hyperthermia systemic

21
HYPERTHERMIA, SYSTEMIC R. WANDA ROWE-HORWEGE University of Texas Medical School Houston, Texas INTRODUCTION Systemic hyperthermia is deliberate heating of the whole body to achieve an elevated core temperature for thera- peutic purposes. Other terms used are whole-body hyperthermia, systemic or whole body thermal therapy, and hyperpyrexia. The goal of systemic hyperthermia is to reproduce the beneficial effects of fever. Typically, core body temperatures of 41–42 8C are induced for 1–2 h, or alternatively 39–40 8C for 4–8 h. Systemic hyperthermia, by virtue of application to the whole body, aims to alleviate systemic disease conditions, in contrast to local or regional hyperthermia that treats only a specific tissue, limb, or body region. HISTORICAL BACKGROUND The use of heat to treat disease goes back to ancient times. Application of fire to cure a breast tumor is recorded in an ancient Egyptian papyrus, and the therapeutic value of elevated body temperature in the form of fever was appre- ciated by ancient Greek physicians. Hippocrates wrote, ‘‘What medicines do not heal, the lance will; what the lance does not heal, fire will,’’ while Parmenides stated, ‘‘Give me a chance to create a fever and I will cure any disease.’’ In the first century AD, Rufus (also written as Refus or Ruphos) of Ephesus advocated fever therapy for a variety of diseases. Hot baths were considered thera- peutic in ancient Egypt, Greece, Rome, China, and India as they still are in many aboriginal cultures today, along with burying diseased individuals in hot sand or mud. Hot baths and saunas are an integral part of health traditions throughout the Orient, in Indian Ayurvedic medicine, as well as in Eastern European and Scandina- vian countries. Following several earlier anecdotal reports, several nineteenth century German physicians observed regression or cure of sarcoma in patients who suffered prolonged, high fevers due to infectious diseases. This led to efforts to induce infectious fevers in cancer patients, for example, by applying soiled bandages or the blood of malaria patients to wounds. The late nineteenth century New York physician, William Coley, achieved cancer cures by administration of erysipelas and other bacterial endotoxins, now known as Coley’s toxins, and attempted to create standardized preparations of these pyrogens (1). At around the same time, treatment of syphilis by placing the patient in a stove-heated room, or a heat box, became commonplace. Successful hyperther- mic treatment of other sexually transmitted diseases, such as gonorrhea, and neurological conditions, such as chorea minor, dementia paralytica, and multiple sclerosis along with arthritis, and asthma were widely reported. Interest- ingly, it was noted by Italian physicians that upon comple- tion of the draining of the Pontine Swamps near Rome by Mussolini in the 1930s, not only was malaria eradicated, but the prevalence of cancer in the area was the same as in the rest of Italy, whereas earlier the whole malaria- infected region was noted for its absence of cancer. It was concluded that the frequent fever attacks common in malaria stimulated the immune system to prevent the development of cancers. The science of hyperthermia became grounded in the first few decades of the twentieth century when some of the biological effects of elevated body temperature were eluci- dated and attempts were made to understand and control the therapeutic application of heat. Numerous devices were developed to produce elevated temperatures of the body, by a variety of physical means. After a shift in focus to local and regional hyperthermia, there is now a resurgence of interest in systemic hyperthermia for treatment of cancer, as well as other systemic diseases. Whole-body hyperther- mia treatment is now carried out at several university centers in the United States, and Europe (Table 1), where controlled clinical trials are being carried out. Numerous private clinics, principally in North America, Germany, Austria, Eastern Europe, Japan, and China also perform systemic hyperthermia, mostly as part of holistic, alterna- tive, treatment regimens. PHYSICS OF SYSTEMIC HYPERTHERMIA As shown schematically in Fig. 1, in order to achieve body temperature elevation, there must be greater deposition of heat energy in the body than heat energy lost from 42 HYPERTHERMIA, SYSTEMIC Encyclopedia of Medical Devices and Instrumentation, Second Edition, edited by John G. Webster Copyright # 2006 John Wiley & Sons, Inc.

Upload: ataraix22

Post on 14-Feb-2016

250 views

Category:

Documents


1 download

DESCRIPTION

Articulo académico

TRANSCRIPT

Page 1: Hyperthermia Systemic

HYPERTHERMIA, SYSTEMIC

R. WANDA ROWE-HORWEGE

University of Texas MedicalSchoolHouston, Texas

INTRODUCTION

Systemic hyperthermia is deliberate heating of the wholebody to achieve an elevated core temperature for thera-peutic purposes. Other terms used are whole-bodyhyperthermia, systemic or whole body thermal therapy,and hyperpyrexia. The goal of systemic hyperthermia is toreproduce the beneficial effects of fever. Typically, corebody temperatures of 41–42 8C are induced for 1–2 h, oralternatively 39–40 8C for 4–8 h. Systemic hyperthermia,by virtue of application to the whole body, aims to alleviatesystemic disease conditions, in contrast to local or regionalhyperthermia that treats only a specific tissue, limb, orbody region.

HISTORICAL BACKGROUND

The use of heat to treat disease goes back to ancient times.Application of fire to cure a breast tumor is recorded in anancient Egyptian papyrus, and the therapeutic value ofelevated body temperature in the form of fever was appre-ciated by ancient Greek physicians. Hippocrates wrote,‘‘What medicines do not heal, the lance will; what thelance does not heal, fire will,’’ while Parmenides stated,

‘‘Give me a chance to create a fever and I will cure anydisease.’’ In the first century AD, Rufus (also written asRefus or Ruphos) of Ephesus advocated fever therapy fora variety of diseases. Hot baths were considered thera-peutic in ancient Egypt, Greece, Rome, China, and Indiaas they still are in many aboriginal cultures today, alongwith burying diseased individuals in hot sand or mud.Hot baths and saunas are an integral part of healthtraditions throughout the Orient, in Indian Ayurvedicmedicine, as well as in Eastern European and Scandina-vian countries. Following several earlier anecdotalreports, several nineteenth century German physiciansobserved regression or cure of sarcoma in patients whosuffered prolonged, high fevers due to infectious diseases.This led to efforts to induce infectious fevers in cancerpatients, for example, by applying soiled bandages or theblood of malaria patients to wounds. The late nineteenthcentury New York physician, William Coley, achievedcancer cures by administration of erysipelas and otherbacterial endotoxins, now known as Coley’s toxins, andattempted to create standardized preparations of thesepyrogens (1). At around the same time, treatment ofsyphilis by placing the patient in a stove-heated room,or a heat box, became commonplace. Successful hyperther-mic treatment of other sexually transmitted diseases, suchas gonorrhea, and neurological conditions, such as choreaminor, dementia paralytica, and multiple sclerosis alongwith arthritis, and asthma were widely reported. Interest-ingly, it was noted by Italian physicians that upon comple-tion of the draining of the Pontine Swamps near Rome byMussolini in the 1930s, not only was malaria eradicated,but the prevalence of cancer in the area was the same as inthe rest of Italy, whereas earlier the whole malaria-infected region was noted for its absence of cancer. Itwas concluded that the frequent fever attacks commonin malaria stimulated the immune system to prevent thedevelopment of cancers.

The science of hyperthermia became grounded in thefirst few decades of the twentieth century when some of thebiological effects of elevated body temperature were eluci-dated and attempts were made to understand and controlthe therapeutic application of heat. Numerous deviceswere developed to produce elevated temperatures of thebody, by a variety of physical means. After a shift in focus tolocal and regional hyperthermia, there is now a resurgenceof interest in systemic hyperthermia for treatment of cancer,as well as other systemic diseases. Whole-body hyperther-mia treatment is now carried out at several universitycenters in the United States, and Europe (Table 1), wherecontrolled clinical trials are being carried out. Numerousprivate clinics, principally in North America, Germany,Austria, Eastern Europe, Japan, and China also performsystemic hyperthermia, mostly as part of holistic, alterna-tive, treatment regimens.

PHYSICS OF SYSTEMIC HYPERTHERMIA

As shown schematically in Fig. 1, in order to achieve bodytemperature elevation, there must be greater depositionof heat energy in the body than heat energy lost from

42 HYPERTHERMIA, SYSTEMIC

Encyclopedia of Medical Devices and Instrumentation, Second Edition, edited by John G. WebsterCopyright # 2006 John Wiley & Sons, Inc.

Page 2: Hyperthermia Systemic

Ta

ble

1.

Cli

nic

al

Aca

dem

ic/R

eg

ion

al

Sy

stem

icH

yp

erth

erm

iaC

en

ters

Cou

ntr

yC

ity

Inst

itu

tion

Pri

nci

pal

Inves

tigato

rH

eat

Typ

e,M

ach

inea

Pro

toco

l(t

ime,

tem

p)

Asi

aC

hin

aB

aod

ing

Sec

ond

Hos

pit

al

ofB

aod

ing

Ch

un

zhu

Yin

RF

3.5

–6

h,

40–40.5

8CC

hin

aC

han

gch

un

Jil

inT

um

orH

osp

ital

Ch

an

ggu

oH

ong

RF

3.5

–6

h,

40–40.5

8CC

hin

aJia

ngm

enG

uan

gd

ong

Jia

ngm

enR

enm

inH

osp

ital

Wen

pin

gW

uIR

Ch

ina

Sh

an

gh

ai

Ch

an

gh

ai

Hos

pit

al,

Sec

ond

Mil

itary

Med

ical

Sch

ool

Yaji

eW

an

gC

hin

aS

han

gh

ai

Dep

art

men

tof

Tu

mor

Hyp

erth

erm

iaC

ente

rK

ai-

shen

gH

ouIR

,E

T-S

pace

extr

aco

rpor

eal

1–2

h,

41.8

–42.5

8C

Ch

ina

Sh

an

gh

ai

Sh

an

gh

ai

Jin

gan

Cen

tral

Hos

pit

al

Wei

pin

gT

ao

IR,

ET

-Sp

ace

extr

aco

rpor

eal

2h

,41.6

8C4

h,

42.1

8C

Ch

ina

Tai’an

Cit

y88th

Hos

pit

al

ofP

LA

Yon

gP

eng

1–2

h,

41.8

8C6

h,

39.5

–408C

Ch

ina

Zh

engzh

ouM

oder

nH

osp

ital,

Zh

engzh

ouD

ingji

uL

iR

F3.5

–6

h,

40–40.5

8CJap

an

Tok

yo

Lu

ke

Hos

pit

al

Ak

ira

Tak

euch

iIR

Eu

rop

eB

elaru

sM

insk

Bel

aru

sC

ente

rfo

rP

edia

tric

On

colo

gy

an

dH

emato

logy

Rei

man

nIs

mail

-zad

eH

FE

M,

Yak

hta

-52

h,

41.8

–42.5

8C1

h,

42.5

–438C

Ger

man

yB

erli

nL

ud

wig

Maxim

ilia

nU

niv

ersi

tyC

hari

teM

edic

al

Cen

ter

Ber

tH

ild

ebra

nd

t,H

an

no

Rie

ss,

Pet

erW

ust

IR,

Irath

erm

1h

,41.8

8C

Ger

man

yF

ran

kfu

rtK

ran

ken

hau

sN

ord

wes

tE

lke

Jager

,A

kin

Atm

ata

IR,

Aqu

ath

erm

1h

,41.8

8CG

erm

an

yM

un

ich

Lu

dw

igM

axim

ilia

nU

niv

ersi

tyH

osp

ital

Cli

nic

Hara

ldS

omm

erIR

,Ir

ath

erm

1h

,41.8

8CH

un

gary

Kec

skem

etIn

stit

ute

ofR

ad

iolo

gy

ofK

ecsk

emet

Mik

los

Szu

csIR

,O

nco

Th

erm

1h

,41.8

8CN

orw

ay

Ber

gen

Un

iver

sity

ofB

ergen

,H

au

kel

an

dU

niv

ersi

tyH

osp

ital

Baard

-Ch

rist

ian

Sch

emIR

,Ir

ath

erm

1h

,41.8

8CR

uss

iaN

ovos

ibir

skS

iber

ian

Sci

enti

fic

Res

earc

hIn

stit

ute

ofH

yp

erth

erm

iaR

oman

Tch

ervov

Wate

rbath

43.5

–44.0

8CR

uss

iaO

bn

insk

Med

ical

Rad

iolo

gic

al

Res

earc

hC

ente

rof

Ru

ssia

nA

cad

emy

ofM

edic

al

Sci

ence

s,O

bn

insk

Yu

riM

ard

yn

sky

HF

EM

,Y

ak

hta

51–2

h,

41.0

–42.3

8C

Nort

hA

mer

ica

Un

ited

Sta

tes

Galv

esto

n,

TX

Un

iver

sity

ofT

exas

Med

ical

Bra

nch

Jos

eph

Zw

isch

enber

ger

extr

aco

rpor

eal

2h

,42.5

8CU

nit

edS

tate

sH

oust

on,

TX

Un

iver

sity

ofT

exas

Med

ical

Sch

ool

Joa

nM

.B

ull

IR,

Hec

kel

6h

,408C

Un

ited

Sta

tes

Bu

ffalo

,N

YR

osw

ell

Park

Can

cer

Inst

itu

teW

illi

am

G.

Kra

ybil

lIR

,H

eck

el6

h,

408C

Un

ited

Sta

tes

Du

rham

,N

CD

uk

eC

omp

reh

ensi

ve

Can

cer

Cen

terb

Zel

jko

Vu

jask

ovic

IR,

Hec

kel

6h

,408C

aR

ad

iofr

equ

ency

¼R

F;

infr

are

IR.

bS

tart

ing

in2006.

