993
Views
52
CrossRef citations to date
0
Altmetric
Original

Hyperthermia, cisplatin and radiation trimodality treatment: A promising cancer treatment? A review from preclinical studies to clinical application

, , , , &
Pages 329-341 | Published online: 09 Jul 2009

Abstract

This review discusses available clinical and experimental data and the underlying mechanisms involved in trimodality treatment consisting of hyperthermia, cisplatin and radiotherapy. The results of phase I/II clinical trials show that trimodality treatment is effective and feasible in various cancer types and sites with tolerable toxicity. Based on these results, phase III trials have been launched to investigate whether significant differences in treatment outcome exist between trimodality and standard treatment. In view of the clinical interest, it is surprising to find so few preclinical studies on trimodality treatment. Although little information is available on the doses of the modalities and the treatment sequence resulting in the largest degree of synergistic interaction, the results from in vivo and in vitro preclinical studies support the use of trimodality treatment for cancer patients. Animal studies show an improvement in treatment outcome after trimodality treatment compared with mono- and bimodality treatment. Studies in different human tumour cell lines show that a synergistic interaction can be obtained between hyperthermia, cisplatin and radiation and that this interaction is more likely to occur in cell lines which are more sensitive to cisplatin.

Introduction

The beneficial effects of hyperthermia when added to radiotherapy have been proven by several randomized clinical trials Citation[1–11]. Combined hyperthermia and radiotherapy regimens are regarded as safe for patients with minor acute side-effects Citation[12]. Hyperthermia can also improve the effectiveness of several chemotherapeutic agents. A widely used agent often combined with hyperthermia treatment is cisplatin. Both in vitro and in vivo experiments have shown that its effectiveness is enhanced by the addition of concomitant hyperthermia Citation[13–20]. In the clinic, combined cisplatin and hyperthermia treatment is successfully used for treatment of several cancer types Citation[21–24]. Combined hyperthermia and cisplatin treatment is well tolerated by patients Citation[24]. Good results have also been obtained combining cisplatin and radiotherapy particularly in cervical cancer patients Citation[25–28]. In vitro, cisplatin has been shown to sensitize tumour cells to ionizing radiation Citation[29–34]. Therefore, combining hyperthermia, cisplatin and radiation may result in even better treatment responses through interactions between the three modalities.

The aim of this review is to discuss available clinical and experimental data and the underlying mechanisms involved in trimodality treatment consisting of hyperthermia, cisplatin and radiotherapy.

Patient studies

Regimens using cisplatin as a part of trimodality treatment have been tested in phase I/II trials in several types of superficial tumours Citation[35], breast carcinomas Citation[36] head and neck and oesophageal carcinomas Citation[37–44] and cervical carcinomas Citation[45–47]. An overview of these studies is shown in . This table shows treatment outcome as represented by the percentage of complete responses (CR) obtained after different trimodality treatment schedules.

Table I.  Phase II clinical studies for trimodality treatment consisting of hyperthermia, cisplatin and radiotherapy.

The first clinical trial was conducted by Herman et al. Citation[35]. In this phase I/II trial the addition of systemic cisplatin to local hyperthermia and radiation was tested to determine the tolerable cisplatin dose and to estimate the therapeutic potential of this treatment in patients with locally advanced superficial malignancies. Tumour types included in this trial were breast adenocarcinoma, head and neck squamous cell carcinoma, non-Hodgkins lymphoma, soft tissue sarcomas and colon and lung cancer. Hyperthermia and cisplatin were given concurrently before irradiation. The treatment was feasible and effective and a cisplatin dose of 30 mg/m2 injected intravenously weekly for 6 weeks appeared tolerable in patients heavily pre-treated with chemotherapy Citation[35]. In patients who had had little or no prior chemotherapy, cisplatin at 50 mg/m2 weekly was tolerable. It is notable, however, that pelvic radiation was not a component of therapy for any of these cases, and may have lowered the tolerable dose of cisplatin, since bone marrow toxicity was dose limiting (Herman, 2007, personal communication). Patients of this study were also included in a retrospective study of 29 patients with local-regional recurrence or advanced carcinoma of the breast Citation[36]. In these patients hyperthermia and radiotherapy were combined with cisplatin alone or cisplatin with etanidazole or bleomycin. This resulted in an overall CR rate of 53% for all treatments. However significant more and severe complications were seen in patients who had a history of previous radiotherapy.

In patients with advanced head and neck/oesophageal carcinoma several phase I/II trials were conducted Citation[37], Citation[38], Citation[40–44]. Various treatment schemes were investigated and all studies show that trimodality treatment with hyperthermia, cisplatin and radiotherapy is feasible in patients with acceptable toxicity. The highest CR rate for head and neck cancer patients was 93% after trimodality treatment compared with 86% in the no-chemotherapy group Citation[38]. However, larger trials and longer follow-up are necessary to prove statistical significance. This was the only study in head and neck carcinomas in which concomitant hyperthermia and low-dose (20 mg/m2) cisplatin treatment preceded radiotherapy. In oesophageal cancer patients the CR was between 30% and 35% in all studies. In two of these studies, concomitant hyperthermia and cisplatin followed radiotherapy; the third study did not report the implemented treatment schedule.

Of all cancer sites, cervical carcinoma is an obvious choice for trimodality treatment because both cisplatin-based chemoradiation and thermoradiotherapy were reported to obtain large improvements in treatment response and overall survival compared with standard radiotherapy Citation[5], Citation[25–28], Citation[48]. The feasibility of trimodality treatment in cervical carcinoma was tested in two phase II studies. In a phase II trial published by Jones et al. Citation[45] concurrent radiotherapy, cisplatin and hyperthermia was tested in patients with locally advanced and locally recurrent cervical cancer. Treatment consisted of weekly cisplatin and hyperthermia after irradiation. The therapy resulted in an excellent clinical response (initial CR of 92% and local control rate of 83%) and was well tolerated in the 12 patients studied. Another publication analysed the data of three independent but similar trials in the USA, Norway and the Netherlands including the patients of the study by Jones et al. Citation[46]. Sixty-eight patients with locally advanced cervical carcinoma received trimodality treatment. The patients received weekly cisplatin alone or concomitant with hyperthermia. Hyperthermia was applied before or after irradiation. The trimodality treatment was feasible and effective. Based on these results, a phase III study comparing the treatment with standard chemoradiation has been launched. Another trimodality study in cervical carcinoma was conducted by Sreenivasa et al. Citation[47] In this study the treatment resulted in high response rates, enabling curative surgery in a high proportion of patients with FIGO IIB-IVA non-resectable cancer.

Trimodality treatment using a chemotherapy regimen consisting of weekly oxaliplatin concomitantly with hyperthermia and folinic acid and 5-fluorouracil was conducted in pre-irradiated patients with locally recurrent rectal cancer Citation[49]. Treatment was performed for 6 consecutive weeks. Radiation was added to chemotherapy and hyperthermia in a subset of patients to elongate the remission interval. In this pilot study it was shown that this treatment was feasible with moderate overall toxicity. Oxaliplatin was used as the chemotherapeutic modality in this study because this is mostly regarded as the standard chemotherapy in rectal cancer patients.

According to the results of phase I/II clinical trials trimodality treatment is feasible and effective in several cancer types with only modest toxicity. Different sequencing could have an effect on the interaction of the three modalities and might therefore be important in determining treatment response. Because several in vitro studies showed that simultaneous hyperthermia and cisplatin result in increased cytotoxicity compared with consecutive treatment, most trials applied hyperthermia and cisplatin concomitantly before or after radiotherapy. Despite few pre-clinical studies, however, no conclusive evidence exists on the optimal regimen for trimodality treatment. This lack of evidence about the optimal treatment schedule and the effectiveness of trimodality treatment vs. standard treatment stressed the need for more phase III trials.

