT, radiotherapy.
a
Datta NR, Bose AK, Kapoor HK, Gupta S. Head and neck cancers: Results of thermoradiotherapy versus ra-
diotherapy. Int J Hypertherm 6:479-486, 1990.
"Valdagni R, Amichetti M: Report of long-term follow-up in a randomized trial comparing radiation therapy and
radiation therapy plus hyperthermia to metastatic lymph nodes in stage IV head and neck patients. Int J Radiat
Oncol Biol Phys 28:163-169, 1994.
c
Overgaard J, Gonzalez Gonzalez D, Hulshof MCCM, et al.: Randomized trial of hyperthermia as adjuvant to
radiotherapy for recurrent or metastatic malignant melanoma. Lancet 345:540-543, 1995.
°Vernon CC, Hand JW, Field SB, et al.: Radiotherapy with or without hyperthermia in the treatment of superfi-
cial localized breast cancer. Results from five randomized controlled triais. Int J Radiat Oncol Biol Phys 35:
731-744, 1996.
e
Sneed PK, Stauffer PR, Diederich CJ, et al.: Survival benefit of hyperthermia in a prospective randomized trial
of brachytherapy boost ± hyperthermia for glioblastoma multiforme. 38th Am. Soc. Therap. Radioi. Oncol., Los An-
'geles, CA, October, 1996, (Abs 1).
Van der Zee J, Gonzalez Gonzalez D, Vernon CC, et al.: Therapeutic gain by hyperthermia added to radio-
therapy. ICRO/OGRO 6, 6th Int. Meeting on Progress in Radio-Oncology, Salzburg, Austria, May 13-17, 1998.

ing the protracted treatment time. Because heat pendent of the method used to produce the
inhibits the repair of sublethal damage, it is heating, some remarkable results have been ob-
logical to predict that the effectiveness of low tained with interstitial heating combined with
dose rate irradiation would be enhanced by the brachytherapy. The reason for this superior re-
simultaneous application of heat. This has been sponse is likely to be the relatively homoge-
investigated in detail in vitro and confirmed in neous heating distribution, in general far better
vivo., with TERs approaching 2. These reports than can be achieved by external heating.
inspired the use of hyperthermia and low dose Figure 28.14 shows the technique of com-
rate irradiation m implants in patients. Inde- bining hyperthermia with a low dose rate im-
HYPERTHERMIA 515

Figure 28.14. The use of a combination of hyperthermia and low dose rate irradiation for the treat-
ment of retrogastric squamous cell carcinoma. Left: Standard 14-gauge brachytherapy catheters
surgically implanted during an exploration laparotomy. Once exteriorized, they function as conduits
through which radioactive iridium-192 wires or microwave antennae can be inserted. Right: Anterior
radiograph showing the microwave antennae in place. (From Coughlin CT, Wong TZ, Strohbehn JW,
et al.: Intraoperative interstitial microwave-induced hyperthermia and brachytherapy. Int J Radiat On-
col Biol Phys 11:1673-1678, 1985, with permission.)

plant of indium-192 wires. Brachytherapy thermia and high dose rate implants. This
catheters are implanted surgically and func- tends to confirm the impression that the suc-
tion as conduits for radioactive sources or mi- cess is a result of the favorable physical dis-
crowave antennae. Results of these combined tributions of both heat and radiation that are
treatments are impressive, although no con- possible with implants rather than of any par-
trolled study has been performed and it is not ticular biologic advantage.
clear whether the results are better than could
have been obtained with a conventional im- POSTSCRIPT ON HYPERTHERMIA
plant of radium needles or iridium-192 wires.
Implants with radioactive sources give good Research in hyperthermia has dropped out
localized dose-distributions. In addition, heat- of the limelight in recent years, despite the
ing by means of implanted electrodes or an- demonstration in several clinical trials sum-
tennae gives a better heat distribution than marized previously that it can enhance the ef-
usually can be obtained by external mi- fect of radiation therapy in a number of sites.
crowave or ultrasonic devices. If the best heat The position can be summed up as follows:
distribution is combined with the best local- 1. The biologic properties of hyperthermia
ized radiation dose distribution, it should make it an attractive modality for the
come as no surprise that the clinical results treatment of cancer, suggesting a useful
are good. More recently, good results have differential between effects on tumors
been claimed for the combination of hyper- and normal tissues.
516 RADIOBIOLOGY FOR THE RADIOLOGIST