43

Page 3: Hyperthermia Systemic

conduction, convection, radiation and evaporation, that is,

Q0depDt>Q0

lossDt ð1Þ

where Q0 ¼ DQ/Dt represents the change in heat energy,Q (measured in Joules or calories), over a time periodDt. Net heat energy deposition in a volume element DVof tissue of density rtis results in an increase in tem-perature DT dependent on the specific heat of the tissue,ctis,

Q0dep

DV�

Q0loss

DV

!Dt ¼ ðrtisDVÞctisDT

DT ¼ ðQ0dep � Q0

lossÞ Dt

rtisctisð2Þ

Heat deposition is the sum of the absorbed power den-sity, Pabs, from external sources and heat generated bymetabolism, Qmet,

DQ0dep

DV¼ Pabsðx; y; z; tÞ þ

DQ0met

DVðx; y; z; tÞ ð3Þ

If the air temperature is higher than the body surfacetemperature, heat is absorbed from air surrounding thebody by the skin, as well as during respiration. Powerdeposition in tissue from external electromagnetic fieldsdepends on the coupling of the radiation field (micro-wave, RF, ultrasound, visible or IR light) with tissue.The body’s metabolic rate depends on the amount ofmuscular activity, the temperature, pressure andhumidity of the environment, and the size of the body.Metabolic rate increases nonlinearly with core bodytemperature, in part due to the exponential increaseof the rate of chemical reactions with temperature(Arrhenius equation). An empirical relationship between

basal metabolic rate and core temperature has beendetermined as

Basal MR ¼ 85 � 1:07ðTcoreÞ

0:5ð4Þ

whichcanbeexploited tomaintainelevatedbodytemperatures(2). At room temperature a human body produces � 84 W,which increases to �162 W at a core temperature of 41.8 8C.

Heat losses from the body are often termed sensible(convective, conductive, radiative) and insensible (evapora-tive, latent). The primary mode of heat loss from the body isby radiation, as described by the Stefan–Boltzmann law,

Q0rad

DV¼ eskinsAskinðTskin � TsÞ4 ð5Þ

where Q0rad/DV is the power radiated, eskin is the emissivity

of the skin (radiating material), s is Stefan’s con-stant ¼ 5.6703 � 10�8 Wm�2/K, Askin is the skin surfacearea, Tskin is the temperature of the skin (radiator), and Ts

is the temperature of the surroundings (e.g., air, water,wax). Human skin is a near perfect radiator in the IR, withan emissivity of 0.97. At room temperature, >50% of theheat generated by metabolism is lost by radiation; a clothedadult loses some 50 W at room temperature. This increasesto �66% at a core temperature of 41.8 8C, as is targeted in anumber of systemic hyperthermia protocols, when the skintemperature rises to 39–40 8C (3).

Direct transfer of body heat to the molecules around thebody (typically air) occurs by conduction, or molecularagitation within a material without any motion of thematerial as a whole, which is described by Fourier’s law,

DQ0cond

DV¼ kAskin

DT

Dxð6Þ

where DQcond is the heat energy transferred per unitvolume in time Dt, k is the thermal conductivity (WmK�1)of the material surrounding the body (air, water), and DT isthe temperature difference across thickness Dx of thematerial. Air is a poor thermal conductor, therefore heatloss by conduction is relatively low. On the other hand,water has a thermal conductivity 20 times that of air at0 8C, increasing further with temperature, therefore dur-ing hyperthermia it is important that any water in contactwith the skin is not at a lower temperature. The relativethermal conductivity of body tissues is important in deter-mining thermal conduction within the body from externalsources of heat. For example, fat is a relative thermalinsulator with a thermal conductivity one third of that ofmost other tissues, therefore fat bodies are slower to heat.

Convective heat transfer involves material movementand occurs principally via blood moving heat to, or from,the skin and other tissues, and air currents (respiratoryand environmental) moving warm air to or from the body.Equation 7 is written for the blood,

DQ0conv

DV¼ rbcb½wbðx; y; z;TÞ ðT � TbÞ þ Ubðx; y; z;TÞ rT�

ð7Þ

where wb is the specific capillary blood flow rate, Ub is thespecific blood flow through other vessels. In the context of

44 HYPERTHERMIA, SYSTEMIC

Figure 1. Schematic of heat balance mechanisms in the humanbody. Body temperature is determined by the balance of metabolicheat production plus heating from external sources, and heatlosses by radiation, evaporation, convection, and conduction.

Page 4: Hyperthermia Systemic

systemic hyperthermia, where a patient is in a closedchamber, environmental air currents can be minimized.Heat loss by respiration, however, can amount to almost10% of metabolic heat generation.

Another route of heat loss from the body is evaporationof perspiration from the skin. Because of the very largeheat of vaporization of water, cooling of the blood in skincapillaries occurs due to evaporation of sweat. Evaporationfrom exhaled moisture also results in cooling of the sur-rounding air.

DQ0evap

DV¼ mw

Lv

Dtð8Þ

where mw is the mass of the water and Lv is the latent heatof vaporization (2.4 � 106 Jkg�1 at 34 8C). In hot condi-tions with maximal rates of evaporation, heat loss throughevaporation of sweat can be as much as 1100 W. Heat lossin the lungs is �10 W.

Combining the heat generation and heat loss termsleads to a general heat transfer equation, an extensionof the classic Pennes bioheat transfer equation.

DQ0met

DVþ Pabs

� ��

�DQ0

rad

DVþDQ0

cond

DVþ DQ0

conv

DVþDQ0

resp

DVþDQ0

evap

DV

� ��¼ rtisctisDT ð9Þ()

into which the expressions given in Eqs. 2–8 may be sub-stituted. Precise solution of this equation for temperaturedistribution is complex and requires a number of simplifyingassumptions which have generated significant controversyin bioheat transfer circles. Modeling of temperature distri-butions within a body subjected to hyperthermia is alsocomplex because of the heterogeneity of thermal character-istics between and within tissue, the directionality of powerapplication, and the dynamic nature of thermoregulation byhuman body. Nonetheless, the factors governing systemicheating of the body can be appreciated.

INDUCTION OF SYSTEMIC HYPERTHERMIA

Apart from the induction of biological fever by pathogens ortoxins, all methods of hyperthermia involve transfer ofheat into the body from an external energy source. Therequired net power to raise the temperature of a 70 kghuman from 37 to 41.8 8C (2) is 400 W (5.7 mW). While theheat absorption from these sources is highly nonuniform,distribution of thermal energy by the vascular systemquickly results in a uniform distribution of temperature.Indeed, systemic hyperthermia is the only way to achieveuniform heating of tissues. Because physiological thermo-regulation mechanisms such as vasodilation and perspira-tion counteract attempts to increase core body temperature,careful attention must be paid to optimizing the physicalconditions for heating such that there is efficient depositionof heat energy in the body and, even more importantly,minimization of heat losses. Wrapping the body in reflectiveblankets, foil, or plastic film to reduce radiative and eva-porative losses, or keeping the surrounding air moist to

minimize losses by perspiration are key techniques forachieving a sustained increase in body temperature.

Noninvasive methods of heating include immersing thebody in hot water or wax, wrapping the body in a blanket orsuit through which heated water is pumped, placing thepatient on a heated water mattress, surrounding the bodywith hot air, irradiating with IR energy, and applying RFor microwave electromagnetic energy. These techniquesmay be applied singly or in combination. For example, thePomp–Siemens cabinet used until recently throughoutEurope, as well as in the United States, a modificationof a device originally developed by Siemens in the 1930s,has the patient lying on a heated water mattress underwhich an inductive loop generates an RF field, all inside achamber through which hot air is circulated. The RussianYakhta-5 system applies a high frequency (13.56 MHz)electromagnetic field through a water-filled mattress topermit whole body heating up to 43.5 8C and simultaneousdeep local hyperthermia through additional applicators pro-viding 40.6 MHz electromagnetic radiation. The majority ofwhole-body hyperthermia systems currently in clinical useemploy IR radiation to achieve systemic heating. Invasiveapproaches to systemic hyperthermia are extracorporealheating of blood, removed from the body via an arteriovenousshunt, prior to returning it to the circulation, as well asperitoneal irrigationwithheatedfluid (4).A useful schematicsummary of whole-body hyperthermia induction techniquesalong with references is provided by van der Zee (5).

All of these approaches involve a period of steadytemperature increase, followed by a plateau or equilibriumphase where the target temperature is maintained for any-where from 30 min to several hours, and finally a cool-downphase. Depending on the method of hyperthermia induction,the patient may be anesthetized, consciously sedated, admi-nistered analgesia, or not given any kind of medication atall. An epidural block is sometimes given to induce orincrease vasodilation. During radiant heat induction, thetemperature of the skin and superficial tissues (includingtumors) is higher than the core (rectal) temperaturewhereas during the plateau (maintenance) phase, theskin–superficial tissue temperature drops below the coretemperature. As already described, heat losses due to phy-siological mechanisms limit the rate of heating that can beachieved. When insulation of the patient with plastic foilwas added to hot air heating, the heating time to 41.8 8C wasdecreased from 230 to 150 min (65%), and further to 110 min(48%) by addition of a warm water perfused mattress (5).The homogeneity of the temperature distribution was alsosignificantly increased by the addition of insulation and thewater mattress. Noninvasive systemic hyperthermia meth-odologies typically produce heating rates of 1–10 8Ch�1

with 2–3 8Ch�1 being most common. More rapid heatingcan be achieved by the invasive techniques, at the expense ofgreater risk of infection and morbidity.

COMMERCIALLY AVAILABLE WHOLE-BODYHYPERTHERMIA SYSTEMS

A number of commercially available devices have resultedfrom the development of these initially experimental

HYPERTHERMIA, SYSTEMIC 45

Page 5: Hyperthermia Systemic

systems. The Siemens–Pomp system has already been men-tioned, but is no longer commercially available. Similarly,neither the radiant heat chamber developed by Robins (3),and marketed as the Aquatherm system, nor the similarEnthermics Medical Systems RHS-7500 radiant heatdevice, both producing far IR radiation (IR C) in a moistair chamber, are currently being sold, though they arestill in use in several centers. A close relative is theIratherm2000 radiant heat chamber originally developedby von Ardenne and co-workers (6). In this device, water-filtered infrared radiators at 2400 8C emit their energy fromabove and below the patient bed, producing near-IR (IR A)radiation that penetrates deeper into tissue than far IRradiation, causing direct heating of the subcutaneous capil-lary bed. Thermal isolation is ensured by reflective foilsplaced around the patient. However, note that significantevaporative heat loss through perspiration can be a problemwith this system. Also with a significant market share is theHeckel HT 2000 radiant heat device in which patients lie ona bed enclosed within a soft-sided rectangular tent whoseinner walls are coated with reflective aluminum foil thatensures that the short wavelength infrared A and B radia-tion emitted by four radiators within the chamber uniformlybathes the body surface. Once the target temperature isreached, the chamber walls are collapsed to wrap around thebody, thereby preventing radiative and evaporative heatloss, and permitting maintenance of the elevated tempera-ture, as shown in Fig. 2.

Another radiant heat device, used mainly in Germany,is the HOT-OncoTherm WBH-2000 whole-body hyperther-mia unit which is a chamber that encloses all but thepatient’s head. Special light-emitting diode (LED) radia-tors deliver computer-generated, alloy-filtered IR A wave-lengths that penetrate the skin to deliver heat to thecapillary bed. The manufacturer claims that these wave-lengths also preferentially stimulate the immune system.Recently, Energy Technology, Inc. of China has releasedthe ET-SPACE whole-body hyperthermia system, which

produces IR A radiation in a small patient chamber intowhich warm liquid is infused to help increase the airhumidity and thereby reduce perspiration losses. A num-ber of low cost, far infrared, or dry, saunas are being soldto private clinics, health clubs, and even individuals fortreatment of arthritis, fibromyalgia, detoxification, andweight loss. Examples are the Smarty HyperthermicChamber, the TheraSauna, the Physiotherm, and theBiotherm Sauna Dome. Table 2 summarizes features ofthese commercially available whole-body hyperthermiadevices.

BIOLOGICAL EFFECTS OF SYSTEMIC HYPERTHERMIA

An understanding of the biological effects of systemichyperthermia is critical to both its successful inductionand to its therapeutic efficacy. Systemic responses to bodyheating, if not counteracted, undermine efforts to raisebody temperature, while cellular effects underlie boththe rationale for the use of hyperthermia to treat specificdiseases, and the toxicities resulting from treatment.Although improved technology has allowed easier andmore effective induction of systemic hyperthermia, mostof the recent clinical advances are due to better under-standing and exploitation of specific biological phenomena.

Physiological Effects of Elevated Body Temperature

The sympathetic nervous system attempts to keep all partsof the body at a constant temperature, tightly controlled bya central temperature ‘set point’ in the preoptic–anteriorhypothalamus and a variety of feedback mechanisms. Thethermostat has a circadian rhythm and is occasionallyreset, for example, during fever induced by infectiousagents and endotoxins, but not in endogenously inducedhyperthermia. Occasionally, it breaks down completely asin malignant hyperthermia or some neurological disordersaffecting the hypothalamus. Ordinarily, when core bodytemperature rises, the blood vessels initially dilate, heartrate rises, and blood flow increases in an effort to transportheat to the body surface where it is lost by radiation,conduction, and convection. Heart rate increases on averageby 11.7 beatsmin�1 8C�1 and typically remains elevated forseveral hours after normal body temperature is regained.Systolic blood pressure increases to drive the blood flow, butdiastolic pressure decreases due to the decreased resistanceof dilated vessels, thus there is an increase in cardiacoutput. Heart rate and blood pressure must therefore bemonitored during systemic hyperthermia, and whole-bodyhyperthermia is contraindicated in most patients withcardiac conditions. Interestingly, hyperthermia increasescardiac tolerance to ischemia/reperfusion injury probablydue to activation of manganese superoxide dismutase(Mn-SOD) and involvement of cytokines.

