1,619
Views
72
CrossRef citations to date
0
Altmetric
Review Article

Hyperthermia combined with radiation therapy for superficial breast cancer and chest wall recurrence: A review of the randomised data

, , , MD, PhD, , , , & show all
Pages 612-617 | Received 03 Feb 2010, Accepted 17 Apr 2010, Published online: 17 Sep 2010

Abstract

Hyperthermia has long been used in combination with radiation for the treatment of superficial malignancies, in part due to its radiosensitising capabilities. Patients who suffer superficial recurrences of breast cancer, be it in their chest wall following mastectomy, or in their breast after breast conservation, typically have poor clinical outcomes. They often develop distant metastatic disease, but one must not overlook the problems associated with an uncontrolled local failure. Morbidity is enormous, and can significantly impair quality of life. There is no accepted standard of care in treating superficial recurrences of breast cancer, particularly in patients that have previously been irradiated. There is a substantial literature regarding the combined use of hyperthermia and radiotherapy for these superficial recurrences. Most of it is retrospective in nature, but there are several larger phase III randomised trials that show an improved rate of clinical complete response in patients treated with both modalities. In this review article, we will highlight the important prospective data that has been published regarding the combined use of hyperthermia and radiation.

Introduction

Patients with chest wall/superficial breast cancer recurrences are a heterogeneous group, but the unifying principle is that they have failed standard therapy. Local recurrence rates after mastectomy range from 5% to 45%, thus prompting the consideration of adjuvant radiation therapy Citation[1–7]. The use of post-mastectomy radiotherapy has been demonstrated to dramatically decrease this risk of failure to 2% to 15%, as well as promote a survival advantage Citation[6], Citation[8], Citation[9]. When patients do fail locally, morbidities include pain, ulceration, bleeding, lymphoedema, brachial plexopathy, as well as the psychological distress of having visible local disease Citation[10], Citation[11]. A significant proportion of patients who experience a chest wall failure will also develop distant metastatic disease, prompting some to treat patients with local recurrence palliatively Citation[5], Citation[6], Citation[17–23]. However, with aggressive local therapy, some patients are able to have long disease-free intervals Citation[17–23].

Hyperthermia (HT) in the clinical context of a radiosensitiser for superficial tumours is defined as temperatures that are above normal physiological conditions, ranging from 40° to 45°C. The earliest report of its use in the treatment of breast cancer was more than 5000 years ago, described on an Egyptian papyrus Citation[24]. In the modern era, its use in conjunction with radiotherapy (RT) is based on several biological principles, including its ability to affect cells in S phase, a portion of the cell cycle where radiation is less effective Citation[25]. In addition to being directly cytotoxic, hyperthermia can inhibit sub-lethal damage repair and improve oxygenation, thus limiting the degree of hypoxia that hampers the effectiveness of radiotherapy Citation[26–28]. Utilising hyperthermia as a radiosensitiser, by definition, would reduce the dose of radiation needed to get the same effect as in its absence Citation[29]; this is especially important in women who received prior irradiation for their breast cancer, when one worries about increased normal tissue toxicity. Furthermore, doses for treating disease in a previously irradiated fields are often more limited, which increases the importance of tumour sensitisation.

Randomised data

There are numerous reports in the literature detailing the results of combined hyperthermia and radiation therapy for chest wall/superficial recurrences of breast cancer Citation[30–42]. Many are single institution retrospective accounts, but there are several large prospective randomised trials with a primary endpoint of percentage achieving a clinical complete response (CR) Citation[30–32], Citation[34] ().

Table I.  Complete response (CR) rate in randomised trials involving irradiation with or without hyperthermia for chest wall recurrence/superficial breast cancer.

