5,536
Views
41
CrossRef citations to date
0
Altmetric
Review

The clinical benefit of hyperthermia in pancreatic cancer: a systematic review

, , , , , , , , , ORCID Icon, , ORCID Icon & show all
Pages 969-979 | Received 23 Dec 2016, Accepted 30 Oct 2017, Published online: 23 Nov 2017

Abstract

Objective: In pancreatic cancer, which is therapy resistant due to its hypoxic microenvironment, hyperthermia may enhance the effect of radio(chemo)therapy. The aim of this systematic review is to investigate the validity of the hypothesis that hyperthermia added to radiotherapy and/or chemotherapy improves treatment outcome for pancreatic cancer patients.

Methods and materials: We searched MEDLINE and Embase, supplemented by handsearching, for clinical studies involving hyperthermia in pancreatic cancer patients. The quality of studies was evaluated using the Oxford Centre for Evidence-Based Medicine levels of evidence. Primary outcome was treatment efficacy; we calculated overall response rate and the weighted estimate of the population median overall survival (mp) and compared these between hyperthermia and control cohorts.

Results: Overall, 14 studies were included, with 395 patients with locally advanced and/or metastatic pancreatic cancer of whom 248 received hyperthermia. Patients were treated with regional (n = 189), intraoperative (n = 39) or whole-body hyperthermia (n = 20), combined with chemotherapy, radiotherapy or both. Quality of the studies was low, with level of evidence 3 (five studies) and 4. The six studies including a control group showed a longer mp in the hyperthermia groups than in the control groups (11.7 vs. 5.6 months). Overall response rate, reported in three studies with a control group, was also better for the hyperthermia groups (43.9% vs. 35.3%).

Conclusions: Hyperthermia, when added to chemotherapy and/or radiotherapy, may positively affect treatment outcome for patients with pancreatic cancer. However, the quality of the reviewed studies was limited and future randomised controlled trials are needed to establish efficacy.

Introduction

The prognosis for pancreatic cancer is very poor and has scarcely improved over the last two decades, with a 5-year overall survival of less than 10% [Citation1,Citation2]. For (borderline) resectable pancreatic cancer, the standard treatment is exploratory laparotomy, with resection and adjuvant chemotherapy when a resection was performed [Citation3–5]. The impact of neoadjuvant chemoradiotherapy is under investigation; its suggested beneficial effect on overall survival has been studied in two randomised trials that were terminated prematurely [Citation6,Citation7], as well as in one recently completed randomised multicenter trial [Citation8]. However, the vast majority of patients present with either locally advanced or metastatic pancreatic cancer [Citation9]. Also, most patients develop recurrent disease after surgery. For both groups, chemotherapy is the treatment of choice [Citation10].

While chemotherapy and/or radiotherapy are successful in many tumour types, pancreatic cancer remains quite resistant to these conventional therapies. This resistance is in large part due to the abundant and dense accumulation of nontumour cells known as the stroma, which obstructs vascularisation and the delivery of chemotherapeutics. In addition, it hampers the delivery of oxygen, causing hypoxia [Citation11]. Such a hypoxic microenvironment diminishes sensitivity to chemotherapeutics and radiation and negatively impacts prognosis [Citation12–16].

To improve outcome, new approaches are being explored. For clinically fit patients with locally advanced (LAPC) or metastatic pancreatic cancer, the multiagent regimen FOLFIRINOX (i.e. leucovorin, fluorouracil, irinotecan and oxaliplatin) has shown promising results [Citation17,Citation18]. The combination of gemcitabine and nab-paclitaxel showed improved survival over gemcitabine monotherapy in patients with metastatic pancreatic cancer [Citation19,Citation20]. For localised disease, encouraging results from stereotactic body radiation therapy (SBRT), radiofrequency ablation (RFA) and irreversible electroporation (IRE) have been reported, though evidence from randomised trials is still lacking [Citation21–24]. However, the aforementioned treatments are often taxing to the patient, have high toxicity rates, or are still lacking evidence of efficacy. Therefore, development of treatment options that may directly reduce resistance to the conventional therapies is an attractive approach, for example, by counteracting the negative impact of the tumour stroma.

Mild hyperthermia (HT), that is, raising the temperature of the tumour to approximately 40–44 °C, typically for one hour, acts as a sensitiser for radiotherapy and chemotherapy [Citation25–27]. In this temperature range, improved drug delivery and dispersion in the tumour enhances the effect of chemotherapy [Citation28–30]. With improved blood flow, hypoxia is decreased, which further enhances radiation effects through an increased production of oxygen radicals. In addition, hyperthermia inhibits DNA repair, thus enhancing tumour cell killing [Citation31,Citation32].

Efficacy of HT, in combination with radiotherapy, chemotherapy or chemoradiotherapy, has been demonstrated for several tumour types [Citation26,Citation33,Citation34]. For pancreatic cancer, the efficacy of HT has been validated in the preclinical setting. For example, in subcutaneous pancreatic cancer xenografts in rats and mice, the combination of gemcitabine and HT was more effective than gemcitabine or HT alone [Citation35–37]. For pancreatic cancer patients, however, data are limited.