2. It is still difficult to achieve uniform heat- branes, intracellular structures, and cyto-
ing of a volume deep within the body, de- skeleton. Heat killing appears to be associated
spite considerable ingenuity displayed in with degradation or denaturation of proteins
designing heating systems. The basic (as judged by the inactivation energies from
laws of physics make the desired end dif- the Arrhenius plots), which is different from
ficult to achieve. that for radiation killing, which clearly in-
3. The clinical value of hyperthermia in the volves primarily damage to DNA.
routine treatment of cancer is still not Although the intermediate steps may be
clear, despite its proven efficacy in a few different, the ultimate cytotoxic effect of both
specific instances. It should be noted that heat and radiation is at the DNA level, espe-
the situations in which hyperthermia ap- cially for cells treated in the S phase of the
pears to be effective involve either com- cell cycle. For cells in vitro, heat induces
bination with external-beam radiotherapy chromosomal aberrations in the S phase, and,
for the treatment of superficial tumors— for a given level of cell killing induced by
in which adequate heating is not so diffi- heat alone or heat plus radiation, the fre-
cult—or combination with brachytherapy, quency and types of chromosomal aberrations
in which heating is induced by implanted correspond closely to those resulting from ra-
electrodes, inevitably involving all the diation alone; that is, one aberration corre-
complications and limitations of accessi- sponds to 37% survival. In organized tissues,
bility. however, heat damage occurs more rapidly
than radiation damage, because differentiated
cells are killed as well as dividing cells. Also,
MECHANISMS OF ACTION OF cells tend to die in apoptosis.
HYPERTHERMIA
The events associated with heat radiosensi-
Hyperthermia induces effects in both the tization involve DNA damage and the inhibi-
nucleus and the cytoplasm. It is unclear tion of its repair. Hyperthermia has little ef-
whether heat effects in the nucleus, such as fect on the amount of radiation-induced DNA
loss of polymerase activities, inhibition of damage in terms of single- or double-strand
DNA synthesis, and chromosomal aberra- breaks. The role of heat appears to be to block
tions, are direct effects of heat within the nu- the process of repair of these radiation-in-
cleus or secondary to the effects on the mem- duced lesions.

SUMMARY OF PERTINENT CONCLUSIONS


Localized hyperthermia using external devices can be induced by microwaves, ultra-
sound, or radiofrequency-induced currents.
The best heat distributions can be obtained by implanting microwave or radiofrequency
sources.
Heat kills: Survival curves for heat are similar in shape to those for radiation, except that
time at the elevated temperature replaces absorbed dose.
The inactivation energy is different above and below a break temperature of 42.5 to 43°C;
that is, change of sensitivity with temperature is more rapid below than above this tem-
perature.
Above the break point, the heating time required to produce a given level of cell killing
is halved for every 1-degree temperature rise; below the break point, the time must be re-
duced by a factor of 4 to 6 for each 1-degree temperature rise.
HYPERTHERMIA 517

