1,249
Views
83
CrossRef citations to date
0
Altmetric
Research Articles

Pathophysiological and vascular characteristics of tumours and their importance for hyperthermia: Heterogeneity is the key issue

&
Pages 211-223 | Received 14 Dec 2009, Accepted 04 Jan 2010, Published online: 26 Mar 2010

Abstract

Tumour blood flow before and during clinically relevant mild hyperthermia exhibits pronounced heterogeneity. Flow changes upon heating are not predictable and are both spatially and temporally highly variable. Flow increases may result in improved heat dissipation to the extent that therapeutically relevant tissue temperatures may not be achieved. This holds especially true for tumours or tumour regions in which flow rates are substantially higher than in the surrounding normal tissues.

Changes in tumour oxygenation tend to reflect alterations in blood flow upon hyperthermia. An initial improvement in the oxygenation status, followed by a return to baseline levels (or even a drop to below baseline at high thermal doses) has been reported for some tumours, whereas a predictable and universal occurrence of sustained increases in O2 tensions upon mild hyperthermia is questionable and still needs to be verified in the clinical setting. Clarification of the pathogenetic mechanisms behind possible sustained increases is mandatory.

High-dose hyperthermia leads to a decrease in the extracellular and intracellular pH and a deterioration of the energy status, both of which are known to be parameters capable of acting as direct sensitisers and thus pivotal factors in hyperthermia treatment. The role of the tumour microcirculatory function, hypoxia, acidosis and energy status is complex and is further complicated by a pronounced heterogeneity. These latter aspects require additional critical evaluation in clinically relevant tumour models in order for their impact on the response to heat to be clarified.

Introduction

Several pathophysiological features of the tumour microenvironment greatly affect the efficacy of anticancer treatments, hyperthermia (HT) included Citation[1], Citation[2]. Amongst these factors, tumour blood flow (TBF) plays a seminal role since (1) it critically determines convective heat dissipation and thus the ability to heat the tissue, and (2) it greatly influences the tumour microenvironment and subsequently the sensitivity of the tumour to heat. A compromised tumour blood flow results in the development of adverse microenvironmental conditions (e.g. hypoxia, acidosis, energy depletion Citation[3]). Cells living under such hostile conditions seem to be more sensitive to therapeutic hyperthermia. TBF and the tumour microenvironment thus play a significant role in determining the response of tumour cells to heat as evidenced in experimental (preclinical) as well as clinical settings. Besides sensitising cancer cells to heat, pathophysiological characteristics of the tumour microenvironment are themselves affected and altered by tissue heating in return, thus creating a complex (and partly self-perpetuating) situation, depending on the direction, extent and duration of individual changes upon heating.

These changes in microenvironmental factors upon heating have been described in detail previously, especially for experimental tumours in the preclinical setting Citation[4–8]. Comparable studies on human tumours are very scarce because most of the data are derived from anecdotal observations Citation[4]. In addition, in many of the investigations, hyperthermia at temperatures above 42.5°C have been used, a situation which is only rarely achievable in the clinical setting, where–in most cases–hyperthermic tissue temperatures of between 39°C and 42.5°C have been reported Citation[9], Citation[10]. Whereas in the preclinical HT studies on experimental rodent tumours tissue temperatures were relatively uniform, in human tumours spatially heterogeneous temperature fields have often been described Citation[4].

In this review, updated data on relevant parameters of the tumour micro-milieu are presented with special emphasis on changes occurring during ‘mild’ or ‘low dose’ hyperthermia, i.e. within the temperature range 39–42.5°C. To provide a basis for this review, a general description of the pathophysiological tumour microenvironment is presented first. Furthermore, particular emphasis is given to the in vivo situation, which is rather more complex than that of in vitro tumour biology.

Pathophysiological characteristics of the tumour microenvironment

The physiology of tumours is uniquely different from that of normal tissues. It is characterised by O2 depletion (hypoxia and anoxia), extracellular acidosis, high lactate levels, glucose deprivation, energy impoverishment, significant interstitial fluid flow and interstitial hypertension Citation[11–22]. The O2 depletion found in tumours is not the result of increased oxygen consumption rates, but instead is due to an imbalance between perfusion and consumption, so that changes in both of these factors must be taken into consideration. Since the hostile environment is largely determined by an abnormal tumour microcirculation (), the features of the tumour microcirculation and changes upon hyperthermia will be discussed first. (The term microcirculation is generally used to describe the functions and structures of microvessels with outer diameters <100 µm, terminal lymphatics included.)

Table I.  Factors characterising the hostile pathophysiological tumour microenvironment (‘crucial Ps’).

Tumour angiogenesis, vascularity and blood flow

When considering the continuous and indiscriminate formation of the vascular network in a growing tumour, five different pathogenetic mechanisms can be discussed: (1) angiogenesis by endothelial sprouting from pre-existing venules, (2) co-option of existing vessels which can keep their normal structure and function, (3) vasculogenesis (de novo vessel formation through incorporation (recruitment) of circulating endothelial precursor cells from bone marrow), (4) intussusception (splitting of the lumen of a vessel into two), and (5) formation of pseudovascular channels lined by tumour cells rather than endothelial cells (‘vascular mimicry’ Citation[3]). The tumour vasculature is characterised by vigorous proliferation leading to immature, structurally defective, and–in terms of perfusion–ineffective microvessels. Consequently, tumour blood flow is chaotic and heterogeneous, both spatially and temporally (‘4D-heterogeneity’ of TBF).

In contrast to co-opted vessels, newly formed microvessels in most solid tumours do not conform to the normal morphology of the host tissue vasculature. Microvessels in solid tumours exhibit a series of severe structural and functional abnormalities. They are often dilated, tortuous (convoluted), elongated, and saccular. There is significant arterio-venous shunt perfusion accompanied by a chaotic vascular organisation that lacks any regulation matched to the metabolic demands or functional status of the tissue. Excessive branching is a common finding, often coinciding with blind vascular endings. Incomplete or even missing endothelial lining, lack of pericytes and interrupted (discontinuous) basement membranes result in an increased vascular permeability with extravasation of blood plasma and of red blood cells expanding the interstitial fluid space and drastically increasing the hydrostatic pressure in the tumour interstitium (interstitial fluid pressure). In solid tumours there is a rise in viscous resistance to flow caused mainly by haemoconcentration. Reductions in red blood cell flexibility brought about by vascular hypoxia, low pH or hyperthermia can also contribute to the increased viscous resistance Citation[23], Citation[24]. Aberrant vascular morphology and a decrease in vessel density are responsible for an increase in geometric resistance to flow, which can lead to an inadequate perfusion.

Substantial spatial heterogeneity in the distribution of tumour vessels and significant temporal heterogeneity in the microcirculation within a tumour Citation[3] may result in a considerably anisotropic distribution of tumour tissue oxygenation and of a number of other factors which are usually closely linked and which define the so-called metabolic microenvironment. Variations in these parameters between tumours are often more pronounced than differences occurring between different locations or microareas within a tumour Citation[3].

Blood flow can vary considerably, ranging from 0.01 to 2.9 mL/g/min, both in the preclinical and clinical setting Citation[3]. Tumours can thus have flow rates similar to those measured in tissues with a high metabolic rate or can exhibit perfusion rates comparable to those of tissues with a low metabolic turnover. Flow data from multiple sites of measurement show marked heterogeneity within individual tumours. However, tumour-to-tumour variability seems to be more pronounced than intratumour heterogeneity.

Angiogenesis, microcirculation and tumour blood flow upon hyperthermia

Hyperthermia can inhibit angiogenesis, a mechanism which has been suggested to play a role in tumour regression caused by HT Citation[25]. These authors reported that angiogenesis inhibition occurs as a result of direct injury to endothelial cells (EC). An inverse relationship was demonstrated between vessel growth and treatment temperature over the range of 41–44°C after 30 min hyperthermia treatment Citation[26]. Following these earlier experiments, Sawaji et al. Citation[27] were able to show that the anti-angiogenic action of HT may be due to the down-regulation of the expression of tumour-derived vascular endothelial growth factor (VEGF) production, thereby inhibiting proliferation of ECs and extracellular matrix remodelling in blood vessels. In addition, HT can activate a specific gene response that involves the transcription of the human plasminogen activator inhibitor-1 (PAI-1), the key regulator of the plasminogen activation pathway Citation[28]. The latter study indicated that the heat-mediated PAI-1 induction may be an important pathway by which HT exerts its antitumour activity.

