2,621
Views
14
CrossRef citations to date
0
Altmetric
Review Article

Hyperthermia and radiotherapy in bladder cancer

, &
Pages 398-406 | Received 21 Nov 2015, Accepted 31 Jan 2016, Published online: 20 Apr 2016

Abstract

Hyperthermia represents a unique, safe, and advantageous methodology for improving therapeutic strategies in the management of bladder cancer. This modality has shown promise in contributing to treatment regimens for both superficial and muscle-invasive disease. Especially in conjunction with intravesical chemotherapy, systemic therapy, and radiotherapy, hyperthermia shows particular synergistic benefit. As such, it should be explored further through clinical use and clinical trial in conjunction with currently available techniques and emerging technologies. However, to conceptualise the way forward, it is particularly important to understand the current challenges to widespread use of non-invasive, bladder-sparing approaches and the current state of bladder cancer care. As such, in the following article, we have focused on not only the rationale for concurrent radiotherapy and hyperthermia, but also the clinical landscape in bladder cancer as a whole.

Bladder cancer – the clinical perspective

Modern oncological therapy has increasingly focused on organ-sparing techniques and modalities. Reducing the invasiveness and post-operative morbidity associated with radical surgeries or aggressive resections has become a particular priority. The management of bladder cancer is no exception, as increasingly patients and practitioners have pursued multimodality therapies with bladder preservation intent. In particular, in well-selected muscle-invasive cancer, many patients today receive concurrent chemoradiotherapy following maximal transurethral resection of the bladder tumor (TURBT), in an attempt to avoid radical cystectomy, and many are cured of their disease without the morbidity of bladder removal. With bladder cancer on the rise, this approach will likely see increased utilisation and clinical relevance [Citation1].

Utilised in T2 (muscle-invasive) or less invasive disease, the first step in bladder preservation, TURBT, is generally a piecemeal, estimated resection, preferably until there is no visual evidence of residual disease. The information gleaned is imperfect but essential for risk-stratifying patients for therapeutic decision-making. For patients with T1 disease, and thus no clear evidence of muscle involvement, various methods have been suggested to prognosticate on or predict for recurrence/progression based upon clinical and pathological findings. Most commonly, studies have cited poorly differentiated, high grade (grade 3) cells as an important risk factor [Citation2–9]. The European Organisation for Research and Treatment of Cancer (EORTC) developed a scoring system for estimating a patient’s risk of both recurrence of disease as well as progression to muscle-invasive disease [Citation10]. For recurrence, the number of tumours present, the size of the tumours, and rate of prior recurrences were most prognostic. For progression, the presence of carcinoma in situ disease, T stage, and grade of disease proved most important. The differences amongst risk groups were vast. Recurrence risk was rated from 0 to 17 with the lowest group (score 0) having a 31% risk of recurrence at 5 years, while the highest risk group (score 10–17) exhibited a 78% chance of recurrence. Progression to muscle-invasive disease (scored 0–23) showed similar trends: lowest risk group (score 0) 0.8% at 5 years, highest risk (score 14–23) 45%.

Patients with very low risk non-muscle-invasive disease are likely the ideal candidates for TURBT with either subsequent observation or adjuvant intravesical chemotherapeutic installation. This remains a standard of care option for this group [Citation11]. However, as suggested in the EORTC risk grouping, there is clearly a group of T1 patients who have an exceedingly high risk of recurrence or progression despite TURBT and intravesical chemotherapy [Citation10]. The authors here also posit that these patients may be good candidates for earlier, more aggressive local therapy such as cystectomy. These patients are thought to have disease with outcomes more akin to their muscle-invasive counterparts, and in truth, under-sampling by TURBT may leave these patients with occult T2 cancer, staged at T1 [Citation12–16].

In muscle-invasive disease, cystectomy remains a standard of care, but this is not without substantial and often life-altering morbidity. Radical cystectomy with pelvic nodal dissection (with strong consideration for neoadjuvant systemic therapy) remains the category 1 recommendation of the National Comprehensive Cancer Network for muscle-invasive disease patients; while these are only an expert panel’s recommendations as to approach to therapy, they are a frequent resource for practitioners looking for guidance [Citation11]. This surgery requires complete removal of the native bladder with prostatectomy in men, and frequently hysterectomy in women. Urinary diversion may be achieved with an orthotopic neobladder, created from a loop of bowel in the original bladder position with urethral anastomosis, or by continent or incontinent conduit with a stoma [Citation17]. Each of these options is accompanied by considerable recovery time, operative risk, potential morbidity, and patient adjustment requirement.

As a result of the toxicity associated with radical surgical intervention, bladder-preservation approaches have been developed and have increased in utilisation in recent years. As previously alluded to, TURBT with maximal safe resection followed by intravesical chemotherapy remains an option to preserve the bladder in non-muscle-invasive disease. This approach has shown efficacy in reducing recurrence rates, especially in previous meta-analysis data [Citation18–22]. However, in muscle-invasive disease or high risk/multiply recurrent non-muscle-invasive disease, the current standard of care alternative to cystectomy is known as trimodality therapy and requires maximal safe TURBT followed by chemoradiation.

The current format of bladder preservation therapy

Trimodality therapy (TURBT, radiotherapy, and chemotherapy) represents a somewhat daunting regimen for patients, but also a vast improvement in efficacy over TURBT alone or TURBT with unimodal adjuvant radiotherapy. Historically, in patients who were poor candidates for cystectomy or who were motivated to preserve natural bladder function, maximal safe TURBT could be followed by consolidative radiotherapy. However, multiple series have demonstrated an improvement in disease control with the further addition of concurrent chemotherapy [Citation23–26]. The rationale for this approach is straightforward. Bladder cancer has a high proclivity for systemic dissemination, as autopsy and clinical series have evinced [Citation27]. The addition of chemotherapy especially in systemic doses, as allowed by cisplatin administration, during the course of local therapy thus has clear advantages. Also, the agents commonly utilised are effective radiosensitisers, and this is demonstrated well by the increased local control and bladder preservation rates in the aforementioned trials [Citation28]. The most common technique applies cisplatin chemotherapy concurrent with a mid-60s Gy dose of radiation, given in conventional fractionation, with or without split course to investigate response.