Different cells have very different sensitivities to heat. No consistent difference in inher-
ent sensitivity exists between normal and malignant cells.
The age-response function for heat complements that for x-rays. S-phase cells that are re-
sistant to x-rays are sensitive to heat.
Cells at low pH and nutritionally deprived (more likely to be in tumors) are more sensi-
tive to heat, although cells can adapt to pH changes in 80 to 100 hours and lose their sen-
sitivity to heat.
Hypoxia does not protect cells from heat as it does from x-rays.
Heat damage in normal tissues and tumors is expressed more rapidly than x-ray damage,
because heat kills differentiated as well as dividing cells, and also cells can die in inter-
phase rather than at the next (or subsequent) mitosis, as in the case of x-rays.
Thermotolerance is the induced resistance to a second heat exposure by prior heating.
Thermotolerance can be induced during a heat exposure at temperatures below about
42.5°C but develops some hours after heating for temperatures above 42.5°C.
The development of thermotolerance may be monitored by the appearance of heat-shock
proteins.
Thermotolerance is a complication in fractionated clinical hyperthermia. With our pre-
sent state of knowledge the best way to deal with it may be to avoid it.
Heat preferentially damages tumor vasculature. After heating, blood flow goes down in
tumors but increases in normal tissues. This may result in an enhanced temperature dif-
ferential between tumors and normal tissues. There is good evidence for this in trans-
plantable animal tumors, but it is less clear in spontaneous human tumors.
Mild hyperthermia can promote tumor reoxygenation; this has been shown in animal ex-
periments and in clinical studies.
Temperature measurement in vivo is difficult but improving with multiple nonmetallic
thermometers. The most successful noninvasive technique involves magnetic resonance
imaging.
Thermal dose is expressed in terms of cumulative equivalent minutes at 43°C.
In experimental animals treated with heat alone, tumor cure correlates best with minimum
temperature. If heat and radiation are combined, however, tumor response correlates best
with some measure of temperature distribution.
Delivered simultaneously, hyperthermia interacts with radiation m a more-than-additive
way and inhibits the repair of sublethal damage.
The thermal enhancement ratio (TER) is the ratio of radiation doses with and without
heat to produce the same biologic effects. TERs of 2 to 4 can be obtained in tumors and
normal tissues m experimental animals.
The therapeutic gam factor is the ratio of the TER in the tumor to the TER in normal tis-
sues.
In a clinical situation, in which modest levels of hyperthermia are attained and small daily
doses of radiation are delivered, the cytotoxic effects of the two modalities are probably
independent and additive.
Hyperthermia potentiates some chemotherapeutic agents. Local hyperthermia "targets"
their action.
Many thousands of patients have been treated with hyperthermia.
The general consensus is that hyperthermia alone is of limited use, except for palliation
or re-treatment of recurrent tumors.
Several clinical studies have shown that the combination of hyperthermia and radiation
produces more complete and partial tumor responses than radiation alone.
518 RADIOBIOLOGY FOR THE RADIOLOGIST

Several phase III randomized trials show a clear and significant benefit for the addition
of hyperthermia to standard radiation therapy. The sites include superficial localized
breast cancer, recurrent or metastatic melanoma, and nodal metastases from head-and-
neck cancer.
Trials involving advanced breast cancer and neck nodes with full-dose radiation therapy
did not show any benefit from the addition of hyperthermia.
Biologic properties of heat appear favorable for its use in cancer therapy. Physical devices
to produce uniform heating at a depth are not well developed, and adequate thermometry
is also difficult.
Microwaves provide good localization at shallow depths, but localization is poor at
greater depths, at which low frequencies are needed.
With ultrasound, deep heating can be achieved with focused arrays, but bones or air cav-
ities cause distortions.
In practice, breast tumors, including recurrences and melanomas, can be heated ade-
quately with microwaves, and deep-seated tumors below the diaphragm can be heated ad-
equately with ultrasound. In any other site present methods of heating pose a problem.
The combination of interstitial implants of radioactive sources with hyperthermia gener-
ated by radiofrequency power applied to the implanted sources appears to be particularly
promising, because a good radiation dose distribution is combined with a good heat dis-
tribution.
Hyperthermia induces effects in both the nucleus and the cytoplasm. Probable mecha-
nisms for heat killing include damage to the plasma membrane and inactivation of pro-
teins, but these mechanisms may be different above and below the break temperature in
the Arrhenius plot.
Heat radiosensitization involves DNA damage and its repair.

