Abstract
Background
Advanced epithelial ovarian cancer (EOC) is an incurable disease with over 75% of the patients developing recurrence in the peritoneum. Hyperthermic intraperitoneal chemotherapy (HIPEC) is a promising treatment option for both first-line therapy and treatment of recurrence. In this article, we review the rationale and current evidence for performing HIPEC and the role of HIPEC in the light of targeted systemic therapies.
Summary
There are few randomized trials and several retrospective studies on the role of HIPEC in the management of EOC. A 12-month-overall survival (OS) benefit of the addition of HIPEC to interval cytoreductive surgery (CRS) was demonstrated in 1 randomized trial following which HIPEC has been included as a treatment option for this indication in several national/international guidelines. One retrospective propensity score-matched analysis showed a 16-month OS benefit of adding HIPEC to primary CRS. One randomized trial showed no benefit of the addition of carboplatin HIPEC to secondary CRS over secondary CRS alone. For patients undergoing primary CRS and secondary CRS for recurrence, the results of ongoing randomized trials are needed to define the role of HIPEC in these situations. All clinical trials have shown that the morbidity of HIPEC performed after CRS is acceptable. Along with the emergence of HIPEC as a promising surgical therapy, targeted therapies like bevacizumab and poly adenosine diphosphate-ribose polymerase inhibitors have been developed that have shown a survival benefit in selected patients. In principle, HIPEC and targeted therapies work in different ways and it is plausible to assume that their benefit could be additive, and their combination should be evaluated in clinical trials. The impact of prognostic factors like the disease extent, pathological response to systemic chemotherapy (SC), the histological subtype and molecular profile on the benefit of HIPEC, and targeted therapies has not been evaluated in clinical trials.
Key Messages
HIPEC is an important therapeutic strategy in the treatment of EOC. While its role in patients undergoing interval CRS has been established, the results of ongoing randomized trials are needed to define its benefit at other time points. The morbidity of HIPEC in addition to CRS is acceptable. More research is needed to define subgroups that benefit most from HIPEC based on the extent of disease, response to SC, histology, and molecular profile. The combination of HIPEC and maintenance therapies should be evaluated in well-designed randomized clinical trials that evaluate not just the survival benefit and morbidity but also the cost-effectiveness of each therapy.
Keywords: Advanced ovarian cancer, Hyperthermic intraperitoneal chemotherapy, Interval cytoreductive surgery, Primary debulking surgery
Introduction
Advanced epithelial ovarian cancer (EOC), despite the advances in surgical and systemic therapies, remains an incurable disease. Over 75% of the patients develop recurrence/progression, and most of these recurrences are in the peritoneum itself [1]. This makes hyperthermic intraperitoneal chemotherapy (HIPEC) an attractive treatment option in both first-line therapy and recurrent disease. In the near past, most of the evidence favoring or disfavoring the use of HIPEC came from retrospective studies comprising heterogeneous groups of patients, with a greater heterogeneity in the treatment protocols, making it difficult to derive significant conclusions [2]. To date, 3 randomized clinical trials (RCTs) have been published, 2 favoring the use of HIPEC at different time points, 1 disfavoring its use [3, 4, 5]. Along with the emergence of HIPEC as a promising surgical therapy, targeted therapies like bevacizumab and poly adenosine diphosphate-ribose polymerase (PARP) inhibitors have been developed that have shown a survival benefit in selected patients in RCTs [6, 7, 8]. The combination of HIPEC with targeted therapies has not been evaluated in any RCT. Since the survival results in many of these trials are similar, there is skepticism regarding the benefit of HIPEC, given the added morbidity and cost. This is despite the fact that targeted therapies carry a significant morbidity and cost as well [9]. The median progression-free survival (PFS) in most clinical trials after first-line therapy ranges from 10 to 18 months and overall survival (OS) from 30 to 50 months (Table 1) [10, 11, 12, 13, 14]. Clearly, more effective treatments are needed to improve on these results. In principle, HIPEC and systemic therapies work in different ways. It is plausible to assume that the benefit of the 2 could be additive, and they should not be considered complementary treatments. HIPEC is unique as it is an adjuvant surgical therapy that is used in combination with cytoreductive surgery (CRS), and the outcomes are influenced greatly by the quality and extent of CRS. This makes it difficult to quantify and demonstrate the benefit of HIPEC in RCTs. In this article, we review the rationale for performing HIPEC, the current evidence for performing it at different time points, and the role of HIPEC in the light of targeted systemic therapies.
Table 1.
PFS and OS in patients with advanced ovarian cancer undergoing surgery and SC
| First author; year [ref] | N | Trial name | Comparator arms | Optimal debulking | CC-0 or complete gross resection | Median PFS | Median OS |
|---|---|---|---|---|---|---|---|
| Vergote et al. [10] | 670 | EORTC 55971/NCIC OV13 | PDS versus IDS | 41.6% (PDS) and 80.6% (IDS) | NR | 12 mo in both arms | 29 mo (PDS) and 30 mo (IDS) |
|
| |||||||
| Kehoe et al. [11] | 550 | CHORUS | PDS versus IDS | 28% (IIIC) 23% (IV) |
28% (IIIC) 30% (IV) |
107 mo (PDS) and 12 mo (IDS) | 22.6 mo (PDS) and 24.1 mo (IDS) |
|
| |||||||
| Fagotti et al. [12] | 171 | SCORPION (NCT01461850) | PDS versus IDS | 34.2% | 61.4% | 15 mo (PDS) and 14 mo (IDS) | 41 mo (PDS) and 43 mo (IDS) |
|
| |||||||
| Chi et al. [13] | 316 | Retrospective study | PDS versus IDS | 24% (PDS) 54% (IDS) | 71% (PDS) 86% (IDS) | 13 mo (IDS) and 17 mo (PDS) | 37 mo (IDS) and 50 mo (PDS) |
|
| |||||||
| Rauh-Hain et al. [14] | 242 | Retrospective study | PDS versus IDS | NR | 7.5% (PDS) and 27% (IDS) | 11 mo (PDS) and 14 mo (IDS) | 29 mo (PDS) and 33 mo (IDS) |
PDS, primary debulking surgery; IDS, interval debulking surgery; mo, months.
Rationale and Pitfalls of HIPEC
HIPEC involves the circulation of a heated chemotherapy solution in the peritoneal cavity for 30–120 min using a roller pump and heat exchanger (or custom-made machines). The theoretical benefits of HIPEC include a direct exposure of tumor cells to the drug, prolonged retention of the drug in the peritoneal cavity resulting in prolonged exposure of the peritoneal surfaces to the drug, and a decreased systemic absorption resulting in reduced systemic toxicity [15, 16]. Thus, higher drug doses can be administered without the resultant systemic toxicity.
Intraperitoneally administered chemotherapeutic agents gain access to peritoneal lymphatics and can act on trans-lymphatic spread [17, 18]. Since these lymphatics are connected sub-peritoneally, the drug can reach different areas of the sub-peritoneal space though the mechanical distribution is uneven, and some areas are underexposed or not exposed at all to the drug [17, 18].
Despite this pharmacokinetic advantage, the depth of penetration of the chemotherapeutic drug into the peritoneum and tumor nodules is not more than 2–3 mm [19, 20]. Thus, HIPEC acts best in the presence of minimal or no residual disease. Following complete CRS, HIPEC can address the free intraperitoneal cancer cells more effectively and reduces the implantation of these cells at the surgical site [21].
There are many theoretical benefits of hyperthermia, but the clinical benefit has not been proven for the treatment of a peritoneal metastasis (PM). Heat itself is cytotoxic; it potentiates the cytotoxicity of the chemotherapeutic drugs and in some instances, helps in overcoming drug resistance [22, 23].
