2,702
Views
20
CrossRef citations to date
0
Altmetric
Review Article

Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for treatment of ovarian cancer

, , , , , , , & show all
Pages 298-310 | Received 20 Oct 2015, Accepted 26 Jan 2016, Published online: 17 Mar 2016

Abstract

Hyperthermic intraperitoneal chemotherapy (HIPEC), a strategy combining maximal cytoreductive surgery and maximal regional chemotherapy, has been applied to treat ovarian cancer resulting in long-term survival rates in selected patients. However, the status of HIPEC in ovarian cancer remains an experimental procedure, given the many variables among the data and trials reviewed, to enable us to derive strong conclusions about its role from this overview. In this review we discuss treatment with HIPEC in patients with ovarian cancer and future prospective of its use in clinical setting. HIPEC is an effective tool in the treatment of selected patients with peritoneal carcinomatosis from ovarian cancer. Unfortunately, due to the lack of randomised trials, the evidence of HIPEC is very limited. Future randomised studies are awaited to define the role and clinical impact of HIPEC in ovarian cancer.

Introduction

Ovarian cancer (OC) is the sixth most common neoplasm in women and accounts for 25% of all malignancies affecting the female genital tract, with age-standardised incidence and mortality rates reaching the highest values in north-western Europe and northern America [Citation1]. OC is currently the seventh most common female cancer worldwide and the eighth most common cause of death worldwide [Citation2]. A fundamental reason for mortality from this disease is due to the lack of specific symptoms and a marked propensity for peritoneal spread, also known as ‘peritoneal carcinomatosis’ (PC) [Citation3]. The peritoneal dissemination from OC may be present at the time of diagnosis of the primary tumour, but it can also arise as a tumour recurrence after radical surgical treatment [Citation4]. Approximately 70% of patients with this type of tumour present an advanced disease (stage III or IV), and approximately 50–75% of women with OC will develop persistent or recurrent disease [Citation5].

The first use of hyperthermic intraperitoneal chemotherapy (HIPEC) for OC was reported in 1994 [Citation6]. Since then, there has been a large body of research evaluating this therapeutic approach associated with cytoreductive surgery (CRS). The rationale for HIPEC is based on 1) direct cytotoxicity of hyperthermia against malignant cells, 2) enhancement of the cytotoxicity of anticancer drugs, and 3) the pharmacokinetic advantages of the intraperitoneal route for chemotherapy [Citation7–9]. However, HIPEC, which does not reach more than 5 mm depth in tissue, requires complete surgical removal of PC [Citation10]. Although the positive impacts of the combination of the peritonectomy with HIPEC in advanced OC have been widely reported in terms of survival effects [Citation4], this procedure remains ‘experimental’ because of lack of randomised trials. This treatment requires not only highly specialised human resources but also complex technological facilities.

In this review we summarise and discuss the current knowledge of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in the treatment of PC in OC.

Timing and patient selection

The natural history of OC presents several time points at which HIPEC can be performed: 1) in primary OC (first-line treatment), 2) as consolidation treatment, and 3) in persistent, progressive and recurrent disease (second-line treatment) [Citation11–13]. Therefore, the heterogeneous group of patients who are candidates for CS and HIPEC range from primary OC or recurrent OC treated with surgery and chemotherapy to chemoresistant OC patients with a history of multiple treatments [Citation5]. This heterogeneity in treatment procedures results in a difficult statement of the correct timing to perform HIPEC. For this reason, to address the optimal therapy response, an adequate patient selection is more clinically relevant than the proper HIPEC timing.

Exclusion criteria for a safe procedure [Citation3,Citation7] are 1) age > 70 years old, 2) serious medical histories (especially cardiorespiratory, neurological or renal), 3) clinical aggravation with systemic chemotherapy, 4) malnutrition, 5) concomitant extra-abdominal metastasis (aggressive locoregional treatment is not appropriate in non-controlled systemic disease), 6) unresectable liver metastases; 7) massive retroperitoneal bulk disease or lymph node involvement; 8) any contraindication for chemotherapy. Additional minor exclusion criteria are 1) no drop markers with neoadjuvant chemotherapy, 2) overweight (BMI > 40), 3) history of pelvic irradiation, 4) carcinomatosis extended at the scanner or clinically significant, 5) more than four surgical procedures, and 6) occlusion. With a particular focus on age, Votanopoulos et al. [Citation14] demonstrated that HIPEC in patients older than 70 years is associated with considerable morbidity and mortality. Additionally, Cascales-Campos et al. [Citation15], in a study on patients older than 75 years submitted to surgical debulking after neoadjuvant chemotherapy plus HIPEC, reported no advantages in terms of prognosis with high post-operative morbidity. Therefore, this age group should be excluded from this therapeutic procedure. However, age alone is not a contraindication for HIPEC. High-volume centres and rigorous patient selection are key factors for prolonged survival.

Preoperative assessment

Preoperative selection of patients is essential to minimise the incidence and the lethality of HIPEC post-operative complications. More recently, although it has been shown that magnetic resonance imaging (MRI) seems more accurately to predict preoperative Peritoneal Cancer Index (PCI) in patients undergoing evaluation for cytoreductive surgery compared with computed tomography (CT) [Citation16], at present, only direct exploration of the peritoneum allows the detection of ideal candidates for HIPEC in OC. Additionally, fluoro- D-glucose (FDG)-PET/CT was investigated for detection of PC before HIPEC in OC. Razzouk-Cadet et al. [Citation17] stated that FDG-PET/CT can be considered an useful tool for the selection of patients for HIPEC without distant metastasis. Nevertheless, the sensitivity of this procedure is limited in regard to the millimetric metastatic spots. Moreover, Fagotti et al. [Citation18] reported encouraging results of FDG-PET combined with staging laparoscopy performed immediately before secondary cytoreductive surgery with HIPEC. Notwithstanding, the current gold standard continues to be direct peritoneal visualisation, either by laparotomy or laparoscopy. However, the debate regarding which staging system is better in determining the prediction of complete cytoreduction is still open. In 1994, Gilly [Citation19] proposed a staging system of PC based on the size and distribution of malignant granulations. Several clinical trials [Citation7,Citation11] demonstrated the validity of this classification system as a prognostic indicator in patients affected by PC, suggesting a correlation with prognosis. The PCI, described by Jacquet and Sugarbaker [Citation20], is a widely cited system of PC staging that is more accurate than the Gilly system in carcinomatosis assessment [Citation19]. The PCI assesses the carcinomatosis quantification and distribution and has demonstrated a high prognostic value [Citation11].

Currently, many new therapeutic options and diagnostic tools are available in current OC oncology [Citation21–24]. For OC staging evaluation, we can also use a new technology that uses invisible light. Near infrared florescence (NIRF)-guided surgery seems to be a future technology that may improve safety and visualisation of cancer spread. NIRF has been a useful tool in many fields of medicine, such as surgery, urology and others [Citation25–32]. In advanced surgical approaches in performing peritonectomy, the need for ureter preservation is a primary issue. NIRF helps the visualisation of the ureter through the fatty tissue [Citation25]. The other usage of NIRF is visualisation of peritoneal metastases [Citation33,Citation34]. Van Dam et al. [Citation33] first proposed intraoperative tumour-specific fluorescence imaging of OC in humans. The fluorescent dye binds to the folate receptor α in OC. As an overexpression of this antigen occurs in most OC cases, the authors considered tumour-specific fluorescence imaging to be appropriate. Many more new foci of peritoneal spread were visible [Citation34]. In a study by Kosaka et al. [Citation35], peritoneal spread from the OC origin was detected in an animal model using IGG alone or IR 800 albumin. These researchers considered the potential use of ICG in clinical practice. This new technology may guide a surgeon for a more efficient cytoreduction in the future, which will lead to an improvement from an oncology perspective. Currently, new trials are ongoing to evaluate the use of NIRF technology in improving detection of PC in various cancers (as NCT02032485, NCT01982227). Other dyes such as 5-aminolevulinic acid (5-ALA) that are visible at various wavelengths could also be considered for the procedures. Kishi et al. [Citation36] reported that during staging laparoscopy, using 5-ALA improved the detection of peritoneal metastases in 21% of patients without laparoscopic signs of PC. Results from the phase I clinical trial were published by Liu et al. using aminolevulinic acid photodynamic visualisation for CRS and additionally HIPEC in patients with OC or primary PC [Citation37]. In 95% of patients (19/20 analysed cases) strong red fluorescence was detected in the analysed tissue invaded by cancer with a specificity of 100%. It is possible this new improved intraoperative visualisation of cancer deposits using fluorescent guided surgery could help in further improvement of CRS for OC in the future.