The objective of this systematic review is to investigate the literature on the effect of hyperthermia combined with radiotherapy and/or chemotherapy for pancreatic cancer. We aim to test the validity of the hypothesis that the addition of hyperthermia as a sensitiser to radiotherapy and/or chemotherapy for locally advanced and metastatic pancreatic cancer improves overall survival.

Methods

Execution of this systematic review was based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) standard guidelines [Citation38]. The review was registered at PROSPERO, the international prospective register of systematic reviews (registration number CRD42017073851).

Search strategy

MEDLINE and Embase were systematically searched from onset using the Ovid interface. Key concepts were “hyperthermia” and “pancreatic cancer”. For each concept, eligible synonyms and controlled terms were identified by a scoping search. The systematic search was supplemented with handsearching (). See Appendix A for a detailed display of the systematic Ovid search strategies.

Figure 1. PRISMA diagram. OS: overall survival; other language = Chinese, Japanese or Russian; a[Citation68] describes same study as included study [Citation57]. bcohort in [Citation67] largely overlaps with cohort in included study [Citation54].

Figure 1. PRISMA diagram. OS: overall survival; other language = Chinese, Japanese or Russian; a[Citation68] describes same study as included study [Citation57]. bcohort in [Citation67] largely overlaps with cohort in included study [Citation54].

Selection

Selection of articles was independently performed by two reviewers (EV and AVDH) using Rayyan, a web application to screen identified articles [Citation39]. In case of discrepancies, consensus was reached in deliberation with a third person (JC). The first selection was based on title and abstract; the second selection was based on the full text of the articles.

Inclusion criteria

Papers were included according to the following criteria: pancreatic cancer; study conducted in humans; number of patients >1; disease stage described; HT treatment aimed at a temperature in the range 40–45 °C in combination with radiotherapy and/or chemotherapy; treatment regimen described (type and dose of chemotherapy; radiation dose; type, temperature and duration of HT); outcome of HT treatment described in terms of overall survival (OS) and/or tumour response; text in English, German, French or Dutch language.

Exclusion criteria

Studies involving hyperthermic intraperitoneal chemotherapy (HIPEC) and thermal ablation were not included. Studies that included patients with a variety of carcinomas were only included when they reported on the subgroup of pancreatic cancer patients separately. Studies without a control cohort were excluded when the treatment regimen was different than commonly prescribed (e.g. combinations of compounds not in current clinical use). Since studies applying hyperthermia in pancreatic cancer were likely to be scarce, no particular level of evidence was required for inclusion.

Data extraction and analysis

For the selected studies, we assessed type of study, patient cohorts, delivered treatments, treatment efficacy and reported toxicity. Therefore, the following data were extracted from the articles (for the study group and, when applicable, also for the control group): study characteristics; number of patients; median age; tumour stage; tumour type and histological confirmation; previous treatments; treatment protocol (HT treatment regimen, complementing chemotherapy and/or radiotherapy treatment regimen); type of HT; type of temperature measurement; treatment protocol of control group, when available; tumour response; overall survival; reported side effects, grade ≥3. When the median overall survival was not stated, but a Kaplan–Meier survival plot was supplied, the median OS was obtained from that plot by determining the time on the x-axis at 50% survival rate.

Evaluation of study quality and risk of bias assessment

The quality of the included studies regarding treatment benefits of HT was evaluated according to the 2011 Oxford Centre for Evidence-Based Medicine (OCEBM) levels of evidence [Citation40].

In addition, we assessed risk of bias based on the following criteria: the presence of control group, randomness of allocation, comparability of prognostic parameters (age, stage, etc.) and follow-up, selective reporting and confounding by indication.

Patients characteristics and treatment regimens

To obtain insight into the patient cohorts included in the studies, we tabulated median patient age as well as tumour stage. For the latter, we distinguished between locally advanced (2012 AJCC stage III [Citation41]) and metastatic disease (stage IV). The definition of the AJCC staging has changed over the years. Therefore, when necessary and possible, we reported staging according to the current AJCC guidelines.

Most pancreatic cancers are pancreatic ductal adenocarcinomas (PDACs) [Citation42]. However, confirmation by means of histology is not always obtained. For each study, we assessed the number of PDAC patients and whether histological and/or cytological confirmation was obtained. As treatments may differ between studies, we presented treatment as well as previously received treatments.

Temperature measurements

In HT, the thermal dose and the resulting temperature obtained in the tumour are crucial. While radiosensitisation by increased perfusion and reoxygenation [Citation43,Citation44] can be observed already at 39 °C, the effect increases substantially with increasing temperature with more direct radiosensitisation due to the inhibition of DNA damage repair machinery [Citation45–47]. Also chemosensitisation starts at relatively low temperatures for many chemotherapy compounds, including drugs frequently used in pancreatic cancer, such as gemcitabine and fluorouracil [Citation48–51]. For other drugs, the effect appears to be additive and only effective at higher temperatures [Citation32].