The first randomised trial was run by the Radiation Therapy Oncology Group (RTOG), protocol 8104, and included 307 patients with superficially measurable tumours, 245 of which had single lesions and were available for analysis. Their treatment consisted of a radiation dose of 32 Gy, given in 4 Gy twice weekly fractions, with two hyperthermia sessions (goal 42.5°C, 45–60 min). Approximately 30% (68 patients) of those included had superficial disease in the breast or chest wall. Their primary endpoint was rate of CR, and overall, they did not find a statistically significant increase in local control with the addition of heat to radiotherapy Citation[30]. On subset analysis, those patients with breast or chest wall/flank lesions had a CR rate of 62% with the addition of heat, compared to 40% without. In addition, patients with a lesion diameter <3 cm had a CR rate of 52% with heat, versus 39% with radiotherapy alone (p = 0.02). A caveat of their results that was pointed out, which has been borne out in other studies, was the quality of treatment delivered. Only 52% in the combined HT/RT group received full RT dose and 8 HT treatments; 42% and 31% of lesions <3 cm and >3 cm received ‘good’ heating, respectively, defined as at least 4 HT sessions of 42.5°C for 45 min at the temperature reference point Citation[30]. The authors noted that the poor survival of their patients (22% alive at 1 year), could also help explain their low control rates, in that patients need to survive long enough for full clinical impact to be appreciated in both arms. All toxicities were similarly balanced in the two groups, except that 30% of the patients who received RT and HT developed thermal blisters, versus 0% in the RT only arm. No mention was made as to the severity of the thermal injury, nor to the methods of treatment if necessary.

Shortly after RTOG 8104 began accruing patients, both Stanford and Thomas Jefferson Universities ran single institution randomised trials to address the question of whether one or two hyperthermia treatments should be given weekly Citation[34], Citation[42]. Both of these trials, which included a heterogeneous group of patients (mostly chest wall), found no difference in rate of CR if HT was given once or twice weekly. Radiotherapy was similar in the two trials for recurrences in the previously unirradiated chest, but when patients with chest wall recurrences required re-irradiation, the fractionation was different. Thomas Jefferson utilised a more hypofractionated regimen (40 Gy in 4 Gy fractions), whereas Stanford used more traditional fractionation (21.6–36 Gy in 1.8–2 Gy fractions). Both trials evaluated similar thermal profiles and both found Tmin to be the only thermal predictor for durable local control. The Stanford trial found a trend for increased local control with a Tmin ≥ 41°C, versus <41°C (p = 0.08), whereas the Thomas Jefferson found a median local control to be 12 months with a Tmin ≤ 39.5°C, versus 23 months for a Tmin > 39.5°C (p = 0.01).

Thermal injury was not statistically different in patients treated with one versus two weekly HT sessions. In the Stanford analysis, only 3/58 patients’ complications required medical treatment, and 2/58 required surgical intervention Citation[34]. Nearly 40% of the patients at Thomas Jefferson were reported to have no skin reactions related to therapy; 11/56 heated fields developed thermal blistering, which was equally balanced between the two groups Citation[42].

The reason this question was posed deals with the phenomenon of thermotolerance; that is, that most mammalian cells become resistant to the effects of heat at temperatures below 43°C, or at 37°C after exposure to temperatures greater than 43°C Citation[43]. As more research has been done in this area, the tolerance to heat has been found to be related to heat shock proteins (HSPs), which are up-regulated by hyperthermia Citation[44]. As a consequence, most advocate an interval between HT sessions of at least 48–72 h, so as to allow adequate time for their removal. However, there is some data to suggest that thermal radiosensitisation is not subject to thermotolerance. Armour et al. found that the protein synthesis inhibitor cycloheximide prohibited the induction of thermotolerance, but did not influence radiation sensitisation Citation[45]. Given this biological finding, a more aggressive hyperthermia fractionation scheme may be warranted, but to date has not been attempted.

The largest collection of prospective data for combining radiotherapy and hyperthermia for the treatment of superficial breast cancer was compiled in a collaborative effort by the UK Medical Research Council, European Society for Hyperthermic Oncology, Dutch Hyperthermia Group and the Princess Margaret Hospital/Ontario Cancer Institute Citation[32]. There were five simultaneous ongoing prospective trials being conducted by the aforementioned groups. Due to poor patient accrual it was decided to pool their data so as to increase statistical power. As one might expect, by combining multiple trials there is a large degree of heterogeneity in the patient populations, as well as the treatment delivered. In this meta-analysis of the European and Canadian series there were three identifiable patient groups: untreated primary inoperable breast cancer, recurrent disease in sites not previously irradiated, and those with recurrences in sites having previously been irradiated; 71% had recurrences on their chest wall Citation[32].