HIPEC Methodology and Factors Influencing the Efficacy of HIPEC
HIPEC is a surgical therapy that is performed by the open, semi-open, or closed methods. There are 3 main components of a HIPEC procedure that could influence its efficacy:
Pharmacological factors having an impact on the efficacy of HIPEC include the pharmacokinetic variables like the drug (combination), dosage, molecular weight, carrier solution and its volume, application time and intra-abdominal pressure and pharmacodynamic variables like tumor nodule size, density, vascularity, interstitial fluid pressure, binding and temperature [25]. The nonpharmacological factors are the extent of peritoneal resection, the method (open or closed), and the chemosensitivity of the tumors cells to the chemotherapeutic agent(s) used.
Even though the mechanical part can be standardized, it is not possible to standardize the other 2 as innumerable permutations and combinations are possible. The drugs with the best pharmacokinetic properties often are not the ones that have the greatest activity against a particular tumor type [26]. Thus, in clinical practice, there is heterogeneity in the HIPEC methodology and the drug/regimen used [26].
Drugs and Drug Regimens for HIPEC in EOC
Cisplatin alone or combined with other drugs is the most commonly used drug for performing HIPEC (Table 2). Cisplatin HIPEC showed a survival benefit in the OVIHIPEC 1 trial and is the drug being used in several other clinical trials [4]. The dose of cisplatin in the OVIHIPEC 1 trial was 100 mg/m2 with half the dose being administered at the beginning of HIPEC and a quarter each at 30 and 60 min.
Table 2.
Various drug regimens used for performing HIPEC in ovarian cancer [4, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37]
| Regimen | IP drugs | IV drugs | Carrier solution | Duration, min | Indications | |
|---|---|---|---|---|---|---|
| OVIHIPEC 1 (and 2) cisplatin regimen [4] | Cisplatin 100 mg/m2 | Sodium thiosulfate for renal protection | Peritoneal dialysis solution | 90 | Advanced ovarian cancer | |
|
| ||||||
| CHIPASTIN/French cisplatin regimen [27] | Cisplatin 70 mg/m2 | Peritoneal dialysis solution | 60 | Advanced ovarian cancer | ||
|
| ||||||
| HIPEC ROC-1 regimen [28] | Cisplatin 100 mg/m2 | Peritoneal dialysis solution | 90 | Recurrent ovarian cancer | ||
|
| ||||||
| Paclitaxel [29] | Paclitaxel 175 mg/m2 | Peritoneal dialysis solution | 90 | Advanced ovarian cancer | ||
|
| ||||||
| Cisplatin and paclitaxel [30] | Paclitaxel 175 mg/m2 and cisplatin 100 mg/m2 | Peritoneal dialysis solution | 90 | Advanced ovarian cancer | ||
|
| ||||||
| Carboplatin regimen [31] | Carboplatin 800–1,200 mg/m2 | Peritoneal dialysis solution | 90 | Advanced and recurrent ovarian cancer | ||
|
| ||||||
| Sugarbaker regimen [32] | Cisplatin (50 mg/m2) adriamycin (15 mg/m2) | Ifosfamide 1300 mg/m2 and mesna 260 mg/m2 | Peritoneal dialysis solution | 90 | Advanced and recurrent ovarian cancer | |
|
| ||||||
| National Cancer Institute Milan regimen [33] | Doxorubicin 15.25 mg/L cisplatin 43 mg/L | 90 | Advanced and recurrent ovarian cancer | |||
|
| ||||||
| High-dose oxaliplatin regimen [34] | Oxaliplatin 460 mg/m2 | Dextrose 5% | 30 | Recurrent | ||
|
| ||||||
| Sugarbaker's gemcitabine-based regimen [35] | Gemcitabine 1,000 mg/m2 | Peritoneal dialysis solution | 90 | Recurrent ovarian cancer | ||
|
| ||||||
| Sugarbaker melphalan-based regimen [36] | Melphalan 50–70 mg/m2 | Peritoneal dialysis solution | 90 | Recurrent ovarian cancer | ||
|
| ||||||
| Sugarbaker liposomal doxorubicin-based regimen [37] | Liposomal doxorubicin 50–100 mg/m2 | Peritoneal dialysis solution | 180 | Recurrent ovarian cancer | ||
Though the pharmacokinetic profile is less favorable, cisplatin is an adequate drug for hyperthermia, and hyperthermia in experimental studies overcomes platinum resistance [38, 39, 40, 41, 42, 43]. The pharmacokinetic profile of carboplatin is similar to that of cisplatin with fewer side effects, but thermal enhancement is poor, and the depth of penetration in tumor nodules is much lower [38, 44, 45]. In a recently published phase II trial, there was no benefit of the addition carboplatin HIPEC to secondary CRS and 5 cycles of systemic chemotherapy (SC) compared to secondary CRS with 6 cycles of SC [5]. Doxorubicin is not an active drug for SC in ovarian cancer but has a highly favorable pharmacokinetic profile and low toxicity that has led to its use for performing HIPEC usually in combination with cisplatin [46, 47, 48].
The taxols, paclitaxel and docetaxel, are large molecules with favorable pharmacokinetic profiles [49, 50, 51]. The main disadvantage is the lack of thermal enhancement [51, 52]. Moreover, taxols are cell-cycle-specific, i.e., it is active only in the mitotic phase of the cell division [51, 52]. An ideal drug for HIPEC should preferably be cell-cycle-nonspecific as HIPEC is a single shot treatment. HIPEC with paclitaxel is being evaluated in the HIPECOVA trial (NCT02681432) for advanced and recurrent EOC both.
Gemcitabine, melphalan, and oxalipatin are the other drugs that have been used for HIPEC in EOC [34, 35, 36]. None of these drugs is being evaluated in RCTs. Currently, 2 clinical trials (NCT01970722 and NCT02567253) are comparing different HIPEC regimens for advanced EOC.
Current Evidence on the Benefit of HIPEC at Different Time Points in the History of Ovarian Cancer
For advanced EOC, surgery is performed during first-line therapy either upfront (primary CRS) or after a few cycles of neoadjuvant chemotherapy (interval CRS). For recurrent disease, it is performed for selected patients with platinum-sensitive (PS) recurrence.
Primary CRS/Debulking
Most of the evidence for performing HIPEC in addition to primary CRS comes from retrospective studies that included patients undergoing surgery for different indications, had a long recruitment period, small number of patients (less than 100 in the majority of studies), and used heterogeneous treatment protocols and SC regimens. The selection criteria were not clearly defined, and several series comprised patients with extensive disease that were referred to specialized units after having undergone unsuccessful prior surgical attempts [2].
There are 3 systematic reviews and meta-analyses; the first one by Chiva et al. [53] included 11 studies, comprising 248 patients. The authors reported no benefit of adding HIPEC to primary CRS [53]. Huo et al. [54] performed a meta-analysis of 9 comparative and 28 noncomparative studies, evaluating the role of HIPEC for primary and recurrent EOC. Contrary to the first meta-analysis, in the primary setting, this meta-analysis performed on patients of 3 studies showed a survival benefit with HIPEC at 2–5 and 8 years. These studies included patients with stages II-III-B EOC as well and did not make a distinction between patients undergoing primary and interval CRS. In a subgroup analysis of patients with stage III and IV alone, the odds ratio for OS was 3.61 (95% CI: 1.55–8.37; p = 0.003) at 3 years, and for OS at 5 years, it was 2.25 (95% CI: 1.07–4.71; p = 0.030), both favoring the use of HIPEC [54]. Zhang et al. [55] performed a similar meta-analysis and demonstrated a benefit of HIPEC for all patients undergoing primary or interval CRS (HR = 0.59, 95% CI: 0.46–0.75 for OS and HR = 0.41, 95% CI: 0.32–0.54 for PFS) as well as those with stage III or IV disease (HR = 0.64, 95% CI: 0.50–0.82 for OS and HR = 0.36, 95% CI: 0.20–0.65 for PFS) [55].