Tissue heterogeneity, perfusion and the presence of (large) blood vessels can greatly affect local temperatures. Therefore, it is paramount that the temperature is properly monitored, preferably in the target volume. However, for pancreatic cancer intratumoral measurements are often not performed, due to the risks associated with such invasive measurements.

Here, we examined whether and how temperature was measured and whether these measurements were intratumoral. Furthermore, we determined per study the intended temperature as well as the achieved temperature or temperature range.

HT efficacy

To investigate efficacy of hyperthermia as adjuvant therapy in pancreatic cancer patients, we analysed response rate, reported as complete response (CR), partial response (PR), stable disease (SD) and progressive disease (PD). From this, we calculated the overall response rate over the studies as (CR + PR)/n, with n the combined total number of patients in those studies.

In addition, we examined the median OS. Unbiased pooled estimates of median survival times cannot be achieved by simple weighted averaging of medians; therefore, we calculated mp, a weighted estimate of the population median overall survival, using [Citation52]: (1) with mi the median OS of study population i, k the number of included studies, and ni/N the weight function with ni the number of patients in study i and N the total number of patients in studies 1–k. For the studies that included a control cohort, we calculated mp for the HT cohorts and the control cohorts. In addition, we calculated mp for the HT cohorts over all studies.

HT toxicity

We investigated whether toxicity was reported according to established guidelines and whether HT-related toxicity was separately reported. For each study, we registered the reported toxicity ≥ grade 3.

Results

Literature search

A total of 1293 potentially eligible articles were identified. After the first and second selection (), 14 studies were included [Citation53–66]. Two patient cohorts were each described in two articles; for these we excluded the oldest [Citation67,Citation68] and included the latest [Citation54,Citation57]. Based on the reported patient characteristics in [Citation56], it was evident there was no overlap of this patient cohort with the cohort in references [Citation57,Citation68]. One included article was written in German [Citation58]; all others were written in English.

The included 14 studies combined 395 pancreatic cancer patients of whom 248 received HT (). Six out of 14 studies used a control population that enabled a comparison between treatments with and without hyperthermia [Citation54,Citation55,Citation57,Citation60,Citation62,Citation66]. In two studies, patients with a variety of carcinomas were included, but data were reported for the pancreatic patients separately [Citation53,Citation59].

Table 1. Patient characteristics. Number of patients, median age, disease stage per study, fraction of adenocarcinomas and whether histological confirmation was obtained.

Study quality and risk of bias

None of the studies were randomised controlled trials; eight were retrospective studies. Six studies had a control group; in one of these studies, the control group was a cohort of patients treated ten years earlier [Citation55]. Thus, the OCEBM level of evidence was level 3 for five studies and level 4 for all others. Risks of bias are described in Supplementary Table S1.

Patient characteristics and treatment regimens

Most of the 395 pancreatic cancer patients had either LAPC (204 patients, 52%) or metastatic disease (175 patients, 44%); in two studies, a total of 16 patients had resectable or borderline resectable pancreatic cancer [Citation54,Citation58], . Seven studies included both patients with locally advanced and patients with metastatic disease. Not every study reported on previous treatments received by the patients. For the 201 patients who received hyperthermia in the 12 studies that did report on previously received treatment, 10% had received resection, 23% chemotherapy, 1% chemoradiotherapy and 66% had received no previous treatment before enrolling in a study. For the patients in the control cohorts, 21 of the 147 patients (14%) had received resection; all others had received none.

In the studies, a wide variety of treatments were used (), with differences in HT treatment (method, temperature, duration, number of sessions) and in (concurrent) chemotherapy (drug, dose) and radiotherapy (dose, fractionation). For the patients who received HT within the studies, this HT was in combination with radiotherapy for 17 patients (7%, two studies) [Citation53,Citation55], with chemoradiotherapy for 82 patients (33%, four studies) [Citation54,Citation60,Citation62,Citation66], and with chemotherapy for 149 patients (60%) in the remaining eight studies. Three types of hyperthermia treatment were applied: locoregional, whole-body and intraoperative hyperthermia.

Table 2. Treatment regimens.

Locoregional hyperthermia was used in eight studies, with heating of tumour and surrounding tissue either radiative (using radiofrequency waves; 63 patients) [Citation62,Citation64] or capacitive (using electrodes; 126 patients) [Citation53,Citation58,Citation60,Citation63,Citation65,Citation66]. Treatment times varied from 40–60 min.

Intraoperative hyperthermia (four studies, 39 patients) was performed using a radiofrequency device to locally heat the tumour and surrounding area for 40–60 min. during surgery [Citation54–57].

Finally, whole-body hyperthermia (two studies) involved heating the entire body to a temperature of 40 °C for 6 h (seven patients) [Citation59] or 41.8 °C for 1 h (13 patients) [Citation61] using a radiant heat device.

