Discrepancy between recommendations regarding hyperthermic intraperitoneal chemotherapy (HIPEC) in ovarian cancer management: a narrative review
Review Article

Discrepancy between recommendations regarding hyperthermic intraperitoneal chemotherapy (HIPEC) in ovarian cancer management: a narrative review

Szymon Piątek1 ORCID logo, Elżbieta Sarnowska2, Joanna Kacperczyk-Bartnik3, Halszka Kołaczkowska1, Piotr Sobiczewski1, Mariusz Bidziński1

1Department of Gynecologic Oncology, The Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; 2Department of Experimental Immunotherapy, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland; 32nd Department of Obstetrics and Gynecology, Medical University of Warsaw, Warsaw, Poland

Contributions: (I) Conception and design: S Piątek; (II) Administrative support: M Bidziński; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: S Piątek; (V) Data analysis and interpretation: S Piątek; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Szymon Piatek, MD, PhD. Department of Gynecologic Oncology, The Maria Sklodowska-Curie National Research Institute of Oncology, 5 Roentgen Street, 02-781 Warsaw, Poland. Email: szymon.piatek@aol.com.

Background and Objective: Hyperthermic intraperitoneal chemotherapy (HIPEC) is still controversial in ovarian cancer (OC) management. Doubts are related mainly to HIPEC effectiveness, but also to its safety. European Society of Medical Oncology and European Society of Gynecologic Oncology do not consider HIPEC as a standard of care. Opposite to European recommendations, National Comprehensive Cancer Network found HIPEC as a treatment option in patients undergoing interval debulking surgery in first-line treatment. This may be confusing for oncologists in clinical practice. The aim of this narrative review is to present literature review focusing on efficacy, confounding factors, complications and immunological issue of HIPEC in OC management.

Methods: PubMed was searched for meta-analyses, randomized trials, observational studies, experimental studies to outline the role of HIPEC in OC management since January 2015 until August 2023. Keywords included “hyperthermic intraperitoneal chemotherapy”, “HIPEC”, “ovarian cancer”, “immune response”. References from full-text articles were screened for additional studies.

Key Content and Findings: Most meta-analyses found that HIPEC improved survival in patients with OC and none of the meta-analyses showed that addition HIPEC to surgery was associated with a worse treatment outcome compared to surgery alone. Positive effect on treatment outcome was found more common in first-line treatment than recurrent disease. Positive effect on treatment outcome was more common in first-line treatment (especially during interval debulking surgery) than recurrent disease. HIPEC efficacy can be affected by patients’ characteristics (BRCA status, platinum sensitivity), cytostatic type and dose, intensity of hyperthermia and peritoneal flow characteristics. Apart from strict cytotoxic effect, HIPEC can induce anti-cancer immune response.

Conclusions: Although factors confounding HIPEC efficacy are not well-defined, survival improvement, related to addition HIPEC to surgery in OC, was observed. Future studies should focus on determining a subgroup of patients, who benefit from HIPEC. This will contribute to the unification of European and American recommendations.

Keywords: Hyperthermic intraperitoneal chemotherapy (HIPEC); ovarian cancer (OC); survival; complications; review


Submitted Dec 09, 2023. Accepted for publication Feb 29, 2024. Published online May 10, 2024.

doi: 10.21037/cco-23-152


Introduction

Background

The lifetime risk of developing ovarian cancer (OC) is 1.3%, which means that 1 in 78 women will be diagnosed with OC (1). Although the highest incidence of OC occurs in Europe and North America, those regions have decreasing trend in incidence and mortality (2,3). The opposite situation is observed in China and African countries, where the incidence of OC is increasing and it is predicted to increase continuously in the next years (4-6). Incomprehensibly, a substantial increase in incidence was observed in younger females (3). The number of asymptomatic ovarian masses has increased with the use of prenatal ultrasonography (7). However, only 5% of ovarian tumors diagnosed during pregnancy are malignant and majority of them are non-epithelial (mainly germ cell tumors and sex cord-stromal tumors) (7). Germ cell tumors differ to epithelial OCs with their earlier age of incidence, faster rate of growth, unilateral localization (95% of cases) and good prognosis (8).

Incidence rates are calculated per 100,000 people, so they do not show the actual number of new cases. Considering constantly growing population around the world, the number of new cases of OC has raised from 152,000 in 1990 to 314,000 in 2020, while the number of deaths due to OC in the same time period increased from 95,000 to 207,000 (9). Moreover, prognosis for next decades is unfavorable. In 2040, the number of new cases and deaths due to OC will be higher—among countries with low human development index (HDI) by 96% and 100%, respectively, while in countries with very high HDI by 19% and 28%, respectively (10). Although the incidence of OC is not at the top among all malignancies, it has the highest mortality rate among gynecological cancers (9). The poor prognosis of OC is confirmed by the ratio of deaths to new cases (65%), which is similar to stomach cancer (70%) and significantly higher than in breast cancer (30%), endometrial cancer (22%), and even cervical cancer (56%) (11).

According to molecular mechanism type I epithelial OCs are proposed to exhibit relatively indolent behavior and genetic stability (12). In contrast, type II epithelial OCs are suggested to be inherently aggressive tumors, which can disseminate even from small primary lesion in the ovary or fallopian tube (12). High-grade serous OC is the most common histological type and constitute approximately 75% of all epithelial OC. It was found that high-grade serous OC arise through the type II pathway and typically harbor mutations in TP53 and BRCA genes. Germline mutations in BRCA1/2 occur in up to 15% of newly diagnosed OC cases and represent the strongest risk factor for epithelial OC (13). BRCA1/2 carriers exhibit a more favorable response to platinum-based chemotherapy and poly (ADP-ribose) polymerase (PARP) inhibitors compared to non-carriers (13). This leads to improved survival, despite the OC typically being diagnosed at an advanced stage (14).

OC frequently involves the peritoneum, however in most cases, it spreads superficially and towards the peritoneal cavity without deep penetration into muscles, bowels and internal organs. The peritoneal barrier plays a role not only in development of peritoneal metastases, but also in transport of different substances, inflammatory response, antigen presentation, fibrosis and fibrinolysis and tissue repair (15). This barrier is a complex structure made up of the peritoneum, parenchymal cells, interstitial cells, interstitial matrix, pericytes, and the endothelial cells that form the microvessels (16). Intravenous (IV) chemotherapy administration could have limited penetration to peritoneal carcinomatosis, especially that tumor clusters are able to grow up to a size of 2 mm without external nutrients and capillaries (17). Therefore, intraperitoneal (IP) route could omit the peritoneal barrier and chemotherapy could directly act to the tumor. Initially, adjuvant IP chemotherapy was extensively studied. Although a Cochrane meta-analysis showed that IP chemotherapy improves survival in advanced OC (18), its use in clinical practice has been limited (19,20). It was found that 58% of patients did not completed adjuvant IP chemotherapy, mainly due to peritoneal catheter complications (21). IP chemotherapy was associated with higher serious complications rate, more intense abdominal discomfort and neurotoxicity compared to IV route (22). IP chemotherapy, as well as extensive abdominal surgery, inflammation and infection may contribute to the formation of the intraabdominal adhesions (23,24). Intraabdominal adhesions may limit distribution of chemotherapy in the peritoneal cavity (25). Finally, GOG-252 study did not confirm any survival benefit of IP chemotherapy compared to IV administration (22). Therefore, adjuvant IP chemotherapy has not been adopted in everyday clinical practice. European Society of Medical Oncology (ESMO) decline IP chemotherapy as a standard of care due to greater toxicity and complexity of its administration (26). In National Comprehensive Cancer Network (NCCN) guidelines IP regimens can be offered to selected patients in stages II and III, but it is always combined with IV chemotherapy so that systemic disease can also be treated (27).