Lei et al. [56] have recently published a retrospective propensity score-matched study including patients with EOC undergoing primary CRS with (N = 425) or without (N = 159) HIPEC [55]. HIPEC was performed with 50 mg/m2 of cisplatin on days 1, 3, and 5, which is not the common practice [56].
At a median follow-up of 42.2 months, the addition of HIPEC led to a nearly 16-month benefit in OS (median OS, 49.8 m; 95% CI, 45.2–60.2 m for CRS and HIPEC vs. 34.0 m; 95% CI, 28.9–42.3 months for CRS alone). After inverse probability of treatment weighting adjustment, primary CRS with HIPEC was still associated with better oncologic outcomes, with a median OS of 49.8 (95% CI, 45.2–60.2) months compared 34.0 (95% CI, 28.9–41.5) months in the primary CRS alone group (weighted HR, 0.64; 95% CI, 0.50–0.82; p < 0.001). This implies that there was a 36% reduction in the risk of death with the use of HIPEC. The grade 3–4 morbidity and hematological toxicity were similar in both groups except electrolyte imbalance which was seen in a significantly higher number of patients in the HIPEC group. All the limitations of a randomized study and a retrospective analysis apply to this study.
In the only RCT reported so far, Lim et al. [57] reported no benefit of the addition of HIPEC to primary CRS. One hundred and eighty-four patients undergoing both primary and interval CRS were randomized to undergo HIPEC or no HIPEC. Though the trial has been completed, the final results have not been reported or published. The small sample size, the inclusion or patients undergoing both primary and interval CRS, and performing HIPEC for patients with optimal debulking (residual disease <1 cm) are some of the limitations of this trial [57]. There are 4 RCTs in progress looking at the role of HIPEC in addition to primary CRS (Table 3): the OVIHIPEC 2 trial in Europe, Australia, and the USA (NCT03772028); the CHIPPI trial in France (NCT03842982); and the HIPECOC trial (NC04280185) and EHTASEOCCS trial (NCT03373058) both in China.
Table 3.
Published and ongoing clinical trials on the role of HIPEC in advanced ovarian cancer
| ClinicalTrials.gov ID | Phase; primary institution/group | Indication | HIPEC regimen(s) | Control arm | Experimental arm | Main outcomes |
|---|---|---|---|---|---|---|
| Van Driel et al. [4] | Phase 3 randomized controlled trial; Netherlands Cancer Institute | Advanced ovarian cancer (interval CRS) | Cisplatin 100 mg/m2 for CRS + SC 90 min | CRS + HIPEC + SC | Benefit in the HIPEC arm in PFS and OS; similar morbidity | |
|
| ||||||
| Lim et al. [56] Only abstract; full results not available | Phase 3 randomized controlled trial; National cancer Centre, Korea | Advanced ovarian cancer (PDS and IDS) | Cisplatin 75 mg/m2 for CRS + SC (92) 90 min | CRS + HIPEC + SC (92) | No benefit of HIPEC in the PDS arm; benefit with HIPEC in PFS and OS after 20 mo of follow-up in the IDS arm | |
|
| ||||||
| NCT03842982 (CHIPPI) | Randomized phase III; Centre Oscar Lambert | Advanced ovarian, FT, and PP cancer | Cisplatin 100 mg/m2 for PDS/IDS + SC 90 min | PDS/IDS + HIPEC + SC | ||
| NCT02349958 | Non randomized; Bay Area Gynecology Oncology, the USA | Advanced ovarian cancer; other primary sites | Various regimens | CRS + HIPEC + SC | ||
|
| ||||||
| NCT02124421 (HOT trial) | Mercy Medical Center, the USA | Advanced ovarian cancer | Carboplatin (AUC 6) for CRS + SC + IPC 90 min | CRS + HIPEC + SC | ||
|
| ||||||
| NCT01970722 | City of Hope Medical center, UC | Advanced ovarian, FT, uterine, and PP cancer | Various regimens | CRS + HIPEC + SC + IPC | ||
|
| ||||||
| NCT02567253 (OvIP1) | Quadruple arm, phase II; University Hospital, Ghent, Belgium | Advanced ovarian cancer | Cisplatin 75 mg/m2 NT or HT for 90 min Cisplatin 100 mg/m2 NT or HT for 90 min | CRS + HIPEC + SC | ||
| NCT01628380 | Randomized, phase III; A.O. Ospedale Papa Giovanni XXIII, Italy | Advanced ovarian cancer | Cisplatin 100 mg/m2 CRS + SC +paclitaxel 175 mg/m2* | CRS + HIPEC + SC | ||
| NCT02681432 | Randomized, phase III; Hospital General de la Ciudad Real, Spain | Advanced ovarian cancer | Paclitaxel 175 mg/m2 for CRS + SC 60 min | CRS + HIPEC + SC | ||
PDS, primary debulking surgery; IDS, interval debulking surgery.
Time not specified.
Interval CRS/Debulking
The OVIHIPEC 1 trial showed a benefit in both PFS and OS with the addition of HIPEC to interval CRS compared to interval CRS alone. [4] The use of HIPEC led to a reduction in the risk of death by 33% (HR: 0.67; 95% CI: 0.48–0.94; p value = 0.02) [4]. Though the morbidity and mortality were similar in both arms, certain complications like infection and thromboembolism and the rate of stoma formation were more in the HIPEC arm. There were many criticisms of this trial [58, 59]. The survival rates (PFS-10 months and OS-41 months) were similar to studies in which no HIPEC was used. This trial had a very stringent follow-up protocol and strict criteria for diagnosis of recurrence due to which the recurrences may have been reported earlier than usual. Second, since randomization was performed intraoperatively, survival was calculated from the date of surgery and not the time of starting neoadjuvant chemotherapy (approximately reduction of 3 m in PFS and OS). The intraoperative randomization was considered improper and fraught with bias. However, HIPEC is performed only in patients with residual disease measuring less than 2.5 mm. This is because only those patients in whom a complete CRS is feasible or has already been achieved can be randomized to evaluate the benefit of HIPEC over CRS alone. The sample size was considered small and the primary end point, i.e., PFS, was not met with. The long recruitment period of 9 years and reduced benefit in patients recruited at the most experienced centers were some other criticisms. Despite these limitations, an OS benefit of 12 months cannot be ignored, and HIPEC has now been included in many national and international guidelines including the NCCN guidelines as a treatment option for patients undergoing interval CRS [60, 61].
In the trial from Korea discussed in the previous section, there was a trend toward a longer PFS and OS with HIPEC in patients who had a follow-up of more than 2 years [57]. The trial has been completed, but the final results have not been disclosed or published. Ongoing clinical trials on HIPEC in addition to interval CRS are listed in Table 3.
Recurrent Ovarian Cancer
In the recurrent setting, the role of secondary CRS itself was put to question by the results of 1 RCT that showed no difference in patients treated with SC and bevacizumab with or without CRS [62]. However, 2 subsequent RCTs have shown a survival benefit of secondary CRS in selected patients with platinum-sensitive recurrence [63, 64]. Similar to the upfront setting, the majority of data evaluating HIPEC in recurrent ovarian cancer with PS and platinum-resistant (PR) disease has been based on retrospective studies [2]. Several case control studies have compared secondary CRS and HIPEC with CRS alone [65, 66, 67, 68, 69, 70]. In 4 of these, there was a statistically significant benefit of CRS and HIPEC over CRS alone (Table 4). Most of these studies have a small number of patients, and 1 study combined patients with advanced and recurrent disease both.