Temperature measurements

Temperature was measured during HT treatment in eight of the 14 studies (). Of these, only three studies measured temperature intratumorally [Citation54,Citation56,Citation57], all using (micro)thermocouples. In the other five studies, gauges were placed (relatively) nearby in, for example, duodenum, bladder or rectum. In the studies in which no temperature was measured, the authors relied on previously established optimal settings for their equipment.

Table 3. Temperature measurements during HT treatment. For each study, it is indicated whether and for how many patients the temperature was measured, how this was done, what the target temperature was and what temperatures were obtained.

Efficacy: response rate and overall survival

Response rate was reported in 11 studies (243 patients; ). Complete response was observed for six patients in three studies [Citation53,Citation58,Citation60]; all six had received HT. For the 11 studies, the combined overall response rate for the 192 patients treated with HT was 31.3% (6 CR, 54 PR). For the three studies that included a control group [Citation54,Citation57,Citation60], the response rate for 41 patients treated with hyperthermia was 43.9% (1 CR, 17 PR), compared with 35.3% (18 PR) in the 51 control patients.

Table 4. Tumour response.

Overall survival rates were available for 12 studies (); two of these studies did not describe median OS for each cohort but did supply Kaplan–Meier curves [Citation54,Citation66]. For the six studies with a control group, the weighted estimate of the population median overall survival mp was 11.7 months for the cohorts receiving HT (median OS range 6–18.6 months), compared with 5.6 months for the control cohorts (4–11 months). All six studies with control patients showed a longer median OS with HT treatment than without. For the 12 cohorts receiving HT for which median OS was available, mp was 10.5 months ().

Table 5. Median overall survival, for patients who received hyperthermia (HT cohort) and patients who did not (control cohort).

Reported toxicity

Twelve studies reported on toxicity (), of which 10 specified the toxicity criteria used (WHO criteria (2 studies) and a version of the NCI-CTCAE (8 studies)). Most studies did not make a distinction between HT-related toxicities and other toxicities. Grade 5 toxicity was not reported in any study; most reported grade 3/4 toxicities were hematologic, likely due to chemotherapy. The only severe hyperthermia-related toxicity was severe subcutaneous fatty burn following intraoperative hyperthermia in one patient [Citation54].

Table 6. Reported toxicity. For each study it is indicated which if any reporting guidelines were used and which graded (grade ≥3) or other toxicities were reported.

Discussion

This is the first systematic review investigating the potential benefit of hyperthermia added to radiotherapy and/or chemotherapy for pancreatic cancer patients. In this review, in which 14 studies were included, we found for HT combined with chemo(radio)therapy a median OS of 6–18.6 months and a combined overall response rate of 31.3%. Results were consistent, showing longer median overall survival for the cohorts treated with HT compared to the respective control cohorts. Also response, reported for three of the studies that included a control cohort, was better with HT added. These results suggest that HT, when added to chemotherapy and/or radiotherapy, may positively affect response and overall survival for patients with pancreatic cancer. However, quality of the studies was limited and none of the studies was randomised. As a consequence, we did not perform a full meta-analysis of the data. Hyperthermia can safely be used, as reported toxicity was limited, with no grade 5 toxicity and only one case of a hyperthermia-related grade 3 burn.

Apart from the possible benefit in response and overall survival, the addition of hyperthermia may also contribute to a better quality of life (QoL). One controlled study included QoL measurements and showed that patients who received hyperthermia had a significantly larger improvement in QoL and a significant reduction in pain compared with those who did not receive hyperthermia [Citation57]. Although these results are compelling, we did not include them in our analysis, as no other studies investigated the effects of hyperthermia on QoL. Including QoL as an endpoint in future studies will be highly valuable.

Thermal dosimetry

Knowing the actual temperature in the tumour during treatment is critical in HT, to verify that the therapeutically required temperature is obtained [Citation69]. However, in only three studies, temperature was measured intratumorally and in six studies temperature was not measured at all. Frequently, heating is decreased when patients report pain. This may bring the temperature below the required temperature, as reported by Yamada et al. [Citation54]; although their target temperature was 42.5 °C, only in five of the 17 patients the tumour was heated to more than the required 42 °C.

In this review, only one subcutaneous fatty burn was reported in one patient. Especially at the interface of muscle and fat, local hot spots can occur [Citation69]. Thus, heating methods that allow for local adjustment of the temperature distribution are preferred, such as radiative heating using a phased array of multiple antennas [Citation70]. Radiative heating also allows for HT treatment planning [Citation71–74], which may yield better temperature distributions and the possibility to avoid hot spots. With temperature being heterogeneous and not constant in time, reporting is preferably done using a set of thermal dose parameters [Citation69]. Such reporting allows for proper thermal dosimetry and enables (future) linking of thermal dose with outcome.