Prognostic scoring systems

Complete cytoreduction is the mainstay of CRS with HIPEC [Citation38,Citation39]. Currently, two classification systems are used to assess the completeness of cytoreduction [Citation7,Citation20]. The definition of optimal cytoreduction, however, remains controversial and often differs among studies. The experience with aggressive cytoreductive surgery and heated intraoperative chemotherapy shows a more restrictive definition of optimal CRS (< 2.5 mm) resulting in a better correlation with outcomes such as survival. Some authors have indicated that optimal cytoreduction requires no visible residual disease [Citation4]. However, it is clear that there is a significant positive correlation between maximal cytoreduction and median survival time. Therefore, the extent of cytoreduction is one of the most crucial prognostic factors to improve OS greatly in all stages of disease [Citation40]. It was estimated that each 10% increase in maximal cytoreduction was associated with a 5.5% increase in median survival time [Citation38]. Several studies have emphasised the relationship between complete cytoreduction and improved outcomes. Ryu et al. [Citation41] reported that a cytoreduction of less than 1 cm conferred a significantly better disease-free interval (hazard ratio 0.355, p = 0.0005) as well as overall survival (hazard ratio 0.38, p = 0.003). Raspagliesi [Citation42] found that the outcomes were positively affected by CCR score in addition to the World Health Organization performance status and the extent of the carcinomatosis. Similarly, Look et al. [Citation43] found a complete cytoreduction (residual disease of < 2.5 mm) was associated with improved median survival (55.9 versus 8 months, p = 0.037) in multivariate analysis. Therefore, only by assuring that the majority of women with OC have access to a maximal primary surgical effort can we expect to realise the population-based survival benefits of cytoreductive surgery. In the recurrent OC setting, several authors demonstrated the undifferentiated histological subtype represented (hazard ratio, 2.57 p < 0.05) an independent factor associated with reduced disease-free survival [Citation44], whereas BRCA status did not affect progression-free survival [Citation45]. Age, performance status, interval from initial treatment to recurrent, PCI, presence of lymph nodes, initial platinum response and completeness of cytoreduction still remains the most important prognostic indicators in OC.

Morbidity and mortality

Many criticisms against procedures combining CRS and HIPEC were due to the high mortality and morbidity rates reported in the medical literature for the treatment of non-gynaecological peritoneal surface malignancies. However, it is still unclear whether the increased rate of morbidity and mortality are related to CRS or HIPEC. Many authors have published their experiences using various criteria for defining the post-operative or post-HIPEC morbidity and mortality. Due to the lack of uniformity in grading the morbidity as well as in reporting the differences in HIPEC administration (eg, variation in the perfused temperature, chemotherapy protocols, open or closed surgical technique, HIPEC devices), the comparison of the results is inaccurate and inadequate [Citation46–49]. In this context, data from two systematic reviews [Citation4,Citation5] suggested that the morbidity and mortality rates are around 0–40% and 0–10% respectively however, it has been demonstrated by a recent systematic review that the mortality and morbidity rates are lower, in the tertiary high volume centres [Citation50].

Nevertheless locoregional administration of chemo-perfusate should reduce the risk of the chemotherapy agent systemic effects, the complications mainly related to the HIPEC procedure are haematological toxic effects (0–31%) due to transitory bone marrow suppression and renal insufficiency (0–7%), mainly in cisplatin-delivered HIPEC [Citation4] even the real influence of intraperitoneal cisplatin concentrations or dose along with the HIPEC duration and temperature on renal or haematological toxicity remains unknown. The maximum tolerated chemotherapy dose has not yet been clarified and further investigations are warranted. In addition, other significant complications were ileus, anastomotic leakage, intestinal perforation, fistula, abscess, sepsis, bleeding and wound infection/dehiscence. Reoperation for complications was required in 0% to 16% of cases [Citation51]. Moreover, the morbidity and mortality in OC patients submitted to CRS plus HIPEC still remains dependent on the patient’s age, performance status, number and type of peritonectomy procedures, and on the duration of HIPEC. In particular, recent studies [Citation3,Citation12] have shown performance status, chemotherapy dose, duration, the extent of surgery, visceral resection and number of digestive anastomoses were correlated with a higher morbidity rate as independent factors [Citation4]. The extent of carcinomatosis and incomplete cytoreduction has also been reported as a predictive factor of morbidity [Citation3,Citation12,Citation52]. Particularly, the complication rate of patients with stage III–IV carcinomatosis is higher than those with stage I–II disease [Citation3,Citation12,Citation52]. Recently, a retrospective Italian multicentre study [Citation53] of 683 patients showed older age and a higher value of PCI were correlated with higher mortality; instead, older age, ovarian origin of tumour, presence of ascites, closed technique and longer operative time were predictors of higher morbidity. In a recently published meta-analysis [Citation54], the post-HIPEC mortality rates for primary and for recurrent OC were equal at 1.8%. The pooled rates for minor morbidities (grades I–II) were for primary OC 40% and for recurrent OC 27.5%, and for grades III–IV 31.3% and 26.2% respectively. Comparing CRS with HIPEC versus CRS alone, the major morbidities were similarly independent of the usage of HIPEC (26.7% versus 28.6%, p = 0.784) as reported by Warschkow et al. [Citation55].

Search strategy

On 1 December 2015 we searched the PubMed database for trials containing the keywords ‘ovarian cancer’, ‘hyperthermic intraperitoneal chemotherapy’ and ‘HIPEC.’

All studies involved patients with OC who were treated with HIPEC during different stages of disease. Studies were excluded from the review if they did not contain a HIPEC-based procedure and/or complete information. Data on HIPEC as a front-line strategy of several selected studies including patients with primary OC treated with HIPEC, as well as interval cytoreduction (during primary chemotherapy) and consolidation treatment, are presented in , while studies of HIPEC as a second-line treatment including HIPEC during ‘secondary surgery’ are reported in . Finally, in , the available ongoing clinical trials that deal with different factors contributing to survival as a front line as well as additional treatment are reported.

Table 1. HIPEC in ovarian cancer studies as first line treatment during primary cytoreductive surgery presented in more relevant publications.

Table 2. HIPEC as a treatment in recurrence OC presented in more relevant publications and ongoing trials.

Table 3. Selected ongoing trials for HIPEC in ovarian cancer.

An overview of HIPEC efficacy

Considering the small number of patients per centre, a number of collaborative experiences of HIPEC in OC have been developed. Recently in the USA, surgical and gynaecological oncologists participated in a collaborative Internet-based registry (HYPER-O) [Citation65], collecting and analysing data from multiple centres to achieve an understanding of current practice and outcomes. In the initial report, 141 women were treated as frontline (n = 26), as interval debulking (n = 19), for consolidation (n = 12), or for recurrence (n = 83). The median duration of HIPEC was 100 min (range 30–120), and the average perfusion temperature was 38.5–43.0 °C (median 41.9°C). The HIPEC drug was based on the administration of cisplatin (n = 72), mitomycin (n = 53), or their combination (n = 14). The median overall survival of the entire population was 30.3 months. In addition, a retrospective cohort multicentre study from French centres was reported [Citation57] including consecutive patients treated with CRS and HIPEC for advanced and/or recurrent OC. The study involved 566 patients from 13 centres who underwent 607 procedures between 1991 and 2010. There were 92 patients with advanced OC (first-line treatment) and 474 patients with recurrent OC, representing altogether one of the largest cohorts of OC patients treated with HIPEC. HIPEC + CRS yielded a median survival of 35.4 months for OC and 45.7 months for recurrent OC. After CRS has been completed, the median survival was 41.5 months in advanced OC and 47.2 months and 51.6 months in chemosensitive and chemoresistant OC, respectively. Interestingly, the result from this analysis was that no significant difference in survival between patients with chemoresistant and chemosensitive subgroups has been noted. Recently, the first randomised study evaluating the role of HIPEC in recurrent OC was conducted. HIPEC was associated with a significantly prolonged OS (26.7 versus 13.4 months in the non-HIPEC group, p = 0.006) [Citation104]. Additionally, in this study, survival was 26.6 months in platinum-sensitive and 26.8 months in platinum-resistant disease. These data have confirmed the recent similar results reported by Bakrin et al. for chemoresistant and chemosensitive patients [Citation57]. Recently, meta-analysis was published [Citation54] analysing data based on available results from studies comparing CRS + chemotherapy with or without the combination of HIPEC in primary or recurrent OC. The authors proved statistically significant benefit in 2-,3-,4-,5- and 8-year survival with comparable morbidity and mortality between the two procedures. In case of recurrent OC, CRS with or without chemotherapy definitely improves long-term results. The combination of HIPEC and CRS along standard chemotherapy had higher 1-, 3- and 5-year overall survival in comparison with chemotherapy only (100%, 91%, 57% versus 83%, 66%, 41%). Analysing the magnitude of effect related to HIPEC alone, the studies comparing HIPEC + CRS + chemotherapy versus CRS + chemotherapy, small benefit of HIPEC for recurrent OC only has been shown in case of 1- and 3-year OS rate (OR:3.48; OR 7.39, p < 0.01)

Front-line therapy

HIPEC during primary cytoreductive surgery

The standard treatment for patients with advanced ovarian disease (stage III or IV) is considered surgical debulking followed by platinum/paclitaxel-based adjuvant therapy [Citation105,Citation106]. The prognostic impact of maximal cytoreductive surgery in the treatment of PC from OC is well established [Citation107]. Although 60–80% of cases diagnosed at an advanced stage respond to platinum-based systemic chemotherapy [Citation4], the prognosis remains poor due to a high rate of recurrence (5-year survival rate < 25%) [Citation5]. HIPEC performed at the time of initial surgery would have several advantages due to the characteristics of patients (best performance status and less toxicities from prior treatments) and to the characteristics of the tumour (less possibility of pharmacological resistances) [Citation108].