Table 4.
Case control studies comparing CRS and HIPEC with CRS alone as second-line therapy
| Ref No | Year of publication | Type of recurrence | N | CRS + HIPEC | CRS | Survival for CRS + HIPEC | Survival for CRS alone | p value |
| Munoz-casarez et al. [65] | 2009 | NS | 26 | 14 | 12 | 58% (5 yr OS) | 17% (5 yr OS) | 0.011 |
| Spillitois et al. [66] | 2011 | NS | 48 | 24 | 24 | 50% (3 yr OS) | 18% (3 yr OS) | <0.01 |
| Fagotti et al. [67] | 2012 | PS | 67 | 30 | 37 | 68% (5 yr OS) | 42% (5 yr OS) | 0.017 |
| Cascales Campos et al. [68] | 2014 | PS | 54 | 32 | 22 | 45 (3 yr DFS) | 23 (3 yr DFS) | 0.078 |
| Le Brun et al. [69] | 2014 | PS | 42 | 23 | 19 | 75.6% (4 yr OS) | 19.4% (4 yr OS) | 0.013 |
| Biaocchi et al. [70] | 2016 | PS | 79 | 29 | 50 | 59.3 mo (median OS) | 58.3 mo (median OS) | 0.95 |
NS, not specified; DFS, disease-free survival; mo, months; yr, years.
There are 2 RCTs that have been published (Table 5). In the first one by Spiliotis et al. [3] that included 120 patients with both platinum-sensitive and PR disease, patients undergoing HIPEC had a significantly longer OS compared to those undergoing CRS alone. This trial had several methodological limitations: use of heterogeneous chemotherapy regimens and the morbidity was not reported.
Table 5.
Published and ongoing clinical trials evaluating the role of HIPEC in recurrent ovarian cancer
| First author [ref] | Phase | Indication | Primary institution/group | HIPEC drugs | Control (N) | Experimental arm (N) | Main outcomes |
| Spiliotis et al [3] | Phase 3 randomized controlled trial | Recurrent ovarian cancer (PS and PR) | Metaxa Cancer Hospital, Greece | PS: cisplatin 100 mg/m2 and paclitaxel 175 mg/m2 for 60 min PR: doxorubicin 35 mg/m2 and (paclitaxel 175 mg/m2 or mitomycin C 15 MG/M2) | CRS + SC (60) | CRS + HIPEC SC (60) | HIPEC resulted in the superior OS; significant difference in the PS group but not in the PR group |
|
| |||||||
| Zivanovic et al [5] | Phase 2 randomized controlled trial | PS recurrent ovarian cancer | Memorial Sloan Kettering Cancer Centre | Carboplatin 800 mg/m2 for 90 min | CRS + 6#SC (49) | CRS + HIPEC + 5# SC (49) | No difference in survival between the 2 arms; similar morbidity |
|
| |||||||
| NCT01539785 (HORSE/MITO 18) | Randomized, phase III | PS recurrence | Catholic University of the Sacred Heart, Italy | Cisplatin 75 mg/m2 for 60 min | CRS + SC | CRS + HIPEC + SC | |
|
| |||||||
| NCT01376752 (CHIPOR) | Randomized, phase III | PS recurrence | UNICANCER Europe | Cisplatin 75 mg/m2 for 60 min | SC + CRS | SC + CRS + HIPEC | |
|
| |||||||
| NCT03220932 (HIPOVA-1) | Randomized, phase III | PR recurrence | UNICANCER Europe | Cisplatin 70 mg/m2 | SC + Bev | SC + Bev + CRS + HIPEC | |
|
| |||||||
| NCT02672098 | Phase 1 | PS recurrence | Loma Linda University Cancer Center, the USA | Carboplatin 80 mg/m2 for 90 min | CRS + HIPEC +SC | ||
Bev, bevacizumab.
In the other trial by Zivanovic et al. [5] that had a pick-the-winner superiority design, 98 patients were randomized to either secondary CRS with carboplatin HIPEC or CRS alone. Patients in the HIPEC arm received only 5 cycles of adjuvant chemotherapy compared to 6 in the CRS alone arm. There was no difference in the median PFS (15.4 vs. 12.3 months; p = 0.17) or median OS (69.2 vs. 53.1 months; p = 0.32) between the 2 arms, and the morbidity and mortality between the 2 was also similar [5]. There were significantly fewer bowel anastomoses in the HIPEC arm, and a low dose of carboplatin was used. The authors stated that the results should be interpreted with caution since the trial design did not allow a direct arm-to-arm comparison, and the conclusions were based on a post hoc analysis. There are 2 phase III trials evaluating the role of HIPEC for PS recurrence − the CHIPOR (NCT01376752) trial in France and the HORSE (NCT01539785) trial in Italy. Another trial being conducted in France, the HIPOVA-1 trial (NCT03220932), is evaluating the role of HIPEC in PR disease.
Clinical End Points for Evaluating the Benefit of HIPEC
The OVIHIPEC 1 trial showed a greater benefit of HIPEC in terms of OS compared to PFS, the primary end point. There are 2 plausible reasons for this unexpected finding. As a locoregional treatment, HIPEC is expected to result in better locoregional control and therefore delayed recurrence. In advanced EOC which is an incurable disease, the time to second-line platinum therapy or the platinum-free interval is an important prognostic factor that has an impact on OS [71, 72]. By delaying recurrence, this interval is prolonged which could result in a longer OS. By eradicating microscopic disease, the response to second-line therapy could be better. PMs lead to ascites and bowel obstruction and bring down the quality of life. For the PM of gastrointestinal origin, obstruction-free survival has been proposed as a suitable end point to demonstrate the benefit of HIPEC [73]. One retrospective study on the patterns of recurrence following interval CRS and HIPEC showed a reduced incidence of peritoneal recurrence compared to previous reports that could be attributed to the use of HIPEC [74]. Theoretically, HIPEC should not just prolong PFS but OS as well.
In addition to aggressive locoregional treatment, the time to recurrence is influenced by another factor, i.e., the response to SC. Böhm et al. [75] demonstrated a 50% longer PFS in patients with complete or near complete response to SC compared to the moderate and poor responders. Contrary to the results obtained with HIPEC, the difference in OS between the different responders was less compared to the difference in PFS between these groups. Locoregional therapies should have a greater impact on the OS compared to the response to SC by reducing the burden of chemotherapy-resistant cells and thus enhancing the response to subsequent therapies. Thus, though HIPEC is a locoregional therapy, it is plausible that OS is a better primary end point compared to PFS for evaluating its benefit in clinical trials. A recent systematic review of clinical trials in ovarian cancer demonstrated that PFS is not an established surrogate for OS in ovarian cancer [76]. The results suggested that when PFS is the primary end point, OS should be a secondary end point.
The Role of HIPEC in BRCA-Mutated Tumors and Patients Receiving PARP Inhibitors and Bevacizumab
Both germ line and somatic BRCA mutations in EOC have been associated with a greater response to SC and a prolonged OS [77]. The use of PARP inhibitors as maintenance therapies in these patients results in a greater benefit in PFS compared to those without such mutations in the first-line and recurrent setting both [9].