BIBLIOGRAPHY Crile G: The effects of heat and radiation on cancers im-


planted on the feet of mice. Cancer Res 23:372-380, 1963
Breasted JH: The Edwin Smith surgical papyrus. In Licht S Dahl O: Hyperthermia and drugs. In Watmougl DJ. Ross
(ed): Therapeutic Heat and Cold, 2nd ed. p 196. New WM (eds): Hyperthermia. Blackie, Glasson, 1986
Haven, CT, Waverly Press, 1930 Datta NR, Bosc AK, Kapoor HK, Gupta S: Head and neck
Brizel DM, Scully SP, Harrelson JM, et al.: Radiation ther- cancers: Results of thermoradiotherapy versus radiother-
apy and hyperthermia improve the oxygenation of human apy. Int J Hypertherm 6:479-486, 1990
soft tissue sarcomas. Cancer Res 56:5347-5350, 1996 Dewey WC, Hopwood LE, Sapareto LA, Gerweck LE: Cel-
Bull JMC: An update on the anticancer effects of a combi- lular responses to combinations of hyperthermia and radi-
nation of chemotherapy and hyperthermia. Cancer Res ation. Radiology 123:463-474, 1977
44(suppl):4853S-4859S. 1984 Dewhirst MW, Prosnitz L, Thrall D, et al.: Hyperthermic
Coley WB: The treatment of malignant tumors by repeated treatment of malignant diseases: Current status and a
inoculation of erysipelas: With a report of ten original view toward the future. Semin Oncol 24:616-625, 1997
cases. Am J Med Sci 105:487, 1 893 Eddy HA, Chmielevvsky G: Effect of hyperthermia, radia-
Corry PM, Spanos WJ, Tilchen EJ. Barlogie B, Barkley HT, tion, and Adriamycin combination on tumor vascular func-
Armour EP: Combined ultrasound and radiation therapy tion. Int .1 Radiat Oncol Biol Phys 8:1 167-1175. 1982
treatment of human superficial tumors. Radiology 145: Emami BH, Perez CA, Leybovich L. Straube W, Von-
165-169.1982 gerichten D: Interstitial thermoradiotherapy in treatment
Cosset JM, Dutreix J. Haie C. Gerbaulet A, Janoray P, of malignant tumours. Int J Hypertherm 3:107-118, 1987
Dewars JA: Interstitial thermoradiotherapy: A technical Field SB, Morris CC: The relationship between heating time
and clinical study of 29 implantations performed at and temperature: Its relevance to clinical hyperthermia.
the Institut Gustave-Roussy. Int J Hypertherm 1:3-13, Radiother Oncol 1:179-186. 1983
1985 Gautherie M. Cosset JM, Gerard JP, et al.: Interstitial hyper-
Coughlin CT, Wong TZ, Strohbehn J\V* et al.: Intraoperative thermia using implanted electrodes: A multicentric pro-
interstitial microwave-induced hyperthermia and brachy- gram of technical evaluation and clinical trials over 96 pa-
therapy. Int J Radial Oncol Biol Phys 11:1673-1678, tients. In SugaharaT (ed): Hyperthermic Oncology 1988,
1985 vol 2. London, Taylor & Francis. 1989
HYPERTHERMIA 519