Although, the best outcome has been observed after complete CRS, considering the lack of randomised trials and the weakness of published studies, it is hazardous to conclude that HIPEC used as a front-line treatment or as a delayed first-line chemotherapy impacts the OS of patients.

HIPEC during interval cytoreductive surgery

A major controversy concerns the optimal time-point in the natural history of OC for the performance of CRS along with the HIPEC [Citation109]. The patient numbers from various studies are small, and the data seems difficult to interpret. Additionally, initial maximal CRS was not always reported. Therefore, neoadjuvant chemotherapy for two or three cycles may reduce disease burden and increase tumour resectability while simultaneously allowing performance status to improve. Many advantages are possible if surgery is performed after neoadjuvant chemotherapy (i.e. less intraoperative blood loss, shorter operative time, fewer intensive care unit admissions, and a shorter duration of hospital stay) [Citation110,Citation111]. No significant differences resulted in the survival rate between patients submitted to HIPEC as a first-line treatment and as a second-line treatment following neo-adjuvant chemotherapy [Citation65]. However, a recent randomised trial showed that primary systemic chemotherapy followed by delayed CRS and subsequent systemic chemotherapy facilitates optimal CRS with a decrease in morbidity without being less effective than primary debulking surgery followed by systemic chemotherapy [Citation112]. Spiliotis et al. [Citation113] are conducting an ongoing trial of laparoscopic-assisted neoadjuvant HIPEC in patients with stage IIIC or IV OC, in combination with systemic chemotherapy followed by interval debulking + HIPEC and then further systemic chemotherapy. However, the experience with this indication is limited.

Consolidation treatment

Despite the high initial response rates to front-line CRS and HIPEC in OC, most patients experienced recurrence. It was shown even after a documented complete pathological response, approximately 60% of the patients with OC will present recurrent disease [Citation114]. Therefore, there is a need for approaches in ‘consolidating’ initial disease control, as additional therapy may be beneficial in maintaining the disease control [Citation115]. However, large randomised comparative studies with prolonged follow-up are needed to support the ‘consolidation treatment’ in a clinical setting after CRS and HIPEC.

Recurrent OC

In patients initially presenting with advanced disease, recurrence is frequent. One of the most important factors affecting survival in patients with recurrence is the time between the initial cancer treatment and the time of recurrence [Citation116,Citation117]. Those patients progressing after front-line therapy or within 6 months of completion have ‘platinum-resistant’ tumours, with response rates of 8–28% and a median overall survival time of 6–12 months, whereas those recurring after 6 months have ‘platinum-sensitive’ tumours, with response rates up to 77% and a median survival duration of 12–40 months [Citation118,Citation119]. Survival of patients with recurrent OC treated by chemotherapy alone tends to be inferior to that reported in secondary CRS. The influence of secondary CRS without HIPEC on survival outcomes has been addressed in a substantial amount of studies and has been recently systematically reviewed [Citation120]. The addition of HIPEC is an interesting and promising treatment in recurrent OC if it is combined with complete cytoreduction. HIPEC has been mainly investigated in the setting of persistent or recurrent disease. However, a significant number of patients presented with early recurrent disease or heavily pre-treated (re)recurrent disease with diffuse PC, and these patients’ characteristics suggest poor outcomes. In most studies, median OS was longer than that reported in secondary CRS only [Citation121]. In the ‘platinum sensitive setting’, HIPEC resulted in a statistically significant improvement in PFS and OS [Citation122]. HIPEC seems to achieve similar survival benefits in patients with chemoresistant or chemosensitive disease.

Critique of current evidence

Although the standard front-line therapy for OC is considered to be CRS followed by intravenous chemotherapy with a platinum and taxane combination, this approach is not curative, and approximately 60% of all patients with OC have recurrence [Citation123–126]. To improve these results along with the encouraging data achieved from intraperitoneal administration of chemotherapeutic drugs, the combination of the intraperitoneal route with intravenous administration in primary stage III OC has subsequently been studied. Therefore, three randomised trials conducted by the Gynaecological Oncology Group (GOG) [Citation127–129] assessed the efficacy and safety of intraperitoneal chemotherapy in advanced OC. In these studies survival data were clearly in favour of the combination if compared to the conventional systemic chemotherapy alone. Based on these results, the National Cancer Institute issued a clinical announcement in January 2006, recommending that women with stage III OC who undergo optimal surgical cytoreduction should be considered for intraperitoneal chemotherapy [Citation130]. However, these trials considered only patients submitted to optimal cytoreduction, and several complaints mainly related to the management of the catheter for intraperitoneal and patient compliance were reported, resulting in less than half of patients completing the treatment [Citation130]. Moreover, normothermic intraperitoneal chemotherapy still presents several limits, which consist of the inability of this technique to penetrate tumour nodules of 3 mm or greater [Citation131]. Therefore, the pharmacokinetics of intraperitoneal platinum compounds with hyperthermic perfusion after CRS have been successively investigated. However, despite the promise of better results, after approximately 20 years from the first report of HIPEC, little is known currently on the real long-term prognostic impact of this treatment modality. The levels of evidence in the published studies are mostly class II or class III (non-randomised comparative studies and observational studies). Additionally, the patients’ cohort treated was heterogeneous and differed in age, previous systemic therapy, tumour size and timing (primary OC, consolidation treatment and recurrent/persistent disease) [Citation5]. The timing of HIPEC differed greatly, including cases with either advanced OC which had or had not undergone previous surgery and chemotherapy, and cases of recurrent OC pre-treated with many types of therapy or second-look surgery. The lack of homogeneity also consists of differences in administering HIPEC, such as variation in the perfused temperature, chemotherapy protocols, surgical approach (open or closed technique) and HIPEC devices, which further influences the differences in complications. Moreover, different criteria and different classifications for complications were used to evaluate the completeness of cytoreduction. Finally, the surgeon experience as a variable parameter leads to an insufficient CRS [Citation132]. Nevertheless, although non-randomised, some studies [Citation41,Citation66,Citation83,Citation88,Citation91,Citation112,Citation133] have compared HIPEC to another arm consisting of no treatment or CRS. However, even if the small group of patients evaluated in these studies do not allow definitive conclusions, HIPEC may be a feasible treatment with a potential benefit and provide superior survival compared to CRS only or no treatment. However, the nature and small number of patients reported in these studies are not enough to increase the level of evidence for HIPEC in OC treatment. Recently, the first randomised study evaluating the role of HIPEC in recurrent OC showed that HIPEC significantly prolonged OS (26.7 versus 13.4 months in the non-HIPEC group, p = 0.006) [Citation104]. However, one study alone is not sufficient to recommend the use of HIPEC in OC patients, and currently neither the relevant international guidelines nor recent consensus has included HIPEC as a standard treatment for primary or recurrent cancer [Citation134–137]. The AGO-OVAR group also stated ‘the use of HIPEC outside of well-designed, prospective and controlled clinical trials is therefore disregarded’ [Citation138]. In this setting, a recent systemic review [Citation139] regarding the published retrospective data on the use of HIPEC for primary advanced OC and for recurrent OC failed to exhibit a clear survival benefit that justifies the use of this technique as a standard daily practice. HIPEC has obtained modest results concerning survival compared to other studies that have examined standard approaches. The weighted median OS was 37.3 months, which has been historically observed in patients who have undergone suboptimal de-bulking or neoadjuvant therapy [Citation140,Citation141] without HIPEC.

The important issue is to keep the temperature at the optimal level in the whole abdomen during the HIPEC procedure for OC. Rettenmaier et al. [Citation142] proved the temperature variations according to the abdominal site and time intervals were minimal so that the effective time of the hyperthermia was achieved in all sites of the abdomen during the whole procedure.