It is assumed that HIPEC may have a lesser or no benefit in such patients given their favorable prognosis. It must be borne in mind however that HIPEC is a surgical therapy and has a different mechanism of action compared to the targeted at systemic therapies. HIPEC acts on microscopic residual disease as well as free cancer cells shed during surgery minimizing their implantation at sites of resection. It could thus be assumed that the opposite is true − HIPEC adds to the benefit of both systemic and targeted therapies in patients with BRCA-mutated tumors, though there is no evidence to support either claim. There are other factors that could be used to define subgroups that are likely to derive maximum benefit from each therapy. The disease extent (as estimated by the peritoneal cancer index [PCI]) has still not been established as a robust prognostic factor in advanced ovarian cancer though several studies have shown inferior survival in patients with a PCI >20 [78, 79]. With more extensive disease, the probability of residual disease is also likely to be higher, and perhaps, this is 1 subgroup in which HIPEC could have a greater role compared to those with a lower PCI even with BRCA-mutated tumors. The other important factor to be considered is the pathological response to SC [75]. Patients who have a complete or near complete response are likely to have less microscopic residual disease as well and could represent a subgroup that derives less benefit from HIPEC. The only drawback is that it is known only after completion of surgery.
Similarly, maintenance therapy with bevacizumab has shown a benefit in PFS but added no benefit to the OS [80, 81]. A benefit in OS was only seen in patients who had suboptimal debulking or residual disease measuring more than 1 cm in size [82, 83]. Bevacizumab is a cytostatic drug [84]. Thus, it can best control the disease but cannot lead to a complete response as demonstrated with PARP inhibitors. No study has looked at the impact of HIPEC in patients receiving bevacizumab.
There are some histological subtypes like the serous and mucinous variety that may derive a greater benefit from HIPEC. Another important factor is the cost-effectiveness of each of these therapies especially in healthcare systems where such treatments are not covered by public or private insurance. A cost-effectiveness study on the OVIHIPEC 1 data demonstrated that HIPEC was cost-effective in the Netherlands for this indication [85]. Contrary to this, a study from the USA published in 2020 concluded that none of the maintenance therapies was cost-effective regardless of the molecular signature [86].
Conclusions
HIPEC is an important therapeutic strategy in the treatment of ovarian cancer. While its role in patients undergoing interval CRS has been established, the results of ongoing randomized trials are needed to define its benefit at other time points. The morbidity of HIPEC in addition to CRS is acceptable. More research is needed to define subgroups that benefit most from HIPEC at different time points based on the extent of disease, response to SC, histology, and molecular profile. The combination of HIPEC and maintenance therapies should be evaluated in well-designed RCTs that evaluate not just the survival benefit and morbidity but also the cost-effectiveness of each therapy.
Conflict of Interest Statement
O. Glehen is a consultant for Gamida. A. Bhatt has no disclosures. The authors have no conflicts of interest to declare.
Funding Sources
The authors received no funding for this study.
Author Contributions
Aditi Bhatt was responsible for conceptualizing this review, performing the review, interpretation, original draft, reviewing, and editing. Olivier Glehen was responsible for conceptualization, reviewing, and editing the manuscript. Both the authors approved the final version of the manuscript.
References
- 1.Colombo N, Lorusso D, Scollo P. Impact of recurrence of ovarian cancer on quality of life and outlook for the future. Int J Gynecol Cancer. 2017 Jul;27((6)):1134–40. doi: 10.1097/IGC.0000000000001023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bhatt A, Glehen O. The role of Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy (HIPEC) in ovarian cancer: a review. Indian J Surg Oncol. 2016 Jun;7((2)):188–97. doi: 10.1007/s13193-016-0501-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Spiliotis J, Halkia E, Lianos E, Kalantzi N, Grivas A, Efstathiou E, et al. Cytoreductive surgery and HIPEC in recurrent epithelial ovarian cancer: a prospective randomized phase III study. Ann Surg Oncol. 2015 May;22((5)):1570–5. doi: 10.1245/s10434-014-4157-9. [DOI] [PubMed] [Google Scholar]
- 4.van Driel WJ, Koole SN, Sikorska K, Schagen van Leeuwen JH, Schreuder HWR, Hermans RHM, et al. Hyperthermic intraperitoneal chemotherapy in ovarian cancer. N Engl J Med. 2018;378((3)):230–40. doi: 10.1056/NEJMoa1708618. [DOI] [PubMed] [Google Scholar]
- 5.Zivanovic O, Chi DS, Zhou Q, Iasonos A, Konner JA, Makker V, et al. Secondary cytoreduction and carboplatin hyperthermic intraperitoneal chemotherapy for platinum-sensitive recurrent ovarian cancer: an MSK Team Ovary Phase II Study. J Clin Oncol. 2021 Aug 10;39((23)):2594–604. doi: 10.1200/JCO.21.00605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Tewari KS, Burger RA, Enserro D, Norquist BM, Swisher EM, Brady MF, et al. Final overall survival of a randomized trial of bevacizumab for primary treatment of ovarian cancer. J Clin Oncol. 2019 Sep 10;37((26)):2317–28. doi: 10.1200/JCO.19.01009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Moore K, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, et al. Maintenance olaparib in patients with newly diagnosed advanced ovarian cancer. N Engl J Med. 2018;379:2495–505. doi: 10.1056/NEJMoa1810858. [DOI] [PubMed] [Google Scholar]
- 8.Ray-Coquard I, Pautier P, Pignata S, Pérol D, González-Martín A, Berger R, et al. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N Engl J Med. 2019 Dec 19;381((25)):2416–28. doi: 10.1056/NEJMoa1911361. [DOI] [PubMed] [Google Scholar]
- 9.Mirza MR, Pignata S, Ledermann JA. Latest clinical evidence and further development of PARP inhibitors in ovarian cancer. Ann Oncol. 2018 Jun 1;29((6)):1366–76. doi: 10.1093/annonc/mdy174. [DOI] [PubMed] [Google Scholar]
- 10.Vergote I, Trope 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. doi: 10.1056/NEJMoa0908806. [DOI] [PubMed] [Google Scholar]
- 11.Kehoe S, Hook J, Nankivell M, Jayson GC, Kitchener H, Lopes T, et al. Primary chemotherapy versus primary surgery for newly diagnosed advanced ovarian cancer (CHORUS): an open-label, randomised, controlled, non-inferiority trial. Lancet. 2015;386:249–57. doi: 10.1016/S0140-6736(14)62223-6. [DOI] [PubMed] [Google Scholar]