Animal studies

Only four papers were found on in vivo experiments concerning cisplatin-containing trimodality treatment; these are shown in . The first in vivo study was conducted by Herman et al. Citation[50]. In this study the effect of the sequence of the different modalities on tumour growth delay (TGD) and tumour cell kill was investigated to determine an optimal schedule for implementation in the clinic. All trimodality treatments resulted in a larger TGD compared with one-modality or two-modality treatment. The largest TGD was obtained by treating with cDDP just prior to hyperthermia with radiation after hyperthermia. This treatment scheme also had the largest cytotoxic effect as determined by excision of the tumours followed by plating of tumour cell suspensions for colony-forming assay. This suggests that the interaction of cDDP with hyperthermia prior to irradiation would improve tumour control, although this endpoint was not studied. Trimodality treatment using cisplatin/hyperthermia/radiation was shown to be significantly more effective (TGD of 25 days) than with etoposide/hyperthermia/radiation, which resulted in a TGD of only 14 days Citation[51]. However, this is only a tentative conclusion since these results are from sequential studies; other treatment sequences might lead to different results.

Table II.  Trimodality treatment consisting of hyperthermia, cisplatin and radiotherapy in animals.

Overgaard et al. investigated the interaction between cisplatin and hyperthermia when applied after irradiation Citation[52]. To investigate the effect on radiosensitivity the endpoint TCD50 was determined. The study design allowed evaluation of the individual effect of the modalities on the subfractions of hypoxic and oxygenated cells. The effect on the fraction of hypoxic cells was regarded as very important because it is this fraction of cells that is radioresistant and eventually determines treatment outcome. Therefore, especially hypoxic cells may benefit from combined treatment. In order to avoid direct interaction with radiation, they applied hyperthermia 4 h after irradiation either ‘simultaneously’ (hyperthermia 15 min after cisplatin) or ‘sequentially’ (hyperthermia 3.75 h after cisplatin) with cisplatin. The authors reasoned that a direct interaction of hyperthermia with radiation would not allow analysis of the extent of radiation modification. In addition to radiosensitivity, the effects of hyperthermia and cisplatin alone or in combination on clonogenic survival of hypoxic and aerobic cells were studied. While simultaneous hyperthermia and cisplatin resulted in significant additional cytotoxicity of well-oxygenated cells, trimodality treatment did not result in significant enhancement of the effect of combined radiation and hyperthermia. This was due to the lack of interaction between hyperthermia and cisplatin in the fraction of hypoxic cells as determined by clonogenic survival. A study by Herman et al. investigating the addition of 2-nitroimidazole radiosensitizers to cisplatin with radiation and with or without hyperthermia in the murine FSaIIC fibrosarcoma also showed a decrease in cell kill in the hypoxic cell fraction compared to the oxygenated cell fraction Citation[53].

Another study by Rao et al. Citation[54] investigated trimodality treatment in a sarcoma-180 mouse model. When compared with treatment with cisplatin, radiation or hyperthermia alone or a combination of two modalities, trimodality treatment had the largest benefit in overall survival and resulted in CR in 100% of the animals. In addition to trimodality treatment using hyperthermia at 43°C for 30 min, a combination with hyperthermia at 42°C for 60 min was investigated. This led to hyperthermia-induced trauma and some stress-induced death in the animals and is therefore too toxic in this experimental setting. The reason for this unexpected toxic effect was unknown.

Treatment with concomitant cisplatin and hyperthermia directly after irradiation was tested by Ressel et al. in nude mice carrying human-derived head and neck squamous cell carcinoma (SCCHN) xenografts Citation[55]. In this study, treatment outcome with trimodality treatment using cisplatin chemotherapy was compared with ifosfamide as the chemotherapeutic agent. Two different hyperthermia regimens were used: one with a target temperature of 41°C and one with a target temperature of 41.8°C. The different temperatures were chosen because 41°C is often used as a mean temperature in clinically applied locoregional hyperthermia Citation[56–59] and 41.8°C is used in whole body hyperthermia Citation[60–72]. The most effective treatment regimen was trimodality treatment containing cisplatin and local hyperthermia at 41.8°C, which resulted in CR in 80% of the animals. The side effects were mild and not different to those associated with single and double treatment combinations.

From these studies it becomes evident that trimodality treatment has a beneficial effect on treatment outcome compared to bi- or monomodality treatment. The effect of trimodality treatment on radiosensitivity was investigated only by Overgaard et al. Citation[52]. They found no beneficial effect on tumour radiosensitivity after addition of cisplatin to hyperthermia and radiation compared to hyperthermia and radiation alone. However, an additional effect may be observed by hyperthermia increasing cisplatin toxicity. No conclusion exists on the optimal timing of the modalities since all studies used different treatment schedules in various models. In addition, little is known about the toxicity of the treatments in animal experiments.

Studies using cultured tumour cell lines in vitro

Only a few studies exist on the interaction of hyperthermia, cisplatin and radiation in vitro. As stated earlier, an important factor determining the effectiveness of trimodality treatment is sequencing of the different modalities. In vitro studies have shown that simultaneous application of cisplatin and hyperthermia result in the largest cytotoxic effect Citation[14], Citation[20], Citation[73], Citation[74]. When cisplatin is combined with radiation, the addition of cisplatin just before irradiation results in the largest radiosensitization in some studies Citation[75], Citation[76]. However, other studies observed the largest degree of radiosensitization when cisplatin is applied after irradiatio Citation[77], Citation[78]. The effect of sequencing in trimodality treatment has been investigated in ovarian carcinoma cell lines with different cisplatin sensitivities Citation[79], Citation[80]. In these studies, treatment with 1 h 40°C hyperthermia and 1 h low concentration cisplatin combined with low dose rate (LDR) or high dose rate (HDR) irradiation was compared. An overview of the studies is shown in . For HDR irradiation, all treatment sequences resulted in a synergistic interaction between the modalities in both the cisplatin sensitive and the resistant cell line. In the cisplatin sensitive cell line the largest effect on cytotoxicity was observed when cisplatin was applied before irradiation with hyperthermia afterwards. In the cisplatin resistant cell line, by contrast, the largest effect was obtained with concomitant hyperthermia and cisplatin after irradiation. However, in this study the sequence of cisplatin and hyperthermia before irradiation has not been tested. For LDR irradiation, a synergistic interaction between the three modalities was only obtained in the cisplatin sensitive parental cell line. No difference was observed between the different treatment sequences.

Table III.  Trimodality treatment consisting of hyperthermia, cisplatin and radiotherapy in cell lines.

In our lab we recently studied the ability of hyperthermia at 41°C or at 43°C to increase cisplatin-induced radiosensitization in two cell lines with different sensitivities to cisplatin Citation[81]. In this study, cisplatin and hyperthermia were applied concomitantly before irradiation. The two different temperatures were chosen because of a recent debate on the effectiveness of mild temperature hyperthermia Citation[82]. The clinical goal to reach a tumour temperature of 43°C is often not accomplished and hyperthermia treatment in the range of 39–42°C can have powerful effects as well Citation[82]. Our results showed that 41°C hyperthermia added to cisplatin and radiation only increased radiosensitivity in cisplatin-sensitive cells. As expected, hyperthermia at 43°C was able to increase radiosensitivity in both cell lines.