Though various radiation schedules and chemotherapy regimens have been employed, it is important to note that trimodality approaches have shown excellent disease response rates in large, prospective phase III series, which were designed to test various bladder preservation strategies [Citation24,Citation29–31]. With prompt salvage cystectomy only as indicated, trimodality therapy yields 5-year overall survival rates between 48–60%, relatively comparable to historical cystectomy series, with significantly improved toxicity profile [Citation32–39]. Complete response rates have ranged from 60–93% in most large series [Citation32–39]. Though there is a lack of head-to-head comparison between bladder preservation and cystectomy strategies, the outcomes, by most assessments, remain similar.

In late stage, locally advanced disease, systemic dissemination may drive survival primarily, but there remains a large demographic of patients who would vastly benefit from further improved complete response rates and bladder preservation rates [Citation24,Citation29,Citation31,Citation40–42].

Quadrimodality therapy

The rationale for thermoradiotherapy

Considering the rates of progression in high-risk T1 disease and the rates of salvage cystectomy (recurrence) in T2 disease, there are evident avenues for improvement of the bladder preservation approach. As this option increases in utilisation and further proves its viability in larger series, it is key that the therapeutic ratio be maximised in favour of efficacy and reduced toxicity to ensure that patients receive optimal care with the greatest chance of preserving their bladders.

With doses relatively maximised for chemotherapy and radiation in this disease, utilising currently widely available technologies and drugs, hyperthermia has arisen as a potential and promising fourth modality for attacking this locally aggressive carcinoma.

Hyperthermia has been recognised as a potential oncological therapeutic strategy since the 1950s. As accompanying chapters in this volume have detailed, hyperthermia alone has anti-tumoural effects by multiple mechanisms including direct and immune-based cytotoxicity. Perhaps most strikingly though, hyperthermia shows a clear synergistic response with selected chemotherapeutic agents – including several routinely utilised in bladder cancer – and especially with radiation therapy through its physiological ramifications. These effects can often elicit tumoural responses greater than the sum of the expected effects of each modality. As temperatures rise within locally heated tissues, blood perfusion increases. Initially, tissue thermoregulation limits local heating, but this generally exhibits a threshold response that may be overcome [Citation43]. Vascularity in and around the tumour becomes more permeable, leading to oedema, vascular stasis, and sometimes haemorrhage. This can result in relatively preferential delivery of drug or nanoparticles into tumour parenchyma.

Aerobic metabolism may be slowed due to greater heat sensitivity as compared to anaerobic mechanisms [Citation44]. Shifting metabolism thusly logically results in increased available unused intratumoural oxygen. Reoxygenation is key in propagating the indirect effects of radiotherapy which are effected through the creation of DNA-damaging oxygen radicals within the target [Citation45]. In fact, hypoxic cells have been estimated to be three times more resistant to radiotherapy than aerobic cells. The range of achievable clinical temperatures in most tumours without significant patient discomfort is particularly suited to eliciting a reoxygenation effect. Evidence from human tumour xenograft testing and canine soft tissue sarcomas () has suggested that vascular damage reverses the oxygenation benefit over 44 °C and that temperatures between 41 and 43 °C may be better for exploiting this effect [Citation45,Citation46]. Though there are varying reports as to how intratumoural oxygen levels vary during the course of thermoradiotherapy, clinical response evidence from breast cancer and sarcoma studies seems to correlate hyperthermia-induced reoxygenation with improved outcomes [Citation46–49]. In fact, reoxygenation after the first fraction of hyperthermia seems to be relatively predictive of pathological and clinical response rates [Citation50]. Especially in the post-operative setting, a prerequisite in the bladder preservation approach, oxygenation may be particularly lacking due to decreased perfusion from disturbed vasculature. Many patients may have progressed from previously non-muscle-invasive disease and thus may have undergone multiple surgeries with subsequent recurrences. Hyperthermia offers logical potential advantages in this setting to improve oxygenation and thus tumour radiosensitivity.

Figure 1. The hypoxic fraction may be significantly reduced utilising moderate hyperthermia (HT) concurrent with radiotherapy as demonstrated in canine soft tissue sarcomas by Vujaskovic et al. [Citation46] (data replotted with permission). However, temperatures above 44 °C seem to elicit worsening hypoxia.

Figure 1. The hypoxic fraction may be significantly reduced utilising moderate hyperthermia (HT) concurrent with radiotherapy as demonstrated in canine soft tissue sarcomas by Vujaskovic et al. [Citation46] (data replotted with permission). However, temperatures above 44 °C seem to elicit worsening hypoxia.

Finally, cell-cycle synergism also seems to play a role in the effectiveness of thermoradiotherapy based on in vitro findings. Radiation therapy is classically most tumouricidal during the G2 and mitotic (M) phases of the cell cycle. For hyperthermia, nuclear fragility lends greater thermosensitivity during the synthesis (S) and M phases, likely secondary to the increased expression of adaptive heat-shock proteins during the gap (G) phases [Citation51]. However, when combined, hyperthermia may induce the S phase into a more radiosensitive mode. In vitro studies have suggested that therapeutic hyperthermia inhibits DNA-polymerases α and β, staving off cellular repair mechanisms and converting sub-lethal radiation changes to lethal damage [Citation52,Citation53].

Hyperthermia and radiation in bladder cancer

Hyperthermia has been successfully and safely integrated into organ-sparing techniques for multiple disease sites and malignancies with excellent results. Several trials have utilised hyperthermia in conjunction with chemoradiotherapy and thus are clear correlates for the potential in bladder cancer. This combined approach has been employed successfully in head and neck cancer, melanoma, breast cancer, gliomas, cervical cancer, and rectal cancer amongst others, with excellent toxicity profiles and tumoural responses [Citation54].