Respiration rate also increases and breathing becomesshallower. Perspiration results in evaporation of sweatfrom the skin and consequent cooling, while the respirationrate increases in order to increase cooling by evaporation ofmoisture from expired air. Weight loss occurs despite fluidintake. There is a decrease in urinary output and the urinehas a high specific gravity, concentrating urates and

46 HYPERTHERMIA, SYSTEMIC

Figure 2. Heckel HT-2000 radiant heat whole body hyperthermiasystem. Unit at the University of Texas Medical School at Houston.Patient is in the heat maintenance phase of treatment, wrapped inthe thermal blankets which form the sides of the chamber duringactive heating.

Page 6: Hyperthermia Systemic

Ta

ble

2.

Co

mm

erc

iall

yA

va

ila

ble

Cli

nic

al

Wh

ole

-Bo

dy

Hy

perth

erm

iaD

ev

ices

Man

ufa

ctu

rer

Web

site

Dev

ice

Nam

eH

eati

ng

Mec

han

ism

Tem

per

atu

reR

an

ge,

8CA

pp

lica

tion

En

ergy

Tec

hn

olog

yh

ttp

://w

ww

.eti

.com

.cn

/EN

/pro

/pro

du

ct2.h

tmE

T-S

PA

CE

Mu

ltip

leIR

rad

iato

rs(I

RA

)39–41.8

On

colo

gy

Hec

kel

Med

izin

tech

nik

Gm

bH

htt

p://w

ww

.hec

kel

-med

izin

tech

nik

.de/

fram

eset

_e.h

tml

HT

2000

M4

300W

IRra

dia

tors

(IR

A,

B)

38.5

–40.5

On

colo

gy,

rheu

mato

logy

Hot

-On

coth

erm

htt

p://w

ww

.hot

-on

coth

erm

.com

/on

coth

erm

ia.h

tmW

BH

-2000

Mu

ltip

leL

ED

rad

iato

rs(I

RA

)37–42

On

colo

gy

Von

Ard

enn

eIn

stit

ut

fur

An

gew

an

dte

Med

izin

isch

eF

orsc

hu

ng,

Gm

bH

htt

p:/

/ww

w.a

rden

ne.

de/

med

_en

g/

Irath

erm

800

4IR

rad

iato

rs(I

RA

)37–38

Ph

ysi

cal

med

icin

e,co

mp

lem

enta

rym

edic

ine,

onco

logy

Irath

erm

1000

6IR

rad

iato

rs(I

RA

)37–39

Irath

erm

2000

10

IRra

dia

tors

(IR

A)

37–42

47

Page 7: Hyperthermia Systemic

phosphates. In endogenously induced hyperthermia, butnot in fever, glomerular filtration, as evidenced by thecreatinine clearance, decreases with increasing tempera-ture. As already mentioned, metabolic rate increases non-linearly with temperature, which leads to an increase inblood sugar, decreased serum potassium levels, andincreased lactic acid production. All the above normalphysiological effects may be enhanced or counteracted byanesthesia or sedation, as well as by disease states such ascancer because of drugs used in treatment or intrinsicpathophysiological consequences of the disease.

At �42.5 8C, the normal thermocompensatory mechan-isms break down and the body displays the symptoms ofadvanced heat stroke, namely, lack of sweating, rapidheart beat, Cheyne–Stokes breathing, central nervoussystem disfunction, and loss of consciousness. Ultimately,breathing ceases despite the continuation of a heart beat.

Cellular Thermal Damage

When temperature is increased by a few degrees Celcious,there is increased efficiency of enzyme reactions (Arrhe-nius equation), leading to increased metabolic rates, but attemperatures > 40 8C molecular conformation changesoccur that lead to destabilization of macromolecules andmultimolecular structures, for example, to the side chainsof amino acids in proteins, which in turn inhibit enzymeaction. Small heat shock proteins (HSP) interact with theunfolding proteins to stabilize them and prevent theiraggregation and precipitation. Eventually, however, at�42 8C, complete denaturation of proteins begins thattotally disrupts many molecular processes, including deox-yribonucleic acid (DNA) repair. Thus systemic hyperther-mia can have significant effects when paired with drugsthat cause DNA damage (e.g., for chemotherapy of cancer).

Membranes are known to be extremely sensitive toheat stress because of their complex molecular composi-tion of lipids and proteins. At a certain temperature, lipidschange from the tightly packed gel phase to the lesstightly packed liquid crystalline phase, and permeabilityof the cell membrane (membrane fluidity) increases. Astemperature increases further, the conformation of proteinsalso becomes affected, eventually resulting in disorderlyrearrangement of the lipid bilayer structure and receptorinactivation or loss. Temperature changes of �5 8C arenecessary to cause measurable changes in normal cellmembrane permeability. Heat-induced cell membrane per-meability can be exploited to increase drug delivery, forexample, transdermally, or into tumor cells. Increased vas-cular permeability due to thermal increase of endothelialgap size also aids drug delivery into tumors. At highertemperatures, heat damage to membranes can cause celldeath, but it will also interfere with therapeutic approachesthat depend on membrane integrity (e.g., receptor targeteddrug delivery, antibodies, etc.). Irreversible disruption ofcytoplasmic microtubule organization and eventual disag-gregation, as well as disruption of actin stress fibers andvimentin filaments, occur at high temperatures (43–45 8C)above those used in whole-body hyperthermia, but thesecytoskeletal effects are of concern with loco-regional hyper-thermia.

A variety of effects in the cell nucleus also occur at hightemperatures (>41 8C) including damage to the nuclearmembrane, increases in nuclear protein content, changesin the structure of nucleoli, inhibition of DNA synthesisand chromosomal damage in S-phase. These changes innuclear structure compromise nuclear function and maycause cell death, though they are unlikely to be significantat the temperatures achieved in systemic hyperthermia.Disaggregation of the spindle apparatus of mitotic cellsmay be responsible for the high thermal sensitivity of cellsin mitosis, as well as in S phase. Hyperthermic inactivationof polymerase b, an enzyme primarily involved in DNArepair, is sensitized by anesthetics and may have a role toplay in the enhancement of the effects of ionizing radiationby systemic hyperthermia, as well as in augmenting thecytotoxic effect of drugs that cause DNA damage.

Metabolic Effects

Moderate increases in temperature lead to increased cel-lular reaction rates, which may be seen as increased oxy-gen consumption and glucose turnover. In consequence,cells may become deprived of nutrients, the intracellularATP concentration falls, accumulation of acid metabolitesincreases pH, and thermal sensitivity increases. Suchconditions are found in tumors and may contribute to theirsensitivity to heat. Further acidifying tumor cells duringhyperthermic treatment seems a promising approach as isdiscussed further below. At high temperatures, the citricacid cycle may be damaged leading to other acidic meta-bolites. Increased plasma acetate has been measured fol-lowing clinical whole-body hyperthermia treatments,which reduces both release of fatty acids from adiposetissue into plasma and subsequent lipid oxidation.

Endocrine Function

Increases in plasma levels of an array of hormones havebeen noted after whole-body hyperthermia. IncreasedACTH levels appear to be accompanied by increased levelsof circulating endorphins. This may explain the sense ofwell-being felt by many patients after systemic hyperther-mia treatment, and the palliative effect of hyperthermiatreatments for cancer. Increased secretion of somatotropichormone after systemic hyperthermia has also been mea-sured (7).

Thermal Tolerance

Thermal tolerance is a temporary state of thermal resis-tance, common to virtually all mammalian cells, whichdevelops after a prolonged exposure to moderate tempera-tures (40–42 8C), or a brief heat shock followed by incuba-tion at 37 8C, and also certain chemicals. The decay ofthermotolerance occurs exponentially and depends on thetreatment time, the temperature, and the proliferativestatus of the cells. Several days are usually required forbaseline levels of heat sensitivity to be regained, which hasimportant implications for fractionated therapy. When pHis lowered, less thermal tolerance develops, and its decay isslower. Thus the long periods at moderate temperatureachieved by clinical systemic hyperthermia systems should

48 HYPERTHERMIA, SYSTEMIC

Page 8: Hyperthermia Systemic

induce thermal resistance in normal cells, while the acidicparts of tumors should be relatively unaffected. This hasnot, however, been studied clinically. The mechanismsinvolved in the induction of thermotolerance are not wellunderstood, but there is mounting evidence that heat shockproteins are involved.

Step-Down Sensitization

Another distinct phenomenon is step-down sensitization inwhich an exposure of cells to temperatures >43 8C resultsin increased sensitivity to subsequent temperatures of42 8C or lower. This can be important clinically for localand regional hyperthermia if there are marked variationsin temperature during the course of treatment, the mag-nitude of the effect depending on the magnitude of thetemperature change. It has been suggested that this phe-nomenon could be exploited clinically by administering ashort, high temperature treatment prior to a prolongedtreatment at a lower temperature, thereby reducing painand discomfort. Since temperatures >43 8C cannot be tol-erated systemically, a local heat boost would be required totakeadvantageof this effect for whole bodyhyperthermia. Sofar, there is no evidence that tumor cells are differentlysensitized by step-down heating than normal cells.

Effect of Hyperthermia on Tumors

It was initially thought that tumor cells have intrinsicallyhigher heat sensitivity than normal cells, but this is notuniversally true. Although some neoplastic cells are moresensitive to heat than their normal counterparts, thisappears to be the case at temperatures higher than thoseused in systemic hyperthermia. Tumors in vivo, on theother hand, often do have a higher thermal sensitivity thannormal tissues because of abnormal vasculature (reducedblood flow), anaerobic metabolism (acidosis), and nutrientdepletion. Due to their tortuous and poorly constructedvasculature, tumors have poor perfusion, thus heat dis-sipation by convection is reduced. At high temperatures(43 8C and up) this means that tumors become a heatreservoir with a consequent rise in temperature, whichif maintained for too long damages the microcirculationand further impairs convective heat loss. Also increasedfibrinogen deposition at damaged sites in the vascular wallleads to clusion of tumor microvessels. Significant heatingof the tumor cells results, which may be directly cytotoxic.Additionally, the impaired blood flow brings about acidosis,increased hypoxia and energy depletion all of whichincrease the heat sensitivity of tumor cells (8). At lowertemperatures, typical of those achieved in whole-bodyhyperthermia, blood flow increases (9) though the mechan-ism is not well understood. For these reasons, along withthe historical evidence for antitumor effects of fever and themetastatic nature of malignant disease, cancer has becomethe main focus of systemic hyperthermia.

Systemic hyperthermia results in increased delivery ofdrugs to tumor sites because of increased systemic bloodflow. It can also increase blood vessel permeability byincreasing the effective pore size between the loosely boundendothelial cells forming tumor microvessels, permittinglarger molecules, such as nanoparticles and gene therapyvectors, to pass into the interstitium (10). Figure 3 showsincreased uptake of 210 nm liposomes in rat breast tumorsafter 1 h of 41.5 8C whole-body hyperthermia. Heat mayalso be toxic to endothelial cells, resulting in a transientnormalization of vascular architecture and improvementin blood flow (11). Another barrier to drug delivery is thehigh interstitial pressure of many tumors. Since whole-body hyperthermia, even at fever-range temperatures,causes cell death (apoptosis and necrosis) within tumorsit reduces the oncotic pressure allowing greater penetrationof large molecules. Table 3 summarizes the interactions ofsystemic hyperthermia which facilitate nanoparticle deliv-ery to tumors.

HYPERTHERMIA, SYSTEMIC 49

0.0

1.0

2.0

100 nm 210 nm

41.5

˚C/3

7˚C

up

take

rat

io

Liposome size

Figure 3. Increase in tumor uptake of large liposomes after 1 hof 41.5 8C whole-body hyperthermia. Systemic heat treatmentincreased the effective pore size from �210 to 240 nm. Becauseof the large pore size in MTLn3 tumors, 100 nm (average diameter)liposomes were able to pass into the tumor equally well at normaland elevated temperatures. The increased effective pore size dueto hyperthermia allowed larger 200 nm liposomes, which werepartially blocked at normal temperatures, to pass more effectivelyinto the tumor.