The role of cellular cisplatin sensitivity in the effect of trimodality treatment containing long duration mild hyperthermia (LDMH) was investigated by Raaphorst et al. Citation[83]. In these studies LDMH of 40°C for 24 h was combined with LDR irradiation during which repair of sublethal damage takes place. This treatment resulted in a supra-additive effect on cell kill in both cisplatin resistant and sensitive ovarian carcinoma cell lines Citation[83]. The largest synergistic effect was seen in the cisplatin resistant cell line, which had a greater capacity to repair sublethal damage. In glioma cells, on the other hand, hyperthermia treatment could not overcome the resistance to cisplatin-induced radiosensitization Citation[84]. In the cisplatin sensitive glioma cell line trimodality treatment had a synergistic effect. Other studies on LDMH combined with radiation show that it can be an effective radiosensitizer in vitro and that the optimal treatment is to heat as long as practically possible Citation[85–93].

The available in vitro data show that trimodality treatment can result in a synergistic interaction between the different modalities depending on the temperature, cisplatin concentration and duration and radiation dose rate. A synergistic interaction is more likely to occur in cell lines that are more sensitive to cisplatin. However, more studies are needed in order to determine which treatment schedule results in the largest degree of synergism.

Discussion

Because hyperthermia, cisplatin and radiation therapies have a variety of molecular targets, a combination of these three modalities might result in a synergistic interaction. Important factors determining the degree of interaction may include tumour temperature, cisplatin dose, radiation dose rate and the sequence of application.

Hyperthermia has been shown to enhance radiation effects by inhibition of DNA repair processes Citation[94–107] or by tumour reoxygenatio Citation[108], Citation[109]. It is well known that an increase in tumour oxygenation results in an increased radiosensitivity of the tumour Citation[108]. In addition, hyperthermia induces direct cell kill, specifically in insufficiently perfused parts of the tumour Citation[110–115]. Hyperthermia has been shown significantly to enhance drug effectiveness. For cisplatin, enhancement takes place even at low temperatures without a threshold Citation[115]. This suggests a positive interaction between the two modalities. Proposed mechanisms of interaction are increased drug uptake, increased DNA and protein damage and pharmacological changes other than drug uptake Citation[115].

Increased drug uptake can be accomplished by hyperthermic damage to cell membranes Citation[116]. When cisplatin has entered the cellular nucleus, it forms DNA adducts, which can lead to strand breaking and hampered DNA replicatio Citation[33], Citation[117]. Cisplatin-induced DNA damage can be increased by the inhibition of DNA repair proteins caused by hyperthermia. Hyperthermia (and radiation) can also decrease the cellular amount of free radical scavengers Citation[116]. As a result, more DNA damage is induced by the production of free radicals by cisplatin Citation[118]. Another mechanism of interaction is through induction of pharmacological changes. One possibility is a decrease in cisplatin-protein binding leading to an increase in the amount of the cytotoxic free form of the drug Citation[116]. Another important pharmacological change is an increase in cisplatin delivery to the tumour by a hyperthermia-induced increase in blood flow Citation[116].

These proposed mechanisms suggest that the interaction between hyperthermia and cisplatin is optimal when both modalities are applied simultaneously. This has been confirmed by several observations Citation[14], Citation[18], Citation[20], Citation[74]. The treatment-enhancing effects of cisplatin decrease when the time between hyperthermia and cisplatin treatment increases Citation[14]. With hyperthermia increasing the effectiveness of cisplatin, one may expect that an even more effective treatment would consist of combining the two modalities with radiation. In addition to increasing the effectiveness of cisplatin, hyperthermia also increases radiosensitivity of cells by inhibition of DNA repair processes and tumour reoxygenation Citation[85], Citation[94–104, 106–109]. Hyperthermia-induced reoxygenation is caused by an increase in blood flow to the tumour, which also enhances delivery of cisplatin to tumour cells. The specific DNA repair pathways inhibited by hyperthermia are not completely known, but the non-homologous end-joining (NHEJ) Citation[119–121], homologous recombination (HR) Citation[121] and base excision repair (BER) pathways Citation[98] have all been suggested. In addition, cisplatin also acts as a radiosensitizer through inhibition of DNA repair Citation[33, 34, 122–124]. Nucleotide excision repair (NER) and NHEJ have both been reported to be inhibited by cisplatin Citation[125].

A tumour temperature of 43°C for 1 h is the clinical goal for externally applied local or locoregional hyperthermia. Therefore, most in vitro studies on the effects of hyperthermia focus on temperatures above 42°C. However, these temperatures are rarely obtained for deep hyperthermia in clinical practice. Lower temperatures (below 42°C) may have a more beneficial effect on blood flow and therefore on drug delivery to the tumour Citation[82]. In addition, cisplatin cytotoxicity is already enhanced at low temperatures Citation[116]. Therefore, there is renewed interest in the effects of temperatures in the range of 40–41°C Citation[82]. Results from Bergs et al. lead to the conclusion that the goal in trimodality treatment has to be to keep tumour temperatures as high as possible in order to radiosensitize tumours with different cisplatin sensitivities (see below). However, more pre-clinical studies are needed to investigate the importance of tumour temperature and its effects on trimodality treatment.

Different heating times at 40°C, cisplatin concentrations and radiation dose rates have been investigated by Raaphorst et al. Citation[79], Citation[80], Citation[83], Citation[84]. A synergistic interaction was obtained by all combined treatments in cisplatin-sensitive cells with cisplatin concentrations equal to or higher than 3 µg/ml. In patients a cisplatin plasma peak concentration of 49.8 µM/litre (or 16.6 µg/ml) can be achieved after administration of the maximal tolerated dose (MTD) of 100 mg/m2 Citation[126]. Heating to 41°C and 43°C in combination with radiation and various dosing regimens of cisplatin was studied by Bergs et al. Citation[81]. The results of this study show that trimodality treatment using low concentration (1 µM) continuous cisplatin incubation and 41°C hyperthermia resulted only in a synergistic effect in a cisplatin-sensitive cell line. In a cisplatin-resistant cell line, trimodality treatment using hyperthermia to 43°C had to be applied in order to achieve radiosensitization.

A synergistic interaction between the three modalities can be obtained using both LDR and HDR irradiation Citation[79], Citation[80], Citation[83], Citation[84]. LDR was combined with LDMH in these studies. In the clinic LDR in the form of brachytherapy could be used together with LDMH and cisplatin in order to increase treatment effectiveness especially in cells resistant to cisplatin. However, no clinical studies on trimodality using LDMH were found and only very few clinical studies on radiotherapy combined with LDMH exist. One study comparing LDMH/LDR thermoradiotherapy with acute-hyperthermia/LDR observed a better tumour response in patients treated with LDMH/LDR therapy than in patients receiving LDR alone. A review on this topic is published by Armour and Raaphorst Citation[85].