Limitations

The largest limitation of this review was the quality of the studies. All studies were cohort studies (prospective or retrospective); none of the studies were randomised controlled trials. Although the total number of included patients was moderate (n = 281), the individual studies were relatively small with the cohorts who received HT ranging from five to 40 patients. Only six out of 14 studies included a control group, but without randomisation, selection bias cannot be excluded. In addition, stratification according to tumour stage was not possible, as many studies included both patients with locally advanced and metastatic disease and did not report outcome per stage separately. The combining of locally advanced and metastatic disease may contribute to the large variation in outcome between studies.

Future work

The typically large stromal compartment of pancreatic cancer is often hypoxic, aiding tumour cells to escape conventional treatments. Hyperthermia can help to overcome the hypoxia, making those cancer cells more sensitive to radiotherapy and/or chemotherapy [Citation12]. Interestingly, although hypoxia is thought to play a large role in the efficacy of HT, none of the studies included in our systematic review reported on the hypoxic state of the pancreatic tumours. This might in part be due to the fact that most patients in the included studies were treated over a decade ago, when non-invasive assessment of tumour hypoxia was in its infancy. Translational studies coupled to randomised trials that focus on the predictive value of hypoxia for efficacy of hyperthermia may supply further insight into the mechanisms behind HT.

Due to the small size and limited quality of the available clinical studies, the evidence for a potential advantage of adding HT to radiotherapy and/or chemotherapy in pancreatic cancer is supporting but weak. Currently, the Hyperthermia European Adjuvant Trial (HEAT) is being conducted, a randomised two-armed open study in patients with R0/R1 resected pancreatic carcinoma (NCT01077427; EudraCT No. 2008–004802-14). This study intends to include 336 patients in total, with patients in one arm treated with gemcitabine alone and in the other arm with gemcitabine plus cisplatin with regional HT. As of September 2017, 100 patients have been included.

Indeed, to establish the role of hyperthermia as an additional modality to the currently applied treatments for pancreatic cancer, large well-conducted randomised controlled trials are necessary. Reporting should be performed by intention to treat and should include overall survival, response, QoL, and tumour stage and toxicity reporting according to well-established criteria, to enable comparison between studies. Also proper thermometry [Citation69], with accurate registration of the temperature throughout the HT treatment, is essential both for quality assurance of the delivered treatment as well as for linking treatment to clinical outcome.

Conclusions

This systematic review suggests that hyperthermia added to chemotherapy and/or radiotherapy may improve median OS and response rate of patients with locally advanced or metastatic pancreatic cancer. Randomised clinical trials are clearly needed to establish potential benefit of hyperthermia, with comprehensive reporting on stage, treatment including temperatures, response, duration of response, overall survival, quality of life and toxicity. Preferably, such trials will include translational studies that can help uncover the underlying mechanisms of the enhancement of chemotherapy and radiotherapy by hyperthermia.

Supplemental material

Supplemental Files

Download Zip (368.8 KB)