Most studies on HT-induced changes in tumour microcirculation date back to the early 1980s Citation[29–43]. Summarising these observations, it can be stated that the effect of heat on tumour microcirculation is temperature- and time-dependent. Whereas mild HT can (temporarily) increase microcirculatory functions, higher temperature and/or longer exposure times can impair the microcirculation and damage the tumour microvasculature. Normal vasculature responds differently to the fragile and chaotic tumour microvasculature with microvasculatory flow increasing as temperature increases even at temperatures that produce a complete shutdown of tumour microvasculature (T > 42.5°C).

The immediate physiological reaction after elevation of tissue temperature is a change in blood flow Citation[44]. The alteration in perfusion upon heating is in general much greater in normal tissue than in tumours. Normal tissues often included into the heating volume are skeletal muscle and skin with flow values of 0.04 mL/g/min (resting state), and 0.08 mL/g/min (under thermoneutral conditions), respectively Citation[45]. Skeletal muscle and skin can increase flow by at least a factor of 15 whereas tumour perfusion may only increase 2-fold Citation[4], starting from a baseline which is often significantly higher than flow values observed in skeletal muscle and skin.

Changes in tumour blood flow upon hyperthermia in experimental tumours have been comprehensively reviewed Citation[4], Citation[39]. On average, blood flow in experimental tumours temporarily increased upon heating and declined thereafter. The extent and time course of the heat-induced initial flow increase depends on the heating-up rate, hyperthermia level, heating time, temperature homogeneity achieved, tumour type, and growth site of the experimental tumour. Not surprisingly, heat-induced flow changes also depend on the absolute blood flow rate of the tumours before treatment. In experimental tumour models, heat-induced changes in blood flow were also seen to be related to tumour volume, with larger tumours exhibiting the lower flow values Citation[4], Citation[5], Citation[39]. Temperature- and time-dependent changes in tumour perfusion have been reported in a large series of isografted mice and rat tumours and in xenografted human tumours.

Kelleher et al. Citation[46] measured red blood cell (RBC) fluxes simultaneously in subcutaneous rat tumours at multiple sites using invasive laser Doppler flow probes. From these measurements the following conclusions could be drawn:

  1. The response of the tumour to heat was dependent on tumour volume: the smaller the tumour (i.e. the higher the initial blood flow values), the less pronounced (and/or less often) the shutdown of microcirculatory function following a slight, initial increase in flow.

  2. The extent and direction of flow changes observed within a tumour did not correlate with the tumour site (peripheral versus central regions) or with the actual tissue temperature at the site of measurement (42.4°C versus 44.5°C).

  3. Variability in RBC flux responses to heat was similar in small and large tumours.

  4. Whereas RBC flux substantially increased by a factor of 2.3 in some tumour areas during the heating period, in others it steadily decreased by up to 75% upon hyperthermia; biphasic changes in flux could often be observed with an initial flow increase in many cases (see ).

Figure 1. Variability of tumour perfusion response to localised 44°C hyperthermia. Curves show relative red blood cell flux at three different sites within a single tumour at comparable temperatures. The shaded bar shows the time of hyperthermia application (modified from Vaupel et al. Citation[47]).

Figure 1. Variability of tumour perfusion response to localised 44°C hyperthermia. Curves show relative red blood cell flux at three different sites within a single tumour at comparable temperatures. The shaded bar shows the time of hyperthermia application (modified from Vaupel et al. Citation[47]).

In general, there was a clear indication from these preclinical experiments, that–at least at the start of heating and during the early phase of steady-state tissue temperatures–the intratumour variability of blood perfusion increased, both spatially and temporally, indicating an increased ‘4D-heterogeneity’ in tumour perfusion, even when heating is rather uniform. Flow values are finally ‘homogenised’ on a lower level upon shutdown of flow at a later phase or after heating. Heat-induced flow changes seen in experimental tumours are not at all predictable due to pronounced heterogeneities.

At least in experimental tumour models, the heating techniques used may have an additional impact on the biological behaviour. In the case of saline bath hyperthermia (caveat: immersion of a tumour in a hypo-osmolar water bath creates an osmotic water shift leading to oedema formation which adds to the heat-induced oedema caused by an increased vascular permeability at higher temperatures), the highest temperatures are to be found in the tumour shell, which in many experimental systems exhibits the highest flow rates before treatment. In contrast, other heating devices lead to a maximum energy deposition in more central parts of the tumours which are often characterised by lower perfusion rates. Thus tumours showing comparable mean tissue temperatures during hyperthermia may show completely different patterns of maximum heat deposition with respect to high- and low-flow regions Citation[5]. Heating rodent tumours at 40–42°C (i.e. to clinically achievable hyperthermia levels Citation[48]) was found to cause an enduring increase in blood flow (and vascular permeability) thus possibly improving the delivery of chemotherapeutic drugs or immunotherapy agents to tumour cells Citation[49], Citation[50].

In animal tumour models, the response of blood vessels to multiple heating differs markedly from that of single heating Citation[39]. Increases seen in tumour blood flow during a second heating following the ‘conditioning of blood vessels’ by the first heating appear to be due to the development of ‘vascular thermotolerance’, provided that the second heating is applied within 48 hours at relatively mild temperatures (data reviewed by Griffin et al. Citation[51]), indicating a considerable impact of the treatment regime (single versus fractionated, HT alone or in combination) (caveat: the definition ‘vascular thermotolerance’ used by Griffin et al. Citation[51] is not compatible with the traditional definition of Gerner and Schneider Citation[52] and Henle et al. Citation[53]).

Temperature-dependent changes in tumour perfusion have also been reported in spontaneous canine tumours 24 h after hyperthermia treatment with increases in flow upon hyperthermia at lower temperatures, whereas high temperature hyperthermia treatments were more likely to cause vascular damage. In addition, the data suggest that the thermal threshold for vascular damage is higher in the clinical setting than in rapidly growing rodent tumours Citation[54]. In a recent paper by Viglianti and colleagues Citation[55] on canine tumours, changes in perfusion-related parameters after thermoradiotherapy persisted for 24 h after treatment. So far, the underlying mechanisms for this effect are, however, not known. The changes found predicted not only for local tumour control, but also for metastasis-free and overall survival.

Since perfusion is crucial for hyperthermia treatment planning in the clinical setting, assessment of flow changes in human tumours upon hyperthermia are of utmost importance. However, as mentioned above, data from studies on human tumours are still scarce and numerous questions still exist regarding whether or not results obtained from fast-growing murine tumours are an appropriate reflection of what occurs clinically in human tumours, especially when the fact that experimental tumours usually exceed 2% of the animal's body weight is taken into account (i.e. these tumours are considered to be ‘bulky’ tumours Citation[56]).