Table 3. Whole-Body Hyperthermia Facilitates Nanoparticle Therapy

Heat Interaction Therapeutic Effect

" Blood flow " Nanoparticle delivery to tumor" In endothelial gap size " Nanoparticles in interstitium" Endothelial cell apoptosis/necrosis ! transient normalization of vasculature " Nanoparticles in interstitium" Tumor cell apoptosis/necrosis # oncotic pressure " Nanoparticles in interstitiumTemperature-dependent " in permeability of liposome bilayer " And synchronization of drug releaseCellular and molecular effects in tumor " Drug in tumor cell " drug efficacyDirect interactions with drug " Drug efficacy

Page 9: Hyperthermia Systemic

Whole-Body Hyperthermia and the Immune System

An increase in ambient temperature can serve as a naturaltrigger to the immune system and it appears that the thermalmicroenvironment plays a critical role in regulating events inthe immune response. The early work of Coley on cancertherapy with infectious pyrogens implicated fever-inducedimmune stimulation as the mediator of tumor responses (1).While there have been numerous in vitro studies of the effectof temperature on components of the immune system, indi-cating that the thermal milieu regulates T lymphocytes,natural killer (NK) cells, and dendritic cells (DC), in vivoexaminations of the immune effects of systemic hyperther-mia are relatively few. Initial animal model studies con-cluded that whole-body hyperthermia resulted inimmunosuppression, but high temperatures were used,tumors were mostly immunogenic, and immune responsewas merely inferred from the incidence of metastatic spreadrather than from measurement of specific markers ofimmune system activation. The majority of in vivo studiesin animals provide evidence of a nonspecific host reactionin response to hyperthermia in which both T and Blymphocytes, as well as macrophages, are involved (12).Although NK cells are intrinsically more sensitive in vitroto heat than B and T cells, their activation by systemichyperthermia has been observed. Microwave inducedwhole-body hyperthermia of unrestrained, unanesthe-tized mice at 39.5–40 8C for 30 min, three or six timesweekly, resulted in increased NK cell activity and reducedpulmonary metastasis in tumor-bearing mice, but none innormal mice (13). Evidence for hyperthermia-inducedhuman tumor lysis by IL-2 stimulated NK cells activatedby HSP72 expression also exists (14). Increased numbersof lymphocyte-like cells, macrophages, and granulocytesare observed in the tumor vasculature and in the tumorstroma of xenografts and syngeneic tumors in mice imme-diately following a mild hyperthermia exposure for 6–8 h.In the SCID mouse/human tumor system tumor cellapoptosis seen following treatment was due largely tothe activity of NK cells. The investigators hypothesizeheat dilatation of blood vessels and increased vessel per-meability may also give immune effector cells greateraccess to the interior of tumors (15). In balb/C mice,fever-range whole-body hyperthermia increased lympho-cyte trafficking, resulting in early responsiveness to anti-gen challenge (16). Thus systemic hyperthermia may bean effective, nontoxic adjuvant to immunotherapy.

A recent clinical study examined the effect of whole-body hyperthermia combined with chemotherapy on theexpression up to 48 h later of a broad range of activationmarkers on peripheral blood lymphocytes, as well as serumcytokines and intracellular cytokine levels in T cells, andthe capacity of these cells to proliferate. Immediately aftertreatment with 60 min of 41.8 8C WBH as an adjunct tochemotherapy, a drastic but transient, increase in periph-eral NK cells and CD56þ cytotoxic T lymphocytes wasobserved in the patients’ peripheral blood. The numberof T cells then briefly dropped below baseline levels, aphenomeonon that has also been observed by others (17).A marked, but short-lived, increase in the patients’ serumlevels of interleukin-6 (IL-6) was also noted. Significantly

increased serum levels of tumor necrosis factor-alpha(TNF-alpha) were found at 0, 3, 5 and 24 h posttreatment.Further immunological consequences of the treatmentconsisted of an increase in the percentage of peripheralcytotoxic T lymphocytes expressing CD56, reaching a max-imum at 48 h post-WBH. Furthermore, the percentage ofCD4+ T cells expressing the T cell activation marker CD69increased nearly twofold over time, reaching its maximumat 48 h. Since similar changes were not observed inpatients receiving chemotherapy alone, this study pro-vided strong evidence for prolonged activation of humanT cells induced by whole-body hyperthermia combined withchemotherapy (18).

Activation of monocytes has been observed following hotwater bath immersion such that response to endotoxinstimulation is enhanced with concomitant release ofTNF-a. Macrophage activation and subsequent lysosomalexocytosis were observed in the case of a patient treated forliver metastases by hyperthermia. Lysosomal exocytosisinduced by heat may be an important basic reaction of thebody against bacteria, viruses, and tumor growth and wasproposed as a new mechanism of thermally induced tumorcell death mediated by an immune reaction (19).

Several investigators have suggested that the immunechanges seen during in vivo whole-body hyperthermia aremediated by elevations in the plasma concentrations ofeither catecholamines, growth hormone, or beta-endorphins.In volunteers immersed in a heated water bath, neitherrecruitment of NK cells to the blood, nor the percentagesor concentrations of any other subpopulations of blood mono-nuclear cells were altered by hormone blockade. However,somatostatin partly abolished the hyperthermia inducedincrease in neutrophil number. Based on these data andprevious results showing that growth hormone infusionincreases the concentration of neutrophils in the blood, itwas suggested that growth hormone is at least partlyresponsible for hyperthermia induced neutrophil increase.A similar study suggested that hyperthermic inductionof T lymphocytes and NK cells is due to increased secre-tion of somatotropic hormone (7).

The peripheral blood level of prostaglandin E2 (PGE2),which may act as an angiogenic switch, transforming alocalized tumor into an invasive one by stimulating newblood vessel growth, and which also has an immunosup-pressive effect, is elevated in patients with tumors com-pared to healthy control subjects. In a clinical study ofcancer patients receiving 1–2 h of 41.8–42.5 8C whole-bodyhyperthermia, or extracorporeal hyperthermia, bloodlevels of PGE2 decreased markedly after treatment andcorrelated with tumor response (20).

In addition to their role as protectors of unfoldingproteins, extracellular heat shock proteins (HSP) can actsimultaneously as a source of antigen due to their ability tochaperone peptides and as a maturation signal for dendri-tic cells, thereby inducing dendritic cells to cross-presentantigens to CD8þ T cells (21). Heat shock proteins can alsoact independently from associated peptides, stimulatingthe innate immune system by eliciting potent proinflam-matory responses in innate immune cells. The heat shockresponse also inhibits cyclooxygenase-2 gene expression atthe transcriptional level by preventing the activation of

50 HYPERTHERMIA, SYSTEMIC

Page 10: Hyperthermia Systemic

nuclear factor-kappaB (NFkB) (22). Thermal upregulationof HSPs (HSP70 and HSP110) is strongest in lymphoidtissues and may relate to the enhanced immune responsesthat are observed during febrile temperatures. It has beenproposed that local necrosis induced by hyperthermictreatment induces the release of HSPs, followed by uptake,processing and presentation of associated peptides by den-dritic cells. By acting as chaperones and as a signal fordendritic cell maturation, HSP70 might efficiently primecirculating T cells. Therefore, upregulating HSP70 andcausing local necrosis in tumor tissue by hyperthermiaoffers great potential as a new approach to directly activatethe immune system, as well as to enhance other immu-notherapies (23,24).

CLINICAL TOXICITIES OF WHOLE-BODY HYPERTHERMIATREATMENT

At fever-range temperatures, adverse effects of systemichyperthermia treatment are minimal however, at highertemperatures they can be significant, even fatal. On theother hand, the teratogenic effects (birth defects, stillbirths, spontaneous abortions) and 8Cular damage (catar-act induction) resulting from electromagnetic fields used inlocal hyperthermia are not seen in systemic hyperthermia.The transient cardiorespiratory effects of elevated tem-perature can, however, lead to severe toxicity. Elevatedheart rate, especially at high temperatures may result inarrythmias or ischemic heart failure, consequentlypatients have to be very carefully screened with regardto their cardiac status. Beta blockade has generally beenfound to be deleterious although infusion of esmolol hasbeen safely carried out (25). Pulmonary hypertension andedema due to capillary leak may also be seen, but like thecardiac effects, these return to baseline a few hours aftertreatment. Increased serum hepatic enzymes have beennoted, but these may be cancer related. All these toxicitiesare less prevalent or less severe with radiant heat systems,particularly at lower temperatures, and when light con-scious sedation is used rather than general anesthesia. Forexample, decreased platelet count, decreased plasma fibri-nogen, and other factors leading to increased blood clottinghave been noted, particularly in extra-corporeal hyper-thermia, but also with other methods of heating carriedout under inhalation-administered anesthesia drugs.On the other hand, with whole-body hyperthermia underconscious sedation there is no evidence of plateletdrops (26) and animal studies even show platelet stimula-tion providing protection against radiation inducedthrombocytopenia.

Since systemic hyperthermia is almost never used as asingle treatment modality, it is important to recognize thatwhole-body hyperthermia combined with radiation andchemotherapy can enhance some of the toxicities asso-ciated with these modalities. For example, the cardiotoxi-city of doxorubicin and both the renal toxicity andhematological toxicity of platinum agents may increaseunder hyperthermia (27), while the muscle and peripheralnervous system effects of radiation and some drugs can alsobe enhanced (28). Bone marrow suppression is the limiting

toxicity of many chemotherapy drugs but there is little datato suggest that whole body hyperthermia exacerbates thiseffect. On the contrary, the synergy of hyperthermia withseveral chemotherapy agents may mean that lower dosescan be used, resulting in less toxicity. For example, sys-temic hyperthermia combined with carboplatin achievestherapeutic results without elevation of myelosuppressionand responses have occurred at lower than normal doses(29). Pressure sores can easily develop at elevatedtemperatures thus care must be taken not only in patientplacement and support, but also with application of mon-itoring devices. If heat dissipation is locally impaired, forexample, at pressure points, hot spots occur that can lead toburns. This is rarely a problem with fever-range whole-body hyperthermia, but in anesthetized patients under-going high heat regimens burns are not uncommon.

Following systemic hyperthermia treatments, malaiseand lethargy are almost universally experienced althoughthese may be counteracted by pain relief and a sense ofwell-being due to released endorphins. However, the fasterthe target temperature is reached, the less the exhaustion(6), thus attention to minimizing heat dissipation duringthe heat-up phase and using efficient heating devices, suchas those that generate heat by several mechanisms (e.g.,radiant heat and EM fields), add a regional heat boost, orproduce near-IR radiation that is preferentially absorbed,is advantageous to patient well being. Fever after treat-ment in the absence of infectious disease is not uncommonand may be associated with an inflammatory response totumor regression. Nausea and vomiting during the firstcouple of days after treatment are also common. Outbreaksof herpes simplex (cold sores) in susceptible individualshave also been noted, but are easily resolved with acyclovir.

THERMAL DOSE

The definition of dose for systemic hyperthermia is proble-matic. An applied dose would be the amount of heat energygenerated or delivered to the body but even if it can bemeasured, this quantity does not predict biological effects.By analogy with ionizing radiation, the absorbed dosewould be amount of thermal energy absorbed per unitmass of tissue (Jkg�1), however, this is not a quantitythat can be readily measured, or controlled, neither wouldit necessarily predict biological effects. As indicated in theprevious sections, the effects of systemic hyperthermiadepend on (1) the temperature, and (2) the duration ofheating, but not on the energy required to produce thetemperature rise. This leads to the concept of time at agiven temperature as a practical measure of dose. Inreality, however, temperature is seldom constant through-out a treatment, even in the plateau phase of systemichyperthermia, so time at temperature is at best a crudemeasure. Nonetheless, it is the one that is used most oftenclinically for whole-body hyperthermia because of its sim-plicity. Ideally, the dose parameter should allow for com-parison of treatments at different temperatures. Based onthe Arrhenius relationship and measured cell growth inhi-bition curves, the heating time at a given temperaturerelative to the heating time at a standard temperature or

HYPERTHERMIA, SYSTEMIC 51

Page 11: Hyperthermia Systemic

thermal dose equivalent (TDE), was defined empirically as,

T1 ¼ t2 RðT1�T2Þ ð10Þ

A discontinuity occurs in the temperature-time curvesbetween 42 and 43 8C for both cells in culture and heatedtissues, thus the value of R changes for temperatures abovethe transition: R � 2< 42.5 8C and R � 5> 42.5 8C in vitrowhile for in vivo heating studies, R ¼ 2.1 below the transi-tion temperature and 6.4 above 42.5 8C. In practice, a finitetime is required for the body or tissue of interest to reach thetarget temperature, temperature fluctuates even after thetarget temperature is reached, and there is a cooling periodafter heating ceases. If the temperature is measured fre-quently throughout treatment, the temperature–timecurves can be integrated to provide the accumulated ther-mal dose that produces an equivalent effect to that resultingfrom holding the cells–tissue at a constant reference tem-perature for a given a period of time:

t43 ¼Zt f

ti

R43�TðtÞdt ð11Þ

where ti and tf are the initial and final times of the heatingprocedure (30). This thermal isoeffect dose (TID) is usuallyexpressed in minutes is sometimes known as the tdm43 orthe cumulative equivalent minutes (CEM 43 8C). While abiological factor has now been built in to the dose measure,and the integrated TID allows for temperature variationsduring heat-up and cool-down phases, it does not take intoaccount thermal tolerance and step-down sensitization. Noris it particularly relevant to clinical whole-body hyperther-mia where multiple physical and biological effects combinein a complex manner although for a given patient, time–temperature profiles are generally reproducible from onetreatment to another. A further modification attempts totake into account temperature inhomogeneity through themeasurement of temperature at multiple sites and definingT90, namely, that temperature exceeded by 90% of themeasurements (or correspondingly 20%: T20; or 50%:T50). The TID is then expressed as cumulative equivalentminutes that T90 is equal to 43 8C (CEM 43 8C T90) (31).

The efficiency of adjuvant hyperthermia in enhancingthe biological effectiveness of other treatments is oftenreported in terms of the thermal enhancement factor(TEF) or thermal enhancement ratio (TER). This quantityis defined in terms of the isoeffect dose as,

TER ¼

dose of treatment to achieve

a given endpoint

dose of treatment with heat

to achieve the same endpoint

ð12Þ

In clinical and laboratory studies, the TER is oftencomputed on the basis of isodose rather than isoeffect,for example, in the case of hyperthermia plus druginduced arrest of tumor growth, TER ¼ TGDHT/TGTRT,where TGDHT is the tumor growth delay due tohyperthermia plus chemotherapy, and TGTRT is thetumor growth delay resulting from chemotherapy at roomtemperature. Similarly, the enhancing effect of hyperther-

mia on radiation treatment may be expressed throughTER ¼ D0HT/D0RT or TER ¼ LD50HT/LD50RT, where D0is the time required to reduce survival to 1/e of its initialvalue, and LD50 is the lethal dose to 50% of cells.