An interesting report was published by a group from the USA [Citation143] based on HIPEC usage in paediatric patients with paediatric ovarian tumours. The study is based on only eight patients aged 4–18 years, which highlights an important usage of this aggressive treatment in the paediatric field of oncology, with good results of OS and RFS of 64% and 62%, respectively. In 2012, the same group proved the safety profile of HIPEC with cisplatin for use in children [Citation144].

In conclusion, the heterogeneity of patient selection, the timing of disease and HIPEC administration preclude a definitive conclusion from the survival results and make of HIPEC an investigational procedure with a low level of evidence.

State of the art and future prospects

In the last few years, various groups of surgeons have become involved in the use of HIPEC for PC from OC. However, only a few centres have reached experience exceeding 100 cases, and the HIPEC procedure has not been standardised yet. Some authors have reported that the performance of at least 130 procedures is necessary to consider the physician an expert in cytoreduction using the ‘Sugarbaker’ technique [Citation141]. Generally, HIPEC seems well tolerated and associated with acceptable morbidity if patient selection is appropriate and adequate experience is gained in a referral centre. However, the most essential question is whether it provides a survival advantage [Citation85]. To resolve this issue, many centres initiated randomised trials for various stages of disease. To the best of our knowledge, few randomised controlled trials are evaluating the effectiveness of HIPEC in primary or recurrent OC; two have just been proposed, whereas five are already on course [Citation74,Citation75,Citation96,Citation97,Citation99,Citation102]. Amongst the ongoing trials, a Korean study (NCT01091636) [Citation74] has evaluated the efficacy of HIPEC in the treatment of primary and recurrent OC. NCT00426257 [Citation96] is a phase III randomised trial studying the efficacy of HIPEC after secondary debulking surgery. Two more trials are related to HIPEC efficacy in recurrent OC: a multicentre phase III trial from France, the CHIPOR study (NCT01376752) [Citation99], and an Italian multicentre phase III trial, the HORSE study (NCT01539785) [Citation97]. This Italian multicentre trial is to study the role of HIPEC after chemotherapy in those patients who responded to treatment. Finally, another randomised study on HIPEC by the Italian Society of Integrated Locoregional Therapy (STILO) looking at the role of HIPEC in persistent OC had to be closed prematurely almost a decade ago due to poor patient accrual [Citation145]. To date, there are no ongoing trials that offer sequential intraperitoneal chemotherapy as a control arm, and intraperitoneal chemotherapy has resulted in the best results that have yet been reported in newly diagnosed patients. A phase I clinical trial currently recruiting for unresectable stage IIIC OC is also trialling the use of post-operative bevacizumab to change effect with efficacy and delay against cancer (NCT02217956) [Citation102]. Antigen for bevacizumab-VEGF is elevated in the peritoneal cavity with malignant ascites and leads to increasing endothelial cell permeability [Citation146].

The field of uncertainty in OC treatment is the presence of malignant ascites. We have to underline that in recurrent OC in the presence of ascites the usage of HIPEC is known as a contraindication of curative treatment. A paper by Ba et al. investigated the efficacy of HIPEC preceded or followed by CRS because of instability of vital signs in 32 patients with malignant ascites caused by OC [Citation147]. HIPEC under ultrasound guidance in unstable patients with delayed CRS after vital sign stability showed the same results in long-term survival as patients treated with HIPEC and CRS. This approach may be interesting, especially as malignant ascites reduce significantly patients’ quality of life, promote metastatic process, and affect respiratory and circulatory functions [Citation148,Citation149]. For current usage with malignant ascites, HIPEC should be selected only in very specific cases. Further studies are needed to prove the role of palliative treatment of malignant ascites by HIPEC not only because of hypothetical survival benefit, but especially to improve quality of life.

Iterative HIPEC procedures

As the nature of peritoneal spread is associated with recurrence, another interesting issue that has to be mentioned is repeated HIPEC procedure. A paper by Wong et al. [Citation150] describes eight patients who underwent repeat HIPEC procedure because of recurrent mesothelioma. No significant differences were observed between initial and repeated HIPEC in terms of blood loss, hospital stay, and operation time. Also median treatment-free time was no different. Statistically significant patients who had repeated HIPEC showed improved median OS (80 versus 27.2 months, p = 0.007). A paper by Vaira et al. [Citation151] describes patients showing recurrence after HIPEC were treated initially because of different malignancies, but it also showed on a small group of OC patients that in a selected group of recurrent diseases repeated HIPEC is a clinical option. No difference was observed in disease-free survival between initial and repeated HIPEC. Worth mentioning is the acceptable rate of complications grade III and IV, at −18.7% of patients.

Finally, the first experience of minimally invasive CRS plus HIPEC in OC has been published. Fagotti et al. [Citation18] retrospectively evaluated a consecutive series of OC patients with isolated platinum-sensitive relapse. Ten cases were treated with laparoscopic/robotic secondary CRS + HIPEC. In all cases, complete debulking was achieved. No grade III/IV surgical, metabolic, or haematological complications occurred. In all patients, post-operative FDG-PET/CT scan was negative, and after a median time of 10 months (6–37) from secondary CRS + HIPEC, no secondary recurrence was observed. Also, Rettenmaier et al. presented their first experience in laparoscopic debulking surgery and HIPEC procedure together with consolidation chemotherapy [Citation70]. Although the small numbers of patients and the retrospective data do not allow definitive conclusions, minimally invasive CRS + HIPEC may be a safe option in selected OC patients, and further studies are required.

Conclusions

CRS and HIPEC represent an aggressive multidisciplinary approach to an aggressive type of cancer such as OC, and it demonstrates a positive emerging trend towards expected survival rates in patients with PC. Despite encouraging survival rates, HIPEC remains an experimental procedure. It is also clear that CRS plus HIPEC is not indicated for all patients with PC, but has an established role in selected patients [Citation152]. As it is fundamental to achieve the related survival advantage, completeness of CRS must be considered the basic prerequisite for the use of HIPEC. In highly specialised treatment centres, mortality and morbidity rates are lower; thus, criticism against the feasibility of this procedure is no longer acceptable. Combined treatment with CRS and HIPEC is currently suggested by the international consensus group as the most likely to affect survival favourably in stage III OC. Centralisation could also give patients the possibility of undergoing real, complete cytoreduction without the risk of residual disease, with a possible positive impact on survival. However, failure of widespread adoption may reflect the lack of convincing evidence because of lack of randomised trials [Citation5,Citation153]. The growing interest and ongoing clinical trials in the field of HIPEC show the importance of this technique in the modern treatment of advanced OC [Citation153]. Moreover, new criteria are needed to include a unique system for peritoneal disease staging, the optimal extension of surgical resection associated with HIPEC, the best method of performing HIPEC, and the type and dose of anticancer drugs to use in HIPEC treatment.

Acknowledgements

All authors contributed equally to this work.

Disclosure statement

The Authors have no conflict of interest or financial ties to disclose. The authors alone are responsible for the content and writing of the paper.