The mechanisms of action of each modality suggest that treatment sequence could be an important factor in determining the degree of interaction. The optimal treatment sequence leading to a synergistic interaction should be determined by in vitro experiments and verified in animal models. To date, no conclusive in vitro evidence exists on the optimal treatment sequence. Moreover, only one animal study by Herman et al. compared the effect of different treatment sequences on treatment outcome Citation[50]. They observed the largest TGD after concomitant cisplatin and hyperthermia applied before irradiation. More studies are needed to compare different treatment sequences in different tumour types using relevant animal models and endpoints. Because no evidence exists on the treatment sequence resulting in the largest degree of (synergistic) interaction, there is no consensus about which sequence to use in the clinic. Based on the evidence of several in vitro studies that simultaneous hyperthermia and cisplatin result in increased cytotoxicity compared with consecutive treatment most phase I/II studies applied hyperthermia and cisplatin concomitantly before or after radiotherapy. Whether hyperthermia should be applied prior to or after radiotherapy has been subject to debate. According to van der Zee et al., the application of hyperthermia after radiotherapy would result in a maximal therapeutic gain with lower radiosensitization in the normal tissue than in the tumour Citation[127]. In addition, hyperthermia before radiotherapy would increase distant metastases. However, Dewhirst et al. Citation[128] argue that it is highly unlikely that hyperthermia applied before radiotherapy will lead to increased toxicity in normal tissue. Instead, treating with hyperthermia prior to radiation may take advantage of heat-induced tumour reoxygenation resulting in radiosensitization. Dewhirst et al. further believe that there is no strong evidence for concluding that administering hyperthermia prior to radiotherapy carries any increased risk for metastases Citation[128].

Another important factor influencing the clinical response to trimodality treatment is the sensitivity of tumour cells to cisplatin. It has been shown that the addition of hyperthermia to cisplatin treatment could reduce cisplatin resistance in different cell lines Citation[129–132]. Therefore one might expect that hyperthermia and cisplatin treatment lead to increased cellular radiosensitivity compared with cisplatin and radiation. However, the effect of combined hyperthermia and cisplatin on radiosensitivity has been shown to vary in different cisplatin-resistant cell lines Citation[79], Citation[80], Citation[83], Citation[84]. The influence of cisplatin-resistant tumour cells on the clinical response of patients is not known. However, a tumour generally consists of cell fractions with differing degrees of cisplatin sensitivity. Therefore, it might be difficult to use cellular cisplatin sensitivity as a predictor for clinical response.

A restriction is that, to date, only phase II clinical trials on trimodality treatment consisting of hyperthermia, cisplatin and radiation have been published. Phase III trials are needed to compare trimodality treatment with other treatment arms. In 2003 a phase III clinical trial has started to compare trimodality treatment with standard chemoradiation in advanced stage cervical carcinoma patients Citation[46]. In this trial a total of 400 patients will be included in 4–5 years with a follow-up of 3 years. Patients are treated by external beam radiotherapy (45–50 Gy) for 5 weeks and brachytherapy with weekly hyperthermia and cisplatin (40 mg/m2). Cisplatin and hyperthermia are preferably administered concomitantly prior to radiotherapy.

Conclusion

In conclusion, trimodality treatment consisting of hyperthermia, cisplatin and radiation is effective and feasible in patients and seems to be promising. It is surprising that few preclinical studies exist on this subject. Several animal studies show that trimodality treatment has a beneficial effect on treatment outcome compared with mono- or dimodality treatment. This beneficial effect can be explained by the synergistic interaction between hyperthermia, cisplatin and radiation as shown by several in vitro studies. More pre-clinical studies are needed to investigate the mechanism of interaction, optimal doses, and sequencing of the modalities.

Acknowledgements

The authors thank Professor J. P. Medema for discussion in the work on trimodality treatment.