Gerner EW, Boon R, Conner WG, Hicks JA, Boon MLM: A Lindholm CE, Kjellen E, Nilsson P, Hertzman S: Mi-
transient thermotolcrant survival response produced by crowave-induced hyperthermia and radiotherapy in hu-
single thermal doses in HeLa cells. Cancer Res 36: man superficial tumours: Clinical results with a compar-
•1035-1040, 1976 . ative study of combined treatment versus radiotherapy
Gerner EW, Schneider Mi: Induced thermal resistance in alone. Int J Hypertherm 3:393^tl 1, 1987
HeLa cells. Nature 256:500-502, 1975 MacFall JR, Prescott DM, Charles HC, et al.: 1H V1RI Phase
Gerweck LE, Nygaard TG, Burlett M: Response of cells to . Thermometry in vivo in canine brain, muscle and tumor
hypothermia under acute and chronic hypoxic condi- tissue.-Med Phys~23:1775-1782, 1996
tions. Cancer Res 39:966-972. 1979 MacFall J, Prescott DM, Fullar E, et al.: Temperature depen-
Gonzalez Gonzalez D, van Dijk JDP, Blank LECM, Rumke dence of canine brain tissue diffusion coefficient mea-
PH: Combined treatment with radiation and hyperthermia sured in vivo with magnetic resonance echo-planar imag-
in metastatic malignant melanoma. Radiother Oncol ing. Int J Hyperthermia 11:73-86, 1995.
6:105-113, 1986 Nauts HC, Fowler GA, Bogatko FA: A review of the influ-
Griffin RJ, Okajima K, Barrios B, Song CW: Mild tempera- ence of bacterial infection and of bacterial products
ture hyperthermia combined with carbogen breathing in- (Corey's toxins) on malignant tumors in man. Acta Med
creases tumor partial pressure of oxygen (pO2) and ra- Scand 276:1-103, 1953
diosensitivity. Cancer Res 56:5590-5593, 1996 Oleson JR, Dewhirst MW, Harrelson JM, Leopold KA,
Hahn GM, Li GC: Thermotolerance and heat shock proteins Samulski TV, Tso CY: Tumor temperature distributions
in mammalian cells. Radiat Res 92:452-457, 1982 predict hyperthermia effect. Int J Radiat Oncol Biol Phys
Hahn GM, Shiu C: Adaptation to low pH modifies thermal 16:559-570, 1989
and thermo-chemical responses of mammalian cells. Int J Oleson J, Heusinkveld A, Manning M: Hyperthermia by mag-
Hyperther 2:379-387, 1986 netic induction: 11. Clinical experience with concentric
Hall EJ, Roizin-Towle L: Biological effects of heat. Cancer electrodes. Int J Radiat Oncol Biol Phys 9:549-566, 1983
Res (Suppl) 44:47O8S-4713S, 1984 Oleson JR, Samulski TV, Leopold KA, et al.: Sensitivity of
Henle KJ: Sensitization to hyperthermia below 43°C in- hyperthermia trial outcomes to temperature and time: Im-
duced in Chinese hamster ovary cells by step-down heat- plications for thermal goals of treatment. Int J Radiat On-
ing. J Natl Cancer Inst 64:1479-1483, 1980 col Biol Phys 25:289-297. 1993
Henle KJ, Kramuz JE, Leeper DB: Induction of thermotol- Overgaard J: Historical perspectives of hyperthermia. In
erance in Chinese hamster ovary cells by high (45°) or Overgaard J (ed): Introduction to Hyperthermic Oncol-
low (40°) hyperthermia. Cancer Res 38:570-574, 1978 ogy, vol 2, New York, Taylor & Francis, 1984
Henle KJ, Leeper DB: Combination of hyperthermia (40°, Overgaard J, Gonzalez Gonzalez D, Hulschof MCCM, et al.:
45°C) with radiation. Radiology 121:451-454, 1976 Randomized trial of hyperthermia as adjuvant to radio-
Hiraoka M, Jo S, Dodo Y, et al.: Clinical results of radiofre- therapy for recurrent or metastatic malignant melanoma.
quency hyperthermia combined with radiation in the treat- Lancet 345:540-543, 1995
ment of radioresistant cancers. Cancer 54:2898-2904, 1984 Overgaard J, Gonzalez Gonzalez D, Hulschof MC, Arcan-
Hornback NB, Shupe RE, Shidnia H, Marshall CU, Lauer T: geli G, Dahl O, Mella O, Bentzen SM: Hyperthermia as
Advanced state MB cancer of the cervix treatment by hy- an adjuvant to radiation therapy of recurrent or metastatic
perthermia and radiation. Gynecol Oncol 23:160-167, 1986 malignant melanoma: A multicentre randomized trial by
Horsman MR, Overgaard J: Can mild hyperthermia improve the European Society for Hyperthermic Oncology. Int J
tumour oxygenation9 Int j Hypertherm 13:141-147, 1997 Hypertherm 12:3-20, 1996
Iwata K, Shakil A. Hur HI et al.: Tumor pO2 can be in- Overgaard J, Overgaard M: Hyperthermia as an adjuvant to
creased markedly by mild hyperthermia. Br J Cancer radiotherapy in the treatment of malignant melanoma. Int
74(suppl):217s-221s, 1996 J Hypertherm 3:483-501, 1987
Kano E: Hyperthermia and drugs. In Overgaard J (ed): Hy- Perez CA, Kuske RR, Emam B, Fineberg B: Irradiation alone
perthennic Oncology, pp 277-282, London, Taylor & or combined with hyperthermia in the treatment of recurrent
Francis, 1985 carcinoma of the breast in the chest wall: A nonradomized
Kapp DS, Cox RS: Thermal treatment parameters are most comparison. Int J Hypertherm 2:179-187, 1986
predictive of outcome in patients with single tumor nod- Robinson JE, Wizenburg MJ: Thermal sensitivity and the ef-
ules per treatment field in recurrent adenocarcinoma of fect of elevated temperatures on the radiation sensitivity
the breast [see comments]. Int J Radiat Oncol Biol Phys of Chinese hamster cells. Acta Radiol 13:241 -248, 1974
33:887-899, 1995 Roizin-Towle L, Hall EJ, Capuano L: Interaction of hyper-
Lam K, Astraham M, Langholtz B, et al.: Interstitial ther- thermia and cytotoxic agents. Natl Cancer Inst Monogr
moradiotherapy for recurrent or persistent tumors. Int J 61:149-151, 1982
Hypertherm 4:259-266. 1988 Sapareto SA, Dewey \VC: Thermal dose determination in
Law MP, Ahier RG, Field SB: The response of the mouse ear cancer therapy. Int J Radiat Oncol Biol Phys 10:787-800,
to heat applied alone or combined with x-rays. Br J Ra- 1984
diol 51:132-138. 1978 Sapareto SA, Hopwood LE, Dewey WC: Combined effects
Leopold KA, Dewhirst M. Tucker JA, et al.: Relationships of x-irradiation and hyperthermia on CHO cells for vari-
among tumor temperature, treatment time, and histo- ous temperatures and orders of application. Radiat Res
pathological outcome using preoperative hyperthermia 43:221-233. 1978
with radiation in soft tissue sarcomas. Int J Radiat Oncol Sherar M, Liu FF, Pintilie M, et al.: Relationship between
Biol Phys 22:989-998, 1992 thermal dose and outcome in thennoradiotherapy treat-
Li GC, Hahn GM: A proposed operational model of ther- ments for superficial recurrences of breast cancer: Data
motolerance based on effects of nutrients and the initial from a phase III trial. Int J Radiat Oncol Biol Phys 39:
treatment temperature. Cancer Res 40:4501-4508, 1980 371-380, 1997
520 RADIOBIOLOGY FOR THE RADIOLOGIST