Radiotherapy was administered ‘radically’ (range of ‘effective’ dose 60–69.3 Gy) if the patient's failure was not in a previously irradiated field, or ‘palliatively’ (range 39.8–47.2 Gy) if it had been Citation[32]. Their goal with respect to hyperthermia was to achieve a minimum intratumour temperature of 43°C (42.5°C in one of the five trials). Their primary endpoint was the rate of CR, which was 59% and 41% with and without hyperthermia, respectively (p < 0.001, OR 2.3, 95%CI 1.4–3.8). In patients that had previously been irradiated, their rate of CR was 57% versus 31% (OR 4.7, 95%CI 2.4–9.5), and this was with ‘palliative’ doses of radiation. This represents a significant improvement in complete response with the addition of hyperthermia to irradiation. While overall survival was not a primary endpoint of this collection of trials, it was evaluated on subset analysis. There was no statistically significant difference in survival in patients irradiated with or without hyperthermia, due largely in part to the high percentage of patients that developed metastatic disease in both groups; median overall survival was 18 months, regardless of randomisation. However, given the natural history of recurrent breast cancer, a local therapy would not necessarily be expected to improve overall survival.

It should be noted that the majority of patients did not reach the minimum intratumour temperature defined at the outset of the trial. Sherar et al. examined the thermal dosimetry of 120 patients that were enrolled, and sought to find reliable treatment parameters that might predict for response to therapy Citation[46]. Five thermal endpoints were evaluated, of which two were found to be correlated with rates of CR; the Max (TDmin), and the Sum (TDmin), with the TDmin being the lowest recorded temperature during treatment. They highlighted that those patients without distant metastatic disease at entry–difficult to extrapolate from, as only one of the five trials vigorously evaluated for distant disease prior to enrolment–had better outcomes, including increased CR rate, local disease-free survival, time to local failure and overall survival, with improved quality of hyperthermia (with cut-offs of ≤10 or >10 min) Citation[46].

With respect to toxicity of HT combined with RT, the authors note that ‘a small number’ of patients did not complete their planned HT secondary to pain; they did not cite exactly how many. They found little difference in the rates of erythema and desquamation with the addition of HT to RT, but reported an 11% rate of thermal blistering in the combined group, versus a 2% rate in the RT alone patients Citation[32]. They noted that acute treatment-related toxicities healed with conservative measures alone. Three late treatment-related complications were identified in patients receiving HT plus RT (bone necrosis, bone fracture and brachial plexopathy), thus underscoring the importance of long-term follow-up of surviving patients Citation[32].

The most recently reported phase III trial of radiotherapy with or without hyperthermia was from Duke University Citation[31]. This study was rigorous in terms of meeting thermometry/thermal dose guidelines and included a ‘test dose’ of hyperthermia before randomisation to ensure that all patients were indeed heatable. Of the 122 patients enrolled, 108 patients were deemed heatable and were randomised. Of those patients, 65% had disease in their breast or chest wall. Overall, the CR rate was 66.1% for patients treated with both heat and radiotherapy, versus 42.3% in the radiotherapy alone arm (p = 0.02, OR 2.7, 95%CI 1.2–5.8). The thermal parameter used was the number of cumulative equivalent minutes at 43°C exceeded by 90% of monitored points within the tumour (CEM 43°C T90), which is similar to one of the two endpoints evaluated by Sherar et al. found to have a significant effect on CR (i.e. sum (TDmin), which is the CEM 43°C T100) Citation[46], Citation[47]. Jones et al. found that the CEM 43°C T90 was a strong predictor for CR, as was found in a Medical Research Council analysis of their data Citation[48]. Like the Vernon meta-analysis Citation[32], the patients that had the greatest benefit were those that had previously received radiation therapy, with CR rates of 68.2% and 23.5%, with and without hyperthermia, respectively. Not surprisingly, there were no differences in overall survival between the two groups.

As was the case with the previous trials, toxicity with the addition of HT to RT was manageable. Thermal burns occurred in 45% of patients randomised to HT plus RT, versus 5.7% in the RT alone group. Nearly half of the burns in the combined modality arm were first degree, with only three patients experiencing third-degree burns Citation[31]. Catheter-related toxicities were reported in six patients, which included pain requiring over the counter analgesics in three, infection requiring antibiotics in two, and wound management for bleeding in one Citation[31].