References

  • Wust P, Hildebrandt B, Sreenivasa G, Rau B, Gellermann J, Riess H, Felix R, Schlag PM. Hyperthermia in combined treatment of cancer. Lancet Oncol 2002; 3: 487–497
  • van der Zee J. Heating the patient: A promising approach?. Ann Oncol 2002; 13: 1173–1184
  • van der Zee J, Treurniet-Donker AD, The SK, Helle PA, Seldenrath JJ, Meerwaldt JH, Wijnmaalen AJ, van den Berg AP, van Rhoon GC, Broekmeyer-Reurink MP. Low dose reirradiation in combination with hyperthermia: A palliative treatment for patients with breast cancer recurring in previously irradiated areas. Int J Radiat Oncol Biol Phys 1988; 15: 1407–1413
  • Overgaard J. The current and potential role of hyperthermia in radiotherapy. Int J Radiat Oncol Biol Phys 1989; 16: 535–549
  • van der Zee J, Gonzalez GD, van Rhoon GC, van Dijk JD, van Putten WL, Hart AA. Comparison of radiotherapy alone with radiotherapy plus hyperthermia in locally advanced pelvic tumours: A prospective, randomised, multicentre trial. Dutch Deep Hyperthermia Group. Lancet 2000; 355: 1119–1125
  • Datta NR, Bose AK, Kapoor HK, Gupta S. Head and neck cancers: Results of thermoradiotherapy versus radiotherapy. Int J Hyperthermia 1990; 6: 479–486
  • Overgaard J, Gonzalez GD, Hulshof MC, Arcangeli G, Dahl O, Mella O, Bentzen SM. Randomised trial of hyperthermia as adjuvant to radiotherapy for recurrent or metastatic malignant melanoma. European Society for Hyperthermic Oncology. Lancet 1995; 345: 540–543
  • Perez CA, Pajak T, Emami B, Hornback NB, Tupchong L, Rubin P. Randomized phase III study comparing irradiation and hyperthermia with irradiation alone in superficial measurable tumors. Final report by the Radiation Therapy Oncology Group. Am J Clin Oncol 1991; 14: 133–141
  • Valdagni R, Amichetti M. Report of long-term follow-up in a randomized trial comparing radiation therapy and radiation therapy plus hyperthermia to metastatic lymph nodes in stage IV head and neck patients. Int J Radiat Oncol Biol Phys 1994; 28: 163–169
  • Vernon CC, Hand JW, Field SB, Machin D, Whaley JB, van der ZJ, van Putten WL, van Rhoon GC, van Dijk JD, Gonzalez GD, et al. Radiotherapy with or without hyperthermia in the treatment of superficial localized breast cancer: results from five randomized controlled trials. International Collaborative Hyperthermia Group. Int J Radiat Oncol Biol Phys 1996; 35: 731–744
  • Harima Y, Nagata K, Harima K, Ostapenko VV, Tanaka Y, Sawada S. A randomized clinical trial of radiation therapy versus thermoradiotherapy in stage IIIB cervical carcinoma. Int J Hyperthermia 2001; 17: 97–105
  • Dahl O. Editorial: Clinical hyperthermia combined with radiation is safe. Int J Hyperthermia 2005; 21: 193–197
  • Alberts DS, Peng YM, Chen HS, Moon TE, Cetas TC, Hoeschele JD. Therapeutic synergism of hyperthermia-cis-platinum in a mouse tumor model. J Natl Cancer Inst 1980; 65: 455–461
  • Haveman J, Bergs JW, Franken NA, van Bree C, Stalpers LJ. Effect of hyperthermia on uptake and cytotoxicity of cisplatin in cultured murine mammary carcinoma cells. Oncol Rep 2005; 14: 561–567
  • Meyn RE, Corry PM, Fletcher SE, Demetriades M. Thermal enhancement of DNA damage in mammalian cells treated with cis-diamminedichloroplatinum(II). Cancer Res 1980; 40: 1136–1139
  • Miyahara T, Ueda K, Akaboshi M, Shimada Y, Imamura M, Utsumi H. Hyperthermic enhancement of cytotoxicity and increased uptake of cis-diamminedichloroplatinum(II) in cultured human esophageal cancer cells. Jpn J Cancer Res 1993; 84: 336–340
  • Ohno S, Siddik ZH, Kido Y, Zwelling LA, Bull JM. Thermal enhancement of drug uptake and DNA adducts as a possible mechanism for the effect of sequencing hyperthermia on cisplatin-induced cytotoxicity in L1210 cells. Cancer Chemother Pharmacol 1994; 34: 302–306
  • Ohtsubo T, Saito H, Tanaka N, Noda I, Saito T, Kano E. Effect of heat-drug sequences on thermoenhancement and uptake of cis-DDP in human pharyngeal carcinoma. Anticancer Res 1996; 16: 297–300
  • Raaphorst GP, Yang DP. The evaluation of thermal cisplatin sensitization in normal and XP human cells using mild hyperthermia at 40 and 41°C. Anticancer Res 2005; 25: 2649–2653
  • Raaphorst GP, Li LF, Yang DP, LeBlanc JM. Cisplatin sensitization by concurrent mild hyperthermia in parental and mutant cell lines deficient in homologous recombination and non-homologous endjoining repair. Oncol Rep 2005; 14: 281–285
  • Richards WG, Zellos L, Bueno R, Jaklitsch MT, Janne PA, Chirieac LR, Yeap BY, Dekkers RJ, Hartigan PM, Capalbo L, et al. Phase I to II study of pleurectomy/decortication and intraoperative intracavitary hyperthermic cisplatin lavage for mesothelioma. J Clin Oncol 2006; 24: 1561–1567
  • Glehen O, Mithieux F, Osinsky D, Beaujard AC, Freyer G, Guertsch P, Francois Y, Peyrat P, Panteix G, Vignal J, et al. Surgery combined with peritonectomy procedures and intraperitoneal chemohyperthermia in abdominal cancers with peritoneal carcinomatosis: A phase II study. J Clin Oncol 2003; 21: 799–806
  • Sugarbaker PH, Yan TD, Stuart OA, Yoo D. Comprehensive management of diffuse malignant peritoneal mesothelioma. Eur J Surg Oncol 2006; 32: 686–691
  • Douwes F, Bogovic J, Douwes O, Migeod F, Grote C. Whole-body hyperthermia in combination with platinum-containing drugs in patients with recurrent ovarian cancer. Int J Clin Oncol 2004; 9: 85–91
  • Keys HM, Bundy BN, Stehman FB, Muderspach LI, Chafe WE, Suggs CL, III, Walker JL, Gersell D. Cisplatin, radiation, and adjuvant hysterectomy compared with radiation and adjuvant hysterectomy for bulky stage IB cervical carcinoma. N Engl J Med 1999; 340: 1154–1161
  • Morris M, Eifel PJ, Lu J, Grigsby PW, Levenback C, Stevens RE, Rotman M, Gershenson DM, Mutch DG. Pelvic radiation with concurrent chemotherapy compared with pelvic and para-aortic radiation for high-risk cervical cancer. N Engl J Med 1999; 340: 1137–1143
  • Peters WA, III, Liu PY, Barrett RJ, Stock RJ, Monk BJ, Berek JS, Souhami L, Grigsby P, Gordon W, Jr., Alberts DS. Concurrent chemotherapy and pelvic radiation therapy compared with pelvic radiation therapy alone as adjuvant therapy after radical surgery in high-risk early-stage cancer of the cervix. J Clin Oncol 2000; 18: 1606–1613
  • Whitney CW, Sause W, Bundy BN, Malfetano JH, Hannigan EV, Fowler WC, Clarke-Pearson DL, Liao SY. Randomized comparison of fluorouracil plus cisplatin versus hydroxyurea as an adjunct to radiation therapy in stage IIB-IVA carcinoma of the cervix with negative para-aortic lymph nodes: A Gynecologic Oncology Group and Southwest Oncology Group study. J Clin Oncol 1999; 17: 1339–1348
  • Loprevite M, Favoni RE, de Cupis A, Pirani P, Pietra G, Bruno S, Grossi F, Scolaro T, Ardizzoni A. Interaction between novel anticancer agents and radiation in non-small cell lung cancer cell lines. Lung Cancer 2001; 33: 27–39
  • Begg AC, van der Kolk PJ, Dewit L, Bartelink H. Radiosensitization by cisplatin of RIF1 tumour cells in vitro. Int J Radiat Biol Relat Stud Phys Chem Med 1986; 50: 871–884