- 12.Fagotti A, Ferrandina MG, Vizzielli G, Pasciuto T, Fanfani F, Gallotta V, et al. Randomized trial of primary debulking surgery versus neoadjuvant chemotherapy for advanced epithelial ovarian cancer (SCORPION- NCT01461850) Int J Gynecol Cancer. 2020;30:1657–64.. doi: 10.1136/ijgc-2020-001640. [DOI] [PubMed] [Google Scholar]
- 13.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 Jan;124((1)):10–4. doi: 10.1016/j.ygyno.2011.08.014. [DOI] [PubMed] [Google Scholar]
- 14.Rauh-Hain JA, Rodriguez N, Growdon WB, Goodman AK, Boruta DM, 2nd, Horowitz NS, et al. Primary debulking surgery versus neoadjuvant chemotherapy in stage IV ovarian cancer. Ann Surg Oncol. 2012 Mar;19((3)):959–65. doi: 10.1245/s10434-011-2100-x. [DOI] [PubMed] [Google Scholar]
- 15.Flessner MF. The transport barrier in intraperitoneal therapy. Am J Physiol Renal Physiol. 2005;288((3)):F433–42. doi: 10.1152/ajprenal.00313.2004. [DOI] [PubMed] [Google Scholar]
- 16.Flessner MF. Intraperitoneal drug therapy: physical and biological principles. Cancer Treat Res. 2007;134:131–52. doi: 10.1007/978-0-387-48993-3_8. [DOI] [PubMed] [Google Scholar]
- 17.Yonemura Y. Trans-lymphatic metastasis. In: Yonemura Y, editor. Atlas and principles of peritonectomy for peritoneal surface malignancy. Kyoto: NPO to Support Peritoneal Surface Malignancy; 2012. pp. p. 188–206. [Google Scholar]
- 18.Yonemura Y, Canbay E, Endou Y, Ishibashi H, Mizumoto A, Miura M, et al. Mechanisms of the formation of peritoneal surface malignancy on omental milky spots from low grade appendiceal mucinous carcinoma. J Clin Exp Oncol. 2014;3:3. [Google Scholar]
- 19.Solass W, Herbette A, Schwarz T, Hetzel A, Sun JS, Dutreix M, et al. Therapeutic approach of human peritoneal carcinomatosis with Dbait in combination capnoperitoneum: proof of concept. Surg Endosc. 2012;26((3)):847–52. doi: 10.1007/s00464-011-1964-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Khosrawipour V, Khosrawipour T, Diaz-Carballo D, Förster E, Zieren J, Giger-Pabst U. Exploring the spatial drug distribution pattern of pressurized Intraperitoneal aerosol chemotherapy (PIPAC) Ann Surg Oncol. 2016;23((4)):1220–4. doi: 10.1245/s10434-015-4954-9. [DOI] [PubMed] [Google Scholar]
- 21.Yonemura Y, Sako S, Wakama S, Ishibashi H, Mizumoto A, Takao N, et al. History of peritoneal surface malignancy treatment in Japan. Indian J Surg Oncol. 2019 Feb;10((Suppl 1)):3–11. doi: 10.1007/s13193-019-00893-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sticca RP, Dach BW. Rationale for hyperthermia with intraoperative intraperitoneal chemotherapy agents. Surg Oncol Clin N Am. 2003;12:689–701. doi: 10.1016/s1055-3207(03)00029-2. [DOI] [PubMed] [Google Scholar]
- 23.Sugarbaker PH. Laboratory and clinical basis for hyperthermia as a component of intracavitary chemotherapy. Int J Hyperthermia. 2007;23:431–42. doi: 10.1080/02656730701455318. [DOI] [PubMed] [Google Scholar]
- 24.Glehen O, Mohamed F, Gilly FN. Peritoneal carcinomatosis from digestive tract cancer: new management by cytoreductive surgery and intraperitoneal chemohyperthermia. Lancet Oncol. 2004 Apr;5((4)):219–28. doi: 10.1016/S1470-2045(04)01425-1. [DOI] [PubMed] [Google Scholar]
- 25.de Bree E, Michelakis D, Stamatiou D, Romanos J, Zoras O. Pharmacological principles of intraperitoneal and bidirectional chemotherapy. Pleura Peritoneum. 2017;2((2)):47–62. doi: 10.1515/pp-2017-0010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Bhatt A, de Hingh I, Van Der Speeten K, Hubner M, Deraco M, Bakrin N, et al. HIPEC methodology and regimens: the need for an expert consensus. Ann Surg Oncol. 2021;28((13)):9098–113. doi: 10.1245/s10434-021-10193-w. [DOI] [PubMed] [Google Scholar]
- 27.Gouy S, Ferron G, Glehen O, Bayar A, Marchal F, Pomel C, et al. Results of a multicenter phase I dose-finding trial of hyperthermic intraperitoneal cisplatin after neoadjuvant chemotherapy and complete cytoreductive surgery and followed by maintenance bevacizumab in initially unresectable ovarian cancer. Gynecol Oncol. 2016;142((2)):237–42. doi: 10.1016/j.ygyno.2016.05.032. [DOI] [PubMed] [Google Scholar]
- 28.Zivanovic O, Abramian A, Kullmann M, Fuhrmann C, Coch C, Hoeller T, et al. HIPEC ROC I: a phase 1 study of cisplatin administered as hyperthermic intraoperative intraperitoneal chemoperfusion followed by postoperative intravenous platinum-based chemotherapy in patients with platinum-sensitive recurrent epithelial ovarian cancer. Int J Cancer. 2015;136:699–708. doi: 10.1002/ijc.29011. [DOI] [PubMed] [Google Scholar]
- 29.De Bree E, Rosing H, Michalakis J, Romanos J, Relakis K, Theodoropoulos PA, et al. Intraperitoneal chemotherapy with taxanes for ovarian cancer with peritoneal dissemination. Eur J Surg Oncol. 2006;32:666–70. doi: 10.1016/j.ejso.2006.03.008. [DOI] [PubMed] [Google Scholar]
- 30.Ansaloni L, Coccolini F, Morosi L, Ballerini A, Ceresoli M, Grosso G, et al. Pharmacokinetics of concomitant cisplatin and paclitaxel administered by hyperthermic intraperitoneal chemotherapy to patients with peritoneal carcinomatosis from epithelial ovarian cancer. Br J Cancer. 2015;112:306–12. doi: 10.1038/bjc.2014.602. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Steller MA, Egorin MJ, Trimble EL, Bartlett DL, Zuhowski EG, Alexander HR, et al. A pilot phase I trial of continuous hyperthermic peritoneal perfusion with high-dose carboplatin as primary treatment of patients with small-volume residual ovarian cancer. Cancer Chemother Pharmacol. 1999;43((2)):106–14. doi: 10.1007/s002800050870. [DOI] [PubMed] [Google Scholar]
- 32.Van der Speeten K, Stuart OA, Mahteme H, Sugarbaker PH. Pharmacokinetic study of perioperative intravenous Ifosfamide. Int J Surg Oncol. 2011;2011:185092. doi: 10.1155/2011/185092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Deraco M, Kusamura S, Virzì S, Puccio F, Macrì A, Famulari C, et al. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy as upfront therapy for advanced epithelial ovarian cancer: multi-institutional phase-II trial. Gynecol Oncol. 2011 Aug;122((2)):215–20. doi: 10.1016/j.ygyno.2011.05.004. [DOI] [PubMed] [Google Scholar]
- 34.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 Jun;36((6)):589–93. doi: 10.1016/j.ejso.2010.04.005. [DOI] [PubMed] [Google Scholar]
- 35.Sugarbaker PH, Stuart OA, Bijelic L. Intraperitoneal gemcitabine chemotherapy treatment for patients with resected pancreatic cancer: rationale and report of early data. Int J Surg Oncol. 2011;2011:161862. doi: 10.1155/2011/161862. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bijelic L, Sugarbaker PH, Stuart OA. Hyperthermic intraperitoneal chemotherapy with melphalan: a summary of clinical and pharmacological data in 34 patients. Gastroenterol Res Pract. 2012;2012:827534. doi: 10.1155/2012/827534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sugarbaker PH, Van der Speeten K. Surgical technology and pharmacology of hyperthermic perioperative chemotherapy. J Gastrointest Oncol. 2016 Feb;7((1)):29–44. doi: 10.3978/j.issn.2078-6891.2015.105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lepock JR. How do cells respond to their thermal environment? Int J Hyperthermia. 2005;21:681–7. doi: 10.1080/02656730500307298. [DOI] [PubMed] [Google Scholar]