There is relatively limited data regarding the concurrent application of hyperthermia and radiation in the treatment of bladder cancer. The technical demands of deep hyperthermia have deterred many clinics with superficial hyperthermia capabilities from adopting deep treatment units. Furthermore, the relatively slow uptake of bladder-sparing techniques that employ radiotherapy in regular clinical practice has stemmed the flow of patients eligible for such an intervention. Finally, methods for accurate heat monitoring and effective hyperthermia dose estimation have proven challenging and elusive. Only recently have enhanced thermal therapy planning systems in conjunction with newer intravesicular and MR-based thermography monitoring techniques improved and simplified this process () [Citation54]. Despite these hurdles, several important studies and two key prospective trials have been completed with promising results favouring thermoradiotherapy in bladder cancer ().

Figure 2. Treatment workflow for delivering bladder hyperthermia including simulation (a), hyperthermia modelling and planning (b), plan evaluation (c), and deep hyperthermia delivery with temperature monitoring (d). (Reproduced from Datta et al. [Citation54] with permission.)

Figure 2. Treatment workflow for delivering bladder hyperthermia including simulation (a), hyperthermia modelling and planning (b), plan evaluation (c), and deep hyperthermia delivery with temperature monitoring (d). (Reproduced from Datta et al. [Citation54] with permission.)

Table 1. Selected experiences utilising radiation therapy with concurrent hyperthermia in bladder cancer as well as intravesical and systemic therapies.

Several Japanese institutions have published their experiences with thermoradiotherapy in bladder cancer from the late 1970s through the early 1990s. Matsui et al. described a Yokohama City University series in 1991 [Citation55]. Fifty-six patients were included in this report. Hyperthermia was delivered by warm water irrigation with bleomycin intravesical chemotherapy incorporated in the irrigation solution. Patients receiving 40 Gy over 4 weeks with concurrent hyperthermia were compared with those receiving radiation alone to between 50 and 70 Gy. Among patients with T2 to T3 disease, combination therapy produced an 84% response rate compared to 56% with radiation alone. Combination therapy was noted to cause some reduction in bladder capacity, though this tended to be reversible. The higher doses of radiotherapy in the monotherapy cohort, instead, often caused irreversible reductions in bladder capacity. The authors very reasonably concluded that concurrent hyperthermia seemed to improve the therapeutic ratio, enhancing efficacy while decreasing toxicity.

This group had previously published their experience through a protocol for treating patients who were either deemed poor surgical candidates, were scheduled for post-therapy cystectomy, or were assessed to be poor TURBT candidates based on multifocal disease [Citation56]. Patients received between 35 and 40 Gy utilising a 60Co machine over 4 weeks. Hyperthermia was administered 12–16 times for 1 h each session at 42–43 °C with intravesical bleomycin. Patients primarily had T1 to T3 disease at diagnosis, except one T4 patient. In total, 73% of patients experienced at least a partial response, with 42% undergoing complete response. Of the eight patients taken to cystectomy shortly after therapy, final pathology demonstrated five of these to have complete responses and the rest to have partial responses. All of the complete responses were in T1 to T2 grade 3 patients. The partial responses included one T2 and two T3 patients each again with grade 3 disease.

Takechi et al. described their employment of a hypofractionated, low dose course utilising 3 Gy per fraction radiotherapy in conjunction with intravesical pirarubicin and hyperthermia [Citation57]. Five patients were treated in this small series with a total radiation dose goal of 15 Gy with pretreatment administration of pirarubicin and post-treatment hyperthermia daily of 42–43 °C for 35 min utilising an 8 MHz radiofrequency machine. One patient discontinued therapy secondary to urinary irritation, three achieved durable complete response, and one recurred by cytological examination.

A similar radiation regimen with 24 Gy in 4 Gy per fraction, three fractions per week, was employed with or without hyperthermia in a phase I/II neoadjuvant thermoradiotherapy trial at Kyoto University [Citation58]. With 49 patients with T1–T4 node-negative disease enrolled, 28 patients underwent combined therapy while 21 were given radiation alone. Regional hyperthermia was delivered twice weekly for four total treatments, each lasting 35 to 60 min. Patients receiving hyperthermia were stratified by average intravesical temperature above or below 41.5 °C. The higher temperature group demonstrated improved tumour response and downstaging, demonstrating that the quality of hyperthermia is of vital importance with this approach. This also suggests a rationale for hyperthermia and radiotherapy as part of a preoperative strategy for downstaging.

In 1992 Noguchi et al. published a series employing a neoadjuvant regimen that included hyperthermia and radiation with or without systemic chemotherapy in patients with Ta–T4 bladder tumours [Citation59]. M-VAC (methotrexate, vinblastine, Adriamycin, and cisplatin) was the chemotherapeutic course in this study, and it was dosed sequentially after neoadjuvant thermoradiotherapy, and adjuvantly after cystectomy. Seventeen patients were given all three neoadjuvant/adjuvant modalities, while 18 received radiation and hyperthermia only. Radiotherapy was delivered as 40 Gy in 10 fractions, twice weekly, with hyperthermia given again with an 8-MHz device. In the radiation and hyperthermia group, 12/18 patients presented with T2 or greater disease, while all but one (16/17) did so in the chemotherapy cohort. There were 12 patients with T3–T4 disease overall. Each patient’s disease was either grade 2 or 3, with the majority high grade. With seven patients unevaluable, the authors reported 64% and 79% (p not significant) response rates at cystectomy with and without chemotherapy, respectively, with each arm having four complete responses. Subcutaneous fat necrosis was experienced in about a sixth of patients and was attributed to hyperthermia. Otherwise, toxicities included tenesmus, diarrhoea, liver dysfunction, leucopenia, anaemia, and nausea/vomiting. Leucopenia, anaemia, and nausea/vomiting were clearly correlated with chemotherapy administration. It was suggested that earlier chemotherapy or concurrent regimens might prove more efficacious.