Conclusions

While there are some differences in the series on superficial hyperthermia and radiation, there seems to be one unifying theme. In a select group of patients, the addition of hyperthermia to radiotherapy increases the eradication of local tumour, with a modest increase in largely self-limited toxicity. While attainment of CR is a worthwhile study endpoint, one must also consider the need to address palliation of symptoms, in that the majority of these patients will ultimately succumb to their distant disease. In the modern era of ‘targeted’ therapy, the issue of local control will increasingly become more important. Future applications of hyperthermia combined with radiotherapy for should include the addition of targeted biological agents in the hopes of increasing the CR rate and hopefully translating into prolonged disease-free survival. Liposomal doxorubicin has been combined with radiotherapy and hyperthermia by one group and warrants further evaluation in the future Citation[49]. Efforts must be taken to provide reproducible, efficacious heating of tumours so that the synergistic effect of combining radiotherapy and hyperthermia can be optimised. With rigorous thermal dosimetry and careful treatment technique, the addition of heat to radiotherapy can result in long-term local control of breast cancer chest wall recurrences.

Declaration of interest: E.L. Jones, MD, PhD, BSD Medical and National Institutes of Health. M.W. Dewhirst is a paid consultant and Chair of the Scientific Advisory Board of Celsion, and is recipient of research contracts from Varian and GlaxoSmithKline. M.W.D. and V.J. are recipients of grants from the National Institutes of Health, all other authors had none. The authors alone are responsible for the content and writing of the paper.