Disclosure statement

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

References

  • Netherlands Cancer Registry: [Figures about Cancer] (in Dutch). Available from: http://www.cijfersoverkanker.nl/selecties/Dataset_1/img59b5566ddcaf0 [last accessed 14 Sep 2017].
  • SEER database. Available from: https://seer.cancer.gov/statfacts/html/pancreas.html [last accessed 9 Sep 2017].
  • Neoptolemos JP, Palmer DH, Ghaneh P, et al. (2017). Comparison of adjuvant gemcitabine and capecitabine with gemcitabine monotherapy in patients with resected pancreatic cancer (ESPAC-4): a multicentre, open-label, randomised, phase 3 trial. Lancet 389:1011–24.
  • Oettle H, Post S, Neuhaus P, et al. (2007). Adjuvant chemotherapy with gemcitabine vs observation in patients undergoing curative-intent resection of pancreatic cancer: a randomized controlled trial. JAMA 297:267–77.
  • Neoptolemos JP, Dunn JA, Stocken DD, et al. (2001). Adjuvant chemoradiotherapy and chemotherapy in resectable pancreatic cancer: a randomised controlled trial. Lancet 358:1576–85.
  • Casadei R, Di Marco M, Ricci C, et al. (2015). Neoadjuvant chemoradiotherapy and surgery versus surgery alone in resectable pancreatic cancer: a single-center prospective, randomized, controlled trial which failed to achieve accrual targets. J Gastrointest Surg 19:1802–12.
  • Golcher H, Brunner TB, Witzigmann H, et al. (2015). Neoadjuvant chemoradiation therapy with gemcitabine/cisplatin and surgery versus immediate surgery in resectable pancreatic cancer: results of the first prospective randomized phase II trial. Strahlenther Onkol 191:7–16.
  • Versteijne E, van Eijck CHJ, Punt CJA, et al. (2016). Preoperative radiochemotherapy versus immediate surgery for resectable and borderline resectable pancreatic cancer (PREOPANC trial): study protocol for a multicentre randomized controlled trial. Trials 17:127.
  • Siegel RL, Miller KD, Jemal A. (2017). Cancer statistics, 2017. CA Cancer J Clin 67:7–30.
  • Burris HA, 3rd, Moore MJ, Andersen J, et al. (1997). Improvements in survival and clinical benefit with gemcitabine as first-line therapy for patients with advanced pancreas cancer: a randomized trial. J Clin Oncol 15:2403–13.
  • Hidalgo M, Von Hoff DD. (2012). Translational therapeutic opportunities in ductal adenocarcinoma of the pancreas. Clin Cancer Res 18:4249–56.
  • Oei AL, Ahire VR, van Leeuwen CM, et al. (2017). Enhancing radiosensitisation of BRCA2-proficient and BRCA2-deficient cell lines with hyperthermia and PARP1-i. Int J Hyperthermia 19:1-10.
  • Ino Y, Yamazaki-Itoh R, Oguro S, et al. (2013). Arginase II expressed in cancer-associated fibroblasts indicates tissue hypoxia and predicts poor outcome in patients with pancreatic cancer. PLoS One 8:e55146.
  • Miyake K, Yoshizumi T, Imura S, et al. (2008). Expression of hypoxia-inducible factor-1alpha, histone deacetylase 1, and metastasis-associated protein 1 in pancreatic carcinoma: correlation with poor prognosis with possible regulation. Pancreas 36:e1–9.
  • Sun HC, Qiu ZJ, Liu J, et al. (2007). Expression of hypoxia-inducible factor-1 alpha and associated proteins in pancreatic ductal adenocarcinoma and their impact on prognosis. Int J Oncol 30:1359–67.
  • Couvelard A, O’Toole D, Leek R, et al. (2005). Expression of hypoxia-inducible factors is correlated with the presence of a fibrotic focus and angiogenesis in pancreatic ductal adenocarcinomas. Histopathology 46:668–76.
  • Suker M, Beumer BR, Sadot E, et al. (2016). A patient-level meta-analysis of FOLFIRINOX for locally advanced pancreatic cancer. Lancet Oncol 17:801–10.
  • Conroy T, Desseigne F, Ychou M, et al. (2011). FOLFIRINOX versus gemcitabine for metastatic pancreatic cancer. N Engl J Med 364:1817–25.
  • Goldstein D, El-Maraghi RH, Hammel P, et al. (2015). nab-Paclitaxel plus gemcitabine for metastatic pancreatic cancer: long-term survival from a phase III trial. J Natl Cancer Inst 107:dju413.
  • Von Hoff DD, Ervin T, Arena FP, et al. (2013). Increased survival in pancreatic cancer with nab-paclitaxel plus gemcitabine. N Engl J Med 369:1691–703.
  • Ansari D, Kristoffersson S, Andersson R, Bergenfeldt M. (2017). The role of irreversible electroporation (IRE) for locally advanced pancreatic cancer: a systematic review of safety and efficacy. Scand J Gastroenterol 52:1165–71.
  • Rombouts SJ, Vogel JA, van Santvoort HC, et al. (2015). Systematic review of innovative ablative therapies for the treatment of locally advanced pancreatic cancer. Br J Surg 102:182–93.
  • Pandya GJ, Shelat VG. (2015). Radiofrequency ablation of pancreatic ductal adenocarcinoma: The past, the present and the future. World J Gastrointest Oncol 7:6–11.
  • Chuong MD, Springett GM, Freilich JM, et al. (2013). Stereotactic body radiation therapy for locally advanced and borderline resectable pancreatic cancer is effective and well tolerated. Int J Radiat Oncol Biol Phys 86:516–22.