TEMPERATURE MEASUREMENT

Since systemic hyperthermia achieves a uniform tempera-ture distribution, except for possible partial sanctuary sites,thermometry for systemic hyperthermia is much less chal-lenging than for regional or intracavitary hyperthermia, butit is still important to prevent adverse effects, especiallyburns. Also, convection can induce steep thermal gradients,especially around major blood vessels, so that careful place-ment of temperature probes is required. Most practitionersof whole-body hyperthermia measure temperature in sev-eral locations, typically the rectum, the esophagus, and atseveral skin sites. During heat-up, the esophageal tempera-ture is usually 1–2 8C higher than the rectal temperature,but during plateau phase it drops to 0.5–1.5 8C below therectal temperature. Continuous and accurate temperaturemeasurement is particularly important when temperatures>418C are to be achieved, as critical, life-threateningchanges can occur in minutes or even seconds and overchanges in temperature of as little as 0.1–0.2 8C because ofthe nonlinear response to temperature. For moderate tem-perature systemic hyperthermia, temperature measure-ment to within 0.1 8C is usually adequate, but a precisionof 0.01 8C is desirable when heating to >41 8C and alsoallows determination of the specific absorption rate from theslope of the temperature versus time curve. The tempera-ture measuring device must be insensitive to all otherinfluences, such as ambient temperature, moisture, nearbyelectromagnetic fields, and so on and satisfying this criter-ion can be difficult. Frequent calibration of thermometers inthe working range of temperatures is important since somethermometers appear fine at 30 8C, but drift substantially at40 8C and above. Stringent quality control of any thermo-metry system is required to monitor accuracy, precision,stability, and response time.

Table 4 summarizes the different types of thermometerprobes available for internal and external body tempera-ture measurements, and their relative merits and disad-vantages for systemic hyperthermia. Thermistors are mostoften used for standard temperature monitoring sites whilethermocouples are used for tumor or other intra-tissuemeasurements. Recently, noninvasive methods of tem-perature measurement have been developed that arebeginning to see application in hyperthermia. Thermogra-phy provides a two-dimensional (2D) map of surface tem-perature by measurement of infrared emission from thebody, though deep-seated hot structures may be visualizedbecause of heat carried by blood flow from the interior heatsource to the skin. It is useful to detect skin hotspots andtherefore in burn prevention. Since temperature-inducedchanges in the mechanical properties of tissue lead toaltered ultrasound propagation velocity, mapping of ultra-sound velocity can also provide a visual map of tempera-ture. Tomographic reconstruction of 2D or 3D temperatureis theoretically possible, but it is difficult in practicebecause of the heterogeneity of tissue characteristics. A

52 HYPERTHERMIA, SYSTEMIC

Page 12: Hyperthermia Systemic

Ta

ble

4.

Tem

pera

tures

Pro

bes

for

Sy

stem

icH

yp

erth

erm

ia

Pro

be

Typ

eM

easu

rem

ent

Pri

nci

ple

Acc

ura

cyS

ensi

tivit

yS

tabil

ity

Ad

van

tages

orD

isad

van

tages

Cli

nic

al

Exp

an

sion

ofm

ercu

ryor

alc

ohol

ingla

ssM

oder

ate

�0.1

8CL

owH

igh

Larg

esi

ze,

infl

exib

le.

Slo

wre

spon

se.

Pla

tin

um

resi

stan

ceth

erm

omet

erL

inea

rre

sist

an

cech

an

ge

wit

hte

mp

eratu

reH

igh�

0.0

28C

Use

das

stan

dard

for

cali

bra

tion

ofot

her

typ

esof

ther

mom

eter

s.

Exp

ensi

ve.

Dif

ficu

ltto

cali

bra

te.

Larg

esi

ze.

Sen

siti

ve

tosh

ock

.

Th

erm

ocou

ple

See

bec

kef

fect

:te

mp

eratu

red

epen

den

tvol

tage

dif

fere

nce

bet

wee

ntw

oco

nd

uct

ors

mad

eof

dif

fere

nt

met

als

Mod

erate

�0.1

8CM

oder

ate

toh

igh

Mod

erate

Sm

all

sen

sor.

Non

lin

ear

vol

tage

chan

ge

wit

hte

mp

.S

ensi

tive

toE

Mfi

eld

s.C

an

’th

an

dle

stee

pte

mp

.gra

die

nts

.

Th

erm

isto

r(e

.g.,

Bow

man

Loo

pL

ars

enp

robe)

Inver

sere

lati

onsh

ipbet

wee

nte

mp

eratu

rean

dse

mic

ond

uct

orre

sist

an

ce

Hig

h<

0.0

58C

Hig

hP

oor

Req

uir

efr

equ

ent

reca

libra

tion

Sh

ort

tim

eco

nst

an

t.N

otin

terc

han

gea

ble

.S

ensi

tive

toE

Mfi

eld

s.

GaA

sT

emp

eratu

resp

ecifi

cabso

rpti

onM

oder

ate

Low

Sm

all

size

Op

tica

l(fi

ber

opti

cp

robe)

:C

han

ge

w/t

emp

.:N

otse

nsi

tive

toE

Mfi

eld

s.S

ma

llsi

ze.

LC

Dbir

efri

ngen

tcr

yst

al

flu

ores

cen

tp

hos

ph

orC

olor

refl

ecta

nce

Low

Low

Un

stable

Ref

ract

ion

ofp

olari

zed

ligh

tL

owD

ecay

offl

uor

esce

nce

Ver

yh

igh

53

Page 13: Hyperthermia Systemic

Ta

ble

5.

Su

mm

ary

of

Cli

nic

al

Tria

lso

fW

ho

le-B

od

yH

yp

erth

erm

iaa

Fir

stA

uth

orP

ubli

cY

ear

Stu

dy

Typ

eN

um

ber

ofP

ati

ents

Dis

ease

Pro

toco

lR

esu

ltof

WB

HR

efer

ence

,P

MID

WB

HA

lon

eK

raybil

l,W

.G.

2002

Ph

ase

IA

dvan

ced

soli

dtu

mor

s3–6

hat

39.5

–40.0

8CW

ell

tole

rate

dN

osi

gn

ifica

nt

ad

ver

seev

ents

#in

circ

ula

tin

gly

mp

hoc

yte

s

16,

12028640

Ste

inh

au

sen

,D

.1994

Ph

ase

I103

Ad

van

ced

refr

act

ory

orre

curr

ent

can

cers

1h

at

41.8

8Cþ

hyp

ergly

cem

iaþ

hyp

erox

emia

Min

imal

sid

eef

fect

s52

resp

onse

s(5

0%

)8023241

WB

Ch

emoth

era

py

Bak

shan

deh

,A

.2003

Ph

ase

II25

Non

met

ast

ati

cm

ali

gn

an

tp

leu

ral

mes

oth

elio

ma

1h

at

41.8

8Cþ

ifos

fam

ide

þca

rbop

lati

etop

osid

eG

rad

eII

I/IV

neu

trop

enia

an

dth

rom

boc

yto

pen

ia5

part

ial

rem

issi

ons

(20%

)

12609573

Bu

ll,

J.M

.1992

Ph

ase

II17

Ad

van

ced

met

ast

ati

csa

rcom

a2

hat

41.8

–42.0

8Cþ

BC

NU

Lim

itin

gto

xic

ity

¼th

rom

boc

yto

pen

ia7

resp

onse

s/S

D(4

1%

)"

surv

ival

33,

1607734

Bu

ll,

J.M

.2002

Ph

ase

I13

Vari

ous

chem

oth

erap

yre

sist

an

tca

nce

rs6

hat

40.0

8Cþ

dox

ilþ

5-F

met

ron

omic

inte

rfer

on-a

Gra

de

III

toxic

itie

s9

resp

onse

s/S

D(6

9%

)

60

Bu

ll,

J.M

.2004

Ph

ase

I33

Ad

van

ced

met

ast

ati

cca

nce

rs(G

I,bre

ast

,h

ead

an

dn

eck

,sa

rcom

a,

neu

roen

doc

rin

e)

6h

at

40.0

8Cþ

cisp

lati

gem

cita

bin

inte

rfer

on-a

20

resp

onse

s/S

D(6

6%

)"

surv

ival"

qu

ali

tyof

life

35

Dou

wes

,F

.2004

Pil

ot21

Ovari

an

can

cer

1-2

hat

41.5

–42.0

8Cþ

cisp

lati

nor

carb

opla

tin

þh

yp

ergly

cem

ia

18

resp

onse

s/S

D(8

6%

)"

qu

ali

tyof

life

15108039

En

gel

hard

t,R

.1990

Pil

ot23

Ad

van

cem

etast

ati

cm

elan

oma

1h

at

41.0

8Cþ

cisp

lati

dox

oru

bic

inS

ligh

t"

inm

yel

otox

icit

y10

resp

onse

s/S

DR

esp

onse

rate

¼th

at

inli

tera

ture

for

chem

oalo

ne

52,

2198312

Gu

an

,J.

2005

Ph

ase

II32

Ad

van

ced

can

cers

94%

resp

onse

s/S

DP

ain

red

uct

ion

inall

pts

.In

crea

sed

KP

SD

ecre

ase

dtu

mor

mark

ers

65

Heg

ewis

ch-B

eck

er,

S.

2002

Ph

ase

II41

Pre

trea

ted

ad

van

ced

met

ast

ati

cco

lore

ctal

can

cer

1h

at

41.8

þox

ali

pla

tin

þle

uco

vor

inþ

5F

UN

oex

cess

toxic

ity

31

resp

onse

s/S

D(7

6%

)55,

12181242

Hil

deb

ran

dt,

B.

2004

Ph

ase

I/II

28

Met

ast

ati

cco

lore

ctal

can

cer

1h

at

41.8

–42.1

8Cþ

hyp

ergly

cem

iaþ

hyp

erox

emia

þfo

lin

icaci

5-F

mit

omyci

nC

Gra

de

III/

IVto

xic

itie

s11

resp

onse

s/S

D(3

9%

)44,

15204528

54

Page 14: Hyperthermia Systemic

Hou

,K

.2004

Ph

ase

II54

Ad

van

ced

can

cers

1–2

hat

41.8

–42.5

8C,

extr

aco

rpor

eal

þch

emot

her

ap

yvs.

chem

oth

erap

yalo

ne

75.3

%re

spon

ses/

SD

72.6

%#

tum

orm

ark

ers

70%

pain

reli

efim

pro

ved

slee

p"

wei

gh

t,ap

pet

ite,

KP

SA

llsi

gn

ifly>

con

trol

Rev

ersi

ble

toxic

itie

s

68

Ism

ael

-Zad

e,R

.S.

2005

Pil

ot5

Ped

iatr

icre

nal

cell

carc

inom

a3

hat

41.8

–42.5

8Cþ

dox

oru

bic

inþ

inte

rfer

on-a

No

com

pli

cati

ons

5re

spon

ses

(100%

)15700247

Ku

rpes

hev

,O

.K.

2005

Ph

ase

II42

Vari

ous

dis

sem

inate

dca

nce

rs1–2

hat

41.0

–42.3

8Cþ

pol

y-c

hem

oth

erap

yR

egre

ssio

nof

met

ast

ase

s.P

ain

red

uct

ion

66

Ric

hel

,O

.2004

Ph

ase

II21

Met

ast

ati

can

dre

curr

ent

cerv

ical

can

cer

1h

at

41.8

8Cþ

carb

opla

tin

Gra

de

III/

IVle

uco

pen

ia,

thro

mbop

enia

,an

emia

,re

nal

toxic

ity

16

resp

onse

s/S

D(7

6%

)

57,

15581981

Rob

ins,

H.I

.1993

Ph

ase

I30

Vari

ous

refr

act

ory

can

cers

1h

at

41.8

8Cþ

carb

opla

tin

Myel

otox

icit

y9

resp

onse

s(3

0%

)53,

8355046

Rob

ins,

H.I

.1997

Ph

ase

I16

Vari

ous

refr

act

ory

can

cers

1h

at

41.8

8Cþ

L-P

AM

Low

erp

late

let

nad

irm

yel

osu

pp

ress

ion

8re

spon

ses/

SD

(50%

)

48,

8996137

Str

obl,

B.

2004

Ph

ase

II7

Met

ast

ati

cce

rvic

al

can

cer

1h

at

41.5

–41.8

8Cþ

pacl

itaxel

þca

rbop

lati

nG

rad

eII

alo

pec

iaG

rad

eII

I/IV

thro

mbop

enia

,n

eutr

open

ia"

surv

ival

58

Wes

term

an

nA

.M.

2001

Ph

ase

II14

Pla

tin

um

resi

stan

tov

ari

an

can

cer

1h

at

41.8

8Cþ

carb

opla

tin

Gra

de

IVth

rom

boc

yto

pen

ia,

gra

de

III

neu

trop

enia

9re

spon

ses/

SD

(64%

)

11378341

Wes

term

an

nA

.M.