In the early 2000s, Ohguri et al. [Citation60] published a three-patient study finally utilising high dose radiotherapy (66–70 Gy), M-VAC chemotherapy, and hyperthermia (3–12 fractions) together in two patients with T2N1 disease and in one with T2N0. All three experienced complete responses and remained free of disease at 16, 18, and 40 months. Despite these promising small series, larger studies with more homogeneous patient cohorts and treatment protocols were lacking until two more concerted efforts were published out of Germany and the Netherlands.

Between 2003 and 2007, the Department of Radio-oncology at the University of Erlangen enrolled 45 patients on a prospective trial employing ‘quadrimodal treatment’ for high risk T1 and T2 bladder cancer [Citation61]. High risk T1 disease was defined as poorly differentiated tumours (grade 3), tumours with associated carcinoma in situ, multifocal disease, tumours with diameters greater than or equal to 5 cm, or tumours that had recurred/proven refractory following repeated TURBT with or without intravesical chemo/immunotherapy instillation. The treatment regimen employed maximal safe TURBT followed by chemoradiotherapy with concurrent deep hyperthermia. Radiotherapy was delivered by three-dimensional conformal radiotherapy (3D-CRT), usually with a traditional four-field box technique to a dose of 50.4 Gy in 1.8 Gy per fraction with an empty bladder set-up. A boost to between 55.8 Gy and 59.4 Gy was utilised dependent upon resection extent prior to therapy. Cisplatin and 5-FU chemotherapies were delivered in the first and fifth weeks of radiation. Hyperthermia was administered as regional deep hyperthermia with the BSD-2000 delivering electromagnetic microwaves from 90 to 100 MHz. Patients received between five and seven weekly hyperthermia treatments within 60 min before each fraction of radiation. Four probes were utilised for thermometry in the bladder cavity, rectum, anal fold, and oral cavity. Thermometry in the rectum and bladder were achieved through intraluminal insertions without invasive needles. Each administration was prescribed for 60–90 min at or above 41.5 °C. Cumulative equivalent minutes at 43 °C (CEM43) was calculated to assess thermal dose. Patients underwent restaging cystoscopy with resection of any residual disease at 6 weeks after completion of therapy. Persistence of tumour at this time-point was considered non-response, while absence of disease on sampling was considered pathological complete response (pCR). If urine cytology was negative as well, patients were observed with regular cystoscopy and cytology.

Wittlinger et al. reported results from this effort at a median follow-up of 34 months [Citation61]. Forty-three of the patients were able to receive chemotherapy with 60% undergoing full dose and all cycles. The median number of hyperthermia treatments was five with two-thirds receiving five or more. The mean CEM43 was 57 min with a mean cumulative overall hyperthermia treatment time of 387 min. Radiation and hyperthermia were very well tolerated with the most significant side effects stemming from chemotherapy-induced haematological toxicity.

At the 6-week mark, 43 of the 45 patients (96%) showed a pCR to this quadrimodal therapy. The two patients that did not experience a pCR had initially T1 and T2 disease. The T2 patient underwent salvage cystectomy with no subsequent recurrence of disease, though the patient died several months later of a non-small cell lung cancer. The T1 patient was managed with two additional TURBT procedures and intravesical therapy without further evidence of progression. This patient achieved local control for 40 months prior to passing of heart disease.

Of the 43 pCR patients, 80% (36 patients) have remained free of disease. The other seven patients with eventual local recurrence were restaged at the time of recurrence with Tx, Ta, or Tis disease. All of these patients were managed with TURBT and instillation therapy, therefore not requiring salvage cystectomy. Of the total nine deaths on the trial, five were related to bladder cancer, and all of these were attributable to distant disease. These patients each had grade 3 disease, and 4 of the 5 had initially T2 disease with R1 resections. For the entire group, disease-free survival and overall survival were 71% and 80% at 3 years. Statistically significant prognostic factors for survival were T stage (p = 0.04), extent of resection (p = 0.001), and whether the patient received at least five hyperthermia fractions (p = 0.036).

From a quality of life standpoint, 80% of the patients treated were at least ‘mostly satisfied’ with their urinary condition. One patient experienced treatment-refractory haematuria requiring cystectomy without evidence of disease. Common Terminology Criteria for Adverse Events (CTCAE) grade 3 and 4 toxicity was relatively low at 24%, mostly relating to reduced bladder capacity and occasional incontinence.

This trial is particularly thought-provoking and hypothesis-generating for future use of concurrent hyperthermia and radiotherapy in bladder cancer. Importantly, it included 26 patients with non-muscle-invasive disease. Compared to TURBT with intravesical chemotherapy, there is a substantial improvement with this approach in local control of T1 disease. It has been estimated that 50–70% of patients with Ta or T1 disease will have a recurrence after TURBT and intravesical chemotherapy [Citation62]. Wittlinger et al. reported only two patients (8%) with T1 disease at presentation who suffered either persistence or recurrence of disease [Citation61]. These results also improve upon those of intravesical chemotherapy given in conjunction with hyperthermia [Citation63–68]. While this approach is addressed in another chapter, this seems to be also a reasonable alternative with promising results.

Further, it is important to note that therapy was arguably not ideal on this trial. Patients received concurrent systemic therapy regardless of T stage or resection extent. In T1 patients it is conceivable that therapy could be de-escalated from this approach with still adequate local control, though further investigation is required for this hypothesis. Additionally, there was a statistically significant improvement in survival in patients receiving at least five treatments of hyperthermia. Twice-weekly hyperthermia treatment has become more common and could be explored as a way to intensify local therapy with limited additional toxicity expected. Finally, the radiation dose delivered here was relatively low compared to bladder cancer standards today. Use of intensity modulated (IMRT) techniques or other modalities (e.g. proton therapy) could allow for higher dose delivery with lower or at least similar toxicity.