  • Horsman MR, Overgaard J. (2016). The impact of hypoxia and its modification of the outcome of radiotherapy. J Radiat Res 57:i90–8.
  • Datta NR, Gómez Ordóñez S, Gaipl US, et al. (2015). Local hyperthermia combined with radiotherapy and-/or chemotherapy: recent advances and promises for the future. Cancer Treat Rev 41:742–53.
  • Overgaard J. (2011). Hypoxic modification of radiotherapy in squamous cell carcinoma of the head and neck-a systematic review and meta-analysis. Radiother Oncol 100:22–32.
  • Kirui DK, Koay EJ, Guo X, et al. (2014). Tumor vascular permeabilization using localized mild hyperthermia to improve macromolecule transport. Nanomedicine 10:1487–96.
  • van der Zee J. (2002). Heating the patient: a promising approach? Ann Oncol 13:1173–84.
  • Falk MH, Issels RD. (2001). Hyperthermia in oncology. Int J Hyperthermia 17:1–18.
  • Oei AL, Vriend LE, Krawczyk PM, et al. (2017). Targeting therapy-resistant cancer stem cells by hyperthermia. Int J Hyperthermia 33:419–27.
  • Hildebrandt B, Wust P, Ahlers O, et al. (2002). The cellular and molecular basis of hyperthermia. Crit Rev Oncol Hematol 43:33–56.
  • Hu Y, Li Z, Mi DH, et al. (2017). Chemoradiation combined with regional hyperthermia for advanced oesophageal cancer: a systematic review and meta-analysis. J Clin Pharm Ther 42:155–64.
  • Datta NR, Puric E, Klingbiel D, et al. (2016). Hyperthermia and radiation therapy in locoregional recurrent breast cancers: a systematic review and meta-analysis. Int J Radiat Oncol Biol Phys 94:1073–87.
  • Bai Z, Shi Y, Wang J, et al. (2017). Intratumoral radiofrequency hyperthermia-enhanced direct chemotherapy of pancreatic cancer. Oncotarget 8:3591–9.
  • Kirui DK, Celia C, Molinaro R, et al. (2015). Mild hyperthermia enhances transport of liposomal gemcitabine and improves in vivo therapeutic response. Adv Healthc Mater 4:1092–103.
  • Zhang T, Zhang F, Meng Y, et al. (2013). Diffusion-weighted MRI monitoring of pancreatic cancer response to radiofrequency heat-enhanced intratumor chemotherapy. NMR Biome 26:1762–7.
  • Moher D, Liberati A, Tetzlaff J, Altman DG. The PRISMA Group (2009). Preferred reporting items for systematic reviews and meta-analyses: the PRISMA Statement. PLoS Med 6:e1000097.
  • Ouzzani M, Hammady H, Fedorowicz Z, Elmagarmid A. (2016). Rayyan-a web and mobile app for systematic reviews. Syst Rev 5:210.
  • Oxford Levels of Evidence Working Group. The Oxford 2011 Levels of Evidence. 2011 Available from: http://www.cebm.net/index.aspx?o=5653
  • American Joint Committee on Cancer. (2010) Exocrine and endocrine pancreas. In: Compton CC, Byrd DR, Garcia-Aguilar J, et al., eds. AJCC Cancer Staging Manual 7th ed. New York: Springer, 241–250.
  • Yu J, Blackford AL, Dal Molin M, et al. (2015). Time to progression of pancreatic ductal adenocarcinoma from low-to-high tumour stages. Gut 64:1783–9.
  • Dewhirst MW, Lee CT, Ashcraft KA. (2016). The future of biology in driving the field of hyperthermia. Int J Hyperthermia 32:4–13.
  • Horsman MR, Overgaard J. (2007). Hyperthermia: a potent enhancer of radiotherapy. Clin Oncol (R Coll Radiol) 19:418–20.
  • van den Tempel N, Horsman MR, Kanaar R. (2016). Improving efficacy of hyperthermia in oncology by exploiting biological mechanisms. Int J Hyperthermia 32:446–54.
  • Oei AL, Vriend LE, Crezee J, et al. (2015). Effects of hyperthermia on DNA repair pathways: one treatment to inhibit them all. Radiat Oncol 10:165.
  • Krawczyk PM, Eppink B, Essers J, et al. (2011). Mild hyperthermia inhibits homologous recombination, induces BRCA2 degradation, and sensitizes cancer cells to poly (ADP-ribose) polymerase-1 inhibition. Proc Natl Acad Sci USA 108:9851–6.
  • Bergs JW, Franken NA, Haveman J, et al. (2007). Hyperthermia, cisplatin and radiation trimodality treatment: a promising cancer treatment? A review from preclinical studies to clinical application. Int J Hyperthermia 23:329–41.
  • van Bree C, Beumer C, Rodermond HM, et al. (1999). Effectiveness of 2′,2′difluorodeoxycytidine (Gemcitabine) combined with hyperthermia in rat R-1 rhabdomyosarcoma in vitro and in vivo. Int J Hyperthermia 15:549–56.
  • Haveman J, Rietbroek RC, Geerdink A, et al. (1995). Effect of hyperthermia on the cytotoxicity of 2′,2′-difluorodeoxycytidine (gemcitabine) in cultured SW1573 cells. Int J Cancer 62:627–30.
  • Urano M. (1994). Thermochemotherapy: from in vitro and in vivo experiments to potential clinical application. In: Urano M, Douple E, eds. Hyperthermia and Oncology, Vol. 4: Chemopotentiation by Hyperthermia. Utrecht: VSP, 169–204.
  • Gillen S, Schuster T, Büschenfelde CMZ, et al. (2010). Preoperative/Neoadjuvant therapy in pancreatic cancer: a systematic review and meta-analysis of response and resection percentages. PLoS Med 7:e1000267.