  • Huang H, Huang SY, Chen TT, Chen JC, Chiou CL, Huang TM. Cisplatin restores p53 function and enhances the radiosensitivity in HPV16 E6 containing SiHa cells. J Cell Biochem 2004; 91: 756–765
  • Nakamoto S, Mitsuhashi N, Takahashi T, Sakurai H, Niibe H. An interaction of cisplatin and radiation in two rat yolk sac tumour cell lines with different radiosensitivities in vitro. Int J Radiat Biol 1996; 70: 747–753
  • Myint WK, Ng C, Raaphorst GP. Examining the non-homologous repair process following cisplatin and radiation treatments. Int J Radiat Biol 2002; 78: 417–424
  • Dolling JA, Boreham DR, Brown DL, Raaphorst GP, Mitchel RE. Cisplatin-modification of DNA repair and ionizing radiation lethality in yeast, Saccharomyces cerevisiae. Mutat Res 1999; 433: 127–136
  • Herman TS, Jochelson MS, Teicher BA, Scott PJ, Hansen J, Clark JR, Pfeffer MR, Gelwan LE, Molnar-Griffin BJ, Fraser SM. A phase I–II trial of cisplatin, hyperthermia and radiation in patients with locally advanced malignancies. Int J Radiat Oncol Biol Phys 1989; 17: 1273–1279
  • Bornstein BA, Zouranjian PS, Hansen JL, Fraser SM, Gelwan LA, Teicher BA, Svensson GK. Local hyperthermia, radiation therapy, and chemotherapy in patients with local-regional recurrence of breast carcinoma. Int J Radiat Oncol Biol Phys 1993; 25: 79–85
  • Amichetti M, Graiff C, Fellin G, Pani G, Bolner A, Maluta S, Valdagni R. Cisplatin, hyperthermia, and radiation (trimodal therapy) in patients with locally advanced head and neck tumors: A phase I–II study. Int J Radiat Oncol Biol Phys 1993; 26: 801–807
  • Schreiber DP, Overett TK. Interstitial hyperthermia and iridium-192 treatment alone vs. interstitial iridium-192 treatment/hyperthermia and low dose cisplatinum infusion in the treatment of locally advanced head and neck malignancies. Int J Radiat Oncol Biol Phys 1995; 33: 429–436
  • Feyerabend T, Steeves R, Wiedemann GJ, Weiss C, Wagner T, Richter E, Robins HI. Local hyperthermia, radiation, and chemotherapy in locally advanced malignancies. Oncology 1996; 53: 214–220
  • Serin M, Erkal HS, Cakmak A. Radiation therapy, cisplatin and hyperthermia in combination in management of patients with recurrent carcinomas of the head and neck with metastatic cervical lymph nodes. Int J Hyperthermia 1999; 15: 371–381
  • Serin M, Erkal HS, Cakmak A. Radiation therapy, cisplatin and hyperthermia in combination in management of patients with carcinomas of the head and neck with N2 or N3 metastatic cervical lymph nodes. Radiother Oncol 1999; 50: 103–106
  • Saeki H, Kawaguchi H, Kitamura K, Ohno S, Sugimachi K. Recent advances in preoperative hyperthermochemoradiotherapy for patients with esophageal cancer. J Surg Oncol 1998; 69: 224–229
  • Morita M, Kuwano H, Araki K, Egashira A, Kawaguchi H, Saeki H, Kitamura K, Ohno S, Sugimachi K. Prognostic significance of lymphocyte infiltration following preoperative chemoradiotherapy and hyperthermia for esophageal cancer. Int J Radiat Oncol Biol Phys 2001; 49: 1259–1266
  • Ohga T, Kimura Y, Futatsugi M, Miyazaki M, Saeki H, Nozoe T. Surgical and oncological advances in the treatment of esophageal cancer. Surgery 2002; 131: S28–S34
  • Jones EL, Samulski TV, Dewhirst MW, Alvarez-Secord A, Berchuck A, Clarke-Pearson D, Havrilesky LJ, Soper J, Prosnitz LR. A pilot Phase II trial of concurrent radiotherapy, chemotherapy, and hyperthermia for locally advanced cervical carcinoma. Cancer 2003; 98: 277–282
  • Westermann AM, Jones EL, Schem BC, van der Steen-Banasik EM, Koper P, Mella O, Uitterhoeve AL, de Wit R, van der, Velden J, Burger C, et al. First results of triple-modality treatment combining radiotherapy, chemotherapy, and hyperthermia for the treatment of patients with stage IIB, III, and IVA cervical carcinoma. Cancer 2005; 104: 763–770
  • Sreenivasa G, Hildebrandt B, Kummel S, Jungnickel K, Cho CH, Tilly W, Bohmer D, Budach V, Felix R, Wust P. Radiochemotherapy combined with regional pelvic hyperthermia induces high response and resectability rates in patients with nonresectable cervical cancer >/=FIGO IIB ‘bulky’. Int J Radiat Oncol Biol Phys 2006; 66: 1159–1167
  • van der Zee J, Gonzalez GD. The Dutch Deep Hyperthermia Trial: Results in cervical cancer. Int J Hyperthermia 2002; 18: 1–12
  • Hildebrandt B, Wust P, Drager J, Ludemann L, Sreenivasa G, Tullius SG, Amthauer H, Neuhaus P, Felix R, Riess H. Regional pelvic hyperthermia as an adjunct to chemotherapy (oxaliplatin, folinic acid, 5-fluorouracil) in pre-irradiated patients with locally recurrent rectal cancer: A pilot study. Int J Hyperthermia 2004; 20: 359–369
  • Herman TS, Teicher BA. Sequencing of trimodality therapy [cis-diamminedichloroplatinum(II)/hyperthermia/radiation] as determined by tumor growth delay and tumor cell survival in the FSaIIC fibrosarcoma. Cancer Res 1988; 48: 2693–2697
  • Pfeffer MR, Teicher BA, Holden SA, al Achi A, Herman TS. The interaction of cisplatin plus etoposide with radiation +/− hyperthermia. Int J Radiat Oncol Biol Phys 1990; 19: 1439–1447
  • Overgaard J, Radacic MM, Grau C. Interaction of hyperthermia and cis-diamminedichloroplatinum(II) alone or combined with radiation in a C3H mammary carcinoma in vivo. Cancer Res 1991; 51: 707–711
  • Herman TS, Teicher BA, Holden SA, Pfeffer MR, Jones SM. Addition of 2-nitroimidazole radiosensitizers to cis-diamminedichloroplatinum(II) with radiation and with or without hyperthermia in the murine FSaIIC fibrosarcoma. Cancer Res 1990; 50: 2734–2740
  • Rao BS, Devi PU. Response of mouse sarcoma-180 to cis-platin in combination with radiation & hyperthermia. Indian J Med Res 1996; 103: 315–322
  • Ressel A, Schmitt O, Weiss C, Feyerabend T. Therapeutic outcome and side-effects after radiotherapy, chemotherapy and/or hyperthermia treatment of head and neck tumour xenografts. Eur J Cancer 2002; 38: 594–601
  • Seegenschmiedt MH, Sauer R, Miyamoto C, Chalal JA, Brady LW. Clinical experience with interstitial thermoradiotherapy for localized implantable pelvic tumors. Am J Clin Oncol 1993; 16: 210–222
  • Gonzalez GD, van Dijk JD, Blank LE. Radiotherapy and hyperthermia. Eur J Cancer 1995; 31A: 1351–1355
  • Prionas SD, Kapp DS, Goffinet DR, Ben Yosef R, Fessenden P, Bagshaw MA. Thermometry of interstitial hyperthermia given as an adjuvant to brachytherapy for the treatment of carcinoma of the prostate. Int J Radiat Oncol Biol Phys 1994; 28: 151–162
  • Fatehi D, van der ZJ, Wielheesen DH, Van Wieringen WN, van Rhoon GC. Intra-luminal thermometry: Is tissue type assignment a necessity for thermal analysis?. Int J Hyperthermia 2006; 22: 463–473
  • Richel O, Zum Vorde Sive Vording PJ, Rietbroek R, van der Velden J, van Dijk JD, Schilthuis MS, Westermann AM. Phase II study of carboplatin and whole body hyperthermia (WBH) in recurrent and metastatic cervical cancer. Gynecol Oncol 2004; 95: 680–685
  • Westermann AM, Grosen EA, Katschinski DM, Jager D, Rietbroek R, Schink JC, Tiggelaar CL, Jager E, Zum Vorde Sive Vording PZ, Neuman A, et al. A pilot study of whole body hyperthermia and carboplatin in platinum-resistant ovarian cancer. Eur J Cancer 2001; 37: 1111–1117