2003

Ph

ase

II95

Met

ast

ati

csa

rcom

a1

hat

41.8

8Cþ

ifos

fam

ide

þca

rbop

lati

etop

osid

eN

eutr

open

ia,

thro

mboc

yto

pen

ia,

infe

ctio

n58

resp

onse

s/S

D(6

1%

)50,

12759526

WB

Ra

dia

tion

Over

gaard

,J.

1995

Ran

dom

ized

mu

ltic

ente

r70

Met

ast

ati

cm

elan

oma

1h

at

438C

þfr

act

ion

ate

dR

Tvs.

FR

Talo

ne

Imp

roved

loca

ltu

mor

con

trol

,"

surv

ival

41,

7776772

Rob

ins,

H.I

.1990

Pil

ot8

Nod

ula

rly

mp

hom

a,

chro

nic

lym

ph

ocyti

cle

uk

emia

41.8

8Cþ

TB

Ivs.

LO

TB

I8

resp

onse

s/S

D"

surv

ival

24,

2182581

aP

ubli

shed

sin

ce1990.

55

Page 15: Hyperthermia Systemic

number of magnetic resonance (MR) techniques have beenused for thermal mapping and BSD Medical and SIEMENSMedical Systems have collaborated to develop a hybridhyperthermia/MRI system, although it is not a whole-bodyhyperthermia machine. Currently, the most widelyaccepted MR technique is the proton resonance frequency(PRF) method that exploits the temperature dependence ofthe chemical shift of water. Unlike the value of the waterspin-lattice relaxation time or the molecular diffusioncoefficient, both of which have been used for MRI tempera-ture measurements, the thermal coefficient relating tem-perature to the water chemical shift has been shown to beessentially independent of tissue type and physiologicalchanges induced by temperature (32). Recently an inter-leaved gradient echo–echo planar imaging (iGE-EPI)method for rapid, multiplanar temperature imaging wasintroduced that provided increased temperature contrast-to-noise and lipid suppression without compromising spa-tio-temporal resolution (33).

CLINICAL EXPERIENCE

Cancer

Systemic hyperthermia has been used mostly for treatmentof cancer because of its potential to treat metastatic dis-ease. Initial treatments aimed to produce direct killing oftumor cells based on the premise, now understood not to beuniversally true, that cancer cells are more susceptible toelevated temperatures than normal cells, and the higherthe temperature the greater the tumor cell kill. Maximallytolerated temperatures of 41.5–42 8C were therefore main-tained for 1–2 h as the sole treatment. Response rates were,however, disappointing. Tumor regressions were observedin less than half the cases, no tumor cures were achieved,and remissions were of short duration. It became apparentthat the heterogeneity of cell populations within tumors,along with micro-environmental factors, such as blood/nutrient supply, pH, and oxygen tension prevent the ther-motoxic results achieved in the laboratory. Consequently,the focus of research on systemic hyperthermia shifted tousing hyperthermia as an adjunct to other cancer thera-pies, principally chemotherapy and radiotherapy. It isimportant to note that because of the experimental statusof systemic hyperthermia treatment for cancer, almost allclinical trials, summarized in Table 5, have been performedon patients with advanced disease for whom whole-bodyhyperthermia, either as a sole therapy, or as an adjunct, isa treatment of last resort. In these cases, any responsewhatsoever is often remarkable. Nonetheless, a number ofhyperthermia centers in Europe have discontinued sys-temic hyperthermia because the high temperature proto-cols required intensive patient care and led to unacceptabletoxicities, especially in light of the efficacy and reducedtoxicities of newer generation chemotherapies. Large, ran-domized, multicenter, Phase III trials are, however, neededto firmly establish the benefits of systemic hyperthermia inconjunction with chemotherapy and radiation. Also, vali-dation and optimization of fever-range temperature proto-cols are much needed.

Systemic Hyperthermia and Chemotherapy. The bene-ficial interaction of hyperthermia with several classes ofchemotherapy agents, acting via several mechanisms assummarized in Table 6, has spurred a variety of thermo-chemotherapy regimens and several clinical trials of sys-temic hyperthermia and chemotherapy are ongoing. Whilethe results have been mixed, elevated response rates wererecorded in the treatment of sarcoma when systemichyperthermia was combined with doxorubicin and cyclo-phosphamide (54) or BCNU (34). Systemic hyperthermia isthe only way to heat the lung uniformly, and impressiveresponse rates and increased durations of response havebeen achieved in both small cell and nonsmall cell lungcancer treated with the combination of whole bodyhyperthermia at 41 8C for 1 h with adriamycin, cyclopho-sphamide, and vincristine (ACO protocol) (34). Neuroendo-crine tumors also appear to have increased sensitivity tosystemic hyperthermia and multidrug chemotherapy (51).

Optimal combination of whole-body hyperthermia withchemotherapy requires an understanding of the mechan-isms of interaction of heat with individual drugs or drugs incombination. Preclinical data is consistent with the conceptthat the timing of chemotherapy during whole-bodyhyperthermia should affect therapeutic index. For exam-ple, Fig. 4 shows the effect on tumor cures in mammarycarcinoma bearing rats of 6 h of 40 8C whole-bodyhyperthermia administered with, or 24 or 48 h after gem-citabine. A synergistic response was obtained whenhyperthermia was begun with gemcitabine administrationor 48 h later. The effect of gemcitabine was completelynegated, however, when hyperthermia was administered24 h after the start of heating, perhaps due to cell cycleeffects. With cisplatin, the greatest therapeutic index isachieved if the drug is given 24 h before the start of whole-body hyperthermia, thereby preventing thermal augmen-tation of cisplatin induced nephrotoxicity (55). In a clinicalinvestigation of multiple cycles of radiant heat whole-bodyhyperthermia combined with carboplatin, Ifosfamide, eto-poside, and granulocyte colony stimulating factor, it wasfound that toxicity was minimized when carboplatin was

56 HYPERTHERMIA, SYSTEMIC

0.0

1.0

2.0

3.0

4.0

5.0

GEM+WBH schedule

TE

R fo

r tu

mo

r cu

res

GEM+WBH(0) GEM+WBH(+24) GEM+WBH(+48)

GEM+WBH(0 h) GEM+WBH(+24 h) GEM+WBH(+48 h)

0.06

2.983.57

Figure 4. Schedule dependence of fever range whole-bodyhyperthermia enhanced gemcitabine tumor cures. A supraaddi-tive cure rate occurred when whole-body hyperthermia (WBH)was given at the same time as gemcitabine administration or 48 hlater. When hyperthermia followed gemcitabine by 24 h the numberof cures droppedtoalmostzero, wellbelow thenumber achieved withgemcitabine alone.

Page 16: Hyperthermia Systemic

given during the plateau phase of WBH, 10 min after targettemperature was reached (56).

A major rationale for whole-body hyperthermia in can-cer treatment is the ability to treat metastases, but this isactually a controversial issue. There have been no clinicalstudies specifically designed to study the effect of systemichyperthermia on either the efficacy against metastaticdisease or prevention of development of metastases.Increased survival in advanced malignancies is often inter-preted to mean a reduction in metastatic disease, but directmeasurement of the incidence and response of metastasesis rare. Based on some animal studies, it has been sug-gested that systemic hyperthermia could actually promotethe metastatic spread of tumor cells, but this has not beenconfirmed. One clinical study found an increase of tumorcells in blood 24 h after 41.8 8C WBH, but there was no

evidence that this caused metastatic spread of disease (57).Several animal experiments do support the efficacy ofwhole-body hyperthermia against metastases. In mousemodels of lung cancer and melanoma, the number of lungmetastases was scored after repeated systemic microwaveheating. It was found that the number of lung metastaseswas significantly reduced, and NK-cell activity was higher,in treated animals. The authors hypothesized that WBHinterferes with the spread of organ metastases, possiblythrough a mechanism involving NK cells (13). Anotherstudy of mouse Lewis lung carcinoma in which the animalswere treated with 60 min of systemic hyperthermia at42 8C, demonstrated a reduction in the number andpercentage of large metastases (>3 mm) on day 20 post-tumor implantation. Addition of radiation led to a reduc-tion to 50% of control of the number of lung metastases as

HYPERTHERMIA, SYSTEMIC 57

Table 6. Chemotherapy Agents Used with Whole-Body Hyperthermia

Class of AgentLikely Mechanismof Heat Interaction

Drugs Used withWBH in Clinical Studies

InvestigatorReferencesa

Alkylating agents Impaired DNA repair Cyclophosphamide (CTX) Parks, 1983 (4)Improved pharmacokinetics Engelhardt, 1988 (34)

Dacarbazine (DTIC) Lange, 1983 (35)Melphalan (L-PAM) Robins, 1997 (36)Ifosfamide (IFO) Engelhardt, 1988 (34)

Issels, 1990 (37)Westermann, 2003 (38)

Nitrosoureas Impaired DNA repair BCNU Parks, 1983 (4)Improved pharmacokinetics Bull, 1992 (39)

Me-CCNU Bull, 1992 (39)Platinum agents Impaired DNA repair Cisplatin (CDDP) Parks, 1983 (4)

Altered plasma protein binding Herman, 1982 (40)Engelhardt, 1990 (41)Robins, 1993 (42)Douwes, 2004 (43)

Carboplatin (CBDCA) Westermann, 2003 (38)Hegewisch-Becker, 2002 (44)Hegewisch-Becker, 2003 (45)Douwes, 2004 (43)Richel, 2004 (46)Strobl, 2004 (47)

Oxaliplatin Elias, 2004 (48)Hegewisch-Becker, 2002 (44)

Anthracyline antibiotics Impaired DNA repair Adriamycin Engelhardt, 1990 (41)Enzyme activation Bull, 2002 (49)

Bleomycin Herman, 1982 (40)Antimetabolites Increased drug transport 5-FU Lange, 1983 (35)

Cell cycle arrest Larkin, 1979 (50)Impaired DNA repair Bull, 2002 (49)

Hegewisch-Becker, 2002 (44)Gemcitabine Bull, 2004 (51)

Antiproliferatives Impaired DNA repair Etoposide (VP-16) Barlogie, 1979 (52)Issels, 1990 (37)Westermann, 2003 (42)

Topoisomerase inhibitors Impaired DNA repair Irinotecan (CPT-11) Hegewisch-Becker, 2003 (45)Elias, 2004 (48)

Taxanes Microtubule disruption Paclitacel Strobl, 2004 (49)Apoptosis Docetaxel Strobl, 2004 (49)

Biological responsemodifiers

Increased anti-viral andantiproliferative activity

Interferon Robins, 1989 (53)

Bull, 2002, 2004 (34,49)

aReferences prior to 1980, or not in English, are not provided in the Bibliography at the end of this article.

Page 17: Hyperthermia Systemic

well as the percent of large metastases on day 20 (58). In abreast cancer ocult metastasis model in rats, 6 h of 40 8Cwhole-body hyperthermia combined with daily, low dose,metronomic irinotecan resulted in delayed onset, andreduced incidence, of axillary lymph node metastases com-pared to control in rats, as did treatment with 40 8C WBHalone. The combination therapy also reduced axillarymetastasis volume. Interestingly, none of the therapiessignificantly affected inguinal lymph node metastases,but lung metastases were decreased in both the combina-tion therapy and WBH alone groups. Rats treated withfever-range whole-body hyperthermia and metronomicirinotecan also survived significantly longer (36%) thancontrol animals (59).

Systemic Hyperthermia and Radiotherapy. The augmen-tation of ionizing radiation induced tumor kill byhyperthermia is well documented for local hyperthermiaand has led to numerous protocols combining whole-bodyhyperthermia with radiation therapy (60,61). Hyperther-mia is complementary to radiation in several regards:ionizing radiation acts predominantly in the M and G1

phases of the cell cycle while hyperthermia acts largelyin S phase; radiation is most effective in alkaline tissueswhereas hyperthermic cytotoxicity is enhanced underacidic conditions; radiation is not effective in hypoxicregions yet hyperthermia is most toxic to hypoxic cells.Thus when hyperthermia is combined with radiotherapy,both the hypoxic, low pH core of the tumor is treated as wellas the relatively well perfused outer layers of the tumor.Furthermore, because of its vascular effects, hyperthermiaenhances tumor oxygenation thus potentiating radiationcell kill. Hyperthermia also increases the production of oxy-gen radicals by radiation, and reduces the repair of DNAdamage caused by ionizing radiation. Thus hyperthermiaand radiotherapy together often have a synergistic effect,and this combination is now well accepted for treatment of anumber of tumors.

Fever-Range WBH. Like systemic hyperthermia alone,combined modality treatments were initially aimed toachieve maximally tolerated temperatures. Such regi-mens, however, carry significant risk to the patient,require general anesthesia, and necessitate experienced,specialist personnel to provide careful monitoring of vitalsigns and patient care during the treatment. Morerecently, it has been appreciated that lower core bodytemperatures (39–40 8C) maintained for a longer time(4–8 h), much like fever, can indirectly result in tumorregression through effects on tumor vasculature, theimmune response, and waste removal (detoxification).The optimum duration and frequency of mild hyperther-mia treatment has, however, not yet been determined.Protocols range from single treatments of 4–6 h, or similarlong duration treatments given once during each cycle ofchemotherapy, to daily treatments of only 1 h. Severalstudies of mild, fever-range, whole-body hyperthermiawith chemotherapy have demonstrated efficacy against abroad range of cancers (34,17) and clinical trials are cur-rently being conducted at the University of Texas HealthScience Center at Houston, Roswell Park Cancer Institute,

New York, and by the German Interdisciplinary WorkingGroup on Hyperthermia (62).