In a first series of tumour blood flow measurements upon local hyperthermia, Waterman et al. Citation[57], Citation[58] described an increase in tumour blood flow by amounts varying from 15% to 250% during the first 20–50 min of heating at 41–45°C, after which it remained relatively constant during the remainder of the treatment session. A sharp reduction in flow was never observed in this study (caveat: heat transport by conduction was neglected in these measurements Citation[59]). Subsequently, Waterman et al. Citation[60] studied the effects of localised heating on tumour blood flow in human patients using thermal wash-out techniques. In these investigations, great emphasis was laid on the importance of thermal conduction in the dissipation of heat energy during local hyperthermia Citation[59], Citation[61]. In the Waterman study Citation[60], the authors were unable to demonstrate a flow drop during individual hyperthermia sessions of 1 h duration with temperatures as high as 45°C. Similar results were reported by Lammertsma et al. Citation[62] who measured blood flow upon HT in breast cancers using positron emission tomography. A study of lung and breast cancer metastases in the lymph axillary nodes showed an increase in tumour blood flow after each hyperthermia session relative to the pretreatment perfusion during the course of fractionated hyperthermia. In some cases, blood flow rises of up to 200% were found Citation[63]. Temperature profile assessments Citation[64] and PET Citation[65] have been used to observe the effects of heating on blood flow in human pelvic tumours. In these experiments, regional hyperthermia was applied for about 60 min at temperatures of between 40°C and 42°C, but significant changes in perfusion upon hyperthermia were not observed (caveat: in rectal cancers, blood flow varied substantially depending on the methods used to estimate perfusion in rectal cancers: 0.09 mL/g/min with temperature profile assessments Citation[64], and 0.36 mL/g/min with PET Citation[65]).

When the microcirculatory function of human tumours was evaluated using the laser Doppler flow technique, four different temporal patterns were observed during hyperthermia which had been applied 1–2 h after irradiation. These ranged from a steady increase in flow to a plateau level (28%), no change in RBC flow (36%) or a steady drop in flux (23%), to an initial substantial increase in laser Doppler flow followed by a slight decrease during hyperthermia (14% of cases Citation[66]). These different patterns did not correlate with the average temperature recorded at the site of measurement (42–44°C for approximately 57 min).

A lack of changes seen in thermal wash-out rates in rectal carcinomas, soft tissue sarcomas and other tumours are indicative of only minor changes in blood flow during fractionated hyperthermia immediately after radiotherapy Citation[67]. The response of human tumour blood flow to fractionated thermoradiotherapy has also been described by Waterman et al. Citation[68]. In this study, blood flow in human tumours decreased during the fractionated course of radiotherapy. The overall percentage drop of blood flow ranged from 50–100%. The steady-state temperature at which blood flow started to decrease ranged from 40–43°C.

Taken together, there is no clear indication of changes in flow in human tumours to match those described in the experimental setting, i.e. a transient or steady increase in tumour blood flow in the early phase or during localised hyperthermia, and an overall decrease in tumour blood flow in the later phase or after heating at temperatures >42.5°C. In human tumours therefore, blood flow change might not be the key mechanism by which hyperthermia exerts its beneficial clinical effects.

In many reviews on the behaviour of tumour blood flow upon hyperthermia, relative changes in perfusion are shown for normal tissue (skin, muscle) in comparison to experimental tumours Citation[37]. In this type of data presentation, tumour blood flow–on average–goes down, whereas blood flow in representative normal tissues goes up. This way of representing relative flow values can, however, be completely misleading and often results in inappropriate conclusions being made concerning the flow behaviour of human tumours upon heat treatment. When absolute flow values are taken into consideration, a totally different picture becomes apparent Citation[56], Citation[69] which does not support the current concept of HT-induced blood flow changes being used to explain the rationale of HT in the clinical setting. For example, blood flow in breast cancers is, on average, significantly higher than in normal breast tissue (e.g. 0.30 mL/g/min in breast cancer and 0.06 mL/g/min in normal breast tissue Citation[14], Citation[62]), and changes in flow upon hyperthermia do not support the notion of HT-induced flow increases Citation[14], Citation[62] since tumours undergoing HT failed to show any flow changes at all, i.e. upon HT, flow values in breast cancers are consistently higher than in normal breast tissue. Blood flow values in cervix and prostate cancer can–at the least–be three times higher than in normal cervix and prostate, making a quasi selective heating of the malignant tissue very difficult Citation[70], Citation[71].

It is important to recognise that perfusion, with respect to nutrient delivery is quite different from that responsible for heat transfer: Thermally significant vessels are generally ‘macrovessels’ (diameter >100 µm), whereas nutrient transport and exchange generally occur in the microcirculation (vessel diameter <100 µm).

Cellular oxygen consumption at elevated temperatures

In general, the actual uptake of O2 and nutrients by a tissue is determined by the respective availabilities in the microcirculation, the diffusional flux in the interstitial compartment, and the metabolic requirements of the cells. This holds true for both normal tissue and solid tumours during normothermia and hyperthermia. Under in vitro conditions, however, where no supply limitations are present, the capacity of the cells to consume oxygen is the limiting factor. The in vivo O2 availability (i.e. microcirculatory function at normoxaemia) is the paramount limiting parameter for many solid tumours. Accordingly, changes in the in vivo O2 uptake during hyperthermia only occur when tumour heating is accompanied by changes in nutritive blood flow Citation[4]. In other words, O2 uptake rates in vivo will parallel changes in nutritive flow in either direction.

The temperature dependency of cellular oxygen consumption rates has been reviewed by Vaupel Citation[4]. On average, isolated tumour cells (exponentially) increase their O2 consumption rate following temperature elevation up to maximum values at 41–42.5°C. DS-sarcoma cells suspended in native ascitic fluid exhibit a maximum O2 consumption rate of 42 µL/g/min at 42°C (). This consumption rate is comparable to that of the brain at 37°C Citation[14].

Figure 2. Temperature dependency of the O2 consumption rate of isolated ascites cells suspended in native ascites fluid (DS-sarcoma) (modified from Vaupel Citation[4]).

Figure 2. Temperature dependency of the O2 consumption rate of isolated ascites cells suspended in native ascites fluid (DS-sarcoma) (modified from Vaupel Citation[4]).

Assuming a temperature rise during hyperthermia from 32° to 42°C (e.g. in a superficial tumour) and taking into account the data presented in , a 2.3-fold increase in the respiration rate is observed, (i.e. the van’t Hoff quotient, Q10, is 2.3), indicating an undisturbed respiration activity of the cell line investigated. These data speak strongly against a mitochondrial dysfunction, which is often thought to be a general hallmark in malignant growth Citation[72].

Tumour tissue oxygenation upon hyperthermia

Over the last two decades, numerous studies have clearly shown that most locally advanced solid tumours contain hypoxic tissue areas [e.g. 3, 73] and as such are a characteristic of malignant growth. The pathogenesis of tumour hypoxia is multifactorial ().

Table II.  Chronic and acute tumour hypoxia: Classification according to different causative mechanisms (selection).

Hypoxic and/or anoxic tissue areas are heterogeneously (both temporally and spatially) distributed within the tumour mass. Moreover, hypoxia has been shown to be a significant negative prognostic factor in many clinical studies, independent of the treatment type Citation[74].

In addition, hypoxia can lead to therapeutic resistance to standard radiotherapy, some forms of chemotherapy and photodynamic therapy Citation[1], Citation[75]. In contrast, hypoxia may increase the therapeutic effects of HT, most probably via indirect effects (e.g. acidosis, energy depletion). There is no convincing evidence so far for a direct effect of hypoxia per se on the efficacy of HT Citation[76], although a study by Overgaard and Bichel suggested a slightly enhanced hyperthermic response under hypoxic conditions Citation[77].

Evidence supports the notion that hypoxia is an important driving force in malignant progression, since it can promote the development of a malignant phenotype Citation[1], Citation[3], Citation[78], Citation[79]. Hypoxia–as an inherent consequence of unregulated growth–can further local invasion, intravasation of cancer cells and finally metastatic spread to distant sites whereby this occurs in a cooperative manner at different mechanistic biological/molecular levels Citation[3], Citation[79], Citation[80].

Tumour cell variants with adaptations favourable to survival under hypoxic stress may have growth advantages over non-adapted cells in the hypoxic microenvironment and may subsequently develop a more aggressive phenotype which in turn is responsible for malignant progression. Cyclic hypoxia and pronounced spatio-temporal heterogeneities (4D-heterogeneities) in hypoxia may be the most powerful factors promoting an aggressive tumour phenotype, which drive local spread and distant metastasis Citation[79–85].