  • Westermann AM, Wiedemann GJ, Jager E, Jager D, Katschinski DM, Knuth A, Sum Vorde Sive Vording PZ, van Dijk JD, Finet J, Neumann A, et al. A Systemic Hyperthermia Oncologic Working Group trial. Ifosfamide, carboplatin, and etoposide combined with 41.8°C whole-body hyperthermia for metastatic soft tissue sarcoma. Oncology 2003; 64: 312–321
  • Hildebrandt B, Drager J, Kerner T, Deja M, Loffel J, Stroszczynski C, Ahlers O, Felix R, Riess H, Wust P. Whole-body hyperthermia in the scope of von Ardenne's systemic cancer multistep therapy (sCMT) combined with chemotherapy in patients with metastatic colorectal cancer: A phase I/II study. Int J Hyperthermia 2004; 20: 317–333
  • Bakhshandeh A, Bruns I, Traynor A, Robins HI, Eberhardt K, Demedts A, Kaukel E, Koschel G, Gatzemeier U, Kohlmann T, et al. Ifosfamide, carboplatin and etoposide combined with 41.8°C whole body hyperthermia for malignant pleural mesothelioma. Lung Cancer 2003; 39: 339–345
  • Kerner T, Hildebrandt B, Ahlers O, Deja M, Riess H, Draeger J, Wust P, Gerlach H. Anaesthesiological experiences with whole body hyperthermia. Int J Hyperthermia 2003; 19: 1–12
  • Hegewisch-Becker S, Gruber Y, Corovic A, Pichlmeier U, Atanackovic D, Nierhaus A, Hossfeld DK. Whole-body hyperthermia (41.8°C) combined with bimonthly oxaliplatin, high-dose leucovorin and 5-fluorouracil 48-hour continuous infusion in pretreated metastatic colorectal cancer: A phase II study. Ann Oncol 2002; 13: 1197–1204
  • Wust P, Riess H, Hildebrandt B, Loffel J, Deja M, Ahlers O, Kerner T, von Ardenne A, Felix R. Feasibility and analysis of thermal parameters for the whole-body-hyperthermia system IRATHERM-2000. Int J Hyperthermia 2000; 16: 325–339
  • Katschinski DM, Wiedemann GJ, Mentzel M, Mulkerin DL, Touhidi R, Robins HI. Optimization of chemotherapy administration for clinical 41.8°C whole body hyperthermia. Cancer Lett 1997; 115: 195–199
  • Robins HI, Rushing D, Kutz M, Tutsch KD, Tiggelaar CL, Paul D, Spriggs D, Kraemer C, Gillis W, Feierabend C, et al. Phase I clinical trial of melphalan and 41.8°C whole-body hyperthermia in cancer patients. J Clin Oncol 1997; 15: 158–164
  • Wiedemann GJ, Robins HI, Gutsche S, Mentzel M, Deeken M, Katschinski DM, Eleftheriadis S, Crahe R, Weiss C, Storer B, et al. Ifosfamide, carboplatin and etoposide (ICE) combined with 41.8°C whole body hyperthermia in patients with refractory sarcoma. Eur J Cancer 1996; 32A: 888–892
  • Robins HI, Cohen JD, Schmitt CL, Tutsch KD, Feierabend C, Arzoomanian RZ, Alberti D, d’Oleire F, Longo W, Heiss C. Phase I clinical trial of carboplatin and 41.8°C whole-body hyperthermia in cancer patients. J Clin Oncol 1993; 11: 1787–1794
  • van der Zee J, van Rhoon GC, Wike-Hooley JL, Faithfull NS, Reinhold HS. Whole-body hyperthermia in cancer therapy: A report of a phase I–II study. Eur J Cancer Clin Oncol 1983; 19: 1189–1200
  • Ohtsubo T, Saito H, Tanaka N, Tsuzuki H, Saito T, Kano E. In vitro effect of hyperthermia on chemoenhancement and uptake of cisplatin in human pharyngeal carcinoma KB cells. Chemotherapy 1997; 43: 43–50
  • Kusumoto T, Maehara Y, Baba H, Takahashi I, Kusumoto H, Ohno S, Sugimachi K. Sequence dependence of the hyperthermic potentiation of carboplatin-induced cytotoxicity and intracellular platinum accumulation in HeLa cells. Br J Cancer 1993; 68: 259–263
  • Wang X, Chow LS, Nicholls JM, Kwong DL, Sham JS, Wong YC, Tsao SW. Significance of scheduling on the cytotoxicity of radiation and cisplatin combination treatment in nasopharyngeal carcinoma cells. Anticancer Drugs 2002; 13: 957–964
  • Raaphorst GP, Cybulski SE, Sobol R, Ng CE. The response of human breast tumour cell lines with altered polymerase beta levels to cisplatin and radiation. Anticancer Res 2001; 21: 2079–2083
  • Kyriazis AP, Kyriazis AA, Yagoda A. Time dependence of the radiation-modifying effect of cis-diamminedichloroplatinum II (cisplatin, DDP) on human urothelial cancer grown in nude mice. Cancer Invest 1986; 4: 217–222
  • Wilkins DE, Heller DP, Raaphorst GP. Inhibition of potentially lethal damage recovery by cisplatin in a brain tumor cell line. Anticancer Res 1993; 13: 2137–2142
  • Raaphorst GP, Miao J, Ng CE. Cisplatin and mild hyperthermia in radiosensitization to low dose rate irradiation in human ovarian carcinoma cells. Anticancer Res 1997; 17: 3469–3472
  • Raaphorst GP, Miao J, Stewart D, Ng CE. Interactions of mild hyperthermia, cisplatin and split dose irradiation in human ovarian carcinoma cells. Cancer Chemother Pharmacol 1998; 41: 491–496
  • Bergs JWJ, Haveman J, ten Cate R, Medema JP, Franken NAP, van Bree C. Effect of 41°C and 43°C on cisplatin-radiosensitization in two human carcinoma cell lines with different sensitivities for cisplatin. Oncol Rep, in press
  • Dewhirst MW, Vujaskovic Z, Jones E, Thrall D. Re-setting the biologic rationale for thermal therapy. Int J Hyperthermia 2005; 21: 779–790
  • Raaphorst GP, Maio J, Ng CE, Stewart DJ. Concomitant treatment with mild hyperthermia, cisplatin and low dose-rate irradiation in human ovarian cancer cells sensitive and resistant to cisplatin. Oncol Rep 1998; 5: 971–977
  • Raaphorst GP, Yang H, Wilkins DE, Ng CE. Cisplatin, hyperthermia and radiation treatment in human cisplatin-sensitive and resistant glioma cell lines. Int J Hyperthermia 1996; 12: 801–812
  • Armour EP, Raaphorst GP. Long duration mild temperature hyperthermia and brachytherapy. Int J Hyperthermia 2004; 20: 175–189
  • Ben Hur E, Bronk BV, Elkind MM. Thermally enhanced radiosensitivity of cultured Chinese hamster cells. Nat New Biol 1972; 238: 209–11
  • Ling CC, Robinson E. Moderate hyperthermia and low dose rate irradiation. Radiat Res 1988; 114: 379–384
  • Wang Z, Armour EP, Corry PM, Martinez A. Elimination of dose-rate effects by mild hyperthermia. Int J Radiat Oncol Biol Phys 1992; 24: 965–973
  • Spiro IJ, McPherson S, Cook JA, Ling CC, DeGraff W, Mitchell JB. Sensitization of low-dose-rate irradiation by nonlethal hyperthermia. Radiat Res 1991; 127: 111–114
  • Raaphorst GP, Heller DP, Bussey A, Ng CE. Thermal radiosensitization by 41°C hyperthermia during low dose-rate irradiation in human normal and tumour cell lines. Int J Hyperthermia 1994; 10: 263–270
  • Raaphorst GP, Bussey A, Heller DP, Ng CE. Comparison of thermoradiosensitization in two human melanoma cell lines and one fibroblast cell line by concurrent mild hyperthermia and low-dose-rate irradiation. Radiat Res 1994; 137: 338–345
  • Armour E, Wang ZH, Corry P, Martinez A. Equivalence of continuous and pulse simulated low dose rate irradiation in 9L gliosarcoma cells at 37 and 41 C. Int J Radiat Oncol Biol Phys 1992; 22: 109–114
  • Raaphorst GP, Ng CE, Shahine B. Comparison of radiosensitization by 41°C hyperthermia during low dose rate irradiation and during pulsed simulated low dose rate irradiation in human glioma cells. Int J Radiat Oncol Biol Phys 1999; 44: 185–188
  • Mitchel RE, Chan A, Smith BP, Child SD, Paterson MC. The effects of hyperthermia and ionizing radiation in normal and ataxia telangiectasia human fibroblast lines. Radiat Res 1984; 99: 627–635