Systemic Hyperthermia and Metabolic Therapy.Increased rates of metabolic reactions lead to rapid turn-over of metabolites, causing cellular energy depletion,acidosis, and consequent metabolic disregulation. Tumors,which have increased metabolic rates [glucose, adenominetriphosphate (ATP)] compared to normal cells, may beparticularly sensitive to thermally induced energy deple-tion and this has been exploited in the Cancer MultistepTherapy developed by von Ardenne, which is a combinedhyperthermia–chemotherapy–metabolic therapy approachto cancer (63). The core of this approach is systemichyperthermia at 40–42 8C, sometimes with added localhyperthermia to achieve high temperatures within thetumor. A 10% solution of glucose is infused into the patientto achieve a high accumulation of lactic acid within thetumor that cannot be cleared because of sluggish blood flowand confers an increased sensitivity to heat to the tumorcells. Administration of oxygen increases the arterial oxy-gen pressure and stimulates lysozymal cytolysis. Finallylow dose chemotherapy is added.

Palliation. Pain relief is reported by many patientsreceiving systemic hyperthermia treatment, whether withchemotherapy or radiation. Indeed, almost all patientsundergoing thermoradiotherapy report pain relief.Immediate pain relief following treatment is likely to stemfrom an increased level of circulating b-endorphins, whilelonger term pain relief may be due to increased blood flow,direct neurological action, and disease resolution, forexample, tumor regression in cancer patients, or detoxifi-cation. Meaningful improvements in quality of life typi-cally result from such pain relief. Localized infraredtherapy using lamps radiating at 2–25 mm is used forthe treatment and relief of pain in numerous medicalinstitutes in China and Japan.

Diseases Other than Cancer. Therapeutic use of heatlamps emitting IR radiation is commonplace throughoutthe Orient for rheumatic, neurological and musculoskele-tal conditions, as well as skin diseases, wound healing, andburns. The improvements reported appear to be largelydue to increased blood flow bringing nutrients to areas ofischemia or blood vessel damage, and removing wasteproducts. Scientific reports of these treatments are, how-ever, difficult to find. Application of heat via hot baths orultrasound has long been standard in physical therapy forarthritis and musculoskeletal conditions, though ice packsare also used to counter inflammatory responses. Heatdecreases stiffness in tendons and ligaments, relaxes themuscles, decreases muscle spasm, and lessens pain. Unfor-tunately, few clinical trials of efficacy have been performed,and methodological differences or lack of rigor in thestudies hinder comparisons (64). A clinical trial inJapan reported a supposedly successful solution for sevenout of seven cases of rheumatoid arthritis treated withwhole-body IR therapy, and it is reported that the King ofBelgium was cured of his rheumatoid arthritis in threemonths due IR treatments. Systemic hyperthermia with

58 HYPERTHERMIA, SYSTEMIC

Page 18: Hyperthermia Systemic

whole-body radiant heat units is being carried out inclinical centers as well as many private clinics in Germanyfor the purpose of alleviating rheumatoid arthritis. It hasbeen proposed that the induction of TNF receptors by WBHmay induce a remission in patients with active rheumatoidarthritis. The use of heat packs has long been standard torelieve the pain of fibromyalgia. Again, the therapeuticeffect is believed to be due to increased circulation flushingout toxins and speeding the healing process. Whole-bodyhyperthermia treatment for fibromyalgia and chronic fati-gue syndrome (CFS) is to be found in a number of privateclinics. Hyperthermia increases the number and activity ofwhite blood cells, stimulating the depressed immune sys-tem of the CFS patient.

Because of its immune stimulating effects, whole-bodyhyperthermia is a strong candidate for treatment of chronicprogressive viral infections, such as HIV and hepatitis C. Aclinical trial at the University Medical Center Utrecht, TheNetherlands has evaluated extracorporeal heating to inducesystemic hyperthermia of 41.8 8C for 120 min under propo-fol anesthesia for treatment of hepatitis C (65). Humanimmunodeficiency virus (HIV)-infected T cells are moresensitive to heat than healthy lymphocytes, and suscept-ibility increases when the cells are presensitized by expo-sure to tumor necrosis factor. Thus, induction of whole-bodyhyperthermia or hyperthermia specifically limited to tissueshaving a high viral load is a potential antiviral therapy foracquired immunodeficiency syndrome (AIDS). An Italianstudy has found treatment of AIDS with beta-carotene andhyperthermia to be synergistic, preventing progression ofearly disease and also increasing the survival time inpatients with severe AIDS. A single treatment of low flowextracorporeal hyperthermia was found effective againstAIDS associated Kaposi’s sarcoma, though there was sig-nificant toxicity. Core temperature was raised to 42 8C andheld for 1 h with extracorporeal perfusion and ex vivoblood heating to 49 8C. Complete or partial regressionswere seen in 20/29 of those treated at 30 days post-treat-ment, with regressions persisting in 14/29 of those treatedat 120 days post-treatment. At 360 days, 4/29 maintainedtumor regressions with 1 patient being in complete remis-sion still at 26 months (66).

THE FUTURE OF SYSTEMIC HYPERTHERMIA

While there is a resurgence of interest in systemichyperthermia, this modality has not yet been adopted asa mainstream therapy, and optimal clinical trials havenot yet been carried out. Well-designed, well-controlled,multicenter clinical trials need to be conducted. In order tounequivocally demonstrate the utility of whole-body hyper-thermia inthetreatmentofcanceraswellasotherdiseases, itwill be important to accrue a sufficiently large number ofpatients who do not have end-stage disease. Thanks to thecommercial availability of systemic hyperthermia systems,the variability between induction techniques at differentinstitutions can be removed. Newer instrumentation, parti-cularlynear-IR radiantheatdevices,alongwithtreatmentatlower temperatures (fever-range thermal therapy) shouldlead to significantly reduced toxicity. Better exploitation of

the narrow window of effective temperatures within whichthe cellular effects of heat can be exploited yet damageremains minimal, and improved understanding of the biolo-gical interactions invivoof systemicheatwithchemotherapyand radiation will be essential to optimize therapy.

The effects of systemic hyperthermia on tumor bloodflow and vascular permeability have the potential toincrease delivery of various small molecules, nanoparticles,and gene therapy vectors to tumors. Ferromagnetic nano-particles can be heated by external magnetic fields and offerthe potential for internal hyperthermia, both locally andsystemically. Thermally sensitive liposomes that releasetheir contents at designated temperatures are also of inter-est. The ability of systemic hyperthermia to aid in systemicdelivery of gene therapy vectors (the holy grail of genetherapy) and enhance transfection of cells with therapeuticgene plasmids is under investigation in several laboratories(67,68), and shows potential along with targeted gene ther-apy via the heat shock response. For example, Fig. 5 shows afourfold hyperthermic increase of therapeutic gene deliveryto tumor when plasmid DNA was injected intravenouslyinto mammary carcinoma bearing rats immediately after 6h of whole-body hyperthermia at 40 8C. Thus systemichyperthermia is likely to see increasing application as anenhancer of drug delivery.

There is a great deal of interest in the immunologicalconsequences of whole-body hyperthermia, and as theybecome better understood, the combination of systemichyperthermia with specific immunotherapies will undoubt-edly be pioneered, not just for cancer but also, by analogywith fever, in a broad range of diseases.

SUMMARY

Systemic hyperthermia is founded on solid physical andbiological principles and shows promise in the treatmentof a number of diseases. Modern whole-body hyperther-mia devices use IR-A radiation sources together witheffective heat loss techniques to achieve a controlled,uniform temperature distribution throughout the bodywith minimal patient toxicity. A shift in paradigm hasoccurred away from achieving direct cell killing with short

HYPERTHERMIA, SYSTEMIC 59

0

1

2

3

4

5

Liver Tumor

40˚C

/37˚

C r

atio

of

HS

Vtk

m

ole

cule

s p

er c

ell

grp78-HSVtk

Figure 5. Fever range whole-body hyperthermia increasestherapeutic gene (grp78-HSVtk) delivery in tumor.

Page 19: Hyperthermia Systemic

bouts of maximally tolerated temperatures, to inducingindirect curative effects through longer duration treatmentsat lower temperatures, and synergy with other modalities,such as radiotherapy. Better understanding of the interac-tions of elevated temperature with metabolic and geneticpathways will allow thermally driven targeted therapies.Of particular promise is the use of systemic hyperthermiaas an immune system stimulator and adjunct to immu-notherapy. Application of systemic hyperthermia to nano-particle delivery and gene therapy is emerging. Whole-bodyhyperthermia is moving from being dubbed an alternativetherapy to becoming a standard treatment and clinicalhyperthermia centers are to be found all over the world.

Useful Websites

http://www.ncbi.nlm.nih.gov/books/bv.fcgi?rid=hstat6.section.40680

Techniques andDevices Usedto ProduceHyperthermia

http://www.ncbi.nlm.nih.gov/books/bv.fcgi?rid=cmed.section.7813

Physics andPhysiology ofHeating

http://www.duke.edu/~dr3/hyperthermia_general.html

Clinical HyperthermiaBackground

http://www.eurekah.com/isbn.php?isbn=1-58706-248-8&bookid=143&catid=50

Online book:LocoregionalRadiofrequency-perfusionaland Whole BodyHyperthermia inCancer Treatment:New Clinical Aspects,E.F. Baronzio andA. Gramaglia (eds.),Eurekah BioscienceDatabase

http://www.esho.info/professionals/

European Society forHyperthermicOncology

http://www.hyperthermie.org/index2.html

GermanInterdisciplinaryWorking group onhyperthermia

http://www.uth.tmc.edu/thermaltherapy/

Systemic ThermalTherapy at theUniversity of Texas

BIBLIOGRAPHY

1. Nauts HC. Bacterial pyrogens: beneficial effects on cancerpatients. In: Gautherie M, Albert E, editors. Biomedical Ther-mology, Progress in Clinical Biological Research. New York:Alan R. Liss; 1982. p 687–696.

2. Law HT, Pettigrew RT. Heat transfer in whole bodyhyperthermia. Ann NY Acad Sci 1980;335:298–310.

3. Robins HI, et al. A non-toxic system for 41.8 8C whole bodyhyperthermia: results of a phase I study using a radiant heatdevice. Cancer Res 1985;45:3937–3944.

4. Parks LC, Smith GV. Systemic hyperthermia by extracorpor-eal induction techniques and results. In: Storm FK, editor.

Hyperthermia in Cancer Therapy. Boston: Hall; 1983.pp 407–446.

5. van der Zee J, van Rhoon GC, Faithfull NS, van den Berg AP.Clinical hyperthermic practice: whole body hyperthermia. In:Field SB, Hand JW, editors. An Introduction to the PracticalAspects of Clinical Hyperthermia. London: Taylor & Francis;1990.

6. Wust P, et al. Feasibility and analysis of thermal parametersfor the whole-body-hyperthermia system IRATHERM-2000.Int J Hyperthermia 2000 Jul–Aug; 16(4):325–339.

7. Blazickova S, Rovensky J, Koska J, Vigas M. Effect ofhyperthermic water bath on parameters of cellular immunity.Int J Clin Pharmacol Res 2000;20(1–2):41–46.

8. Vaupel P, Kallinowski F. Physiological effects of hyperthermia. In:Streffer C, editor. Hyperthermia and the Therapy of MalignantTumors. Berlin/Heidelberg, Germany: Springer-Verlag; 1987.

9. Dudar TE, Jain RK. Differential response of normal and tumormicrocirculation to hyperthermia. Cancer Res 1984;44(2):605–612.

10. Kong G, Braun RD, Dewhirst MW. Characterization of theeffect of hyperthermia on nanoparticle extravasation fromtumor vasculature. Cancer Res 2001;61:3027–3032.

11. Jain RK. Normalization of tumor vasculature: an emerging con-cept in antiangiogenic therapy. Science 2005;307(5706): 58–62.

12. Urano M, et al. Effect of whole body hyperthermia on cellsurvival, metastasis frequency, and host immunity in moder-ately and weakly immunogenic murine tumors. Cancer Res1983;43:1039–1043.

13. Shen RN, et al. Whole-body hyperthermia decreases lungmetastases in lung tumor-bearing mice, possibly via amechanism involving natural killer cells. J Clin Immunol1987;7(3):246–253.

14. Roigas J, Wallen ES, Loening SA, Moseley PL. Heat shockprotein (HSP72) surface expression enhances the lysis of ahuman renal cell carcinoma by IL-2 stimulated NK cells. AdvExp Med Biol 1998;451:225–229.

15. Burd R, et al. Tumor cell apoptosis, lymphocyte recruitmentand tumor vascular changes are induced by low temperature,long duration (fever-like) whole body hyperthermia. J CellPhysiol 1998;177(1):137–147.

16. Ostberg JR, Gellin C, Patel R, Repasky EA. Regulatory poten-tial of fever-range whole body hyperthermia on Langerhanscells and lymphocytes in an antigen-dependent cellularimmune response. J Immunol 2001;167(5):2666–2670.

17. Kraybill WG, et al. A phase I study of low temperature, longduration (fever-range) whole body hyperthermia (WBH) inpatients with advanced solid tumors: correlation with mousemodels. Int J Hyperthermia 2002;18:253–266.

18. Atanackovic D, et al. 41.8 degrees C whole body hyperthermiaas an adjunct to chemotherapy induces prolonged T cell acti-vation in patients with various malignant diseases. CancerImmunol Immunother Epub 2002 Oct 18 2002;51(11–12):603–613.

19. Barni S, et al. Lysosomal exocytosis induced by hyperthermia:a new model of cancer death. III. effect on liver metastasis.Biomed Pharmacother 1996;50:79–84.