Another source of evidence with a similar approach came earlier from the Dutch Deep Hyperthermia Group in 2000 [Citation69]. Between 1990 and 1996, 358 patients were enrolled on a multicentre trial in the Netherlands. Patients with T2, T3, and T4 non-metastatic bladder cancer were included, though this trial also enrolled patients with cervical and rectal cancers. In total, 101 patients with bladder cancer were included, though most had T3 or T4 disease (T2 10 patients, T3 41 patients, T4 50 patients). All of these patients received 66–70 Gy of radiotherapy including a larger field encompassing regional draining lymphatics to 40 Gy. Patients were randomised as to whether to receive concurrent hyperthermia.

Hyperthermia in this trial was delivered on a once-weekly basis with institutionally variable applicators. It was delivered between 1 and 4 h after radiotherapy for a duration of 60 to 90 min with a goal of 60 min above 42 °C. The primary end points of this trial were complete response and local control. Of the 182 patients in the hyperthermia group, 135 (74%) were able to undergo at least four hyperthermia sessions, again speaking to the tolerability of this modality.

For bladder cancer patients, 73% (38 of 52 patients) and 51% (25 of 49 patients) (p = 0.01) showed complete responses to initial therapy with and without hyperthermia, respectively. At 3 years, local control was 42% and 33% (p not significant) in these two groups. However, it is important to note that 13 patients experienced early distant failure without further local investigation and thus were counted as local failures without direct evidence. This convention could very well explain the lack of difference in local control considering the small size of the bladder cancer cohort in this study. Overall survival was not different based on application of hyperthermia in bladder cancer (28% versus 22%, p = 0.33), though there was a large difference in cervical cancer (51% versus 27%, p = 0.009).

Treatment was well tolerated in this trial, though there were two grade 5 toxicities (one with and one without hyperthermia). Acute grade 3–4 toxicity was limited (4%) with slightly greater prevalence in the radiotherapy alone arm. Late toxicity was similar between the arms.

Results from this effort are, again, promising for the addition of hyperthermia in the management of muscle-invasive and even locally advanced bladder cancer. However, there may be several methods by which to further improve outcomes. It is likely that a well-selected cohort would benefit more from aggressive local approaches. The inclusion of higher stage disease patients in this trial may have masked significant differences based on higher rates of occult dissemination at presentation. Adding systemic chemotherapy to this regimen would likely lessen distant metastasis rates as well as intensify local therapy. Additionally, the administration of more hyperthermia sessions may further contribute to tumoural response rates and disease control.

Improved radiotherapy techniques with concurrent hyperthermia

Hyperthermia’s increased utilisation in recent years concurrent with radiotherapy has followed improvements in the delivery of radiation and techniques for treatment. Certainly, 3-dimensional planning approaches have been vital in improving the sparing of normal tissue while escalating tumoural/target dose. IMRT has permitted radiation oncologists to sculpt dose more sharply, especially reducing the high dose exposure to nearby critical structures. This has proven particularly useful in the pelvis where the rectum, bowel, uninvolved bladder, prostate, uterus, vagina, and genitalia are all at risk and in relatively close proximity.

Emerging technologies such as particle therapy may allow for even further improvements in the therapeutic ratio. Proton therapy has a rapidly expanding footprint especially in the USA and Europe. Proton radiotherapy has the added benefit of largely eliminating exit dose. With pencil-beam scanning techniques (also known as spot scanning), very conformal dose distributions may be achieved even above and beyond passive scattering proton techniques and IMRT. This further improvement may allow for dose escalation of radiotherapy without increased toxicity, paving the way for bladder-sparing approaches in higher burdens of disease or unresectable/non-operative patients [Citation70]. Additionally, it has been argued that the radiobiological equivalence (RBE) of proton therapy may be greatly and disproportionately augmented by the addition of hyperthermia [Citation71]. Potentially, this may raise its RBE to the level of carbon-ion therapy, which remains limited in its availability and challenging in its widespread implementation. It should be noted, however, that there are numerous challenges in delivering technically sound particle therapy to the bladder caused by the organ’s ability to shift within the body and to be variably filled.

Conclusions

Thermoradiotherapy, or concurrent hyperthermia and radiation therapy, has demonstrated promising results, though data is limited in bladder cancer. This represents an exceedingly well-tolerated regimen with little added toxicity over radiotherapy alone. Quadrimodality approaches may offer a particularly efficacious bladder-sparing option to patients with muscle-invasive or locally advanced disease.

Additional investigation is clearly indicated, preferably in the form of clinical trials. With the resurgence of interest in hyperthermia amongst radiation oncologists, this may now be logistically feasible. However, multi-institutional efforts will likely be required to achieve reasonable enrolment for the several questions to be asked. Hyperthermia in conjunction with radiation therapy should be examined in both non-muscle-invasive and muscle-invasive disease. It may also have roles in locally advanced disease, potentially as neoadjuvant therapy. Intermixed with improved radiotherapy techniques such as IMRT or proton radiotherapy, hyperthermia may show particular benefit.

Disclosure statement

The authors report no conflicts of interest. The authors alone are responsible for the content and writing of the paper.