Rationale and knowledge gap

Hyperthermic intraperitoneal chemotherapy (HIPEC) used immediately after cytoreductive surgery, omits problems related to the catheter, multiple drug administration and high rate of treatment discontinuation. An additional advantage of HIPEC compared to IP adjuvant chemotherapy is excellent exposure of the peritoneal lesions, lack of peritoneal adhesions and the possibility to reach all regions of the peritoneal cavity. The clinical significance of HIPEC in OC is currently extensively researched. Although it is not a new therapeutic method, its role in the treatment of OC has not yet been precisely defined, and recommendations for the use of HIPEC are not consistent (26-28). European Society of Gynecologic Oncology (ESGO) and ESMO do not recommend HIPEC in the treatment of primary or recurrent OC (26,28). European experts considered HIPEC to be an experimental method and should not be considered as standard therapy. As opposed to this, NCCN guidelines found that HIPEC with cisplatin (100 mg/m2) can be considered during interval cytoreduction in patients with stage III disease according to protocol used in OV-HIPEC-01 trial, i.e.: an intraabdominal temperature of 40 ℃, perfusion with cisplatin at a dose of 100 mg per square meter, flow rate of 1 liter per minute (with 50% of the dose perfused initially, 25% at 30 minutes, and 25% at 60 minutes) and 90-minute perfusion period.

Objective

Due to the opposite statement of ESGO/ESMO and NCCN regarding HIPEC in OC management, we present this narrative review to analyze the effectiveness, potential confounding factors, complications and immunological issues related to HIPEC. We present this article in accordance with the Narrative Review reporting checklist (available at https://cco.amegroups.com/article/view/10.21037/cco-23-152/rc).


Methods

PubMed was searched using “hyperthermic intraperitoneal chemotherapy”, “HIPEC”, “ovarian cancer”, “immune response” to find relevant articles to this review (Table 1). Full-text articles published in English, including basic science, observational studies, randomized trials and meta-analyses published between January 1, 2015 and August 30, 2023 were included. References from articles were screened for additional studies. There were no exclusion criteria for the literature search. The authors provide a narrative summary of the findings and implications of these studies. All authors contributed to literature search and approved the selected literature.

Table 1

The search strategy summary

Items Specification
Date of search September 1st, 2023
Databases and other sources searched PubMed
Search terms used “hyperthermic intraperitoneal chemotherapy”, “HIPEC”, “ovarian cancer”, “immune response”
Timeframe 01.01.2015–30.08.2023
Inclusion and exclusion criteria Inclusion criteria:
• To assess HIPEC efficacy and complications: observational studies, randomized trials, systematic reviews, meta-analyses were analyzed
• To assess factors that could confound HIPEC efficacy: observational studies, randomized trials, experimental studies were analyzed
• To assess immune response in HIPEC: experimental studies were analyzed
Exclusion criteria:
• Papers published in language other than English
Selection process All authors contributed and reviewed the selected literature. Literature search was conducted independently by all authors. Articles were included in this review if they were selected by at least three authors. In other cases, appropriateness of each paper was discussed by all authors, and the decision about article inclusion was made jointly by all authors

HIPEC, hyperthermic intraperitoneal chemotherapy.


Efficacy of HIPEC in OC

European guidelines, that do not recommend HIPEC in OC management, were supported by a meta-analysis (29) and a randomized trial (30). Chiva et al. showed no benefit in survival with an increased risk of side effects (29). Lim et al. in OV-HIPEC-01 trial also found that addition of HIPEC to cytoreductive surgery did not improve progression-free and overall survival (OS) in patients with advanced epithelial OC. However, in a subgroup analysis, in patients with interval debulking surgery (IDS), HIPEC improved OS and progression-free survival (PFS) (30). Those results confirmed outcomes of a previously published randomized trial by van Driel et al., who found that addition of HIPEC to IDS resulted in longer recurrence-free survival and OS than surgery alone (31). Improved survival in patients undergoing IDS + HIPEC vs. IDS alone was confirmed in several recently published meta-analyses (32-35) (Table 2). The significance of HIPEC during cytoreductive surgery in recurrent disease is not clear, while results of meta-analysis were discordant (Table 2).

Table 2

List of meta-analyses assessing HIPEC in ovarian cancer management

Year Author Primary disease Recurrent disease
PDS + HIPEC + CTH vs. PDS + CTH IDS + HIPEC + CTH vs. IDS + CTH Surgery + HIPEC + CTH vs. surgery + CTH
OS PFS/DFS OS PFS/DFS OS/PFS/DFS
2023 Della Corte (36) No difference No difference Improved OS in HIPEC group Improved PFS in HIPEC group No difference
2023 Llueca (32) Improved OS in HIPEC group Improved PFS in HIPEC group
2023 Xia (33) Improved OS in HIPEC group No difference Improved OS in HIPEC group Improved DFS in HIPEC group No difference
2023 Li (37) Improved DFS and OS (no distinction between PDS and IDS) Improved OS and PFS in HIPEC group
2022 Filis (34) No difference No difference Improved OS in HIPEC group Improved OS in HIPEC group No difference
2020 Cianci (35) Improved OS in HIPEC group
2019 Kim (38) Improved DFS and OS in HIPEC group (no distinction between PDS and IDS) Improved OS, but not DFS in HIPEC group
2019 Zhang (39) Improved DFS and OS in HIPEC group (no distinction between PDS and IDS) Improved OS, but not PFS in HIPEC group
2019 Wang (40) Improved DFS and OS in HIPEC group (no distinction between PDS and IDS) Improved OS, but not DFS in HIPEC group
2019 Wu (41) Improved DFS and OS in HIPEC group (no distinction between PDS and IDS) Improved PFS and OS
2018 Kireeva (42) Improved OS in HIPEC group
2015 Huo (43) Improved OS in HIPEC group Improved OS in HIPEC group

HIPEC, hyperthermic intraperitoneal chemotherapy; PDS, primary debulking surgery; CTH, chemotherapy; IDS, interval debulking surgery; OS, overall survival; PFS, progression-free survival; DFS, disease-free survival.