- 39.De Bree E, Tsiftsis DD. Experimental and pharmacokinetic studies in intraperitoneal chemotherapy: from laboratory bench to bedside. Recent Results Cancer Res. 2007;169:53–73. doi: 10.1007/978-3-540-30760-0_5. [DOI] [PubMed] [Google Scholar]
- 40.Los G, Verdegaal EM, Mutsaers PH, McVie JG. Penetration of carboplatin and cisplatin into rat peritoneal tumor nodules after intraperitoneal chemotherapy. Cancer Chemother Pharmacol. 1991;28:159–65. doi: 10.1007/BF00685503. [DOI] [PubMed] [Google Scholar]
- 41.Van De Vaart PJ, Van Der Vange N, Zoetmulder FA, Van Goethem AR, Van Tellingen O, Ten Bokkel Huinink WW, et al. Intraperitoneal cisplatin with regional hyperthermia in advanced ovarian cancer: pharmacokinetics and cisplatin-DNA adduct formation in patients and ovarian cancer cell lines. Eur J Cancer. 1998;34:148–54. doi: 10.1016/s0959-8049(97)00370-5. [DOI] [PubMed] [Google Scholar]
- 42.Hettinga JVE. Reduction of cisplatin resistance by hyperthermia. University of Groningen; 1996. [Google Scholar]
- 43.Hetting VE, Lemstra W, Meijer C, Dam WA, Uges DR, Konings AW, et al. Mechanism of hyperthermic potentiation of cisplatin action in cisplatin sensitive and -resistant tumour cells. Br J Cancer. 1997;75:1735–43. doi: 10.1038/bjc.1997.297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Mikkelsen MS, Blaakaer J, Petersen LK, Schleiss LG, Iversen LH. Pharmacokinetics and toxicity of carboplatin used for hyperthermic intraperitoneal chemotherapy (HIPEC) in treatment of epithelial ovarian cancer. Pleura Peritoneum. 2020;5((4)):20200137. doi: 10.1515/pp-2020-0137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Los G, Van Vugt MJ, Den Engelse L, Pinedo HM. Effects of temperature on the interaction of cisplatin and carboplatin with cellular DNA. Biochem Pharmacol. 1993;46:1229–37. doi: 10.1016/0006-2952(93)90472-9. [DOI] [PubMed] [Google Scholar]
- 46.Salvatorelli E, De Tursi M, Menna P, Carella C, Massari R, Colasante A, et al. Pharmacokinetics of pegylated liposomal doxorubicin administered by intraoperative hyperthermic intra- peritoneal chemotherapy to patients with advanced ovarian cancer and peritoneal carcinomatosis. Drug Metab Dispos. 2012;40:2365–73. doi: 10.1124/dmd.112.047480. [DOI] [PubMed] [Google Scholar]
- 47.Harrison LE, Bryan M, Pliner L, Saunders T. Phase I trial of pegylated liposomal doxorubicin with hyperthermic intraperitoneal chemotherapy in patients undergoing cytoreduction for advanced intra-abdominal malignancy. Ann Surg Oncol. 2008;15:1407–13. doi: 10.1245/s10434-007-9718-8. [DOI] [PubMed] [Google Scholar]
- 48.Ning S, Macleod K, Abra RM, Huang AH, Hahn GM. Hyperthermia induces doxorubicin release from long-circulating liposomes and enhances their anti-tumor efficacy. Int J Radiat Oncol Biol Phys. 1994;29:827–34. doi: 10.1016/0360-3016(94)90572-x. [DOI] [PubMed] [Google Scholar]
- 49.De Bree E, Theodoropoulos PA, Rosing H, Michalakis J, Romanos J, Beijnen JH, et al. Treatment of ovarian cancer using intraperitoneal chemotherapy with taxanes: from laboratory bench to bedside. Cancer Treat Rev. 2006;32:471–82. doi: 10.1016/j.ctrv.2006.07.006. [DOI] [PubMed] [Google Scholar]
- 50.De Bree E, Rosing H, Filis D, Romanos J, Melissourgaki M, Daskalakis M, et al. Cytoreductive surgery and intraoperative hyperthermic intraperitoneal chemotherapy with paclitaxel:a clinical and pharmacokinetic study. Ann Surg Oncol. 2008;15:1183–92. doi: 10.1245/s10434-007-9792-y. [DOI] [PubMed] [Google Scholar]
- 51.De Bree E, Katsougkri D, Polioudaki H, Angaridou ETS, Michelakis D, Zoras O, et al. Hyperthermia during intraperitoneal chemotherapy with paclitaxel or docetaxel for ovarian cancer: is there any benefit? Anticancer Res. Dec 2020;40((12)):6769–80. doi: 10.21873/anticanres.14700. [DOI] [PubMed] [Google Scholar]
- 52.Rietbroek RC, Katschinski DM, Reijers MH, Robins HI, Geerdink A, Tutsch K, et al. Lack of thermal enhancement for taxanes in vitro. Int J Hyperthermia. 1997 Sep–Oct;13((5)):525–33. doi: 10.3109/02656739709023551. [DOI] [PubMed] [Google Scholar]
- 53.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. doi: 10.1016/j.ygyno.2014.11.072. [DOI] [PubMed] [Google Scholar]
- 54.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 Dec;41((12)):1578–89. doi: 10.1016/j.ejso.2015.08.172. [DOI] [PubMed] [Google Scholar]
- 55.Zhang G, Zhu Y, Liu C, Chao G, Cui R, Zhang Z. The prognosis impact of hyperthermic intraperitoneal chemotherapy (HIPEC) plus cytoreductive surgery (CRS) in advanced ovarian cancer: the meta-analysis. J Ovarian Res. 2019 Apr 17;12((1)):33. doi: 10.1186/s13048-019-0509-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lei Z, Wang Y, Wang J, Wang K, Tian J. Zhao Y, et al. Evaluation of cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy for stage III epithelial ovarian cancer. JAMA Netw Open. 2020;3((8)):e2013940. doi: 10.1001/jamanetworkopen.2020.13940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Lim MC, Chang S-J, Yoo HJ, Nam BH, Bristow R, Park SY, et al. Randomized trial of hyperthermic intraperitoneal chemotherapy (HIPEC) in women with primary advanced peritoneal, ovarian, and tubal cancer. J Clin Oncol. 2017;35:5520. [Google Scholar]
- 58.Spriggs DR, Zivanovic O. Ovarian cancer treatment: are we getting warmer? N Engl J Med. 2018 Jan 18;378((3)):293–4. doi: 10.1056/NEJMe1714556. [DOI] [PubMed] [Google Scholar]
- 59.Vergote I, Chiva L, du Bois A. Hyperthermic intraperitoneal chemotherapy in ovarian cancer. N Engl J Med. 2018 Apr 5;378((14)):1362–3. doi: 10.1056/NEJMc1802033. [DOI] [PubMed] [Google Scholar]
- 60.National Comprehensive Cancer Network® . NCCN guidelines. Available from: www.nccn.org/professionals/physician_gls/pdf/ovarian.pdf Accessed 2021 Aug 12. [Google Scholar]
- 61.Bakrin N, Gladieff L. [Malignant epithelial ovarian cancer: Role of intra-peritoneal chemotherapy and hyperthermic intra peritoneal chemotherapy(HIPEC): Article drafted from the French Guidelines in oncology entitled “Initial management of patients with epithelial ovarian cancerˮ developed by FRANCOGYN, CNGOF, SFOG, GINECO-ARCAGY under the aegis of CNGOF and endorsed by INCa] Gynecol Obstet Fertil Senol. 2019 Feb;47((2)):214–21. doi: 10.1016/j.gofs.2019.01.001. [DOI] [PubMed] [Google Scholar]