References

  • Siegel RL, Miller KD, Jemal A. Cancer statistics, 2015. CA Cancer J Clin 2015;65:5–29.
  • Gattegno B. T1G3 bladder cancer: conservative management or cystectomy? Eur Urol 2004;45:399–400.
  • Serretta V, Pavone C, Ingargiola GB, Daricello G, Allegro R, Pavone-Macaluso M. TUR and adjuvant intravesical chemotherapy in T1G3 bladder tumors: recurrence, progression and survival in 137 selected patients followed up to 20 years. Eur Urol 2004;45:730–5.
  • Malavaud B. T1G3 bladder tumours: the case for radical cystectomy. Eur Urol 2004;45:406–10.
  • Thalmann GN, Markwalder R, Shahin O, Burkhard FC, Hochreiter WW, Studer UE. Primary T1G3 bladder cancer: organ preserving approach or immediate cystectomy? J Urol 2004;172:70–5.
  • Turner W. T1G3 bladder tumours: the case for conservative treatment. Eur Urol 2004;45:401–5.
  • Patard JJ, Rodriguez A, Leray E, Rioux-Leclercq N, Guillé F, Lobel B. Intravesical Bacillus Calmette-Guerin treatment improves patient survival in T1G3 bladder tumours. Eur Urol 2002;41:635–41.
  • Manoharan M, Soloway MS. Optimal management of the T1G3 bladder cancer. Urol Clin North Am 2005;32:133–45.
  • Shahin O, Thalmann GN, Rentsch C, Mazzucchelli L, Studer UE. A retrospective analysis of 153 patients treated with or without intravesical bacillus Calmette-Guerin for primary stage T1 grade 3 bladder cancer: recurrence, progression and survival. J Urol 2003;169:96–100.
  • Sylvester RJ, van der Meijden AP, Oosterlinck W, Witjes JA, Bouffioux C, Denis L, et al. Predicting recurrence and progression in individual patients with stage Ta T1 bladder cancer using EORTC risk tables: a combined analysis of 2596 patients from seven EORTC trials. Eur Urol 2006;49:466–5.
  • Clark PE, Agarwal N, Biagioli MC, Eisenberger MA, Greenberg RE, Herr HW, et al. Bladder cancer. J Natl Compr Canc Netw 2013;11:446–75.
  • Ark JT, Keegan KA, Barocas DA, Morgan TM, Resnick MJ, You C, et al. Incidence and predictors of understaging in patients with clinical T1 urothelial carcinoma undergoing radical cystectomy. BJU Int 2014;113:894–9.
  • Stenzl A, Cowan NC, De Santis M, Jakse G, Kuczyk MA, Merseburger AS, et al. The updated EAU guidelines on muscle-invasive and metastatic bladder cancer. Eur Urol 2009;55:815–25.
  • Madersbacher S, Hochreiter W, Burkhard F, Thalmann GN, Danuser H, Markwalder R, et al. Radical cystectomy for bladder cancer today – a homogeneous series without neoadjuvant therapy. J Clin Oncol 2003;21:690–6.
  • Hautmann RE, Gschwend JE, de Petriconi RC, Kron M, Volkmer BG. Cystectomy for transitional cell carcinoma of the bladder: results of a surgery only series in the neobladder era. J Urol 2006;176:486–92.
  • Rosenkrantz AB, Mussi TC, Melamed J, Taneja SS, Huang WC. Bladder cancer: utility of MRI in detection of occult muscle-invasive disease. Acta Radiol 2012;53:695–9.
  • Kassouf W, Hautmann RE, Bochner BH, Lerner SP, Colombo R, Zlotta A, et al. A critical analysis of orthotopic bladder substitutes in adult patients with bladder cancer: is there a perfect solution? Eur Urol 2010;58:374–83.
  • Böhle A, Bock PR. Intravesical bacille Calmette-Guérin versus mitomycin C in superficial bladder cancer: formal meta-analysis of comparative studies on tumor progression. Urology 2004;63:682–6.
  • Malmström PU, Sylvester RJ, Crawford DE, Friedrich M, Krege S, Rintala E, et al. An individual patient data meta-analysis of the long-term outcome of randomised studies comparing intravesical mitomycin C versus bacillus Calmette-Guérin for non-muscle-invasive bladder cancer. Eur Urol 2009;56:247–56.
  • Sylvester RJ, van der Meijden AP, Witjes JA, Kurth K. Bacillus Calmette-Guerin versus chemotherapy for the intravesical treatment of patients with carcinoma in situ of the bladder: a meta-analysis of the published results of randomized clinical trials. J Urol 2005;174:86–91.
  • Sylvester RJ, van der Meijden AP, Lamm DL. Intravesical bacillus Calmette-Guerin reduces the risk of progression in patients with superficial bladder cancer: a meta-analysis of the published results of randomized clinical trials. J Urol 2002;168:1964–70.
  • Lamm DL, Blumenstein BA, Crissman JD, Montie JE, Gottesman JE, Lowe BA, et al. Maintenance bacillus Calmette-Guerin immunotherapy for recurrent TA, T1 and carcinoma in situ transitional cell carcinoma of the bladder: a randomized Southwest Oncology Group Study. J Urol 2000;163:1124–9.
  • Coppin CM, Gospodarowicz MK, James K, Tannock IF, Zee B, Carson J, et al. Improved local control of invasive bladder cancer by concurrent cisplatin and preoperative or definitive radiation. The National Cancer Institute of Canada Clinical Trials Group. J Clin Oncol 1996;14:2901–7.
  • James ND, Hussain SA, Hall E, Jenkins P, Tremlett J, Rawlings C, et al. Radiotherapy with or without chemotherapy in muscle-invasive bladder cancer. N Engl J Med 2012;366:1477–88.
  • International Collaboration of Trialists; Medical Research Council Advanced Bladder Cancer Working Party (now the National Cancer Research Institute Bladder Cancer Clinical Studies Group); European Organisation for Research and Treatment of Cancer Genito-Urinary Tract Cancer Group; Australian Bladder Cancer Study Group; National Cancer Institute of Canada Clinical Trials Group; Finnbladder, et al. International phase III trial assessing neoadjuvant cisplatin, methotrexate, and vinblastine chemotherapy for muscle-invasive bladder cancer: long-term results of the BA06 30894 trial. J Clin Oncol 2011;29:2171–7.