In a meta-analysis by Kim et al., the effectiveness of HIPEC was assessed depending on chemotherapy administration (44). It was found, that, chemotherapy given before surgery played a key role in the effectiveness of HIPEC, regardless of primary or recurrent OC. Patients who received chemotherapy up to 6 months before HIPEC had improved survival (44). Finding a group of patients who will benefit from HIPEC is crucial. Jian et al. analyzed all OC patients together, irrespective of primary and recurrent disease (45). This meta-analysis did not demonstrate any improvement in OS nor disease-free survival (45).

OC is now considered as a chronic disease with periods of remission and relapse. Reduction in the number of relapses (even without OS improvement) is important because every treatment of relapse is associated with deterioration of the quality of life, including intensification of pain, limitation of physical activity and emotional tension or anxiety (46). OC recurrence frequently is located in the peritoneal cavity. It has been shown that 50% of disease recurrences in patients undergoing HIPEC are located outside the peritoneal cavity (47). Shifting the site of recurrence outside the peritoneal cavity after HIPEC may reduce the incidence of gastrointestinal obstruction and the need for a palliative stoma, which may lead to improvement in quality of life.

Factors affecting HIPEC efficacy

Chemotherapy

Apart from proper qualification to HIPEC, there are many factors that may influence the effectiveness of the procedure (Table 3). The most frequently used cytostatic agent in advanced OC is single-drug chemotherapy using cisplatin at a dose of 75 mg/m2 (50–100 mg/m2) (44). However, the choice of the optimal cytostatic agent in HIPEC procedure has not been yet determined (40). The effectiveness of HIPEC may vary depending on the chemotherapy used and its dose. The study conducted by Chambers et al. showed that the use of paclitaxel and cisplatin during HIPEC significantly improves PFS compared to the use of cisplatin alone (48). Although the OS data are not yet available, it appears that the use of multi-drug chemotherapy during HIPEC may provide better treatment outcomes than single-drug regimen. The mechanism of action of cisplatin and carboplatin is similar, but their pharmacokinetics are different and may lead to different therapeutic results. Zivanovic et al. showed that carboplatin was well tolerated but did not lead to improved oncological outcomes (49).

Table 3

Factors affecting HIPEC efficacy in ovarian cancer patients

Patient characteristics (primary vs. recurrent disease; primary vs. interval debulking surgery; platinum-sensitive vs. platinum-resistant, BRCA mutation)
Type of cytostatic
Dose of cytostatic
Peritoneal perfusion (procedure duration, speed flow)
Temperature of hyperthermia
Type and volume of solvent
HIPEC method (open vs. closed, number of drains)

HIPEC, hyperthermic intraperitoneal chemotherapy.

Although most patients initially respond to platinum-based chemotherapy delivered intravenously, resistance develops over time. Platinum resistance is associated with poor prognosis and therapeutic options are limited. However, it has been shown that the use of HIPEC may be effective in platinum-resistant patients. In gynecologic patients with platinum-resistant disease, either paclitaxel or Mitomycin C can be considered, but there is still advantage of using cisplatin. Bakrin et al. conducted a study in 246 patients with recurrent epithelial OC. It included both platinum-sensitive and platinum-resistant cases. All of them underwent optimal cytoreduction and HIPEC (50). There was no statistically significant difference in OS between platinum-sensitive and platinum-resistant patients (52 vs. 48 months, P=0.568). In addition, Spiliotis et al. demonstrated improved survival in patients undergoing HIPEC during secondary cytoreduction (51). The survival time in patients who underwent HIPEC did not differ between the platinum-resistant and platinum-sensitive groups (26.6 vs. 26.8 months), while in patients who did not undergo HIPEC it was significantly shorter in platinum-resistant patients (15.2 vs. 10.2 months). These results indicate that HIPEC can overcome platinum resistance in recurrent OC. Similar observations were presented by Costales et al., who demonstrated the effectiveness of secondary cytoreduction with HIPEC in the treatment of recurrent OC regardless of platinum sensitivity (52). The ongoing KOV-HIPEC-02 (NCT05316181) trial will provide more information regarding significance of HIPEC in platinum-resistant OC. In this two-armed, phase III trial, patients are randomized to the experimental group (cytoreductive surgery + HIPEC + physician-choice chemotherapy) or control group (physician-choice chemotherapy). HIPEC is carried out using the open or closed technique with infusing 41.5–42.0 ℃ doxorubicin 35 mg/m2 and mitomycin 15 mg/m2 for 90 minutes. PFS, a primary objective of the trial, is expected to be analyzed within a few years. OS, safety and quality of life are secondary endpoints and will be completed after 2029 year.

Hyperthermia

Increased temperature up to 40.5–43 ℃ enhances the cytotoxic effect of chemotherapy drugs, leading to greater destruction of cancer cells (Figure 1) (53). Improved effectiveness of chemotherapy may also result from the effect of hyperthermia on altered blood flow through the tumor tissue (54). Hyperthermia reverses hypoxic tumor environment by increasing vascular permeability, improving tissue oxygenation and normalizing pH. This promotes better distribution of the chemotherapy drug into the tumor. The molecular mechanism of synergy between hyperthermia and chemotherapy involves the induction of reactive oxygen species (ROS), which leads to increased drug uptake. ROS damage tumor cells in two ways: (I) by directly damaging DNA and (II) by increasing the synthesis of p53 protein, which results in cell cycle arrest and initiates apoptosis (55). Moreover, it has been shown that cancer cells can be selectively destroyed under the influence of hyperthermia (41–43 ℃) (56).

Figure 1 Relationship between surgery, HIPEC and immune system. HIPEC, hyperthermic intraperitoneal chemotherapy.

To assess the relationship between chemotherapy effectiveness and temperature, the thermal enhancement ratio (TER) was introduced. It is defined as the ratio of the dose necessary to induce a therapeutic effect at 37 ℃ to the dose required to achieve the same therapeutic effect at an elevated temperature (57). The effectiveness of heat enhancement chemotherapy depends not only on the type of cytostatic agent and temperature, but also on the type and origin of cancer cells (58). Platins show different TERs at higher temperatures, while mitomycin and 5-fluorouracil do not show any enhancement of the effect with increasing temperature (Table 4) (58). The high TER value for platinum derivatives is closely related to their increased uptake in hyperthermia. The degree of heat enhancement for individual cytostatics can be both linear and threshold, which further affects the effectiveness of chemotherapy.

Table 4

Relationship between cytotoxic effect of antineoplastic drugs and temperature

Drug TER for 37–43 ℃*
Cisplatin 1.7–6
Carboplatin 1.4–7.2
Oxaliplatin 1.7–3.3
5-fluorouracil 1
Mitomycin C 1

*, dose required to achieve cytotoxic effect at 37 ℃ and dose required to achieve the same cytotoxic effect at increased temperature. TER, thermal enhancement ratio.