  • Mitchel RE, Smith BP, Wheatly N, Chan A, Child S, Paterson MC. Sensitivity of hyperthermia-treated human cells to killing by ultraviolet or gamma radiation. Radiat Res 1985; 104: 234–241
  • Mitchel RE, Birnboim HC. Triggering of DNA strand breaks by 45°C hyperthermia and its influence on the repair of gamma-radiation damage in human white blood cells. Cancer Res 1985; 45: 2040–2045
  • Dynlacht JR, Xu M, Pandita RK, Wetzel EA, Roti Roti JL. Effects of heat shock on the Mre11/Rad50/Nbs1 complex in irradiated or unirradiated cells. Int J Hyperthermia 2004; 20: 144–156
  • Kampinga HH, Dynlacht JR, Dikomey E. Mechanism of radiosensitization by hyperthermia (≥43°C) as derived from studies with DNA repair defective mutant cell lines. Int J Hyperthermia 2004; 20: 131–139
  • Seno JD, Dynlacht JR. Intracellular redistribution and modification of proteins of the Mre11/Rad50/Nbs1 DNA repair complex following irradiation and heat-shock. J Cell Physiol 2004; 199: 157–170
  • Larsson C, Ng CE. p21+/+ (CDKN1A+/+) and p21−/− (CDKN1A−/−) human colorectal carcinoma cells display equivalent amounts of thermal radiosensitization. Radiat Res 2003; 160: 205–209
  • Kampinga HH, Dikomey E. Hyperthermic radiosensitization: Mode of action and clinical relevance. Int J Radiat Biol 2001; 77: 399–408
  • Raaphorst GP, Ng CE, Yang DP. Thermal radiosensitization and repair inhibition in human melanoma cells: A comparison of survival and DNA double strand breaks. Int J Hyperthermia 1999; 15: 17–27
  • Sakkers RJ, Filon AR, Brunsting JF, Kampinga HH, Konings AW, Mullenders LH. Selective inhibition of repair of active genes by hyperthermia is due to inhibition of global and transcription coupled repair pathways. Carcinogenesis 1995; 16: 743–748
  • Raaphorst GP, Feeley MM. Hyperthermia radiosensitization in human glioma cells comparison of recovery of polymerase activity, survival, and potentially lethal damage repair. Int J Radiat Oncol Biol Phys 1994; 29: 133–139
  • Deorukhakar VV, Anjaria KB, Rao BS. Modification of radiation-induced damage by hyperthermia–role of repair processes. Int J Hyperthermia 1993; 9: 803–810
  • Raaphorst GP, Azzam EI, Feeley M. Potentially lethal radiation damage repair and its inhibition by hyperthermia in normal hamster cells, mouse cells, and transformed mouse cells. Radiat Res 1988; 113: 171–182
  • Roti Roti JL. Heat-induced cell death and radiosensitization: Molecular mechanisms. Natl Cancer Inst Monogr 1982; 61: 3–10
  • Song CW, Shakil A, Osborn JL, Iwata K. Tumour oxygenation is increased by hyperthermia at mild temperatures. Int J Hyperthermia 1996; 12: 367–373
  • 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
  • Wallen CA, Colby TV, Stewart JR. Cell kill and tumor control after heat treatment with and without vascular occlusion in RIF-1 tumors. Radiat Res 1986; 106: 215–223
  • Rofstad EK, Solesvik OV, Brustad T. Tumour growth delay, cell inactivation and vascular damage following hyperthermic treatment of a human melanoma xenograft. Eur J Cancer Clin Oncol 1984; 20: 1295–1305
  • Rofstad EK, Brustad T. Primary and secondary cell death in human melanoma xenografts following hyperthermic treatment. Cancer Res 1986; 46: 355–361
  • Molls M. Hyperthermia—the actual role in radiation oncology and future prospects. Part I. Strahlenther Onkol 1992; 168: 183–190
  • Armour EP, McEachern D, Wang Z, Corry PM, Martinez A. Sensitivity of human cells to mild hyperthermia. Cancer Res 1993; 53: 2740–2744
  • van der Zee J, van Rhoon GC. Hyperthermia is effective in improving clinical radiotherapy results. Int J Radiat Oncol Biol Phys 2006; 66: 633–634
  • Dahl O. Mechanisms of thermal enhancement of chemotherapeutic cytotoxicity. Hyperthermia and Oncology, M Urano, E Douple, Utrecht, VSP BV 1994; 4: 9–29
  • Crul M, van Waardenburg RC, Beijnen JH, Schellens JH. DNA-based drug interactions of cisplatin. Cancer Treat Rev 2002; 28: 291–303
  • Rybak LP, Whitworth CA. Ototoxicity: Therapeutic opportunities. Drug Discov Today 2005; 10: 1313–1321
  • Zhu WG, Seno JD, Beck BD, Dynlacht JR. Translocation of MRE11 from the nucleus to the cytoplasm as a mechanism of radiosensitization by heat. Radiat Res 2001; 156: 95–102
  • Xu M, Myerson RJ, Hunt C, Kumar S, Moros EG, Straube WL, Roti Roti JL. Transfection of human tumour cells with Mre11 siRNA and the increase in radiation sensitivity and the reduction in heat-induced radiosensitization. Int J Hyperthermia 2004; 20: 157–162
  • Yin HL, Suzuki Y, Matsumoto Y, Tomita M, Furusawa Y, Enomoto A, Morita A, Aoki M, Yatagai F, Suzuki T, et al. Radiosensitization by hyperthermia in the chicken B-lymphocyte cell line DT40 and its derivatives lacking nonhomologous end joining and/or homologous recombination pathways of DNA double-strand break repair. Radiat Res 2004; 162: 433–441
  • Bergs JW, Franken NA, ten Cate R, van Bree C, Haveman J. Effects of cisplatin and gamma-irradiation on cell survival, the induction of chromosomal aberrations and apoptosis in SW-1573 cells. Mutat Res 2006; 594: 148–154
  • Lawrence TS, Blackstock AW, McGinn C. The mechanism of action of radiosensitization of conventional chemotherapeutic agents. Semin Radiat Oncol 2003; 13: 13–21
  • Haveman J, Castro KN, Rodermond HM, van Bree C, Franken NA, Stalpers LJ, Zdzienicka MZ, Peters GJ. Cellular response of X-ray sensitive hamster mutant cell lines to gemcitabine, cisplatin and 5-fluorouracil. Oncol Rep 2004; 12: 187–192
  • Huang X, Okafuji M, Traganos F, Luther E, Holden E, Darzynkiewicz Z. Assessment of histone H2AX phosphorylation induced by DNA topoisomerase I and II inhibitors topotecan and mitoxantrone and by the DNA cross-linking agent cisplatin. Cytometry A 2004; 58: 99–110
  • van Hennik MB, van der Vijgh WJ, Klein I, Elferink F, Vermorken JB, Winograd B, Pinedo HM. Comparative pharmacokinetics of cisplatin and three analogues in mice and humans. Cancer Res 1987; 47: 6297–6301
  • van der Zee J, de Bruijne M, van Rhoon GC. Letter to the editor. Int J Hyperthermia 2006; 22: 433–447
  • Dewhirst MW, Thrall D, Vujaskovic Z. In reply to van der Zee et al. Int J Hyperthermia 2006; 22: 437–444
  • Raaphorst GP, Chabot P, Doja S, Wilkins D, Stewart D, Ng CE. Effect of hyperthermia on cisplatin sensitivity in human glioma and ovarian carcinoma cell lines resistant and sensitive to cisplatin treatment. Int J Hyperthermia 1996; 12: 211–222
  • Hettinga JV, Lemstra W, Meijer C, Dam WA, Uges DR, Konings AW, De Vries EG, Kampinga HH. Mechanism of hyperthermic potentiation of cisplatin action in cisplatin-sensitive and–resistant tumour cells. Br J Cancer 1997; 75: 1735–1743
  • Beketic-Oreskovic L, Jaksic M, Oreskovic S, Osmak M. Hyperthermic modulation of resistance to cis-diamminedichloroplatinum (II) in human larynx carcinoma cells. Int J Hyperthermia 1997; 13: 205–214
  • Hettinga JV, Lemstra W, Meijer C, Mulder NH, Konings AW, De Vries EG, Kampinga HH. Hyperthermic potentiation of cisplatin toxicity in a human small cell lung carcinoma cell line and a cisplatin resistant subline. Int J Hyperthermia 1994; 10: 795–805

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.