Due to this crucial role of tumour hypoxia in the clinical setting, the tumour oxygenation status has attracted the interest of numerous researchers over the last two decades Citation[73]. Since tumour hypoxia has been considered to be an indirect sensitiser for HT, spatial and temporal changes of tumour oxygenation have also been evaluated upon tumour heating.

When the oxygen status is assessed for characterisation of solid tumours at elevated temperatures in the preclinical setting, there is clear evidence that low thermal doses can improve tissue oxygenation provided that tumour perfusion exhibits an increase under these conditions Citation[4], Citation[5]. At higher thermal doses, tissue oxygenation deteriorates severely, reflecting the breakdown of tumour microcirculation under these conditions Citation[40], Citation[89–93]. From these experiments it has been concluded that changes in tissue oxygenation upon HT may directly reflect simultaneous alterations in tissue perfusion.

There is some controversy concerning the duration of the improvement in tissue oxygenation upon mild hyperthermia, with a series of studies reporting a rapid return to a normal oxygenation status in tumours after the cessation of heating, i.e. the effects observed are rather short-lived () Citation[89–93]. In this context it has also been noticed that with higher tissue temperatures the initial increase and the following drop in tissue oxygenation were more pronounced (). In contrast to these data, other studies have shown that the improvement in tumour oxygenation can actually last for up to 24–48 h after heating using low-dose HT Citation[38], Citation[49], Citation[94–98].

Figure 3. Tumour pO2 values measured continuously before, during and after 40°C, 41.8°C or 43°C hyperthermia. Each data point indicates the mean value (±SEM) for the number of tumours indicated. The shaded area indicates the time of heating (modified from Kelleher and Vaupel Citation[93]).

Figure 3. Tumour pO2 values measured continuously before, during and after 40°C, 41.8°C or 43°C hyperthermia. Each data point indicates the mean value (±SEM) for the number of tumours indicated. The shaded area indicates the time of heating (modified from Kelleher and Vaupel Citation[93]).

Figure 4. Comparison of mean pO2 values before, during (at t = 35 min after reaching the set temperature) and after localised hyperthermia. Each data point indicates the mean value (±SEM) for the number of tumours indicated. * = p < 0.01 (modified from Kelleher and Vaupel Citation[93]).

Figure 4. Comparison of mean pO2 values before, during (at t = 35 min after reaching the set temperature) and after localised hyperthermia. Each data point indicates the mean value (±SEM) for the number of tumours indicated. * = p < 0.01 (modified from Kelleher and Vaupel Citation[93]).

However, long-lasting improvements in the tumour oxygenation status following mild hyperthermia treatments are difficult to explain because the perfusion changes that most probably account for the enhanced tumour oxygenation during mild HT should return to baseline at or soon after the cessation of heating Citation[2]. If the data showing a long-lasting improvement in the oxygenation status could be confirmed by other groups, using different tumour models and other heating techniques, the sustained increase in the oxygenation status could enhance the response of tumours to radiotherapy and O2-dependent chemotherapy/radiochemotherapy. A sustained increase in blood flow (as suggested by the improvement in tumour oxygenation) could probably enhance delivery of anticancer agents. However, since clinically relevant increases in the tissue oxygenation which endure beyond the heating period are not consistently seen in preclinical studies, irrespective of the tumour temperature level Citation[93], an improvement in the efficacy of O2-dependent cancer therapy is unlikely to be achieved in the post-hyperthermia period if these contradictory results can be translated to the clinical setting.

There are clinical observations that seem to support the finding of a prolonged improvement in oxygenation at mild HT temperatures. These data were obtained in the clinical setting during fractionated hyperthermia and radiation therapy Citation[10], Citation[54], Citation[99], hyperthermia treatment and chemotherapy Citation[100] or thermochemoradiotherapy Citation[48]. These findings, however are in contrast to those reported by Zywietz et al. Citation[101] who observed a progressive deterioration in the oxygenation status with combined fractionated radiation and hyperthermia (43°C, 1 h) which was followed by a continuous shrinkage of the tumours and most probably a decline in the overall tumour O2 consumption resulting from heat-induced cell death.

It is unlikely that increased tumour blood flow produced by low thermal doses represents the only mechanism related to improvement of oxygenation. This is supported by the fact that no relationship between changes in perfusion and comparable alterations in tumour oxygenation in the clinical setting Citation[54] and in experimental tumours Citation[92] were seen.

Another likely cause for the improved tumour oxygenation might be a heat-induced decrease in oxygen consumption. However, the respiration rates of tumour cells are not significantly different at 37°C and 43°C (see ). A maximum uptake of O2 is to be expected at 42°C. Therefore, it cannot be concluded that a decrease in O2 consumption together with an increase in tumour blood flow may be the general mechanisms for the improvement of tumour oxygenation after hyperthermia at moderate thermal dose as suggested by Vujaskovic and Song Citation[44]. Further investigations will be necessary if clearer insights into the relationship between thermal parameters, tumour perfusion and oxygenation upon hyperthermia are to be obtained.

Tumour pH upon heat treatment

Warburg's classic work in the 1920s showed that cancer cells intensively convert glucose to lactic acid even in the presence of oxygen. Because of this excessive lactic acid production it was assumed for many decades that tumours are acidic Citation[14], Citation[18], Citation[102–108]. However, the unfolding story of tumour pH and its consequences have become clearer over the last two decades, due to the availability of techniques capable of preferentially measuring intra- or extracellular pH in malignancies. Under many conditions it has now been confirmed that the intracellular pH in tumour cells is neutral to alkaline as long as tumours are not oxygen- and energy-deprived Citation[22], Citation[109]. Tumour cells have efficient mechanisms for exporting protons into the extracellular space, which represents the acidic compartment in tumours Citation[13],Citation[110–112]. For this reason, a pH gradient exists across the cell membrane in tumours (pHi > pHe). Interestingly, this gradient is the reverse of that found in normal tissues where pHi is lower than pHe Citation[3], Citation[14], Citation[22], Citation[113–116]. In spontaneous canine tumours assessed by Prescott et al., the magnitude of the pH gradient varied widely and individual tumours had both positive and negative pH gradients Citation[117].

As already mentioned, hypoxic cancer cells intensively split glucose to lactic acid. There are no longer any reasons to ascribe the aerobic glycolysis found as being specific to malignant growth although the increased capacity for glycolysis still remains a key feature of tumours. Other relevant pathogenetic mechanisms yielding an intensified tissue acidosis are based on substantial ATP hydrolysis, glutaminolysis, ketogenesis and CO2/carbonic acid production.

pH values measured with invasive electrodes preferentially reflect the acid-base status of the extracellular space (pHe) which occupies approximately 45% of the total tissue volume in malignant tumours. Using pH microsensors, a remarkable variability in measured values could be documented which exceeds the heterogeneity observed within tumours. The results of pH measurements using 31P-nuclear magnetic resonance spectroscopy yield pH values which mainly reflect the pH found in the cytosol Citation[14], Citation[118].

Reduced pH values (acidosis) can sensitise tumour cells to single heat treatments and to fractionated hyperthermia Citation[119]. Based on this knowledge, a series of measurements has been performed on the impact of localised heating on intra- and extracellular pH values Citation[4], Citation[5], Citation[37], Citation[119]. Summarising the relevant data, it can be stated that tumour temperatures >42.5°C and appropriate heating times (>30 min) can reduce both intracellular and extracellular pH, which may further sensitise tumour cells to hyperthermia in the sense of a positive feedback mechanism.

Relevant pathogenetic mechanisms leading to an intensified acidosis upon heat treatment (which is reversible after HT) are:

  1. an increased glycolytic rate with accumulation of lactic acid,

  2. an intensified ATP-hydrolysis,

  3. an increased ketogenesis with accumulation of acetoacetic acid and β-hydroxybutyric acid,

  4. an increase in CO2 partial pressures,

  5. changes in chemical equilibria of the intra- and extracellular buffer systems (ΔpH/ΔT = −0.016 pH units/°C), and

  6. an inhibition of the Na+/H+ antiporter in the cell membrane Citation[5].