References

  • Fisher B, Slack NH, Cavanaugh PJ, Gardner B, Ravdin RG. Postoperative radiotherapy in the treatment of breast cancer: Results of the NSABP clinical trial. Ann Surg 1970; 172: 711–732
  • Lee MC, Jagsi R. Postmastectomy radiation therapy: Indications and controversies. Surg Clin North Am 2007; 87: 511–526
  • Taghian A, Jeong JH, Mamounas E, Anderson S, Bryant J, Deutsch M, Wolmark N. Patterns of locoregional failure in patients with operable breast cancer treated by mastectomy and adjuvant chemotherapy with or without tamoxifen and without radiotherapy: Results from five National Surgical Adjuvant Breast and Bowel Project Randomized Clinical trials. J Clin Oncol 2004; 22: 4247–4254
  • Katz A, Strom EA, Buchholz TA, Thames HD, Smith CD, Jhingran A, Hortobagyi G, Buzdar AU, Theriault R, Singletary SE, et al. Locoregional recurrence patterns after mastectomy and doxorubicin-based chemotherapy: Implications for postoperative irradiation. J Clin Oncol 2000; 18: 2817–2827
  • Nielsen HM, Overgaard M, Grau C, Jensen AR, Overgaard J. Study of failure pattern among high-risk breast cancer patients with or without postmastectomy radiotherapy in addition to adjuvant systemic therapy: Long-term results from the Danish Breast Cancer Cooperative Group DBCG 82 b and c randomized studies. J Clin Oncol 2006; 24: 2268–2275
  • Ragaz J, Olivotto IA, Spinelli JJ, Phillips N, Jackson SM, Wilson KS, Knowling MA, Coppin CM, Weir L, Gelmon K, et al. Locoregional radiation therapy in patients with high-risk breast cancer receiving adjuvant chemotherapy: 20-year results of the British Columbia randomized trial. J Natl Cancer Inst 2005; 97: 116–126
  • MacDonald SM, Abi-Raad RF, Alm El-Din MA, Niemierko A, Kobayashi W, McGrath JJ, Goldberg SI, Powell S, Smith B, Taghian AG. Chest wall radiotherapy: Middle ground for treatment of patients with one to three positive lymph nodes after mastectomy. Int J Radiat Oncol Biol Phys 2009; 75: 1297–1303
  • Overgaard M, Hansen PS, Overgaard J, Rose C, Andersson M, Bach F, Kjaer M, Gadeberg CC, Mouridsen HT, Jensen MB, et al. Postoperative radiotherapy in high-risk premenopausal women with breast cancer who receive adjuvant chemotherapy. Danish Breast Cancer Cooperative Group 82b Trial. N Engl J Med 1997; 337: 949–955
  • Overgaard M, Jensen MB, Overgaard J, Hansen PS, Rose C, Andersson M, Kamby C, Kjaer M, Gadeberg CC, Rasmussen BB, et al. Postoperative radiotherapy in high-risk postmenopausal breast-cancer patients given adjuvant tamoxifen: Danish Breast Cancer Cooperative Group DBCG 82c randomised trial. Lancet 1999; 353: 1641–1648
  • Perez LA, Brady LW. Breast cancer: Locally advanced, inflammatory and recurrent tumors. Lippincott, Philadelphia 2003
  • De Vita V, Hellman S, Rosenberg SA. Cancer of the breast. Cancer, V De Vita, S Hellman, SA Rosenberg. Lippincott, Philadelphia 2001
  • Dunst J, Steil B, Furch S, Fach A, Lautenschlager C, Diestelhorst A, Lampe D, Kolbl H, Richter C. Prognostic significance of local recurrence in breast cancer after postmastectomy radiotherapy. Strahlenther Onkol 2001; 177: 504–510
  • Bedwinek JM, Lee J, Fineberg B, Ocwieza M. Prognostic indicators in patients with isolated local-regional recurrence of breast cancer. Cancer 1981; 47: 2232–2235
  • Chu FC, Lin FJ, Kim JH, Huh SH, Garmatis CJ. Locally recurrent carcinoma of the breast. Results of radiation therapy. Cancer 1976; 37: 2677–2681
  • Bedwinek J. Natural history and management of isolated local-regional recurrence following mastectomy. Semin Radiat Oncol 1994; 4: 260–269
  • Beck TM, Hart NE, Woodard DA, Smith CE. Local or regionally recurrent carcinoma of the breast: Results of therapy in 121 patients. J Clin Oncol 1983; 1: 400–405
  • Willner J, Kiricuta IC, Kolbl O. Locoregional recurrence of breast cancer following mastectomy: Always a fatal event? Results of univariate and multivariate analysis. Int J Radiat Oncol Biol Phys 1997; 37: 853–863
  • Chagpar A, Meric-Bernstam F, Hunt KK, Ross MI, Cristofanilli M, Singletary SE, Buchholz TA, Ames FC, Marcy S, Babiera GV, et al. Chest wall recurrence after mastectomy does not always portend a dismal outcome. Ann Surg Oncol 2003; 10: 628–634
  • Kamby C, Sengelov L. Pattern of dissemination and survival following isolated locoregional recurrence of breast cancer. A prospective study with more than 10 years of follow up. Breast Cancer Res Treat 1997; 45: 181–192
  • Halverson KJ, Perez CA, Kuske RR, Garcia DM, Simpson JR, Fineberg B. Survival following locoregional recurrence of breast cancer: Univariate and multivariate analysis. Int J Radiat Oncol Biol Phys 1992; 23: 285–291
  • Hsi RA, Antell A, Schultz DJ, Solin LJ. Radiation therapy for chest wall recurrence of breast cancer after mastectomy in a favorable subgroup of patients. Int J Radiat Oncol Biol Phys 1998; 42: 495–499
  • Schmoor C, Sauerbrei W, Bastert G, Schumacher M. Role of isolated locoregional recurrence of breast cancer: Results of four prospective studies. J Clin Oncol 2000; 18: 1696–1708