References

  • Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin 2011;61:69–90.
  • Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J, Jemal A. Global cancer statistics, 2012. CA Cancer J Clin 2015;65:87–108.
  • Al-Shammaa HA, Li Y, Yonemura Y. Current status and future strategies of cytoreductive surgery plus intraperitoneal hyperthermic chemotherapy for peritoneal carcinomatosis. World J Gastroenterol 2008;14:1159–66.
  • Bijelic L, Jonson A, Sugarbaker PH. Systematic review of cytoreductive surgery and heated intraoperative intraperitoneal chemotherapy for treatment of peritoneal carcinomatosis in primary and recurrent ovarian cancer. Ann Oncol 2007;18:1943–50.
  • Chua TC, Robertson G, Liauw W, Farrell R, Yan TD, Morris DL. Intraoperative hyperthermic intraperitoneal chemotherapy after cytoreductive surgery in ovarian cancer peritoneal carcinomatosis: Systematic review of current results. J Cancer Res Clin Oncol 2009;135:1637–1645.
  • Loggie BW, Sterchi JM, Rogers AT, Lentz S, Holmesley H, Charles D, Sundberg D. Intraperitoneal hyperthermic chemotherapy for advanced gastrointestinal and ovarian cancers, Reg Cancer Treat 1995;7:78–81.
  • Glehen O, Mohamed F, Gilly FN. Peritoneal carcinomatosis from digestive tract cancer: New management by cytoreductive surgery and intraperitoneal chemohyperthermia. Lancet Oncol 2004;5:219–28.
  • Sugarbaker PH. Intraperitoneal chemotherapy and cytoreductive surgery for the prevention and treatment of peritoneal carcinomatosis and sarcomatosis. Semin Surg Oncol 1998;14:254–61.
  • Van der Speeten K, Stuart OA, Sugarbaker PH. Pharmacokinetics and pharmacodynamics of perioperative cancer chemotherapy in peritoneal surface malignancy. Cancer J 2009;15:216–24.
  • El-Kareh AW, Secomb TW. A theoretical model for intraperitoneal delivery of cisplatin and the effect of hyperthermia on drug penetration distance. Neoplasia 2004;6:117–27.
  • Stewart JH IV, Shen P, Levine EA. Intraperitoneal hyperthermic chemotherapy for peritoneal surface malignancy: Current status and future directions. Ann Surg Oncol 2005;12:765–77.
  • AJCC. Ovary and primary peritoneal carcinoma. In: AJCC cancer staging handbook, 7th ed. Springer; 2009. pp 501–6.
  • Helm CW. The role of hyperthermic intraperitoneal chemotherapy (HIPEC) in ovarian cancer. Oncologist 2009;14:683–94.
  • Votanopoulos KI, Newman NA, Russell G, Ihemelandu C, Shen P, Stewart JH, Levine EA. Outcomes of cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) in patients older than 70 years: survival benefit at considerable morbidity and mortality. Ann Surg Oncol 2013. Available from http://www.ncbi.nlm.nih.gov/pmc/articles/
  • Cascales-Campos P, Gil J, Gil E, Feliciangeli E, López V, Gonzalez AG, et al. Cytoreduction and HIPEC after neoadjuvant chemotherapy in stage IIIC–IV ovarian cancer. Critical analysis in elderly patients. Eur J Obstet Gynecol Reprod Biol 2014;179:88–93.
  • Low RN, Barone RM, Lucero J. Comparison of MRI and CT for predicting the peritoneal cancer index (PCI) preoperatively in patients being considered for cytoreductive surgical procedures. Ann Surg Oncol 2015;22:1708–15.
  • Razzouk-Cadet M, Zwarthoed C, Desantis M, Viau P, Mahamat A, Mounier N, et al. Diagnostic value of FDG-PET/CT for peritoneal carcinomatosis (PC) before hyperthermic intraperitoneal chemotherapy (HIPEC) in ovarian cancer. J Clin Oncol 2012;30(Suppl):e15574.
  • Fagotti A, Petrillo M, Costantini B, Fanfani F, Gallotta V, Chiantera V, et al. Minimally invasive secondary cytoreduction plus HIPEC for recurrent ovarian cancer: a case series. Gynecol Oncol 2014;132:303–6.
  • Gilly FN, Carry PY, Sayag AC. Regional chemotherapy with mitomycin C and intraoperative hyperthermia for digestive cancers with peritoneal carcinomatosis. Hepatogastroenterology 1994;41:124–9.
  • Jacquet P, Sugarbaker PH. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. In: Sugarbaker PH, editor Peritoneal carcinomatosis: Principles of management. Boston, MA: Kluwer Academic Publishers; 1996. pp 359–74.
  • Fischerova D, Burgetova A. Imaging techniques for the evaluation of ovarian cancer. Best Pract Res Clin Obstet Gynaecol 2014;28:697–720.
  • Perez CA, Mutic S. Advances and future of radiation oncology. Rep Pract Oncol Radiother 2013;18:329–32.
  • Isa N. Evidence based radiation oncology with existing technology. Rep Pract Oncol Radiother 2013;19:259–66.
  • Micci F, Haugom L, Abeler VM, Davidson B, Tropé CG, Heim S. Genomic profile of ovarian carcinomas. BMC Cancer 2014;14:315.
  • Polom W, Markuszewski M, Rho YS, Matuszewski M. Use of invisible near infrared light fluorescence with indocyanine green and methylene blue in urology. Part 2. Cent Eur J Urol 2014;67:310–13.
  • de Boer E, Harlaar NJ, Taruttis A, Nagengast WB, Rosenthal EL, Ntziachristos V, van Dam GM. Optical innovations in surgery. Br J Surg 2015;102:e56–72.
  • Polom W, Markuszewski M, Rho YS, Matuszewski M. Usage of invisible near infrared light (NIR) fluorescence with indocyanine green (ICG) and methylene blue (MB) in urological oncology. Part 1. Cent Eur J Urol 2014;67:142–8.
  • Schaafsma BE, Mieog JS, Hutteman M, van der Vorst JR, Kuppen PJ, Löwik CW, et al. The clinical use of indocyanine green as a near-infrared fluorescent contrast agent for image-guided oncologic surgery. J Surg Oncol 2011;104:323–32.
  • Murawa D, Polom K, Rho YS, Murawa P. Developments in near-infrared-guided hepatobiliary, pancreatic and other upper gastrointestinal surgery. Contrast Media Mol Imaging 2013;8:211–19.
  • Polom K, Murawa D, Nowaczyk P, Rho YS, Murawa P. Breast cancer sentinel lymph node mapping using near infrared guided indocyanine green and indocyanine green – human serum albumin in comparison with gamma emitting radioactive colloid tracer. Eur J Surg Oncol 2012;38:137–42.
  • Mitsumori N, Nimura H, Takahashi N, Kawamura M, Aoki H, Shida A, et al. Sentinel lymph node navigation surgery for early stage gastric cancer. World J Gastroenterol 2014;20:5685–93.
  • Polom K, Murawa D, Rho YS, Spychala A, Murawa P. Skin melanoma sentinel lymph node biopsy using real-time fluorescence navigation with indocyanine green and indocyanine green with human serum albumin. Br J Dermatol 2012;166:682–3.
  • van Dam GM, Themelis G, Crane LM, Harlaar NJ, Pleijhuis RG, Kelder W, et al. Intraoperative tumor-specific fluorescence imaging in ovarian cancer by folate receptor-α targeting: First in-human results. Nat Med 2011;17:1315–19.
  • van Oosten M, Crane LM, Bart J, van Leeuwen FW, van Dam GM. Selecting potential targetable biomarkers for imaging purposes in colorectal cancer using target selection criteria (TASC): A novel target identification tool. Transl Oncol 2011;4:71–82.
  • Kosaka N, Mitsunaga M, Longmire MR, Choyke PL, Kobayashi H. Near infrared fluorescence-guided real-time endoscopic detection of peritoneal ovarian cancer nodules using intravenously injected indocyanine green. Int J Cancer 2011;129:1671–7.
  • Kishi K, Fujiwara Y, Yano M, Motoori M, Sugimura K, Ohue M, et al. Diagnostic laparoscopy with 5-aminolevulinic-acid-mediated photodynamic diagnosis enhances the detection of peritoneal micrometastases in advanced gastric cancer. Oncology 2014;87:257–65.
  • Liu Y, Endo Y, Fujita T, Ishibashi H, Nishioka T, Canbay E, et al. Cytoreductive surgery under aminolevulinic acid-mediated photodynamic diagnosis plus hyperthermic intraperitoneal chemotherapy in patients with peritoneal carcinomatosis from ovarian cancer and primary peritoneal carcinoma: Results of a phase I trial. Ann Surg Oncol 2014;21:4256–62.
  • Bristow RE, Tomacruz RS, Armstrong DK, Trimble EL, Montz FJ. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: A meta-analysis. J Clin Oncol 2002;20:1248–59.
  • Chi DS, Eisenhauer EL, Zivanovic O, Sonoda Y, Abu-Rustum NR, Levine DA, et al. Improved progression-free and overall survival in advanced ovarian cancer as a result of a change in surgical paradigm. Gynecol Oncol 2009;114:26–31.