  • Kakehi M, Ueda K, Mukojima T, et al. (1990). Multi-institutional clinical studies on hyperthermia combined with radiotherapy or chemotherapy in advanced cancer of deep-seated organs. Int J Hyperthermia 6:719–40.
  • Yamada S, Takai Y, Nemoto K, et al. (1992). Intraoperative radiation therapy combined with hyperthermia for pancreatic carcinoma. Int J Oncol 1:795–8.
  • Ashayeri E, Bonney G, DeWitty RL, et al. (1993). Preliminary survivorship report on combined intraoperative radiation and hyperthermia treatments for unresectable pancreatic adenocarcinoma. J Natl Med Assoc 85:36–40.
  • Kouloulias VE, Nikita KS, Kouvaris JR, et al. (2001). Cytoreductive surgery combined with intraoperative chemohyperthermia and postoperative radiotherapy in the management of advanced pancreatic adenocarcinoma: feasibility aspects and efficacy. J Hepatobiliary Pancreat Surg 8:564–70.
  • Kouloulias VE, Kouvaris JR, Nikita KS, et al. (2002). Intraoperative hyperthermia in conjunction with multi-schedule chemotherapy (pre-, intra- and post-operative), by-pass surgery, and post-operative radiotherapy for the management of unresectable pancreatic adenocarcinoma. Int J Hyperthermia 18:233–52.
  • Douwes FR. (2006). Thermo-chemotherapie des fortgeschrittenen Pankreaskarzinoms. Biologische Medizin 36:126–30.
  • Bull JMC, Scott GL, Strebel FR, et al. (2008). Fever-range whole-body thermal therapy combined with cisplatin, gemcitabine, and daily interferon-α: a description of a phase I-II protocol. Int J Hyperthermia 24:649–62.
  • Ohguri T, Imada H, Yahara K, et al. (2008). Concurrent chemoradiotherapy with gemcitabine plus regional hyperthermia for locally advanced pancreatic carcinoma: initial experience. Radiat Med 26:587–96.
  • Bakshandeh-Bath A, Stoltz AS, Homann N, et al. (2009). Preclinical and clinical aspects of carboplatin and gemcitabine combined with whole-body hyperthermia for pancreatic adenocarcinoma. Anticancer Res 29:3069–77.
  • Maluta S, Schaffer M, Pioli F, et al. (2011). Regional hyperthermia combined with chemoradiotherapy in primary or recurrent locally advanced pancreatic cancer: an open-label comparative cohort trial. Strahlenther Onkol 187:619–25.
  • Ishikawa T, Kokura S, Sakamoto N, et al. (2012). Phase II trial of combined regional hyperthermia and gemcitabine for locally advanced or metastatic pancreatic cancer. Int J Hyperthermia 28:597–604.
  • Tschoep-Lechner KE, Milani V, Berger F, et al. (2013). Gemcitabine and cisplatin combined with regional hyperthermia as second-line treatment in patients with gemcitabine-refractory advanced pancreatic cancer. Int J Hyperthermia 29:8–16.
  • Volovat C, Volovat SR, Scripcaru V, Miron L. (2014). Second-line chemotherapy with gemcitabine and oxaliplatin in combination with loco-regional hyperthermia (EHY-2000) in patients with refractory metastatic pancreatic cancer – Preliminary results of a prospective trial. Rom Rep Phys 66:166–74.
  • Maebayashi T, Ishibashi N, Aizawa T, et al. (2017). Treatment outcomes of concurrent hyperthermia and chemoradiotherapy for pancreatic cancer: Insights into the significance of hyperthermia treatment. Oncol Lett 13:4959–64.
  • Yamada S, Takai Y, Nemoto K, et al. (1992). Intraoperative radiation therapy combined with hyperthermia against pancreatic carcinoma. Tohoku J Exp Med 166:395–401.
  • Kouloulias VE, Nikita KS, Kouvaris JR, et al. (2002). Intraoperative hyperthermia and chemoradiotherapy for inoperable pancreatic carcinoma. Eur J Cancer Care (Engl) 11:100–7.
  • Bruggmoser G, Bauchowitz S, Canters R, et al. (2011). Quality assurance for clinical studies in regional deep hyperthermia. Strahlenther Onkol 187:605–10.
  • Bruggmoser G, Bauchowitz S, Canters R, et al. (2012). Guideline for the clinical application, documentation and analysis of clinical studies for regional deep hyperthermia: quality management in regional deep hyperthermia. Strahlenther Onkol 188[Suppl 2]:198–211.
  • Kok HP, Kotte ANTJ, Crezee J. (2017). Planning, optimisation and evaluation of hyperthermia treatments. Int J Hyperthermia 33:593–607.
  • Crezee H, van Leeuwen CM, Oei AL, et al. (2016). Thermoradiotherapy planning: integration in routine clinical practice. Int J Hyperthermia 32:41–9.
  • Kok HP, Wust P, Stauffer PR, et al. (2015). Current state of the art of regional hyperthermia treatment planning: a review. Radiat Oncol 10:196.
  • Paulides MM, Stauffer PR, Neufeld E, et al. (2013). Simulation techniques in hyperthermia treatment planning. Int J Hyperthermia 29:346–57.

Appendix. Ovid search strategies

Ovid MEDLINE(R) Epub Ahead of Print, In-Process & Other Non-Indexed Citations, Ovid MEDLINE(R)

Daily and Ovid MEDLINE(R) < 1946 to Present>.

Search date: 28 June 2017

Embase Classic + Embase <1947 to 2017 June 26>. Ovid interface.

Search date: 28 June 2017