- 62.Coleman RL, Spirtos NM, Enserro D, Herzog TJ, Sabbatini P, Armstrong DK, et al. Secondary surgical cytoreduction for recurrent ovarian cancer. N Engl J Med. 2019 Nov 14;381((20)):1929–39. doi: 10.1056/NEJMoa1902626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Du Bois A, Sehouli J, Vergote I, Ferron G, Reuss A, Meier W, et al. Randomized phase III study to evaluate the impact of secondary cytoreductive surgery in recurrent ovarian cancer. Final analysis of AGO DESKTOP III/ENGOT-ov20. J Clin Oncol. 2020;38((15 Suppl)):6000. [Google Scholar]
- 64.Shi T, Zhu J, Feng Y, Tu D, Zhang Y, Zhang P, et al. Secondary cytoreduction followed by chemotherapy versus chemotherapy alone in platinum-sensitive relapsed ovarian cancer (SOC-1): a multicentre, open-label, randomised, phase 3 trial. Lancet Oncol. 2021 Apr;22((4)):439–49. doi: 10.1016/S1470-2045(21)00006-1. [DOI] [PubMed] [Google Scholar]
- 65.Munoz-Casares FC, Rufian S, Rubio MJ, Díaz CJ. Díaz R, Casado A, et al. The role of hyper-thermic intraoperative intraperitoneal chemotherapy (HIPEC) in the treatment of peritoneal carcinomatosis in recurrent ovarian cancer. Clin Transl Oncol. 2009;11:753–9. doi: 10.1007/s12094-009-0438-3. [DOI] [PubMed] [Google Scholar]
- 66.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. [PubMed] [Google Scholar]
- 67.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. doi: 10.1016/j.ygyno.2012.09.020. [DOI] [PubMed] [Google Scholar]
- 68.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((3)):987–93. doi: 10.1245/s10434-014-4049-z. [DOI] [PubMed] [Google Scholar]
- 69.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((11)):3621–7. doi: 10.1245/s10434-014-3693-7. [DOI] [PubMed] [Google Scholar]
- 70.Baiocchi G, Ferreira FO, Mantoan H, da Costa AA, Faloppa CC, Kumagai LY, et al. Hyperthermic intraperitoneal chemotherapy after secondary cytoreduction in epithelial ovarian cancer: a single-center comparative analysis. Ann Surg Oncol. 2016;23((4)):1294–301. doi: 10.1245/s10434-015-4991-4. [DOI] [PubMed] [Google Scholar]
- 71.Bamias A, Bamia C, Zagouri F, Kostouros E, Kakoyianni K, Rodolakis A, et al. Improved survival trends in platinum-resistant patients with advanced ovarian, fallopian or peritoneal cancer treated with first-line paclitaxel/platinum chemotherapy: the impact of novel agents. Oncology. 2013;84((3)):158–65. doi: 10.1159/000341366. [DOI] [PubMed] [Google Scholar]
- 72.Wilson MK, Pujade-Lauraine E, Aoki D, Mirza MR, Lorusso D, Oza AM, et al. Fifth ovarian cancer consensus conference of the gynecologic cancer interGroup: recurrent disease. Ann Oncol. 2017 Apr 1;28((4)):727–32. doi: 10.1093/annonc/mdw663. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Moran BJ, Tzivanakis A. The concept of “Obstruction-Free Survivalˮ as an outcome measure in advanced colorectal cancer management. Pleura Peritoneum. 2018 Feb 28;3((1)):20180101. doi: 10.1515/pp-2018-0101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Sinukumar S, Damodaran D, Ray M, Mehta S, Paul L, Bhatt A. Pattern of recurrence after interval cytoreductive surgery and HIPEC following neoadjuvant chemotherapy in primary advanced stage IIIC/IVA epithelial ovarian cancer. Eur J Surg Oncol. 2021 Jun;47((6)):1427–33. doi: 10.1016/j.ejso.2021.01.013. [DOI] [PubMed] [Google Scholar]
- 75.Böhm S, Faruqi A, Said I, Lockley M, Brockbank E, Jeyarajah A, et al. Chemotherapy response score: development and validation of a system to quantify histopathologic response to neoadjuvant chemotherapy in tubo-ovarian high-grade serous carcinoma. J Clin Oncol. 2015 Aug 1;33((22)):2457–63. doi: 10.1200/JCO.2014.60.5212. [DOI] [PubMed] [Google Scholar]
- 76.Paoletti X, Lewsley LA, Daniele G, Cook A, Yanaihara N, Tinker A, et al. Assessment of progression-free survival as a surrogate end point of overall survival in first-line treatment of ovarian cancer: a systematic review and meta-analysis. JAMA Netw Open. 2020 Jan 3;3((1)):e1918939. doi: 10.1001/jamanetworkopen.2019.18939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.DiSilvestro P, Colombo N, Scambia G, Kim BG, Oaknin A, Friedlander M, et al. Efficacy of maintenance olaparib for patients with newly diagnosed advanced ovarian cancer with a BRCA mutation: subgroup analysis findings from the SOLO1 trial. J Clin Oncol. 2020 Oct 20;38((30)):3528–37. doi: 10.1200/JCO.20.00799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Jónsdóttir B, Lomnytska M, Poromaa IS, Silins I, Stålberg K. The peritoneal cancer index is a strong predictor of incomplete cytoreductive surgery in ovarian cancer. Ann Surg Oncol. 2021;28:244–51. doi: 10.1245/s10434-020-08649-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Llueca A, Climent MT, Escrig J, Carrasco P, Serra A, MUAPOS Working Group (Multidisciplinary Unit of Abdominal Pelvic Oncology Surgery) Validation of three predictive models for suboptimal cytoreductive surgery in advanced ovarian cancer. Sci Rep. 2021;11:8111. doi: 10.1038/s41598-021-86928-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Marchetti C, Muzii L, Romito A, Benedetti Panici P. First-line treatment of women with advanced ovarian cancer: focus on bevacizumab. Onco Targets Ther. 2019 Feb 8;12:1095–103. doi: 10.2147/OTT.S155425. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Ray-Coquard I, Pautier P, Pignata S, Pérol D, González-Martín A, Berger R, et al. Olaparib plus bevacizumab as first-line maintenance in ovarian cancer. N Engl J Med. 2019;381:2416–28. doi: 10.1056/NEJMoa1911361. [DOI] [PubMed] [Google Scholar]
- 82.Perren TJ, Swart AM, Pfisterer J, Ledermann JA, Pujade-Lauraine E, Kristensen G, et al. A phase 3 trial of bevacizumab in ovarian cancer. N Engl J Med. 2011;365:2484–96. doi: 10.1056/NEJMoa1103799. [DOI] [PubMed] [Google Scholar]
- 83.Tewari KS, Burger RA, Enserro D, Norquist BM, Swisher EM, Brady MF, et al. Final overall survival of a randomized trial of bevacizumab for primary treatment of ovarian cancer. J Clin Oncol. 2019;37:2317–28. doi: 10.1200/JCO.19.01009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Byrne AT, Ross L, Holash J, Nakanishi M, Hu L, Hofmann JI, et al. Vascular endothelial growth factor-trap decreases tumor burden, inhibits ascites, and causes dramatic vascular remodeling in an ovarian cancer model. Clin Cancer Res. 2003;9:5721–8. [PubMed] [Google Scholar]
- 85.Koole SN, van Lieshout C, van Driel WJ, van Schagen E, Sikorska K, Kieffer JM, et al. Cost effectiveness of interval cytoreductive surgery with hyperthermic intraperitoneal chemotherapy in stage III ovarian cancer on the basis of a randomized phase III trial. J Clin Oncol. 2019 Aug 10;37((23)):2041–50. doi: 10.1200/JCO.19.00594. [DOI] [PubMed] [Google Scholar]
- 86.Penn CA, Wong MS, Walsh CS. Cost-effectiveness of maintenance therapy based on molecular classification following treatment of primary epithelial ovarian cancer in the United States. JAMA Netw Open. 2020 Dec 1;3((12)):e2028620. doi: 10.1001/jamanetworkopen.2020.28620. [DOI] [PMC free article] [PubMed] [Google Scholar]