  • Mora EM, Singletary SE, Buzdar AU, Johnston DA. Aggressive therapy for locoregional recurrence after mastectomy in stage II and III breast cancer patients. Ann Surg Oncol 1996; 3: 162–168
  • Breasted JH. The Edwin Smith surgical papyrus. University of Chicago Oriental Institute Publications, Chicago 1930
  • Westra A, Dewey WC. Variation in sensitivity to heat shock during the cell-cycle of Chinese hamster cells in vitro. Int J Radiat Biol Relat Stud Phys Chem Med 1971; 19: 467–477
  • 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
  • Dewhirst MW. Concepts of oxygen transport at the microcirculatory level. Semin Radiat Oncol 1998; 8: 143–150
  • Hall EJ, Giaccia AJ. Radiobiology for the radiologist. Lippincott, Philadelphia 2006
  • 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. Am J Clin Oncol 1991; 14: 133–141
  • 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
  • Vernon CC, Hand JW, Field SB, Machin D, Whaley JB, van der Zee J, van Putten WLJ, van Rhoon GC, van Dijk JDP, Gonzalez Gonzalez D, et al. Radiotherapy with or without hyperthermia in the treatment of superficial localized breast cancer: Results from five randomized controlled trials. Int J Radiat Oncol Biol Phys 1996; 35: 731–744
  • Lee HK, Antell AG, Perez CA, Straube WL, Ramachandran G, Myerson RJ, Emami B, Molmenti EP, Buckner A, Lockett MA. Superficial hyperthermia and irradiation for recurrent breast carcinoma of the chest wall: Prognostic factors in 196 tumors. Int J Radiat Oncol Biol Phys 1998; 40: 365–375
  • Kapp DS, Petersen IA, Cox RS, Hahn GM, Fessenden P, Prionas SD, Lee ER, Meyer JL, Samulski TV, Bagshaw MA. Two or six hyperthermia treatments as an adjunct to radiation therapy yield similar tumor responses: Results of a randomized trial. Int J Radiat Oncol Biol Phys 1990; 19: 1481–1495
  • Perez CA, Nussbaum G, Emami B, Von Gerichten D. Clinical results of irradiation combined with local hyperthermia. Cancer 1983; 52: 1597–1603
  • Oleson JR, Sim DA, Manning MR. Analysis of prognostic variables in hyperthermia treatment of 161 patients. Int J Radiat Oncol Biol Phys 1984; 10: 2231–2239
  • Van der Zee J, van Putten WLJ, van den Berg AP, van Rhoon GC, Hooley JL, Broekmeyer-Reurink MP, Reinhold HS. Retrospective analysis of the response of tumours in patients treated with a combination of radiotherapy and hyperthermia. Int J Hyperthermia 1986; 2: 337–349
  • Rafla S, Parikh K, Tchelebi M, Youssef E, Selim H, Bishay S. Recurrent tumors of the head and neck, pelvis, and chest wall: Treatment with hyperthermia and brachytherapy. Radiology 1989; 172: 845–850
  • Seegenschmiedt HM, Karlsson UL, Sauer R, Brady LW, Jr, Herbst M, Amendola BE, Markoe AM, Fisher SA, Micaily B. Superficial chest wall recurrences of breast cancer: Prognostic treatment factors for combined radiation therapy and hyperthermia. Radiology 1989; 173: 551–558
  • Hehr T, Lamprecht U, Glocker S, Classen J, Paulsen F, Budach W, Bamberg M. Thermoradiotherapy for locally recurrent breast cancer with skin involvement. Int J Hyperthermia 2001; 17: 291–301
  • Phromratanapongse P, Steeves RA, Severson SB, Paliwal BR. Hyperthermia and irradiation for locally recurrent previously irradiated breast cancer. Strahlenther Onkol 1991; 167: 93–97
  • Engin K, Tupchong L, Moylan DJ, Alexander GA, Waterman, Komarnicky L, Nerlinger RE, Leeper DB. Randomized trial of one versus two adjuvant hyperthermia treatments per week in patients with superficial tumors. Int J Hyperthermia 1993; 9: 327–430
  • Henle KJ, Leeper DB. Interaction of hyperthermia and radiation in CHO cells: Recovery kinetics. Radiat Res 1976; 66: 505–518
  • Barnes JA, Dix DJ, Collins BW, Luft C, Allen JW. Expression of inducible Hsp70 enhances the proliferation of MCF-7 breast cancer cells and protects against the cytotoxic effects of hyperthermia. Cell Stress Chaperones 2001; 6: 316–325
  • Armour E, Wang Z, Corry P, Martinez A. Thermotolerance and radiation sensitizing effects of long-duration, mild temperature hyperthermia. Int J Hyperthermia 1994; 10: 315–324
  • Sherar M, Liu F, Pintilie M, Levin W, Hunt J, Hill R, Hand J, Vernon C, van Rhoon G, van der Zee J, et al. Relationship between thermal dose and outcome in thermoradiotherapy treatments for superficial recurrences of breast cancer: Data from a phase III trial. Int J Radiat Oncol Biol Phys 1997; 2: 371–380
  • Saparetto SA, Dewey WC. Thermal dose determination in cancer therapy. Int J Radiat Oncol Biol Phys 1984; 10: 787–800
  • Hand JW, Machin D, Vernon CC, Whaley JB. Analysis of thermal parameters obtained during phase III trials of hyperthermia as an adjunct to radiotherapy in the treatment of breast carcinoma. Int J Hyperthermia 1997; 13: 343–364
  • Kouloulias VE, Koukourakis GV, Petridis AK, Kouvaris I, Gouliamos AD. The efficacy of Caelyx and hyperthermia for anticancer treatment. Recent Pat Anticancer Drug Discov 2007; 2: 246–250

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.