The significance of the baseline pHe and pHi values and changes occurring during hyperthermia in clinical studies still remains unclear, since–in contrast to in vitro and in vivo data derived from experimental tumours treated with hyperthermia alone–the results are equivocal so far. There are contradictory results in the clinical setting concerning the direction of heat-induced changes in pH (increase versus decrease), the prognostic power of pHi or pHe before treatment using HT or thermoradiotherapy, etc. Citation[54], Citation[106], Citation[120–124]. In the study of Lora-Michiels et al. Citation[122], the pH changes seen predicted for metastasis-free and overall survival. Further clarification of these effects is urgently needed.

Bioenergetic status of tumours upon hyperthermia

In recent years the bioenergetic status of solid tumours has been assessed by 31P-nuclear magnetic resonance (31P-NMR, 31P-MRS), high performance liquid chromatography (HPLC) and by quantitative bioluminescence Citation[3], Citation[14]. Consistently found changes are: decreased ATP and PCr concentrations, increased inorganic phosphate (Pi), ADP, AMP and (extracellular) adenosine concentrations and lower PCr/Pi and ATP/Pi ratios, which have been reported in solid tumours as compared to their tissues of origin. For example, ATP content of soft tissue sarcomas was on average 0.5 mM compared to approximately 5 mM in normal skeletal muscle Citation[14]. This situation has been repeatedly described for tumours with median pO2 values <10 mmHg, mean glucose concentrations <2 mM and mean lactate levels in the tumour tissue >10 mM Citation[3], Citation[125].

Cellular ATP depletion (most probably caused by substrate deprivation) is thought to sensitise tumour cells to hyperthermia Citation[76], Citation[126], Citation[127]. Upon heating at temperatures >42.5°C and heating times >30 min, a further drop in the ATP and PCr concentrations has consistently been reported Citation[4], Citation[5], whereas the levels of Pi, ADP, AMP and adenosine increased further under HT conditions. Accordingly, ATP/Pi and PCr/Pi ratios substantially dropped at high thermal doses which would probably result in a sensitisation of tumours to heat treatment. At 41°C for 15 min, the ATP/Pi ratio only fell by approx. 30% for approximately 6.5 h Citation[128].

The ATP decline observed upon heat treatment is mostly due to

  1. an increased ATP turnover rate (i.e. intensified ATP hydrolysis). As a result of an increased ATP degradation, an accumulation of purine catabolites (adenosine included) has to be expected together with a formation of H+ ions and reactive oxygen species at several stages during degradation to the final product uric acid Citation[3],

  2. a poorer ATP yield (on a molar basis) as a consequence of a shift from oxidative glucose breakdown to glycolysis.

In many human tumours (other than brain tumours), 31P-NMR has often shown high concentrations of phosphomonoesters (PME) and phosphodiesters (PDE). The PME signal primarily includes membrane phospholipid precursors. The PDE peak was identified to be largely a result of membrane phospholipid decomposition products. Upon hyperthermia treatment (>42.5°C, ≥30 min) the PME/ATP ratio significantly increased, whereas the PME/Pi and PDE/Pi ratios decreased following heating Citation[129].

Interestingly, PME/Pi and PDE/Pi ratios have been described as being significant predictive factors for treatment outcome upon thermoradiotherapy in addition to the oxygenation status (pathological cure rate), and the PCr/PDE, as well as the PME/PDE ratios (overall survival) Citation[130].

Conclusions

Thermal sensitivity has been shown to greatly depend on the efficacy of tumour blood flow and parameters defining the metabolic microenvironment such as hypoxia, acidosis, substrate deprivation, accumulation of metabolic waste products, and energy depletion. If recent experimental data are critically evaluated, there is some evidence that besides microcirculatory function, intracellular pH and the bioenergetic status may be decisive factors ultimately modulating the thermosensitivity of cancer cells.

Results obtained from patient malignancies (canine, human) treated with adjuvant mild hyperthermia (T < 42°C) suggest that therapeutically relevant changes in these microenvironmental parameters may be quite different from those seen in fast-growing, low-flow rodent tumours upon heat treatment. Thus, biological principles that seem favourable in experimental rodent tumours may not necessarily hold for tumours (or tumour areas) with high perfusion rates. In addition, the pronounced heterogeneity between tumours and within tumours may lead to a non-uniform response to heat treatment, especially when hyperthermia is used as an adjuvant to established treatment modalities (radio- and/or chemotherapy).

Acknowledgements

The valuable editorial assistance of Anne Deutschmann-Fleck in preparing this manuscript is greatly appreciated.