  • Pollack A, Zagars GK, Swanson DA. Muscle-invasive bladder cancer treated with external beam radiotherapy: prognostic factors. Int J Radiat Oncol Biol Phys 1994;30:267–77.
  • Stein JP, Lieskovsky G, Cote R, Groshen S, Feng AC, Boyd S, et al. Radical cystectomy in the treatment of invasive bladder cancer: long-term results in 1,054 patients. J Clin Oncol 2001;19:666–75.
  • Bentzen SM, Harari PM, Bernier J. Exploitable mechanisms for combining drugs with radiation: concepts, achievements and future directions. Nat Clin Pract Oncol 2007;4:172–80.
  • Tunio MA, Hashmi A, Qayyum A, Mohsin R, Zaeem A. Whole-pelvis or bladder-only chemoradiation for lymph node-negative invasive bladder cancer: single-institution experience. Int J Radiat Oncol Biol Phys 2012;82:e457–62.
  • Shipley WU, Winter KA, Kaufman DS, Lee WR, Heney NM, Tester WR, et al. Phase III trial of neoadjuvant chemotherapy in patients with invasive bladder cancer treated with selective bladder preservation by combined radiation therapy and chemotherapy: initial results of Radiation Therapy Oncology Group 89-03. J Clin Oncol 1998;16:3576–83.
  • Housset M, Maulard C, Chretien Y, Dufour B, Delanian S, Huart J, et al. Combined radiation and chemotherapy for invasive transitional-cell carcinoma of the bladder: a prospective study. J Clin Oncol 1993;11:2150–7.
  • Efstathiou JA, Spiegel DY, Shipley WU, Heney NM, Kaufman DS, Niemierko A, et al. Long-term outcomes of selective bladder preservation by combined-modality therapy for invasive bladder cancer: the MGH experience. Eur Urol 2012;61:705–11.
  • Mitin T, Hunt D, Shipley WU, Kaufman DS, Uzzo R, Wu CL, et al. Transurethral surgery and twice-daily radiation plus paclitaxel-cisplatin or fluorouracil-cisplatin with selective bladder preservation and adjuvant chemotherapy for patients with muscle invasive bladder cancer (RTOG 0233): a randomised multicentre phase 2 trial. Lancet Oncol 2013;14:863–72.
  • Kotwal S, Choudhury A, Johnston C, Paul AB, Whelan P, Kiltie AE. Similar treatment outcomes for radical cystectomy and radical radiotherapy in invasive bladder cancer treated at a United Kingdom specialist treatment center. Int J Radiat Oncol Biol Phys 2008;70:456–63.
  • Hautmann RE, de Petriconi RC, Pfeiffer C, Volkmer BG. Radical cystectomy for urothelial carcinoma of the bladder without neoadjuvant or adjuvant therapy: long-term results in 1100 patients. Eur Urol 2012;61:1039–47.
  • Culp SH, Dickstein RJ, Grossman HB, Pretzsch SM, Porten S, Daneshmand S, et al. Refining patient selection for neoadjuvant chemotherapy before radical cystectomy. J Urol 2014;191:40–7.
  • Kachnic LA, Kaufman DS, Heney NM, Althausen AF, Griffin PP, Zietman AL, et al. Bladder preservation by combined modality therapy for invasive bladder cancer. J Clin Oncol 1997;15:1022–9.
  • Kaufman DS, Winter KA, Shipley WU, Heney NM, Chetner MP, Souhami L, et al. The initial results in muscle-invading bladder cancer of RTOG 95-06: phase I/II trial of transurethral surgery plus radiation therapy with concurrent cisplatin and 5-fluorouracil followed by selective bladder preservation or cystectomy depending on the initial response. Oncologist 2000;5:471–6.
  • Weiss C, Engehausen DG, Krause FS, Papadopoulos T, Dunst J, Sauer R, et al. Radiochemotherapy with cisplatin and 5-fluorouracil after transurethral surgery in patients with bladder cancer. Int J Radiat Oncol Biol Phys 2007;68:1072–80.
  • Lagrange JL, Bascoul-Mollevi C, Geoffrois L, Beckendorf V, Ferrero JM, Joly F, et al. Quality of life assessment after concurrent chemoradiation for invasive bladder cancer: results of a multicenter prospective study (GETUG 97-015). Int J Radiat Oncol Biol Phys 2011;79:172–8.
  • Gogna NK, Matthews JH, Turner SL, Mameghan H, Duchesne GM, Spry N, et al. Efficacy and tolerability of concurrent weekly low dose cisplatin during radiation treatment of localised muscle invasive bladder transitional cell carcinoma: a report of two sequential phase II studies from the Trans Tasman Radiation Oncology Group. Radiother Oncol 2006;81:9–17.
  • Zapatero A, Martin De Vidales C, Arellano R, Ibañez Y, Bocardo G, Perez M, et al. Long-term results of two prospective bladder-sparing trimodality approaches for invasive bladder cancer: neoadjuvant chemotherapy and concurrent radio-chemotherapy. Urology 2012;80:1056–62.
  • Dewhirst MW, Sim D, Gross J. Effects of heating rate on normal and tumor microcirculatory function. In: Diller K, Roemer RB, eds. Heat and mass transfer in the microcirculation of thermally significant vessels. Anaheim, CA: ASME 1986:75–80.
  • Streffer C. Metabolic changes during and after hyperthermia. Int J Hyperthermia 1985;1:305–19.
  • Oleson JR. Eugene Robertson Special Lecture. Hyperthermia from the clinic to the laboratory: a hypothesis. Int J Hyperthermia 1995;11:315–22.
  • Vujaskovic Z, Poulson JM, Gaskin AA, Thrall DE, Page RL, Charles HC, et al. Temperature-dependent changes in physiologic parameters of spontaneous canine soft tissue sarcomas after combined radiotherapy and hyperthermia treatment. Int J Radiat Oncol Biol Phys 2000;46:179–85.