  • Halkia E, Spiliotis J, Sugarbaker P. Diagnosis and management of peritoneal metastases from ovarian cancer. Gastroenterol Res Pract 2012;2012:541842.
  • Ryu KS, Kim JH, Ko HS, Kim JW, Ahn WS, Park YG, et al. Effects of intraperitoneal hyperthermic chemotherapy in ovarian cancer. Gynecol Oncol 2004;94:325–32.
  • Raspagliesi F, Kusamura S, Campos Torres JC, de Souza GA, Ditto A, Zanaboni F, et al. Cytoreduction combined with intraperitoneal hyperthermic perfusion chemotherapy in advanced/recurrent ovarian cancer patients: The experience of National Cancer Institute of Milan. Eur J Surg Oncol 2006;32:671–5.
  • Look M, Chang D, Sugarbaker PH. Long-term results of cytoreductive surgery for advanced and recurrent epithelial ovarian cancers and papillary serous carcinoma of the peritoneum. Int J Gynecol Cancer 2004;14:35–41.
  • Cascales-Campos PA, Gil J, Feliciangeli E, Gil E, González-Gil A, López V, et al. The role of hyperthermic intraperitoneal chemotherapy using paclitaxel in platinum-sensitive recurrent epithelial ovarian cancer patients with microscopic residual disease after cytoreduction. Ann Surg Oncol 2015;22:987–93.
  • Safra T, Grisaru D, Inbar M, Abu-Abeid S, Dayan D, Matceyevsky D, et al. Cytoreduction surgery with hyperthermic intraperitoneal chemotherapy in recurrent ovarian cancer improves progression-free survival, especially in BRCA-positive patients – a case-control study. J Surg Oncol 2014;110:661–5.
  • Koga S, Hamazoe R, Maeta M. Treatment of implanted peritoneal cancer in rats by continuous hyperthermic peritoneal perfusion in combination with anticancer drugs. Cancer Res 1984;44:1840–2.
  • Fujimoto S, Shrestha RD, Kokubun M. Intraperitoneal hyperthermic perfusion combined with surgery effective for gastric cancer patients with peritoneal seedings. Ann Surg 1988;208:36–41.
  • Beaujard AC, Glehen O, Caillot JL, Francois Y, Bienvenu J, Panteix G, et al. Intraperitoneal chemohyperthermia with mitomycin C for digestive tract cancer patients with peritoneal carcinomatosis. Cancer 2000;88:2512–19.
  • Sadeghi B, Arvieux C, Glehen O, Beaujard AC, Rivoire M, Baulieux J, et al. Peritoneal carcinomatosis from non-gynecologic malignancies: Results of the EVOCAPE 1 multicentric prospective study. Cancer 2000;88:358–63.
  • Chua TC, Yan TD, Saxena A, Morris DL. Should the treatment of peritoneal carcinomatosis by cytoreductive surgery and hyperthermic intraperitoneal chemotherapy still be regarded as a highly morbid procedure? A systematic review of morbidity and mortality. Ann Surg 2009;249:900–7.
  • Fagotti A, B. Costantini, G. Vizzielli, Perelli F, Ercoli A, Gallotta V, et al. HIPEC in recurrent ovarian cancer patients: Morbidity-related treatment and long-term analysis of clinical outcome. Gynecol Oncol 2011;122:221–5.
  • Königsrainer I, Horvath P, Struller F, Grischke EM, Wallwiener D, Königsrainer A, Beckert S. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in recurrent epithelial ovarian cancer with peritoneal metastases: A single centre experience. Langenbecks Arch Surg 2014;399:589–94.
  • Macrì A, Arcoraci V, Belgrano V, Caldana M, Cioppa T, Costantini B, et al. Short-term outcome of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy: preliminary analysis of a multicentre study. Anticancer Res 2014;34:5689–93.
  • Huo YR, Richards A, Liauw W, Morris DL. Hyperthermic intraperitoneal chemotherapy (HIPEC) and cytoreductive surgery (CRS) in ovarian cancer: A systematic review and meta-analysis. Eur J Surg Oncol 2015;41:1578–89.
  • Warschkow R, Tarantino I, Lange J, Müller SA, Schmied BM, Zünd M, Steffen T. Does hyperthermic intraoperative chemotherapy lead to improved outcomes in patients with ovarian cancer? A single center cohort study in 111 consecutive patients. Patient Saf Surg 2012;6:12.
  • Ansaloni L, Agnoletti V, Amadori A, Catena F, Cavaliere D, Coccolini F, et al. Evaluation of extensive cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with advanced epithelial ovarian cancer. Int J Gynecol Cancer 2012;22:778–85.
  • Bakrin N, Bereder JM, Decullier E, Classe JM, Msika S, Lorimier G, et al. Peritoneal carcinomatosis treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) for advanced ovarian carcinoma: A French multicentre retrospective cohort study of 566 patients Eur J Surg Oncol 2013;39:1435–43.
  • Carrabin N, Mithieux F, Meeus P, Trédan O, Guastalla JP, Bachelot T, et al. Hyperthermic intraperitoneal chemotherapy with oxaliplatin and without adjuvant chemotherapy in stage IIIC ovarian cancer. Bull Cancer 2010;97:E23–32.
  • Cascales Campos PA, Gil Martínez J, Galindo Fernández PJ, Gil Gómez E, Martínez Frutos IM, Parrilla Paricio P. Perioperative fast track program in intraoperative hyperthermic intraperitoneal chemotherapy (HIPEC) after cytoreductive surgery in advanced ovarian cancer. Eur J Surg Oncol 2011;37:543–8.
  • Cascales-Campos PA, Gil J, Gil E, Feliciangeli E, González-Gil A, Parrilla JJ, Parrilla P. Treatment of microscopic disease with hyperthermic intraoperative intraperitoneal chemotherapy after complete cytoreduction improves disease-free survival in patients with stage IIIC/IV ovarian cancer. Ann Surg Oncol 2014;21:2383–9.
  • Coccolini F, Campanati L, Catena F, Ceni V, Ceresoli M, Jimenez Cruz J, et al. Hyperthermic intraperitoneal chemotherapy with cisplatin and paclitaxel in advanced ovarian cancer: A multicenter prospective observational study. J Gynecol Oncol 2015;26:54–61.
  • Deraco M, Kusomura S, Virzi S, Puccio F, Macrì A, Famulari C, et al. Cytoreductive surgery and hyperthermic intra-peritoneal chemotherapy as upfront therapy for advanced EOC: Multi-institutional phase II study. Gynecol Oncol 2011;122:215–20.
  • Di Giorgio A, Naticchioni E, Biacchi D, Sibio S, Accarpio F, Rocco M, et al. Cytoreductive surgery (peritonectomy procedures) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) in the treatment of diffuse peritoneal carcinomatosis from ovarian cancer. Cancer 2008;113:315–25.
  • Frenel JS, Leux C, Pouplin L, Ferron G, Berton Rigaud D, Bourbouloux E, et al. Oxaliplatin-based hyperthermic intraperitoneal chemotherapy in primary or recurrent epithelial ovarian cancer: A pilot study of 31 patients. J Surg Oncol 2011;103:10–16.
  • Helm CW, Richard SD, Pan J, Bartlett D, Goodman MD, Hoefer R, et al. HIPEC in ovarian cancer: first report of HYPER-O registry. Int J Gynecol Cancer 2010;20:61–9.
  • Kim JH, Lee JM, Ryu KS, Lee YS, Park YG, Hur SY, et al. Consolidation hyperthermic intraperitoneal chemotherapy using paclitaxel in patients with epithelial ovarian cancer. J Surg Oncol 2010;101:149–55.
  • Lim MC, Kang S, Choi J, Song YJ, Park S, Seo SS, Park SY. Hyperthermic intraperitoneal chemotherapy after extensive cytoreductive surgery in patients with primary advanced epithelial ovarian cancer: interim analysis of a phase II study. Ann Surg Oncol 2009;16:993–1000.
  • Pavlov MJ, Kovacevic PA, Ceranic MS, Stamenkovic AB, Ivanovic AM, Kecmanovic DM. Cytoreductive surgery and modified heated intraoperative intraperitoneal chemotherapy (HIPEC) for advanced and recurrent ovarian cancer – 12-year single center experience. Eur J Surg Oncol 2009;35:1186–91.
  • Pomel C, Ferron G, Lorimier G, Rey A, Lhomme C, Classe JM, et al. Hyperthermic intra-peritoneal chemotherapy using oxaliplatin as consolidation therapy for advanced epithelial ovarian carcinoma. Results of a phase II prospective multicentre trial. CHIPOVAC study. Eur J Surg Oncol 2010;36:589–93.
  • Rettenmaier MA, Mendivil AA, Abaid LN, Brown Iii JV, Wilcox AM, Goldstein BH. Consolidation hyperthermic intraperitoneal chemotherapy and maintenance chemotherapy following laparoscopic cytoreductive surgery in the treatment of ovarian carcinoma. Int J Hyperthermia 2015;31:8–14.