Declaration of interest: Both authors gratefully acknowledge project grants from the Erwin Braun Foundation, Basel (Switzerland). The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Vaupel P. Physiological mechanisms of treatment resistance. The Impact of Tumor Biology on Cancer Treatment and Multidisciplinary Strategies, M Molls, P Vaupel, C Nieder, MS Anscher. Springer, Berlin, Heidelberg, New York 2009; 273–290
  • Horsman MR, Overgaard J. Hyperthermia: A potent enhancer of radiotherapy. Clin Oncol 2007; 19: 418–426
  • Vaupel P. Pathophysiology of solid tumors. The Impact of Tumor Biology on Cancer Treatment and Multidisciplinary Strategies, M Molls, P Vaupel, C Nieder, MS Anscher. Springer, Berlin, Heidelberg, New York 2009; 51–92
  • Vaupel P. Pathophysiological mechanisms of hyperthermia in cancer therapy. Biological Basis of Oncologic Thermotherapy, M Gautherie. Springer, Berlin, Heidelberg, New York 1990; 73–134
  • Vaupel P, Kelleher DK. Metabolic status and reaction to heat of normal and tumor tissue. Thermoradiotherapy and Thermochemotherapy, MH Seegenschmiedt, P Fessenden, CC Vernon. Springer, Berlin, Heidelberg New York 1995; 1: 157–176
  • Vaupel P, Kallinowski F. Physiological effects of hyperthermia. Rec Res Cancer Res 1987; 104: 71–109
  • Vaupel P, Kallinowski F, Kluge M. Pathophysiology of tumors in hyperthermia. Rec Res Cancer Res 1988; 107: 65–75
  • Gerweck LE. Modifiers of thermal effects: Environmental factors. Hyperthermia and Oncology, M Urano, E Douple. VSP, Utrecht 1988; 1: 83–98
  • Jones EL, Oleson JR, Prosnitz LR, Samulski TV, Vujaskovic Z, Yu D, Sanders LL, Dewhirst MW. Randomized trial of hyperthermia and radiation for superficial tumors. J Clin Oncol 2005; 23: 3079–3085
  • Thrall DE, Larue SM, Pruitt AF, Case B, Dewhirst MW. Changes in tumour oxygenation during fractionated hyperthermia and radiation therapy in spontaneous canine sarcomas. Int J Hyperthermia 2006; 22: 365–373
  • Sutherland RM. Cell and environment interactions in tumor microregions: The multicell spheroid model. Science 1988; 240: 177–184
  • Bussink J, The Tumor Microenvironment and Effects of Hypoxia Modification. Nijmegen: Proefschrift, Katholieke Universiteit; 2000.
  • Goode JA, Chadwick DJ, The Tumour Microenvironment: Causes and Consequences of Hypoxia and Acidity. Novartis Foundation Symposium 240, Chichester, New York: John Wiley; 2001.
  • Vaupel P, Kallinowski F, Okunieff P. Blood flow, oxygen and nutrient supply, and metabolic microenvironment of human tumors: A review. Cancer Res 1989; 49: 6449–6465
  • Vaupel P. Tumor microenvironmental physiology and its implications for radiation oncology. Sem Radiat Oncol 2004; 14: 198–206
  • Vaupel P. Blood flow and metabolic microenvironment of brain tumors. J Neuro-Oncol 1994; 22: 261–267
  • Vaupel P, Kelleher DK, Tumor Hypoxia. Stuttgart: Wissenschaftliche Verlagsges; 1999.
  • Vaupel P, Jain RK, Tumor Blood Supply and Metabolic Microenvironment. Characterization and Implications for Therapy. Stuttgart: Fischer; 1991.
  • Vaupel P, Höckel M. Blood supply, oxygenation status and metabolic micromilieu of breast cancers: Characterization and therapeutic relevance. Int J Oncol 2000; 17: 869–879
  • Molls M, Vaupel P, Blood Perfusion and Microenvironment of Human Tumors. Berlin, Heidelberg, New York: Springer; 2000.
  • Vaupel PW, Blood Flow, Oxygenation, Tissue pH Distribution, and Bioenergetic Status of Tumors. Lecture 23. Berlin: Ernst Schering Research Foundation; 1994.
  • Vaupel P. Physiological properties of malignant tumours. NMR Biomed 1992; 5: 220–225
  • Barnikol WKR. Temperatur, pH-Wert, Säurebelastung und Filtrierbarkeit normaler menschlicher Erythrozyten: In-vitro-Untersuchungen. Mögliche Bedeutung für die hypertherme hyperazidotische Tumortherapie [Temperature, pH value, acid load and filtrability of normal human erythrocytes: In vitro studies. Possible significance for hyperthermic hyperacidotic tumor therapy]. Arch Geschwulstforsch 1989; 59: 11–17
  • Kavanagh BD, Coffey BE, Needham D, Hochmuth RM, Dewhirst MW. The effect of flunarizine on erythrocyte suspension viscosity under conditions of extreme hypoxia, low pH, and lactate treatment. Br J Cancer 1993; 67: 734–741
  • Fajardo LF, Prionas SD. Endothelial cells and hyperthermia. Int J Hyperthermia 1994; 10: 347–353
  • Fajardo LF, Prionas SD, Kowalski J, Kwan HH. Hyperthermia inhibits angiogenesis. Radiat Res 1988; 114: 297–306
  • Sawaji Y, Sato T, Takeuchi A, Hirata M, Ito A. Anti-angiogenic action of hyperthermia by suppressing gene expression and production of tumour-derived vascular endothelial growth factor in vivo and in vitro. Br J Cancer 2002; 86: 1597–1603
  • Roca C, Primo L, Valdembri D, Cividalli A, Declerck P, Carmeliet P, Gabriele P, Bussolino F. Hyperthermia inhibits angiogenesis by a plasminogen activator inhibitor 1-dependent mechanism. Cancer Res 2003; 63: 1500–1507
  • Bicher HI, Sandhu TS, Vaupel P, Hetzel FW. Effect of localized microwave hyperthermia on physiological responses. J Natl Cancer Inst 1980; 61: 217–219
  • Eddy HA. Alterations in tumor microvasculature during hyperthermia. Radiology 1980; 137: 515–521
  • Emami B, Nussbaum GH, Ten Haken RK, Hughes WL. Physiological effects of hyperthermia: Response of capillary blood flow and structure to local tumor heating. Radiology 1980; 137: 805–809
  • Emami B, Song CW. Physiological mechanisms in hyperthermia: A review. Int J Radiat Oncol Biol Phys 1984; 10: 289–295
  • Reinhold HS, Endrich B. Tumour microcirculation as a target for hyperthermia. Int J Hyperthermia 1986; 2: 111–137
  • Reinhold HS, Van den Berg-Blok AP. Effects of hyperthermia on blood flow and metabolism. An Introduction to the Practical Aspects of Clinical Hyperthermia, SB Field, JW Hand. Taylor & Francis, London, New York 1990; 77–107
  • Reinhold HS. Physiological effects of hyperthermia. Rec Res Cancer Res 1988; 107: 32–43
  • Song CW. Physiological factors in hyperthermia. J Natl Cancer Inst 1980; 61: 169–176
  • Song CW. Effect of local hyperthermia on blood flow and microenvironment. Cancer Res 1984; 44: S4721–S4730
  • Song CW, Park H, Griffin RJ. Improvement of tumor oxygenation by mild hyperthermia. Radiat Res 2001; 155: 512–528
  • Song CW, Choi IB, Nah BS, Sahu SK, Osborn JL. Microvasculature and perfusion in normal tissues and tumors. Medical Radiology-Diagnostic Imaging and Radiation Oncology, MH Seegenschmiedt, P Fessenden, CC Vernon. Springer, Heidelberg 1995; 139–156
  • Vaupel P, Frinak S, Mueller-Klieser W, Bicher HI. Impact of localized hyperthermia on the cellular microenvironment in solid tumors. Natl Cancer Inst Monogr 1982; 61: 207–209
  • Endrich B, Hammersen F, Messmer K. Hyperthermia-induced changes in tumor microcirculation. Rec Res Cancer Res 1988; 107: 44–59
  • Endrich B. Hyperthermie und Tumormikrozirkulation. Eine kritische Wertung experimenteller und klinischer Befunde [Hyperthermia and tumor microcirculation. A critical evaluation of experimental and clinical data.]. Beitrg Onkol 1988; 31: 1–138
  • Dudar TE, Jain RK. Differential response of normal and tumor microcirculation to hyperthermia. Cancer Res 1984; 44: 605–612
  • Vujaskovic Z, Song CW. Physiological mechanisms underlying heat-induced radiosensitization. Int J Hyperthermia 2004; 20: 163–174
  • Thews G, Vaupel P. Vegetative Physiologie. Springer, Auflage. Heidelberg 2005; 5
  • Kelleher DK, Engel T, Vaupel P. Changes in microregional perfusion, oxygenation, ATP and lactate distribution in subcutaneous rat tumours upon water-filtered IR-A hyperthermia. Int J Hyperthermia 1995; 11: 241–255