20. Hou K. The level changes of peripheral blood PGE2 and clinicalsignificance in patients with malignant tumor before and afterwhole body hyperthermia. Proceedings of the 26th Congress ofthe International Clinical Hyperthermia Society, September9–12, 2004, Shenzhen, China, 2004; p 66.

21. Manjili MH, et al. Subjeck, Cancer immunotherapy: stressproteins and hyperthermia. Int J Hyperthermia 2002;18(6):506–520.

22. Ialenti A, et al. Inhibition of cyclooxygenase-2 gene expressionby the heat shock response in J774 murine macrophages. EurJ Pharmacol 2005;509(2–3):89–96.

60 HYPERTHERMIA, SYSTEMIC

Page 20: Hyperthermia Systemic

23. Repasky EA, Issels R. Physiological consequences ofhyperthermia: Heat, heat shock proteins and the immuneresponse. Int J Hyperthermia 2002;18:486–489.

24. Ostberg JR, Repasky EA. Emerging evidence indicates thatphysiologically relevant thermal stress regulates dendritic cellfunction. Cancer Immunol Immunother [Epub ahead of print];Apr 28, 2005.

25. Berry JM, Michalsen A, Nagle V, Bull JM. The use of esmololin whole-body hyperthermia: cardiovascular effects. Int JHyperthermia 1997;13(3):261–268.

26. Robins HI, et al. Adjunctive therapy (whole body hyperther-mia versus Ionidamine) to total body irradiation for the treat-ment of favorable B-cell neoplasms: a report of two pilotclinical trials and laboratory investigations. Int J Radiat OncoBiophys 1990;18:909–920.

27. Ohno S, et al. Haematological toxicity of carboplatin andcisplatin combined with whole body hyperthermia in rats.Br J Cancer 1993;68:469–474.

28. Haveman J, et al. Effects of hyperthermia on the peripheralnervous system: a review. Int J Hyperthermia 2004;20(4):371–391.

29. Calvert AH, et al. Early clinical studies with cis-diammine-1,1-cyclobutane dicarboylate platinum II. Cancer ChemotherPharmacol 1982;9:140–147.

30. Sapareto SA, Dewey WC. Thermal dose determination in cancertherapy. Int J Radiat Oncol Biol Phys 1984;10(6):787–800.

31. Thrall DE, et al. Using units of CEM 43 degrees C T90, localhyperthermia thermal dose can be delivered as prescribed. IntJ Hyperthermia 2000;16(5):415–428.

32. Webb AG. Temperature measurement using nuclear magneticresonance. Ann Reports NMR Spectrosc 2001;45:1–67.

33. Stafford RJ, Hazle JD, Glover GH. Monitoring of high-inten-sity focused ultrasound-induced temperature changes in vitrousing an interleaved spiral acquisition. Magn Reson Med2000;43(6):909–912.

34. Engelhardt R. Summary of recent clinical experience in whole-body hyperthermia combined with chemotherapy. RecentResults Cancer Res 1988;107:200–224.

35. Lange J, Zanker KS, Siewert JR, Eisler K, Landauer B, KolbE, Blumel G. and Remy, W. Extracorporeally induced whole-body hyperthermia in conventionally incurable malignanttumor patients Med Wochenschr. 1983;108(13):504–509.

36. Robins HI, et al. Phase I clinical trial of melphalan and 41.8degrees C whole-body hyperthermia in cancer patients. J ClinOncol 1997;15:158–164.

37. Issels RD, Wilmanns W, editors. Recent Results in CancerResearch, Vol. 107: Application of Hyperthermia in theTreatment of Cancer.Berlin/Heidelberg: Springer Verlag; 1988.

38. Westermann AM, et al. Systemic Hyperthermia OncologicWorking Group, A Systemic Hyperthermia Oncologic WorkingGroup trial, Ifosfamide, carboplatin, and etoposide combinedwith 41.8 degrees C whole-body hyperthermia for metastaticsoft tissue sarcoma. Oncology 2003;64(4):312–321.

39. Bull JM, et al. Chemotherapy resistant sarcoma treated withwhole body hyperthermia (WBH) combined with 1-3-bis(2-chloroethyl)-1-nitrosourea (BCNU). Int J Hyperthermia 1992;8(3):297–304.

40. Herman TS, Sweets CC, White DM, Gerner EW. Effectof heating on lethality due to hyperthermia and selectedchemotherapeutic drugs. J Natl Cancer Inst 1982;68(3):487–491.

41. Engelhardt R, et al. Treatment of disseminated malignantmelanoma with cisplatin in combination with whole-bodyhyperthermia and doxorubicin. Int J Hyperthermia 1990;6(3):511–515.

42. Robins HI, et al. Phase I clinical trial of carboplatin and 41.8degrees C whole-body hyperthermia in cancer patients. J ClinOncol 1993;11:1787–1794.

43. Douwes F, et al. Whole-body hyperthermia in combinationwith platinum-containing drugs in patients with recurrentovarian cancer. Int J Clin Oncol 2004;9(2):85–91.

44. Hegewisch-Becker S, et al. Whole-body hyperthermia(41.8 8C) combined with bimonthly oxaliplatin, high-dose leu-covorin and 5-fluorouracil 48-hour continuous infusion inpretreated metastatic colorectal cancer: a phase II study.Ann Onc 2002;13(8):1197–1204.

45. Hegewisch-Becker S, et al. Whole body hyperthermia (WBH,41.8 8C)combinedwithcarboplatinandetoposide inadvancedbiliarytract cancer. Proc Am Soc Clin Oncol 2003; (abstr. 1247), 22:311.

46. Richel O, et al. Phase II study of carboplatin and whole bodyhyperthermia (WBH) in recurrent and metastatic cervicalcancer. Gynecol Oncol 2004;95(3):680–685.

47. Strobl B, et al. Whole body hyperthermia combined with carbo-platin/paclitaxel in patients with ovarian carcinoma–Phase-II-study. J Clin Oncol -Proc Am Soc Clin Oncol 2004;22(14S).

48. Elias D, et al. Heated intra-operative intraperitoneal oxali-platin plus irinotecan after complete resection of peritonealcarcinomatosis: pharmacokinetics, tissue distribution and tol-erance. Ann Oncol 2004;15:1558–1565.

49. Bull JM, et al. Phase I study of long-duration, low-tempera-ture whole-body hyperthermia (LL-WBH) with liposomal dox-orubicin (Doxil), 5-fluorouracil (5-FU), & interferon-a (IFN-a),Proc Amer. Soc Clin Oncol 2002;(Abst. 2126).

50. Larkin JM, A clinical investigation of total-body hyperthermiaas cancer therapy. Cancer Res, 1979;39(6 Pt 2):2252–2254.

51. Bull JM, et al. Update of a phase I clinical trial using fever-range whole-body hyperthermia (FR-WBH) þ cisplatin (CIS) þgemcitabine (GEM) þ metronomic, low-dose interferon-alpha(IFN-alpha), Proceedings of the International Congress onHyperthermic Oncology, 20.-24.04., St. Louis, (Session 21and Poster 689); 2004.

52. Barlogie B, Corry PM, Lip E, Lippman L, Johnston DA,Tenczynski TF, Reilly E, Lawson R, Dosik G, Rigor B,Hankenson R, Freireich EJ Total-body hyperthermia withand without chemotherapy for advanced human neoplasms.Cancer Res 1979;39(5):1481–1489.

53. Robins HI, et al. Phase I trial of human lymphoblastoidinterferon with whole body hyperthermia in advanced cancer.Cancer Res 1989;49(6):1609–1615.

54. Gerad H, van Echo DA, Whitacre M, Ashman M, Helrich M, FoyJ, Ostrow S, Wiernik PH, Aisner J. Doxorubicin, cyclophospha-mide, and whole body hyperthermia for treatment of advancedsoft tissue sarcoma. Cancer. 1984 Jun 15;53(12):2585–91.

55. Baba H, et al. Increased therapeutic gain of combined cis-diamminedichloroplatinum (II) and whole body hyperthermiatherapy by optimal heat/drug scheduling. Cancer Res1989;49(24 Pt. 1):7041–7044.

56. Katschinski DM, et al. Optimization of chemotherapy admin-istration for clinical 41.8 degrees C whole body hyperthermia.Cancer Lett 1997;115(2):195–199.

57. Hegewisch-Becker S, et al. Effects of whole body hyperthermia(41.8 degrees C) on the frequency of tumor cells in the per-ipheral blood of patients with advanced malignancies. ClinCancer Res 2003;9(6):2079–2084.

58. Teicher BA, Holden SA, Ara G, Menon K. Whole-bodyhyperthermia and lonidamine as adjuvant therapy to treat-ment with cisplatin with or without local radiation in mousebearing the Lewis lung carcinoma. Int J Hyperthermia1995;11(5):637–645.

59. Sumiyoshi K, Strebel FR, Rowe RW, Bull JM. The effectof whole-body hyperthermia combined with ‘metronomic’ che-motherapy on rat mammary adenocarcinoma metastases. IntJ Hyperthermia 2003;19(2):103–118.

60. Overgaard J, et al. Randomised trial of hyperthermiaas adjuvant to radiotherapy for recurrent or metastatic

HYPERTHERMIA, SYSTEMIC 61

Page 21: Hyperthermia Systemic

malignant melanoma, European Society for HyperthermicOncology. Lancet 1995;345(8949):540–543.

61. Hehr T, Wust P, Bamberg M, Budach W. Current and poten-tial role of thermoradiotherapy for solid tumours. Onkologie2003;26(3):295–302.

62. Hildebrandt B, et al. Current status of radiant whole-bodyhyperthermia at temperatures > 41.5 degrees C and practicalguidelines for the treatment of adults. The German Interdis-ciplinary Working Group on Hyperthermia. Int J Hyperther-mia 2005;21(2):169–183.

63. Hildebrandt B, et al. Whole-body hyperthermia in the scope ofvon Ardenne’s systemic cancer multistep therapy (sCMT)combined with chemotherapy in patients with metastaticcolorectal cancer: a phase I/II study. Int J Hyperthermia2004;20(3):317–333.

64. Robinson V, et al. Thermotherapy for treating rheumatoidarthritis. Cochrane Database Syst Rev 1: CD002826; 2002.

65. van Soest H, van Hattum J. New treatment options for chronichepatitis C. Adv Exp Med Biol 2003;531:219–226.

66. Pontiggia P, Rotella GB, Sabato A, Curto FD. Therapeutichyperthermia in cancer and AIDS: an updated survey. JEnviron Pathol Toxicol Oncol 1996;15(2–4):289–297.

67. Li CY, Dewhirst MW. Hyperthermia-regulated immunogenetherapy. Int J Hyperthermia 2002;18(6):586–596.

68. Okita A, et al. Efficiency of lipofection combined with hyperther-mia in Lewis lung carcinoma cells and a rodent pleural dissemi-nation model of lung carcinoma. Oncol Rep 2004;11:1313–1318.

Further Reading

Bakhshandeh A, et al. Year 2000 guidelines for clinical practice ofwhole body hyperthermia combined with cytotoxic drugs fromthe University of Lubeck and the University of Wisconsin. JOncol Pharm Practice 1999;5(3):131–134.

Field SB, Hand JW, editors. An Introduction to the Practical Aspectsof Clinical Hyperthermia. London: Taylor & Francis; 1990.

Gautherie M, editor. Methods of External Hyperthermic Heating.Berlin/Heidelberg: Springer Verlag; 1990.

Gautherie M, editor. Whole Body Hyperthermia: Biological andClinical Aspects. Berlin/Heidelberg: Springer Verlag; 1992.

Hahn GM. Hyperthermia and Cancer. New York: Plenum Press, 1982.Hildebrandt B, et al. Current status of radiant whole-body

hyperthermia at temperatures > 41.5 degrees C and practicalguidelines for the treatment of adults. The German Interdisci-plinary Working Group on Hyperthermia. Int J Hyperthermia2005;21(2):169–183.

Issels RD, Wilmanns W, editors. Recent Results in CancerResearch, Vol. 107: Application of Hyperthermia in theTreatment of Cancer. Berlin/Heidelberg: Springer Verlag;1988.

Nussbaum GH, editor. Physical Aspects of Hyperthermia. AmericanAssociationofPhysicists inMedicineMedicalPhysicsMonographNo. 8. New York: American Institute of Physics; 1982.

Guan J, et al. The clinical study of whole-body hyperthermia(WBH) improving the survival state of patients with advancedcancer. Proc 26th Congress of the International ClinicalHyperthermia Society, Sept. 9–12, 2004, Shenzhen, China;2004; p 66.

Kurpeshev OK, Tsyb AF, Mardynsky YS. Whole-body hyperthermiafor treatment of patients with disseminated tumors- Phase II. In:P.H. Rehak, K.H. Tscheliessnigg, editors. Proceedings 22nd.Annual Meeting of the European Society for HyperthermicOncology, June 8–11, 2005, Graz, Austria, 2005; p 103.

Hou K. Assessment of the effects and clinical safety of the treat-ment of advanced malignant tumor with extracorporeal wholebody hyperthermia. Proceedings of the 26th Congress of the

International Clinical Hyperthermia Society, Sept. 9–12, 2004,Shenzhen, China; 2004 p 71.

See also BIOHEAT TRANSFER; HEAT AND COLD, THERAPEUTIC; HYPERTHER-

MIA, INTERSTITIAL; HYPERTHERMIA, ULTRASONIC; RADIATION DOSIMETRY FOR

ONCOLOGY.

62