Other factors related to HIPEC technique

Various types of substances are used as solvents for cytostatics: 0.9% NaCl, 1.5% dextrose, 5% glucose. The impact of these substances on the effectiveness of HIPEC has not been analyzed in the published literature so far. However, some of them have been found to contribute to electrolyte disturbances. It has been shown that the use of 1.5% dextrose is associated with the occurrence of hyponatremia, hypokalemia and reduced serum pH (59).

There are three methods to perform HIPEC: open, semi-open and closed techniques. No study has been published directly comparing different methods in OC. Therefore, no method can be recommended as preferred option in clinical practice (60).

BRCA status

Koole et al. performed a post hoc analysis of data retrieved from 200 (82%) patients enrolled in the OVHIPEC-1 trial to assess the predictive effect of homologous recombination deficiency and BRCA mutation status on response to HIPEC treatment (61). Patients were assigned to three subgroups depending on the tumour genomic profile: BRCA1/2 mutated, homologous recombination deficient with BRCA wild-type (HRD/BRCAwt) and homologous recombination proficient. The authors demonstrated in a multivariable analysis a significant beneficial effect on the OS for treatment with HIPEC and for having BRCA1/2 mutations. Most benefit from addition of HIPEC to cytoreductive surgery was observed in the subgroup with HRD/BRCAwt tumors.

Insights from a retrospective case-control study by Ghirardi et al. indicate that patients with BRCA wild-type tumors might benefit from addition of HIPEC after primary debulking surgery due to increase in chemosensitivity (62). Similar findings were suggested in a study by Schaaf et al. as authors stated that hyperthermia may impair the repair of DNA damage caused by chemotherapy agents resulting in an effect similar to that of PARP inhibitors in homologous recombination proficient tumors (63).

In the future, more information regarding BRCA status and HIPEC efficacy may be served by CHIPOR randomized trial (64).

Role of minimally invasive surgery (MIS)

Petrillo et al. hypothesize that MIS may enhance cisplatin uptake to the peritoneum and plasma due to integrity of the abdominal wall and higher intraabdominal pressure (65). They compared cisplatin pharmacokinetic profile in women with recurrent OC treated with MIS vs. laparotomy. It was found that cisplatin concentration in peritoneal tissue and serum was higher in patients treated with MIS (65). However, the role of MIS and HIPEC has not been established due to lack of randomized data. Feasibility of laparoscopy/robotic surgery and HIPEC was assessed in several retrospective studies (66,67). Fagotti et al. presented a case series of 10 patients with isolated platinum sensitive recurrent OC, who underwent MIS and HIPEC (66). No serious complications were found. Post-operative 18F-fludeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) scan did not reveal any residual disease and no secondary recurrence was observed. Morton et al. assessed peri-operative outcomes in primary OC after IDS with MIS or laparotomy (67). Decision regarding MIS or open technique was based on patients’ characteristics. No differences were found in adverse perioperative outcomes and rate of complete resections, while hospitalization was shorter in MIS group (67). Laparotomy remains main technique preceding HIPEC. However, MIS and HIPEC may be feasible in properly selected patients without multifocal disease or peritoneal carcinomatosis.


Immune response in HIPEC

The use of hyperthermia in cancer treatment occurs after observation of spontaneous tumor regression in patients with persistent fever. Fever has stimulating effects on the innate and adaptive immune system through multiple pathways, where interleukin-6 (IL-6) seems to play a central role (68). Hyperthermia has been reported to cause the alteration in the expression of thousands of genes which are involved in different cellular processes including protein folding, cell death and cell cycle (69). Hyperthermia activates the main regulator of the heat shock response the heat shock factor 1 (HSF1), which together with hypoxia-inducible factor 1 (HIF-1) and other partners activates the production of heat shock proteins (HSPs) (70). The HSPs belong to chaperone proteins and their function is to protect intracellular proteins from misfolding or aggregation and inhibit cell death. According to their molecular weight, HSP family consists of HSP110, HSP90, HSP70, HSP60, HSP40, and other small HSPs. In fact, the overexpression of HSPs is one of the features of many solid tumors, and is known that HSPs have a tumor protective function. Therefore, the activation of HSPs by hyperthermia and its effect on immune response can be a double blade sword. The HSPs may activate the different immune cells type including monocytes, macrophages, B cells, dendritic cells (DC) via binding the tumor antigens, which in consequence may lead to activation of cytotoxic T-cell (71), but HSF1 can also enhance the expression of programmed death-ligand 1 (PD-L1) in breast cancer cells. Moreover, the HSPs can be also released by normal cells in response to stress, transported into the extracellular compartment as extracellular HSPs (eHSPs) which may promote the remodeling of extracellular matrix, promote stromal cell activation like cancer-associated fibroblasts (CAFs), which secrete pro-tumor cytokines (72). eHSPs also can bind directly to the different receptors on the cancer cells surface where have a cancer protective function (73).

The described effects may not apply to HIPEC method where mild hyperthermia is used. The mild hyperthermia used in HIPEC increases in vascular adhesion molecules, may enhance immune cells recruitment and tumor infiltration (74). Moreover, the use of mild hyperthermia in animal models, showed the enhancement of natural killer, CD4 and CD8 T cells infiltration and activation, reduction of tumor growth and improved animal survival. Treatment of melanoma with 43 ℃ during 30 min, in mice model, resulted in activation of DCs and cytotoxic CD8 T cells in the draining lymph node. Those results clearly demonstrated the promising potential of mild, local hyperthermia in induction of immune response that may reduce the risk of recurrence and tumor metastasis (75).

Another author found that in peritoneal metastases of colorectal carcinoma in mice model, HIPEC together with mitomycin C led to increased infiltration of CD4, CD8 T cells and B cells into omental and visceral metastases. The HIPEC significantly increased the expression of HSP-90 mRNA, which strongly suggests the HSPs involvement in these processes (76). In the same cancer model, it was found that combining HIPEC with immune checkpoint inhibitor (anti-PD-1) treatment improved animals survival compared with HIPEC alone (77). In OC mice model the combined chemotherapy with hyperthermia led to the conclusion that hyperthermia increased the number of IP macrophages and DCs compared to chemotherapy group alone (78). Moreover, Huang and colleagues in OC mice model compared the effects of HIPEC and conventional IP chemotherapy on bone marrow and neutrophil recovery. They found that hyperthermia increased the neutrophil-recruiting cytokine levels in the serum and reduced the magnitude of chemotherapy-induced neutropenia. This strongly suggests that HIPEC compared with conventional IP chemotherapy improved neutrophil and bone marrow recovery (79). Zunino et al. observed that vaccine composed of CT26 cancer cells treated with HIPEC or mitomycin C, and after injected into mice may induce efficient immune response, which is mediated via HSP90 protein (80).

The effects of hyperthermia on the peritoneal immune environment have not been studied in detail, and the exact mechanism of HIPEC is still unclear and need to be further elucidated.


Complications

One of the main issues against introducing HIPEC into clinical practice was the concern regarding the potential high toxicity and increased risk of delaying adjuvant chemotherapy due to complications after HIPEC.