  • Vaupel P, Kelleher DK, Thews O. Microtopology of local perfusion, oxygenation, metabolic and energetic status, and interstitial pH in malignant tumors: techniques and characterization. Exptl Oncol 2000; 22: 15–25
  • Jones EL Prosnitz LR, Dewhirst MW, Marcom PK, Hardenbergh PH, Marks LB, Brizel DM, Vujaskovic Z. Thermochemoradiotherapy improves oxygenation in locally advanced breast cancer. Clin Cancer Res 2004; 10: 4287–4293
  • Shakil A, Osborn JL, Song CW. Changes in oxygenation status and blood flow in a rat tumor model by mild hyperthermia. Int J Radiat Oncol Biol Phys 1999; 43: 859–865
  • Song CW, Park HJ, Lee CK, Griffin R. Implications of increased tumor blood flow and oxygenation caused by mild temperature hyperthermia in tumor treatment. Int J Hyperthermia 2005; 21: 761–767
  • Griffin RJ, Dings RPM, Jamshidi-Parsian A, Song CW. Mild temperature hyperthermia and radiation therapy: role of tumor vascular thermotolerance and relevant physiological factors. Int J Hyperthermia 2010, in press
  • Gerner EW, Schneider MJ. Induced thermal resistance in HeLa cells. Nature 1975; 256: 500–502
  • Henle KJ, Kramuz JE, Leeper DB. Induction of thermotolerance in Chinese hamster ovary cells by high (45°) or low (40°) hyperthermia. Cancer Res 1978; 38: 570–574
  • Vujaskovic Z, Poulson JM, Gaskin AA, Thrall DE, Page RL, Charles HC, MacFall JR, Brizel DM, Meyer RE, Prescott DM, et al. Temperature-dependent changes in physiological parameters of spontaneous canine soft tissue sarcomas after combined radiotherapy and hyperthermia treatment. Int J Radiat Oncol Biol Phys 2000; 46: 179–185
  • Viglianti BL, Lora-Michiels M, Poulson JM, Lan L, Yu D, Sanders L, Craciunescu O, Vujaskovic Z, Thrall DE, MacFall J, et al. Dynamic contrast-enhanced magnetic resonance imaging as a predictor of clinical outcome in canine spontaneous soft tissue sarcomas treated with thermoradiotherapy. Clin Cancer Res 2009; 15: 4993–5001
  • Vaupel, PW. Effects of physiological parameters on tissue response to hyperthermia: New experimental facts and their relevance to clinical problems. Hyperthermic Oncology 1992, EW Gerner, TC Cetas. Arizona Board of Regents, Tucson 1993; 17–23
  • Waterman FM, Fazekas J, Nerlinger RE, Leeper DB. Blood flow rates in human tumors during hyperthermia treatments as indicated by thermal washout. Radiat Res 1982; 91: 426–427
  • Waterman FM, Nerlinger RE, Moylan DJ, III, Leeper DB. Response of human tumor blood flow to local hyperthermia. Int J Radiat Oncol Biol Phys 1987; 13: 75–82
  • Groebe K, Kallinowski F, Vaupel P. Is the division of heated tissue into temperature equivalent zones suitable for estimation of tumor blood flow from thermal clearance curves?. Int J Radiat Oncol Biol Phys 1987; 13: 917–920
  • Waterman FM, Tupchong L, Nerlinger R, Matthews J. Blood flow in human tumors during local hyperthermia. Int J Radiat Oncol Biol Phys 1991; 20: 1255–1262
  • Waterman FM, Tupchong L, Matthews J, Nerlinger R. Mechanisms of heat removal during local hyperthermia. Int J Radiat Oncol Biol Phys 1989; 17: 1049–1055
  • Lammertsma AA, Wilson CB, Jones T. In vivo physiological studies in human tumors using positron emission tomography. Funktionsanalyse biolog Systeme 1991; 20: 319–325
  • Bowman HF, Martin GT, Newman WH, Kumar S, Welch C, Bornstein B, Herman TS. Human tumor perfusion measurements during hyperthermia therapy. Hyperthermic Oncology 1992, EW Gerner. Arizona Board of Regents, Tucson 1992; I: A17
  • Rau B, Wust P, Tilly W, Gellermann J, Harder C, Riess H, Budach V, Felix R, Schlag PM. Preoperative radiochemotherapy in locally advanced or recurrent rectal cancer: Regional radiofrequency hyperthermia correlates with clinical parameters. Int J Radiat Oncol Biol Phys 2000; 48: 381–391
  • Lüdemann L, Sreenivasa G, Amthauer H, Michel R, Gellermann J, Wust P. Use of H215O-PET for investigating perfusion changes in pelvic tumors due to regional hyperthermia. Int J Hyperthermia 2009; 25: 299–308
  • Acker JC, Dewhirst MW, Honoré GM, Samulski TV, Tucker JA, Oleson JR. Blood perfusion measurements in human tumours: Evaluation of laser Doppler methods. Int J Hyperthermia 1990; 6: 287–304
  • Molls M, Feldmann HJ. Clinical investigations of blood flow in malignant tumors of the pelvis and the abdomen in patients undergoing thermoradiotherapy. Funktionsanalyse biolog Systeme 1991; 20: 143–153
  • Waterman FM, Komarnicky L, Leeper DB. The response of human tumour blood flow to a fractionated course of thermoradiotherapy. Int J Hyperthermia 1998; 14: 421–434
  • Vaupel P. Pathophysiological effects of hyperthermia in solid tumors and their clinical implication. Isolated Hyperthermic Limb Perfusion, GH Omlor, P Vaupel, Chr Alexander. Springer, Berlin, Heidelberg, New York 1995; 9–45
  • Lyng H, Vorren AO, Sundfør K, Taksdal I, Lien HH, Kaalhus O, Rofstad EK. Intra- and intertumor heterogeneity in blood perfusion of human cervical cancer before treatment and after radiotherapy. Int J Cancer 2001; 96: 182–190
  • Kershaw LE, Logue JP, Hutchinson CE, Clarke NW, Buckley DL. Late tissue effects following radiotherapy and neoadjuvant hormone therapy of the prostate measured with quantitative resonance imaging. Radiother Oncol 2008; 88: 127–134
  • Warburg O. On respiratory impairment in cancer cells. Science 1956; 124: 269–270
  • Vaupel P, Höckel M, Mayer A. Detection and characterization of tumor hypoxia using pO2 histography. Antioxid Redox Signal 2007; 9: 1221–1235
  • Vaupel P, Mayer A. Hypoxia in cancer: Significance and impact on clinical outcome. Cancer Metastasis Rev 2007; 26: 225–239
  • Vaupel P, Thews O, Hoeckel M. Treatment resistance of solid tumors: Role of hypoxia and anemia. Med Oncol 2001; 18: 243–259
  • Gerweck LE, Urano M, Koutcher J, Fellenz MP, Kahn J. Relationship between energy status, hypoxic cell fraction, and hyperthermic sensitivity in a murine fibrosarcoma. Radiat Res 1989; 117: 448–458
  • Overgaard J, Bichel P. The influence of hypoxia and acidity on the hyperthermic response of malignant cells in vitro. Radiology 1977; 123: 511–514
  • Vaupel P. The role of hypoxia-induced factors in tumor progression. Oncologist 2004; 9: S10–S17
  • Giaccia AJ. Hypoxic stress proteins: Survival of the fittest. Semin Radiat Oncol 1996; 6: 46–58
  • Höckel M, Vaupel P. Tumor hypoxia: Definitions and current clinical, biologic and molecular aspects. J Natl Cancer Inst 2001; 93: 266–276
  • Rofstad EK, Sundfør K, Lyng H, Trope CG. Hypoxia-induced treatment failure in advanced squamous cell carcinoma of the uterine cervix is primarily due to hxpoxia-induced radiation resistance rather than hypoxia-induced metastasis. Br J Cancer 2000; 83: 354–359
  • Rofstad EK, Galappathi K, Mathiesen B, Ruud EB. Fluctuating and diffusion-limited hypoxia in hypoxia-induced metastasis. Clin Cancer Res 2007; 13: 1971–1978
  • Cairns RA, Hill RP. Acute hypoxia enhances spontaneous lymph node metastasis in an orthotopic murine model of human cervical carcinoma. Cancer Res 2004; 64: 2054–2061
  • Le QT, Denko NC, Giaccia AJ. Hypoxic gene expression and metastasis. Cancer Metastasis Rev 2007; 26: 333–339
  • Chan DA, Giaccia AJ. Hypoxic gene expression and metastasis. Cancer Metastasis Rev 2004; 23: 293–310
  • Bicher HI, Hetzel FW, Sandhu TS, Frinak S, Vaupel P, O’Hara MD, O’Brien T. Effects of hyperthermia on normal and tumor microenvironment. Radiology 1980; 137: 523–530
  • Vaupel P, Ostheimer K, Mueller-Klieser W. Circulatory and metabolic responses of malignant tumors during localized hyperthermia. J Cancer Res Clin Oncol 1980; 98: 15–29
  • Vaupel P, Otte J, Manz R. Changes in tumor oxygenation after localized microwave hyperthermia. Progr Clin Biol Res 1982; 107: 65–74