  • 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;37:155–62.
  • Brizel DM, Scully SP, Harrelson JM, Layfield LJ, Dodge RK, Charles HC, et al. Radiation therapy and hyperthermia improve the oxygenation of human soft tissue sarcomas. Cancer Res 1996;56:5347–50.
  • 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;22:365–73.
  • Jones EL, Prosnitz LR, Dewhirst MW, Marcom PK, Hardenbergh PH, Marks LB, et al. Thermochemoradiotherapy improves oxygenation in locally advanced breast cancer. Clin Cancer Res 2004;10:4287–93.
  • Vaupel P, Kallinowski F, Kluge M. Pathophysiology of tumors in hyperthermia. Recent Results Cancer Res 1988;107:65–75.
  • Dewey WC, Hopwood LE, Sapareto SA, Gerweck LE. Cellular responses to combinations of hyperthermia and radiation. Radiology 1977;123:463–74.
  • Oei AL, Vriend LE, Crezee J, Franken NA, Krawczyk PM. Effects of hyperthermia on DNA repair pathways: one treatment to inhibit them all. Radiat Oncol 2015;10:165.
  • Datta NR, Ordóñez SG, Gaipl US, Paulides MM, Crezee H, Gellermann J, et al. Local hyperthermia combined with radiotherapy and-/or chemotherapy: Recent advances and promises for the future. Cancer Treat Rev 2015;41:742–53.
  • Matsui K, Takebayashi S, Watai K, Kakehi M, Kubota Y, Yao M, et al. Combination radiotherapy of urinary bladder carcinoma with chemohyperthermia. Int J Hyperthermia 1991;7:19–26.
  • Kubota Y, Shuin T, Miura T, Nishimura R, Fukushima S, Takai S. Treatment of bladder cancer with a combination of hyperthermia, radiation and bleomycin. Cancer 1984;53:199–202.
  • Takechi T, Kawamura M, Nakagawa H, Inatsuki S, Akiyama K, Yano M, et al. Combined treatment in patients with carcinoma in situ of the urinary bladder using intravesical pirarubicin, irradiation and hyperthermia. Nihon Gan Chiryo Gakkai Shi 1990;25:2724–7.
  • Masunaga SI, Hiraoka M, Akuta K, Nishimura Y, Nagata Y, Jo S, et al. Phase I/II trial of preoperative thermoradiotherapy in the treatment of urinary bladder cancer. Int J Hyperthermia 1994;10:31–40.
  • Noguchi S, Kubota Y, Miura T, Shuin T, Hosaka M. Use of methotrexate, vinblastine, Adriamycin, and cisplatin in combination with radiation and hyperthermia as neo-adjuvant therapy for bladder cancer. Cancer Chemother Pharmacol 1992;30:S63–5.
  • Ohguri T, Imada H, Nomoto S, Kato F, Yahara K, Morioka T, et al. Initial experience of bladder preservation therapy using chemoradiotherapy with regional hyperthermia for muscle-invasive bladder cancer. Jpn J Hyperthermic Oncol 2005:21:151–8.
  • Wittlinger M, Rödel CM, Weiss C, Krause SF, Kühn R, Fietkau R, et al. Quadrimodal treatment of high-risk T1 and T2 bladder cancer: transurethral tumor resection followed by concurrent radiochemotherapy and regional deep hyperthermia. Radiother Oncol 2009;93:358–63.
  • McDougal WS, Shipley WU, Kaufman DS, Dahl DM, Michaelson MD, Zietman AL. Cancer of the bladder, ureter, and renal pelvis. In: DeVita VT, Hellman S, Rosenberg SA, eds. Cancer: Principles and Practice of Oncology, 9th ed. Philadelphia: Lippincott Williams & Wilkins; 2011, pp. 1192–211.
  • Colombo R, Da Pozzo LF, Lev A, Freschi M, Gallus G, Rigatti P. Neoadjuvant combined microwave induced local hyperthermia and topical chemotherapy versus chemotherapy alone for superficial bladder cancer. J Urol 1996;155:1227–32.
  • Gofrit ON, Shapiro A, Pode D, Sidi A, Nativ O, Leib Z, et al. Combined local bladder hyperthermia and intravesical chemotherapy for the treatment of high-grade superficial bladder cancer. Urology 2004;63:466–71.
  • Colombo R, Salonia A, Da Pozzo LF, Naspro R, Freschi M, Paroni R, et al. Combination of intravesical chemotherapy and hyperthermia for the treatment of superficial bladder cancer: preliminary clinical experience. Crit Rev Oncol Hematol 2003;47:127–39.
  • Moskovitz B, Meyer G, Kravtzov A, Gross M, Kastin A, Biton K, et al. Thermo-chemotherapy for intermediate or high-risk recurrent superficial bladder cancer patients. Ann Oncol 2005;16:585–9.
  • Colombo R, Da Pozzo LF, Salonia A, Rigatti P, Leib Z, Baniel J, et al. Multicentric study comparing intravesical chemotherapy alone and with local microwave hyperthermia for prophylaxis of recurrence of superficial transitional cell carcinoma. J Clin Oncol 2003;21:4270–6.
  • Van der Heijden AG, Kiemeney LA, Gofrit ON, Nativ O, Sidi A, Leib Z, et al. Preliminary European results of local microwave hyperthermia and chemotherapy treatment in intermediate or high risk superficial transitional cell carcinoma of the bladder. Eur Urol 2004;46:65–71.
  • Van der Zee J, González González D, 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(9210):1119–25.
  • Hata M, Miyanaga N, Tokuuye K, Saida Y, Ohara K, Sugahara S, et al. Proton beam therapy for invasive bladder cancer: a prospective study of bladder-preserving therapy with combined radiotherapy and intra-arterial chemotherapy. Int J Radiat Oncol Biol Phys 2006;64:1371–9.
  • Datta NR, Puric E, Schneider R, Weber DC, Rogers S, Bodis S. Could hyperthermia with proton therapy mimic carbon ion therapy? Exploring a thermo-radiobiological rationale. Int J Hyperthermia 2014;30:524–30.

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.