It was estimated that in HIPEC, serious complications occurred in 19.3–20.5% of patients, and 30-day mortality was 1.1–2.3% (81,82). Xia et al. showed that the overall number of serious complications in the case of HIPEC was not higher than in cytoreduction alone (33). HIPEC has not been found to increase the risk of intestinal anastomotic leak (83). There was no increased risk of blood transfusion, intestinal obstruction, peritoneal effusion or thromboembolic events (33). In preliminary results of CHIPOR randomized trial, it was found that addition of HIPEC was related to two fold higher serious (grades 3 and 4) complications compared to patients without HIPEC (64). There was also a higher risk of HIPEC-dependent complications, such as nausea, leukopenia, neutropenia, electrolyte disturbances and hypoalbuminemia (33). However, the risk of constipation after HIPEC was lower than after cytoreduction alone.

Acute kidney injury (AKI) is one of the most serious complications related to HIPEC with potentially increased mortality rate and risk of progression to chronic kidney disease (84). The incidence of AKI varies greatly but can be as high as 66% (85). Among the cytostatics used in HIPEC, cisplatin is characterized by the highest nephrotoxicity. The mechanism of cisplatin nephrotoxicity is diverse, and the risk of AKI may be increased by intraoperative blood loss, dehydration, or insufficient intraoperative hydration (Table 5).

Table 5

AKI after HIPEC

Increased risk of AKI (84-87) Decreased risk of AKI (86)
Preoperative kidney impairment (eGFR ≤60 mL/min) Sodium thiosulfate (9 g/m2 before HIPEC, then 12 g/m2 6 h after HIPEC)
Time since previous chemotherapy cycle (≤7 days) Intravenous fluids
Low intravenous fluid volume (≤77 mL/kg) Magnesium supplementation
Extensive peritonectomy
Cisplatin dose ≥70 mg/m2
angiotensin-converting enzyme inhibitors and sartans
Hypertension

AKI, acute kidney injury; HIPEC, hyperthermic intraperitoneal chemotherapy; eGFR, estimated glomerular filtration rate.

Cisplatin also has a toxic effect directly on the nephron. It accumulates in epithelial cells, leading to DNA damage. Consequently, cytokines and inflammatory cells are recruited to the kidneys. The accumulation of mast cells, neutrophils, macrophages, NK cells and T lymphocytes further exacerbates kidney damage. Sodium thiosulfate as a chelating agent has been shown to have a nephroprotective effect (86). It binds to platinum and can deactivate it. This leads to a reduction in the excretion of cisplatin by the kidneys and, consequently, reduces the necrosis of renal tubular cells. In addition, sodium thiosulfate prevents cisplatin-stimulated magnesium loss.

Quality of life (QoL)

Toxicity of HIPEC may negatively impact on quality of life. Koole et al. evaluated the effect of HIPEC on patient’s health-related quality of life in the OVHIPEC trial (88). Patients filled EORTC QLQ-C30 questionnaires at baseline, after surgery, after end of treatment, and every three months thereafter. No significant difference in QoL was observed in patients with HIPEC compared to those without HIPEC (88). Similar results were seen in patients enrolled to KOV-HIPEC-01 trial (89). There were no statistically significant differences in functional scales and symptom scales between HIPEC and control group. Moreover, addition of HIPEC to cytoreductive surgery decreased postoperative pain. Patients in HIPEC group reported lower pain in early postoperative period, which led to lesser consumption of analgetics (90).


Conclusions

Survival improvement related to addition HIPEC to surgery in OC was observed in most meta-analyses. Previously published randomized trials indicated longer PFS and OS in patients undergoing HIPEC after IDS compared to those without HIPEC (30,91). Nevertheless, HIPEC is not widely accepted in clinical practice. This may be changed by the results of two ongoing randomized trials evaluating addition of HIPEC to IDS: KOV-HIPEC-04 (NCT05827523) and HOTT (NCT05659381). In those studies, patients randomized to HIPEC arm received a dose of cisplatin 75 and 100 mg/m2, respectively. Apart from survival, quality of life, cost-effectiveness and effect of homologous recombination repair deficiency will be assessed. Despite this, several issues need to be explained. Future studies should focus on determining a subgroup of patients, who mostly benefit from HIPEC. At the same time, this will limit the exposure of non-responding patients to side effects related to HIPEC. This will lead to the unification of European and American recommendations.

Another issue regarding HIPEC in OC management is determining its role and place in the era of Poly (ADP-ribose) polymerase (PARP) inhibitors and other evolving treatment modalities. This applies to both first-line treatment and recurrent disease management.


Acknowledgments

Funding: None.


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at https://cco.amegroups.com/article/view/10.21037/cco-23-152/rc

Peer Review File: Available at https://cco.amegroups.com/article/view/10.21037/cco-23-152/prf

Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://cco.amegroups.com/article/view/10.21037/cco-23-152/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Torre LA, Trabert B, DeSantis CE, et al. Ovarian cancer statistics, 2018. CA Cancer J Clin 2018;68:284-96. [Crossref] [PubMed]
  2. Wojtyla C, Bertuccio P, Wojtyla A, et al. European trends in breast cancer mortality, 1980-2017 and predictions to 2025. Eur J Cancer 2021;152:4-17. [Crossref] [PubMed]
  3. Huang J, Chan WC, Ngai CH, et al. Worldwide Burden, Risk Factors, and Temporal Trends of Ovarian Cancer: A Global Study. Cancers (Basel) 2022;14:2230. [Crossref] [PubMed]
  4. Feng J, Xu L, Chen Y, et al. Trends in incidence and mortality for ovarian cancer in China from 1990 to 2019 and its forecasted levels in 30 years. J Ovarian Res 2023;16:139. [Crossref] [PubMed]
  5. Gizaw M, Parkin DM, Stöter O, et al. Trends in the incidence of ovarian cancer in sub-Saharan Africa. Int J Cancer 2023;152:1328-36. [Crossref] [PubMed]
  6. Chen X, Du J, Wang X, et al. Trends and projections of ovarian cancer incidence in Hong Kong: A population-based study. Acta Obstet Gynecol Scand 2023;102:942-9. [Crossref] [PubMed]
  7. Boussios S, Moschetta M, Tatsi K, et al. A review on pregnancy complicated by ovarian epithelial and non-epithelial malignant tumors: Diagnostic and therapeutic perspectives. J Adv Res 2018;12:1-9. [Crossref] [PubMed]
  8. Saani I, Raj N, Sood R, et al. Clinical Challenges in the Management of Malignant Ovarian Germ Cell Tumours. Int J Environ Res Public Health 2023;20:6089. [Crossref] [PubMed]
  9. Ferlay J, Colombet M, Soerjomataram I, et al. Cancer statistics for the year 2020: An overview. Int J Cancer 2021; Epub ahead of print. [Crossref] [PubMed]
  10. Cabasag CJ, Fagan PJ, Ferlay J, et al. Ovarian cancer today and tomorrow: A global assessment by world region and Human Development Index using GLOBOCAN 2020. Int J Cancer 2022;151:1535-41. [Crossref] [PubMed]
  11. Sung H, Ferlay J, Siegel RL, et al. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J Clin 2021;71:209-49. [Crossref] [PubMed]
  12. Pavlidis N, Rassy E, Vermorken JB, et al. The outcome of patients with serous papillary peritoneal cancer, fallopian tube cancer, and epithelial ovarian cancer by treatment eras: 27 years data from the SEER registry. Cancer Epidemiol 2021;75:102045. [Crossref] [PubMed]
  13. Shah S, Cheung A, Kutka M, et al. Epithelial Ovarian Cancer: Providing Evidence of Predisposition Genes. Int J Environ Res Public Health 2022;19:8113. [Crossref] [PubMed]
  14. Bolton KL, Chenevix-Trench G, Goh C, et al. Association between BRCA1 and BRCA2 mutations and survival in women with invasive epithelial ovarian cancer. JAMA 2012;307:382-90. [Crossref] [PubMed]
  15. van Baal JOAM, van Noorden CJF, Nieuwland R, et al. Development of Peritoneal Carcinomatosis in Epithelial Ovarian Cancer: A Review. J Histochem Cytochem 2018;66:67-83. [Crossref] [PubMed]
  16. Flessner MF. The transport barrier in intraperitoneal therapy. Am J Physiol Renal Physiol 2005;288:F433-42. [Crossref] [PubMed]
  17. Brooks PC. Cell adhesion molecules in angiogenesis. Cancer Metastasis Rev 1996;15:187-94. [Crossref] [PubMed]
  18. Jaaback K, Johnson N, Lawrie TA. Intraperitoneal chemotherapy for the initial management of primary epithelial ovarian cancer. Cochrane Database Syst Rev 2011;CD005340. [PubMed]
  19. Monk BJ, Chan JK. Is intraperitoneal chemotherapy still an acceptable option in primary adjuvant chemotherapy for advanced ovarian cancer? Ann Oncol 2017;28:viii40-5. [Crossref] [PubMed]
  20. Gourley C, Walker JL, Mackay HJ. Update on Intraperitoneal Chemotherapy for the Treatment of Epithelial Ovarian Cancer. Am Soc Clin Oncol Educ Book 2016;35:143-51. [Crossref] [PubMed]
  21. Walker JL, Armstrong DK, Huang HQ, et al. Intraperitoneal catheter outcomes in a phase III trial of intravenous versus intraperitoneal chemotherapy in optimal stage III ovarian and primary peritoneal cancer: a Gynecologic Oncology Group Study. Gynecol Oncol 2006;100:27-32. [Crossref] [PubMed]
  22. Walker JL, Brady MF, Wenzel L, et al. Randomized Trial of Intravenous Versus Intraperitoneal Chemotherapy Plus Bevacizumab in Advanced Ovarian Carcinoma: An NRG Oncology/Gynecologic Oncology Group Study. J Clin Oncol 2019;37:1380-90. [Crossref] [PubMed]
  23. Tabibian N, Swehli E, Boyd A, et al. Abdominal adhesions: A practical review of an often overlooked entity. Ann Med Surg (Lond) 2017;15:9-13. [Crossref] [PubMed]
  24. Akgol G, Yildiz C, Karakus S, et al. The effects of intraperitoneal chemotherapeutic agents on adhesion formation. Eur J Gynaecol Oncol 2016;37:781-5. [PubMed]
  25. Guardiola E, Delroeux D, Heyd B, et al. Intra-operative intra-peritoneal chemotherapy with cisplatin in patients with peritoneal carcinomatosis of ovarian cancer. World J Surg Oncol 2009;7:14. [Crossref] [PubMed]
  26. González-Martín A, Harter P, Leary A, et al. Newly diagnosed and relapsed epithelial ovarian cancer: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2023;34:833-48. [Crossref] [PubMed]
  27. Ovarian Cancer Continue Including Fallopian Tube Cancer and Primary Peritoneal Cancer. NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines®). Version 2.2023. 2023 Jun 2.
  28. Colombo N, Sessa C, du Bois A, et al. ESMO-ESGO consensus conference recommendations on ovarian cancer: pathology and molecular biology, early and advanced stages, borderline tumours and recurrent disease. Ann Oncol 2019;30:672-705. [Crossref] [PubMed]
  29. 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. [Crossref] [PubMed]
  30. Lim MC, Chang SJ, Park B, et al. Survival After Hyperthermic Intraperitoneal Chemotherapy and Primary or Interval Cytoreductive Surgery in Ovarian Cancer: A Randomized Clinical Trial. JAMA Surg 2022;157:374-83. [Crossref] [PubMed]
  31. van Driel WJ, Koole SN, Sikorska K, et al. Hyperthermic Intraperitoneal Chemotherapy in Ovarian Cancer. N Engl J Med 2018;378:230-40. [Crossref] [PubMed]
  32. Llueca M, Ibañez MV, Climent MT, et al. Effectiveness of Hyperthermic Intraperitoneal Chemotherapy Associated with Cytoreductive Surgery in the Treatment of Advanced Ovarian Cancer: Systematic Review and Meta-Analysis. J Pers Med 2023;13:258. [Crossref] [PubMed]
  33. Xia Y, Wang H, Zhang J, et al. Prognostic value and adverse events of cytoreductive surgery with hyperthermic intraperitoneal chemotherapy in primary advanced and platinum-sensitive recurrent epithelial ovarian cancer: a systematic review and meta-analysis. Int J Hyperthermia 2023;40:2165729. [Crossref] [PubMed]