  • Roviello F, Pinto E, Corso G, Pedrazzani C, Caruso S, Filippeschi M, et al. Safety and potential benefit of hyperthermic intraperitoneal chemotherapy (HIPEC) in peritoneal carcinomatosis from primary or recurrent ovarian cancer. J Surg Oncol 2010;102:663–70.
  • Rufian S, Munoz-Casares FC, Briceno J, Díaz CJ, Rubio MJ, Ortega R, et al. Radical surgery – peritonectomy and intra-operative intraperitoneal chemotherapy for the treatment of peritoneal carcinomatosis in recurrent or primary ovarian cancer. J Surg Oncol 2006;94:316–24.
  • www.clinicaltrials.gov/ct2/show/NCT02124421
  • www.clinicaltrials.gov/ct2/show/NCT01091636
  • www.clinicaltrials.gov/ct2/show/NCT01628380
  • www.clinicaltrials.gov/ct2/show/NCT02328716
  • www.clinicaltrials.gov/ct2/show/NCT01709487
  • www.clinicaltrials.gov/ct2/show/NCT02249013
  • www.clinicaltrials.gov/ct2/show/NCT02199171
  • www.clinicaltrials.gov/ct2/show/NCT02217956
  • www.clinicaltrials.gov/ct2/show/NCT02567253
  • Argenta PA, Sueblinvong T, Geller MA, Jonson AL, Downs LS Jr, Carson LF, et al. Hyperthermic intraperitoneal chemotherapy with carboplatin for optimally-cytoreduced, recurrent, platinum-sensitive ovarian carcinoma: A pilot study. Gynecol Oncol 2013;129:81–5.
  • Bae JH, Lee JM, Ryu KS, Lee YS, Park YG, Hur SY, et al. Treatment of ovarian cancer with paclitaxel- or carboplatin-based intraperitoneal hyperthermic chemotherapy during secondary surgery. Gynecol Oncol 2007;106:193–200.
  • Ceelen WP, Van Nieuwenhove Y, Van Belle S, Denys H, Pattyn P. Cytoreduction and hyperthermic intraperitoneal chemoperfusion in women with heavily pretreated recurrent ovarian cancer. Ann Surg Oncol 2012;19:2352–9.
  • Cotte E, Glehen O, Mohamed F, Lamy F, Falandry C, Golfier F, et al. Cytoreductive surgery and intraperitoneal chemo-hyperthermia for chemoresistant and recurrent advanced epithelial ovarian cancer: Prospective study of 81 patients. World J Surg 2007;31:1813–20.
  • de Bree E, Romanos J, Michalakis J, Relakis K, Georgoulias V, Melissas J, Tsiftsis DD. Intraoperative hyperthermic intraperitoneal chemotherapy with docetaxel as second-line treatment for peritoneal carcinomatosis of gynaecological origin. Anticancer Res 2003;23:3019–27.
  • Deraco M, Virzì S, Iusco DR, Puccio F, Macrì A, Famulari C, et al. Secondary cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for recurrent epithelial ovarian cancer: A multi-institutional study. BJOG 2012;119:800–9.
  • Gori J, Castaño R, Toziano M, Häbich D, Staringer J, De Quirós DG, Felci N. Intraperitoneal hyperthermic chemotherapy in ovarian cancer. Int J Gynecol Cancer 2005;15:233–9.
  • Helm CW, Randall-Whitis L, Martin RS III, Metzinger DS, Gordinier ME, Parker LP, Edwards RP. Hyperthermic intraperitoneal chemotherapy in conjunction with surgery for the treatment of recurrent ovarian carcinoma. Gynecol Oncol 2007;105:90–6.
  • Le Brun JF, Campion L, Berton-Rigaud D, Lorimier G, Marchal F, Ferron G, et al. Survival benefit of hyperthermic intraperitoneal chemotherapy for recurrent ovarian cancer: A multi-institutional case control study. Ann Surg Oncol 2014;21:3621–7.
  • Muñoz-Casares FC, Rufián S, Rubio MJ, Díaz CJ, Díaz R, Casado A, et al. The role of hyperthermic intraoperative intraperitoneal chemotherapy (HIPEC) in the treatment of peritoneal carcinomatosis in recurrent ovarian cancer. Clin Transl Oncol 2009;11:753–9.
  • Massari R, Barone M, Basilico R, Carella C, Colasante A, De Tursi M, et al. Peritonectomy and hyperthermic chemotherapy in patients with advanced or recurrent epithelial ovarian cancer: A single center cohort study. Minerva Chir 2014;69:17–26.
  • Piso P, Dahlke MH, Loss M, Schlitt HJ. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in peritoneal carcinomatosis from ovarian cancer. World J Surg Oncol 2004;2:21.
  • Robella M, Vaira M, Marsanic P, Mellano A, Borsano A, Cinquegrana A, et al. Treatment of peritoneal carcinomatosis from ovarian cancer by surgical cytoreduction and hyperthermic intraperitoneal chemotherapy (HIPEC). Minerva Chir 2014;69:27–35.
  • Zanon C, Clara R, Chiappino I, Bortolini M, Cornaglia S, Simone P, et al. Cytoreductive surgery and intraperitoneal chemohyperthermia for recurrent peritoneal carcinomatosis from ovarian cancer. World J Surg 2004;28:1040–5.
  • www.clinicaltrials.gov/ct2/show/NCT00426257
  • www.clinicaltrials.gov/ct2/show/NCT01539785
  • www.clinicaltrials.gov/ct2/show/NCT01767675
  • www.clinicaltrials.gov/ct2/show/NCT01376752
  • www.clinicaltrials.gov/ct2/show/NCT01970722
  • www.clinicaltrials.gov/ct2/show/NCT01588964
  • www.clinicaltrials.gov/ct2/show/NCT02217956
  • www.clinicaltrials.gov/ct2/show/NCT02567253
  • Spiliotis J, Halkia E, Lianos E, Kalantzi N, Grivas A, Efstathiou E, Giassas S. Cytoreductive surgery and HIPEC in recurrent epithelial ovarian cancer: A prospective randomized phase III study. Ann Surg Oncol 2014;22:570–5.
  • Bristow RE, Tomacruz RS, Armstrong DK, Trimble EL, Montz FJ. Survival effect of maximal cytoreductive surgery for advanced ovarian carcinoma during the platinum era: A meta-analysis. J Clin Oncol 2002;20:1248–59.
  • Verleye L, Ottevanger PB, Vergote I. Surgical program building in advanced ovarian cancer: European perspective. Gynecol Oncol 2009;114:S10–14.
  • Bertelsen K. Tumor reduction surgery and long-term survival in advanced ovarian cancer: A DACOVA study. Gynecol Oncol 1990;38:203–9.
  • de Bree E, Helm CW. Hyperthermic intraperitoneal chemotherapy in ovarian cancer: Rationale and clinical data. Expert Rev Anticancer Ther 2012;12:895–911.
  • Deraco M, Baratti D, Laterza B, Balestra MR, Mingrone E, Macrì, et al. Advanced cytoreduction as surgical standard of care and hyperthermic intraperitoneal chemotherapy as promising treatment in epithelial ovarian cancer. Eur J Surg Oncol 2011;37:4–9.
  • Surwit E, Childers J, Atlas I, Nour M, Hatch K, Hallum A, Alberts D. Neoadjuvant chemotherapy for advanced ovarian cancer. Int J Gynecol Cancer 1996;6:356–61.
  • Huober J, Meyer A, Wagner U, Wallwiener D. The role of neoadjuvant chemotherapy and interval laparotomy in advanced ovarian cancer. J Cancer Res Clin Oncol 2002;128:153–60.
  • Vergote I, Tropé CG, Amant F, Kristensen GB, Ehlen T, Johnson N, et al. Neoadjuvant chemotherapy or primary surgery in stage IIIC or IV ovarian cancer. N Engl J Med 2010;363:943–53.
  • Spiliotis J, Halkia E, Efstathiou E. Peritoneal carcinomatosis 2011. It’s about time for chemosurgery. J BUON 2011;16:400–8.
  • Rubin SC, Randall TC, Armstrong KA, Chi DS, Hoskins WJ. Ten-year follow-up of ovarian cancer patients after second-look laparotomy with negative findings. Obstet Gynecol 1999;93:21–4.
  • Markman M, Liu PY, Wilczynski S, Monk B, Copeland LJ, Alvarez RD, et al. Phase III randomized trial of 12 versus 3 months of maintenance paclitaxel in patients with advanced ovarian cancer after complete response to platinum and paclitaxel-based chemotherapy: A Southwest Oncology Group and Gynecologic Oncology Group trial. J Clin Oncol 2003;21:2460–5.
  • Gore ME, Fryatt I, Wiltshaw E, Dawson T, Robinson BA, Calvert AH. Cisplatin/carboplatin cross-resistance in ovarian cancer. Br J Cancer 1989;60:767–9.
  • Markman M, Rothman R, Hakes T, Reichman B, Hoskins W, Rubin S, et al. Second-line platinum therapy in patients with ovarian cancer previously treated with cisplatin. J Clin Oncol 1991;9:389–93.
  • Berek JS. Epithelial ovarian cancer. In: Berek JS, Hacker NF, editors Practical gynecologic oncology, 3rd ed. Philadelphia: Lippincott Williams and Wilkins; 2000. p 500.
  • Munkarah AR, Coleman RL. Critical evaluation of secondary cytoreduction in recurrent ovarian cancer. Gynecol Oncol 2004;95:273–80.
  • Galaal K, Naik R, Bristow RE, Patel A, Bryant A, Dickinson HO. Cytoreductive surgery plus chemotherapy versus chemotherapy alone for recurrent epithelial ovarian cancer. Cochrane Database Syst Rev 2010;6:CD007822.