Sneed PK, Stauffer PR, Diederich CH, et al.: Survival bene- diation carcinogenesis: Effect on early and late appearing
fit of hyperthennia in a prospective randomized trial of reactions Int J Radiat Oncol Biol Phys 15:159-166, 1988
brachytherapy boost ± hvperthermia for glioblastoma mul- Valdagni R, Amichetti M: Report of long-term follow-up in
tiforme. Int J Radiat Oncol Biol Phys 40:287-295, 1998 a randomized trial comparing radiation therapy and radi-
Song CW: Effect of local hyperthennia on blood flow and ation therapy plus hyperthermia to metastatic lymph
microenvironment: A review. Cancer Res 44(suppl): nodes in stage IV head and neck patients. Int J Radiat On-
472IS-4730S, 1984 col Biol Phys 28:163-169, 1994
Song CW, Shakil A, Osborn JL, Iwata K: Tumour oxygena- Valdagni R, Kapp DS, Valdagm C: N3 (TNM-UICC) metasta-
tion is increased by hyperthennia at mild temperatures. tic neck nodes managed by combined radiation therapy and
Int J Hyperthermia 12:367-373, 1996 hyperthennia: Clinical results and analysis of treatment pa-
Steeves RA, Severson SB, Paliwal BR, Anderson S, Robins rameters. Int J Hyperthermia 2:189-200, 1986
HI: Matched-pair analysis of response to local hyperther- Vernon CC, Hand ]Vv, Field SB, et al.: Radiotherapy with or
mia and megavoltage electron therapy for superficial hu- without hyperthermia in the treatment of superficial local-
man tumors. Endocuriether Hyperther Oncol 2:163-170, ized breast cancer: Results from five randomized controlled
1986 trials. Int J Radiat Oncol Biol Phys 35:731-744, 1996
Subjeck JR, Sciandra JJ, Chao CF, Johnson RJ: Heat shock- Vora N, Shaw S, Forell B, et al.: Primary radiation combined
proteins and biological response to hyperthermia. Br J with hyperthermia for advanced (stage III-IV) and in-
Cancer 45(supplV):127-131. 1982 flammatory carcinoma of breast. Endocuriether Hyper-
Urano M: Kinetics of thermotolerance in normal and tumor ther Oncol 2:101-106, 1986
tissues: A review. Cancer Res 46:474-482. 1986 Westra A, Dewey WC: Variation in sensitivity to heat shock
Urano M, Kenton A, Kahn J: The effect of hyperthennia on the during the cell cycle of Chinese hamster cells in vitro. Int
early and late appearing mouse foot reactions and on the ra- J RadlatBiol 19:467-477, 1971

You might also like