  34. Filis P, Mauri D, Markozannes G, et al. Hyperthermic intraperitoneal chemotherapy (HIPEC) for the management of primary advanced and recurrent ovarian cancer: a systematic review and meta-analysis of randomized trials. ESMO Open 2022;7:100586. [Crossref] [PubMed]
  35. Cianci S, Riemma G, Ronsini C, et al. Hyperthermic intraperitoneal chemotherapy (HIPEC) for ovarian cancer recurrence: systematic review and meta-analysis. Gland Surg 2020;9:1140-8. [Crossref] [PubMed]
  36. Della Corte L, Conte C, Palumbo M, et al. Hyperthermic Intraperitoneal Chemotherapy (HIPEC): New Approaches and Controversies on the Treatment of Advanced Epithelial Ovarian Cancer-Systematic Review and Meta-Analysis. J Clin Med 2023;12:7012. [Crossref] [PubMed]
  37. Li H, Liao LJ. Efficacy of hyperthermic intraperitoneal chemotherapy plus cytoreductive surgery for advanced or recurrent ovarian cancer: a systematic evaluation and meta-analysis. Eur Rev Med Pharmacol Sci 2023;27:8135-43. [Crossref] [PubMed]
  38. Kim SI, Cho J, Lee EJ, et al. Selection of patients with ovarian cancer who may show survival benefit from hyperthermic intraperitoneal chemotherapy: A systematic review and meta-analysis. Medicine (Baltimore) 2019;98:e18355. [Crossref] [PubMed]
  39. Zhang G, Zhu Y, Liu C, et al. The prognosis impact of hyperthermic intraperitoneal chemotherapy (HIPEC) plus cytoreductive surgery (CRS) in advanced ovarian cancer: the meta-analysis. J Ovarian Res 2019;12:33. [Crossref] [PubMed]
  40. Wang Y, Ren F, Chen P, et al. Effects of CytoReductive surgery plus hyperthermic IntraPEritoneal chemotherapy (HIPEC) versus CytoReductive surgery for ovarian cancer patients: A systematic review and meta-analysis. Eur J Surg Oncol 2019;45:301-9. [Crossref] [PubMed]
  41. Wu Q, Wu Q, Xu J, et al. Efficacy of hyperthermic intraperitoneal chemotherapy in patients with epithelial ovarian cancer: a meta-analysis. Int J Hyperthermia 2019;36:562-72. [Crossref] [PubMed]
  42. Kireeva GS, Gafton GI, Guseynov KD, et al. HIPEC in patients with primary advanced ovarian cancer: Is there a role? A systematic review of short- and long-term outcomes. Surg Oncol 2018;27:251-8. [Crossref] [PubMed]
  43. Huo YR, Richards A, Liauw W, et al. 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. [Crossref] [PubMed]
  44. Kim SI, Kim JH, Lee S, et al. Hyperthermic intraperitoneal chemotherapy for epithelial ovarian cancer: A meta-analysis. Gynecol Oncol 2022;167:547-56. [Crossref] [PubMed]
  45. Jian C, Mou H, Zhang Y, et al. Survival and complications of cytoreductive surgery with hyperthermic intraperitoneal chemotherapy in patients with intra-abdominal malignancies: A meta-analysis of randomized controlled trials. Front Pharmacol 2023;14:1094834. [Crossref] [PubMed]
  46. 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;27:1134-40. [Crossref] [PubMed]
  47. Chambers LM, Yao M, Morton M, et al. Patterns of recurrence in women with advanced and recurrent epithelial ovarian cancer treated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Gynecol Oncol 2021;161:389-95. [Crossref] [PubMed]
  48. Chambers LM, Chau D, Yao M, et al. Efficacy of hyperthermic intraperitoneal chemotherapy and interval debulking surgery in women with advanced uterine serous carcinoma. Gynecol Oncol Rep 2021;38:100876. [Crossref] [PubMed]
  49. Zivanovic O, Chi DS, Zhou Q, 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;39:2594-604. [Crossref] [PubMed]
  50. Bakrin N, Classe JM, Pomel C, et al. Hyperthermic intraperitoneal chemotherapy (HIPEC) in ovarian cancer. J Visc Surg 2014;151:347-53. [Crossref] [PubMed]
  51. Spiliotis J, Halkia E, Lianos E, et al. Cytoreductive surgery and HIPEC in recurrent epithelial ovarian cancer: a prospective randomized phase III study. Ann Surg Oncol 2015;22:1570-5. [Crossref] [PubMed]
  52. Costales AB, Chambers L, Chichura A, et al. Effect of platinum sensitivity on the efficacy of hyperthermic intraperitoneal chemotherapy (HIPEC) in recurrent epithelial ovarian cancer. J Gynecol Obstet Hum Reprod 2021;50:101844. [Crossref] [PubMed]
  53. Issels RD. Hyperthermia adds to chemotherapy. Eur J Cancer 2008;44:2546-54. [Crossref] [PubMed]
  54. Dunne M, Regenold M, Allen C. Hyperthermia can alter tumor physiology and improve chemo- and radio-therapy efficacy. Adv Drug Deliv Rev 2020;163-164:98-124. [Crossref] [PubMed]
  55. Yi GY, Kim MJ, Kim HI, et al. Hyperthermia Treatment as a Promising Anti-Cancer Strategy: Therapeutic Targets, Perspective Mechanisms and Synergistic Combinations in Experimental Approaches. Antioxidants (Basel) 2022;11:625. [Crossref] [PubMed]
  56. Overgaard J. Effect of hyperthermia on malignant cells in vivo. A review and a hypothesis. Cancer 1977;39:2637-46. [Crossref] [PubMed]
  57. Overgaard J. Formula to estimate the thermal enhancement ratio of a single simultaneous hyperthermia and radiation treatment. Acta Radiol Oncol 1984;23:135-9. [Crossref] [PubMed]
  58. Helderman RFCPA, Löke DR, Verhoeff J, et al. The Temperature-Dependent Effectiveness of Platinum-Based Drugs Mitomycin-C and 5-FU during Hyperthermic Intraperitoneal Chemotherapy (HIPEC) in Colorectal Cancer Cell Lines. Cells 2020;9:1775. [Crossref] [PubMed]
  59. Rubio-López JD, Durán-Martínez M, Moreno-Blázquez A, et al. Intraoperative metabolic changes associated with cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. Langenbecks Arch Surg 2023;408:34. [Crossref] [PubMed]
  60. Neuwirth MG, Alexander HR, Karakousis GC. Then and now: cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (HIPEC), a historical perspective. J Gastrointest Oncol 2016;7:18-28. [PubMed]
  61. Koole SN, Schouten PC, Hauke J, et al. Effect of HIPEC according to HRD/BRCAwt genomic profile in stage III ovarian cancer: Results from the phase III OVHIPEC trial. Int J Cancer 2022;151:1394-404. [Crossref] [PubMed]
  62. Ghirardi V, De Felice F, D'Indinosante M, et al. Hyperthermic intraperitoneal chemotherapy (HIPEC) after primary debulking surgery in advanced epithelial ovarian cancer: Is BRCA mutational status making the difference? Cancer Treat Res Commun 2022;31:100518. [Crossref] [PubMed]
  63. Schaaf L, Schwab M, Ulmer C, et al. Hyperthermia Synergizes with Chemotherapy by Inhibiting PARP1-Dependent DNA Replication Arrest. Cancer Res 2016;76:2868-75. [Crossref] [PubMed]