  • Bristow RE, Puri I, Chi DS. Cytoreductive surgery for recurrent ovarian cancer: A meta-analysis. Gynecol Oncol 2009;112:265–74.
  • Spiliotis J, Vaxevanidou A, Sergouniotis F, Lambropoulou E, Datsis A, Christopoulou A. The role of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in the management of recurrent advanced ovarian cancer: A prospective study. J BUON 2011;16:74–9.
  • McGuire WP, Hoskins WJ, Brady MF, Kucera PR, Partridge EE, Look KY, et al.Cyclophosphamide and cisplatin versus paclitaxel and cisplatin: A phase III randomized trial in patients with suboptimal stage III/IV ovarian cancer (from the Gynecologic Oncology Group). Semin Oncol 1996;23(suppl12):40–7.
  • Muggia FM, Braly PS, Brady MF, Sutton G, Niemann TH, Lentz SL, Alvarez RD, et al. Phase III randomized study of cisplatin versus paclitaxel versus cisplatin and paclitaxel in patients with suboptimal stage III or IV ovarian cancer: a Gynecologic Oncology Group study. J Clin Oncol 2000;18:106–15.
  • Ozols RF, Bundy BN, Greer BE, Fowler JM, Clarke-Pearson D, Burger RA, et al. Gynecologic Oncology Group. Phase III trial of carboplatin and paclitaxel compared with cisplatin and paclitaxel in patients with optimally resected stage III ovarian cancer: A Gynecologic Oncology Group study. J Clin Oncol 2003;21:3194–200.
  • Alberts DS, Green S, Hannigan EV, O’Toole R, Stock-Novack D, Anderson P, et al. Improved therapeutic index of carboplatin plus cyclophosphamide versus cisplatin plus cyclophosphamide: Final report by the Southwest Oncology Group of a phase III randomized trial in stages III and IV ovarian cancer. J Clin Oncol 1992;10:706–17. Erratum in J Clin Oncol 1992;10:1505.
  • Alberts DS, Liu PY, Hannigan EV, O’Toole R, Williams SD, Young JA, et al. Intraperitoneal cisplatin plus intravenous cyclophosphamide versus intravenous cisplatin plus intravenous cyclophosphamide for stage III ovarian cancer. N Engl J Med 1996;335:1950–5.
  • Markman M, Bundy BN, Alberts DS, Fowler JM, Clark-Pearson DL, Carson LF, et al. Phase III trial of standard-dose intravenous cisplatin plus paclitaxel versus moderately high-dose carboplatin followed by intravenous paclitaxel and intraperitoneal cisplatin in small-volume stage III ovarian carcinoma: An intergroup study of the Gynecologic Oncology Group, Southwestern Oncology Group, and Eastern Cooperative Oncology Group. J Clin Oncol 2001;19:1001–7.
  • Armstrong DK, Bundy B, Wenzel L, Huang HQ, Baergen R, Lele S, et al. Intraperitoneal cisplatin and paclitaxel in ovarian cancer. N Engl J Med 2006;354:34–43.
  • National Cancer Institute. Comprehensive Cancer Information. Available at www.cancer.gov
  • Trimble EL, Christian MC. Intraperitoneal chemotherapy for women with advanced epithelial ovarian carcinoma. Gynecol Oncol 2006;100:3–4.
  • Eisenkop SM, Spirtos NM, Lin WC. Optimal cytoreduction for advanced epithelial ovarian cancer: A commentary. Gynecol Oncol 2006;103:329–35.
  • Fagotti A, Costantini B, Petrillo M, Vizzielli G, Fanfani F, Margariti PA, et al. Cytoreductive surgery plus HIPEC in platinum-sensitive recurrent ovarian cancer patients: A case-control study on survival in patients with two year follow-up. Gynecol Oncol 2012;127:502–5.
  • Stuart GC, Kitchener H, Bacon M, duBois A, Friedlander M, Ledermann J, et al. 2010 Gynecologic Cancer InterGroup (GCIG) consensus statement on clinical trials in ovarian cancer: report from the Fourth Ovarian Cancer Consensus Conference. Int J Gynecol Cancer 2011;73:750–5.
  • Morgan Jr RJ, Alvarez RD, Armstrong DK, Burger RA, Chen LM, Copeland L, et al. National comprehensive cancer networks. Ovarian cancer, version 2. 2013. J Natl Compr Canc Netw Oct 1 2013;11:1199–209.
  • González Martín A, Redondo A, Jurado M, De Juan A, Romero I, Bover I, et al. GEICO (Spanish Group for Investigation on Ovarian Cancer) treatment guidelines in ovarian cancer 2012. Clin Transl Oncol 2013;15:509–25.
  • Harter P, Mahner S, Hilpert F, Runnebaum I, Ortmann O, Mustea A, et al. Statement by the Kommission OVAR of the AGO Study Group on the use of HIPEC (hyperthermic intraperitoneal chemotherapy) to treat primary and recurrent ovarian cancer. Geburtshilfe Frauenheilkd 2013;73:221–3.
  • Chiva LM, Gonzalez-Martin A. A critical appraisal of hyperthermic intraperitoneal chemotherapy (HIPEC) in the treatment of advanced and recurrent ovarian cancer. Gynecol Oncol 2015;136:130–5.
  • du Bois A, Reuss A, Pujade-Lauraine E, Harter P, Ray-Coquard I, Pfisterer J. Role of surgical outcome as prognostic factor in advanced epithelial ovarian cancer: A combined exploratory analysis of 3 prospectively randomized phase 3 multicenter trials: by the Arbeitsgemeinschaft Gynaekologische Onkologie Studiengruppe Ovarialkarzinom (AGO-OVAR) and the Groupe d'Investigateurs Nationaux Pour les Etudes des Cancers de l'Ovaire (GINECO). Cancer 2009;115:1234–44.
  • Chi DS, Musa F, Dao F, Zivanovic O, Sonoda Y, Leitao MM, et al. An analysis of patients with bulky advanced stage ovarian, tubal, and peritoneal carcinoma treated with primary debulking surgery (PDS) during an identical time period as the randomized EORTC-NCIC trial of PDS vs neoadjuvant chemotherapy (NACT). Gynecol Oncol 2012;124:10–14.
  • Spiliotis JD, Rogdakis A, Vaxevanidou A, Datsis A, Zacharis G, Christopoulou A. Morbidity and mortality of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in the management of peritoneal carcinomatosis. J BUON 2009;14:259–64.
  • Rettenmaier MA, Mendivil AA, Gray CM, Chapman AP, Stone MK, Tinnerman EJ, Goldstein BH. Intra-abdominal temperature distribution during consolidation hyperthermic intraperitoneal chemotherapy with carboplatin in the treatment of advanced stage ovarian carcinoma. Int J Hyperthermia 2015;31:396–402.
  • Hayes-Jordan A, Lopez C, Green HL, Xiao LC, Huh W, Herzog CE. Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) in pediatric ovarian tumors: A novel treatment approach. Pediatr Surg Int 2015.
  • Hayes-Jordan A, Green H, Ludwig J, Anderson P. Toxicity of hyperthermic intraperitoneal chemotherapy (HIPEC) in pediatric patients with sarcomatosis/carcinomatosis: early experience and phase 1 results. Pediatr Blood Cancer 2012;59:395–7.
  • Deraco M, Raspagliesi F, Kusamura S. Management of peritoneal surface component of ovarian cancer. Surg Oncol Clin N Am 2003;12:561–83.
  • Kitayama J. Intraperitoneal chemotherapy against peritoneal carcinomatosis: Current status and future perspective. Surg Oncol 2014;23:99–106.
  • Ba M, Long H, Zhang X, Tang Y, Wu Y, Yu F, et al. Different sequential approaches of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in treating ovarian cancer with malignant ascites. J Cancer Res Clin Oncol 2014;140:1497–506.
  • Woopen H, Sehouli J. Current and future options in the treatment of malignant ascites in ovarian cancer. Anticancer Res 2009;29:3353–9.
  • Becker G, Galandi D, Blum HE. Malignant ascites: Systematic review and guideline for treatment. Eur J Cancer 2006;42:589–597.
  • Wong J, Koch AL, Deneve JL, Fulp W, Tanvetyanon T, Dessureault S. Repeat cytoreductive surgery and heated intraperitoneal chemotherapy may offer survival benefit for intraperitoneal mesothelioma: A single institution experience. Ann Surg Oncol 2014;21:1480–6.
  • Vaira M, Robella M, Mellano A, Sottile A, De Simone M. Iterative procedures combining cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for isolated peritoneal recurrence. Int J Hyperthermia 2014;30:565–9.
  • Roviello F, Roviello G, Petrioli R, Marrelli D. Hyperthermic intraperitoneal chemotherapy for the treatment of ovarian cancer: A brief overview of recent results. Crit Rev Oncol Hematol 2015;95:297–305.
  • Cihoric N, Tsikkinis A, van Rhoon G, Crezee H, Aebersold DM, Bodis S, et al. Hyperthermia-related clinical trials on cancer treatment within the ClinicalTrials.gov registry. Int J Hyperthermia 2015;31:609–14.

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.