  • Horsman MR, Overgaard J. Can mild hyperthermia improve tumour oxygenation?. Int J Hyperthermia 1997; 13: 141–147
  • Sun X, Li X-F, Russell J, Xing L, Urano M, Li GC, Humm JL, Ling CC. Changes in tumor hypoxia induced by mild temperature hyperthermia as assessed by dual-tracer immunohistochemistry. Radiother Oncol 2008; 88: 269–276
  • Horsman MR. Tissue physiology and the response to heat. Int J Hyperthermia 2006; 22: 197–203
  • Thews O, Li Y, Kelleher DK, Chance B, Vaupel P. Microcirculatory function, tissue oxygenation, microregional redox status and ATP distribution in tumors upon localized infrared-A hyperthermia at 42°C. Adv Exp Med Biol 2003; 530: 237–247
  • Kelleher DK, Vaupel P. No sustained improvement in tumor oxygenation after localized mild hyperthermia. Adv Exp Med Biol 2010; 662: 393–398
  • Song CW, Shakil A, Griffin RJ, Okajima K. Improvement of tumor oxygenation status by mild temperature hyperthermia alone or in combination with carbogen. Semin Oncol 1997; 24: 626–632
  • Song CW, Shakil A, Osborn JL, Iwata K. Tumor oxygenation is increased by hyperthermia at mild temperatures. Int J Hyperthermia 2009; 25: 91–95
  • Song C, Park H, Lee CK, Griffin R. Implications of increased tumor blood flow and oxygenation caused by mild temperature hyperthermia in tumor treatment. Int J Hyperthermia 2005; 21: 761–767
  • Iwata K, Shakil A, Hur WJ, Makepiece CM, Griffin RJ, Song CW. Tumour pO2 can be increased markedly by mild hyperthermia. Br J Cancer Suppl 1996; 27: S217–S221
  • Hetzel FW, Chopp M, Dereski MO. Variations in pO2 and pH response to hyperthermia: Dependence on transplant site and duration of treatment. Radiat Res 1992; 131: 152–156
  • Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Dodge RK, Charles HC, Samulski TV, Prosnitz LR, Dewhirst MW. Radiation therapy and hyperthermia improve the oxygenation of human soft tissue sarcomas. Cancer Res 1996; 56: 5347–5350
  • Vujaskovic Z, Rosen EL, Blackwell KL, Jones EL, Brizel DM, Prosnitz LR, Samulski TV, Dewhirst MW. Ultrasound guided pO2 measurement of breast cancer reoxygenation after neoadjuvant chemotherapy and hyperthermia treatment. Int J Hyperthermia 2003; 19: 498–506
  • Zywietz F, Reeker W, Kochs E. Changes in tumor oxygenation during a combined treatment with fractionated irradiation and hyperthermia: An experimental study. Int J Radiat Oncol Biol Phys 1997; 37: 155–162
  • Warburg O. Über den Stoffwechsel der Carcinomzelle. Klin Wschr 1925; 4: 534–536
  • Warburg O. The Metabolism of Tumours. Constable, London 1930
  • Thistlethwaite AJ, Leeper DB, Moylan DJ, Nerlinger RE. pH distribution in human tumors. Int J Radiat Oncol Biol Phys 1985; 11: 1647–1652
  • van den Berg AP, Wike-Hooley JL, van den Berg-Blok AE, van der Zee J, Reinhold HS. Tumour pH in human mammary carcinoma. Eur J Cancer Clin Oncol 1982; 18: 457–462
  • Wike-Hooley JL, Haveman J, Reinhold HS. The relevance of tumour pH to the treatment of malignant disease. Radiother Oncol 1984; 2: 343–366
  • Kallinowski F, Vaupel P. pH distributions in spontaneous and isotransplanted rat tumors. Br J Cancer 1988; 58: 314–321
  • Kallinowski F, Vaupel P. Factors governing hyperthermia-induced pH changes in malignant tumors. Int J Hyperthermia 1989; 5: 641–652
  • Vaupel P, Schaefer C, Okunieff P. Intracellular acidosis in murine fibrosarcomas coincides with ATP depletion, hypoxia, and high levels of lactate and total Pi. NMR Biomed 1994; 7: 128–136
  • Stubbs M, McSheehy PMJ, Griffiths JR, Bashford CL. Causes and consequences of tumour acidity and implications for treatment. Mol Med Today 2000; 6: 15–19
  • Tannock IF, Rotin D. Acid pH in tumors and its potential for therapeutic exploitation. Cancer Res 1989; 49: 4373–4384
  • Newell K, Tannock I. Regulation of intracellular pH and viability of tumor cells. Funktionsanalyse biolog Systeme 1991; 20: 219–234
  • Griffiths JR. Are cancer cells acidic?. Br J Cancer 1991; 64: 425–427
  • Gerweck LE. Tumor pH: Implications for treatment and novel drug design. Sem Radiat Oncol 1998; 8: 176–182
  • Song CW, Lyons JC, Luo Y. Intra- and extracellular pH in solid tumors: Influence on therapeutic response. Drug Resistance in Oncology, BA Teicher. Marcel Dekker, New York, Basel, Hong Kong 1993; 25–51
  • Song CW, Park H, Ross BD. Intra- and extracellular pH in solid tumors. Antiangiogenic Agents in Cancer Therapy, BA Teicher. Humana Press, Totowa 1999; 51–64
  • Prescott DM, Charles CH, Poulson JM, Page RL, Thrall DE, Vujaskovic Z, Dewhirst MW. The relationship between intracellular and extracellular pH in spontaneous canine tumors. Clin Cancer Res 2000; 6: 2501–2505
  • Stubbs M, Bhujwalla ZM, Tozer GM, Rodrigues LM, Maxwell RJ, Morgan R, Howe FA, Griffiths JR. An assessment of 31P-MRS as a method of measuring pH in rat tumours. NMR Biomed 1992; 5: 351–359
  • Gerweck LE. Modifiers of thermal effects: Environmental factors. Hyperthermia and Oncology, M Urano, E Douple. VSP, Utrecht 1988; 1: 83–98
  • van der Zee J, Broekmeyer-Reurink MP, van den Berg AP, van Geel BN, Jansen RMF, Kroon BBR, van Wyk J, Hagenbeck A. Temperature distribution and pH changes during hyperthermic regional isolation perfusion. Eur J Cancer Clin Oncol 1989; 25: 1157–1163
  • van de Merwe SA, van den Berg AP, van der Zee J, Reinhold HS. Measurement of tumor pH during microwave induced experimental and clinical hyperthermia with a fiber optic pH measurement system. Int J Radiat Oncol Biol Phys 1990; 18: 51–57
  • Lora-Michiels M, Yu D, Sanders L, Poulson JM, Azuma C, Case B, Vujaskovic Z, Thrall DE, Charles HC, Dewhirst MW. Extracellular pH and P-31 magnetic resonance spectroscopic variables are related to outcome in canine soft tissue sarcomas treated with thermoradiotherapy. Clin Cancer Res 2006; 12: 5733–5740
  • Engin K, Leeper DB, Thistlethwaite AJ, Tupchong L, McFarlane JD. Tumor extracellular pH as a prognostic factor in thermoradiotherapy. Int J Radiat Oncol Biol Phys 1994; 30: 125–132
  • van den Berg AP, Wike-Hooley JL, Broekmeyer-Reurink MP, van der Zee J, Reinhold HS. The relationship between the unmodified initial tissue pH of human tumours and the response to combined radiotherapy and local hyperthermia treatment. Eur J Cancer Clin Oncol 1989; 25: 73–78
  • Vaupel P, Kelleher DK, Engel T. Stable bioenergetic status despite substantial changes in blood flow and tissue oxygenation in a rat tumour. Br J Cancer 1994; 69: 46–49
  • Gerweck LE, Dahlberg WK, Epstein LF, Shimm D. Influence of nutrient and energy deprivation on cellular response to single and fractionated heat treatments. Radiat Res 1984; 99: 573–581
  • Koutcher JA, Barnett D, Kornblith AB, Cowburn D, Brady TJ, Gerweck LE. Relationship of changes in pH and energy status to hypoxic cell fraction and hyperthermia sensitivity. Int J Radiat Oncol Biol Phys 1990; 18: 1429–1435
  • Sijens PE, Bovee WMMJ, Koole P, Schipper J. Phosphorus NMR study of the response of a murine tumour to hyperthermia as a function of treatment time and temperature. Int J Hyperthermia 1989; 5: 351–357
  • Vaupel P, Okunieff P, Neuringer LJ. In vivo 31P-NMR spectroscopy of murine tumours before and after localized hyperthermia. Int J Hyperthermia 1990; 6: 15–31
  • Dewhirst MW, Poulson JM, Yu D, Sanders L, Lora-Michiels M, Vujaskovic Z, Jones EL, Samulski TV, Powers BE, Brizel DM, et al. Relation between pO2, 31P magnetic resonance spectroscopy parameters and treatment outcome in patients with high-grade soft tissue sarcomas treated with thermoradiotherapy. Int J Radiat Oncol Biol Phys 2005; 61: 480–491

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.