  64. Classe JM, Meeus P, Leblanc E, et al. Hyperthermic intraperitoneal chemotherapy in platinum-sensitive relapsed epithelial ovarian cancer: The CHIPOR randomized phase III trial. J Clin Oncol 2023;41:5510. [Crossref]
  65. Petrillo M, Zucchetti M, Cianci S, et al. Pharmacokinetics of cisplatin during open and minimally-invasive secondary cytoreductive surgery plus HIPEC in women with platinum-sensitive recurrent ovarian cancer: a prospective study. J Gynecol Oncol 2019;30:e59. [Crossref] [PubMed]
  66. Fagotti A, Petrillo M, Costantini B, et al. Minimally invasive secondary cytoreduction plus HIPEC for recurrent ovarian cancer: a case series. Gynecol Oncol 2014;132:303-6. [Crossref] [PubMed]
  67. Morton M, Chambers LM, Costales AB, et al. Assessing feasibility and perioperative outcomes with minimally invasive surgery compared with laparotomy for interval debulking surgery with hyperthermic intraperitoneal chemotherapy for advanced epithelial ovarian cancer. Gynecol Oncol 2021;160:45-50. [Crossref] [PubMed]
  68. Evans SS, Repasky EA, Fisher DT. Fever and the thermal regulation of immunity: the immune system feels the heat. Nat Rev Immunol 2015;15:335-49. [Crossref] [PubMed]
  69. Scutigliani EM, Lobo-Cerna F, Mingo Barba S, et al. The Effects of Heat Stress on the Transcriptome of Human Cancer Cells: A Meta-Analysis. Cancers (Basel) 2022;15:113. [Crossref] [PubMed]
  70. Minnaar CA, Szasz A. Forcing the Antitumor Effects of HSPs Using a Modulated Electric Field. Cells 2022;11:1838. [Crossref] [PubMed]
  71. Cyran AM, Zhitkovich A. Heat Shock Proteins and HSF1 in Cancer. Front Oncol 2022;12:860320. [Crossref] [PubMed]
  72. Seclì L, Fusella F, Avalle L, et al. The dark-side of the outside: how extracellular heat shock proteins promote cancer. Cell Mol Life Sci 2021;78:4069-83. [Crossref] [PubMed]
  73. Javid H, Hashemian P, Yazdani S, et al. The role of heat shock proteins in metastatic colorectal cancer: A review. J Cell Biochem 2022;123:1704-35. [Crossref] [PubMed]
  74. Scutigliani EM, Liang Y, Crezee H, et al. Modulating the Heat Stress Response to Improve Hyperthermia-Based Anticancer Treatments. Cancers (Basel) 2021;13:1243. [Crossref] [PubMed]
  75. Toraya-Brown S, Sheen MR, Zhang P, et al. Local hyperthermia treatment of tumors induces CD8(+) T cell-mediated resistance against distal and secondary tumors. Nanomedicine 2014;10:1273-85. [Crossref] [PubMed]
  76. Nevo N, Lee Goldstein A, Bar-David S, et al. Immunological effects of heated intraperitoneal chemotherapy can be augmented by thymosin α1. Int Immunopharmacol 2023;116:109829. [Crossref] [PubMed]
  77. Geva R, Alon G, Nathanson M, et al. PD-1 Blockade Combined with Heated Intraperitoneal Chemotherapy Improves Outcome in Experimental Peritoneal Metastases from Colonic Origin in a Murine Model. Ann Surg Oncol 2023;30:2657-63. [Crossref] [PubMed]
  78. Wu CC, Hsu YT, Chang CL. Hyperthermic intraperitoneal chemotherapy enhances antitumor effects on ovarian cancer through immune-mediated cancer stem cell targeting. Int J Hyperthermia 2021;38:1013-22. [Crossref] [PubMed]
  79. Huang WC, Wu CC, Hsu YT, et al. Effect of hyperthermia on improving neutrophil restoration after intraperitoneal chemotherapy. Int J Hyperthermia 2019;36:1255-63. [Crossref] [PubMed]
  80. Zunino B, Ricci JE. Hyperthermic intra-peritoneal chemotherapy and anticancer immune response. Oncoimmunology 2015;5:e1060392. [Crossref] [PubMed]
  81. Acs M, Gerken M, Schmitt V, et al. Role of HIPEC after Complete Cytoreductive Surgery (CRS) in Peritoneal Recurrence of Platinum-Sensitive Recurrent Ovarian Cancer (OC): The Aim for Standardization at Two Reference Centers for CRS. Cancers (Basel) 2023;15:405. [Crossref] [PubMed]
  82. Piso P, Nedelcut SD, Rau B, et al. Morbidity and Mortality Following Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy: Data from the DGAV StuDoQ Registry with 2149 Consecutive Patients. Ann Surg Oncol 2019;26:148-54. [Crossref] [PubMed]
  83. Fornasiero M, Geropoulos G, Kechagias KS, et al. Anastomotic Leak in Ovarian Cancer Cytoreduction Surgery: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022;14:5464. [Crossref] [PubMed]
  84. Sin EI, Chia CS, Tan GHC, et al. Acute kidney injury in ovarian cancer patients undergoing cytoreductive surgery and hyperthermic intra-peritoneal chemotherapy. Int J Hyperthermia 2017;33:690-5. [Crossref] [PubMed]
  85. Liesenfeld LF, Quiring E, Al-Saeedi M, et al. Extensive Peritonectomy is an Independent Risk Factor for Cisplatin HIPEC-Induced Acute Kidney Injury. Ann Surg Oncol 2023;30:2646-56. [Crossref] [PubMed]
  86. Kurreck A, Gronau F, Alberto Vilchez ME, et al. Sodium Thiosulfate Reduces Acute Kidney Injury in Patients Undergoing Cytoreductive Surgery Plus Hyperthermic Intraperitoneal Chemotherapy with Cisplatin: A Single-Center Observational Study. Ann Surg Oncol 2022;29:152-62. [Crossref] [PubMed]
  87. Bai Y, Du Y, Ye P, et al. Acute kidney injury after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in patients with advanced ovarian cancer. Front Oncol 2023;13:1094410. [Crossref] [PubMed]
  88. Koole SN, Kieffer JM, Sikorska K, et al. Health-related quality of life after interval cytoreductive surgery with or without hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with stage III ovarian cancer. Eur J Surg Oncol 2021;47:101-7. [Crossref] [PubMed]
  89. Kim JH, Lee DE, Lee Y, et al. Quality of life outcomes from the randomized trial of hyperthermic intraperitoneal chemotherapy following cytoreductive surgery for primary ovarian cancer (KOV-HIPEC-01). J Gynecol Oncol 2022;33:e54. [Crossref] [PubMed]
  90. Kim JH, Park KN, Park EY, et al. Impact of warm saline irrigation, hyperthermic intraperitoneal chemotherapy on postoperative pain in primary ovarian cancer from the KOV-HIPEC-01 randomized trial. Gynecol Oncol 2023;177:32-7. [Crossref] [PubMed]
  91. Aronson L, Lopez-Yurda MI, Koole SN, et al. Final survival analysis of the phase III OVHIPEC-1 trial of hyperthermic intraperitoneal chemotherapy in ovarian cancer after ten year follow-up. J Clin Oncol 2023;41:5509. [Crossref]
Cite this article as: Piątek S, Sarnowska E, Kacperczyk-Bartnik J, Kołaczkowska H, Sobiczewski P, Bidziński M. Discrepancy between recommendations regarding hyperthermic intraperitoneal chemotherapy (HIPEC) in ovarian cancer management: a narrative review. Chin Clin Oncol 2024;13(3):37. doi: 10.21037/cco-23-152

Download Citation