Foregut Surg. 2024 Jan;4(1):6-16. English.
Published online Jan 17, 2024.
Copyright © 2024. Korean Society of Gastrointestinal Surgery
Review

Intraperitoneal paclitaxel for gastric cancer with peritoneal metastasis

Jong-Han Kim
    • Department of Surgery, Korea University College of Medicine, Korea University Guro Hospital, Seoul, Republic of Korea.
Received December 11, 2023; Revised January 09, 2024; Accepted January 09, 2024.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Peritoneal seeding is the most prevalent and grave form of metastatic gastric cancer (GC), and is a poor prognostic factor that is unresponsive to conventional systemic chemotherapy. Peritoneal metastasis (PM) in GC can be treated using several methods, including systemic chemotherapy, intraperitoneal (IP) chemotherapy, and cytoreductive surgery with hyperthermic IP chemotherapy for treating PM in GC. Diverse treatment outcomes of IP chemotherapy for GC with PM have been reported. Therefore, comprehensive therapeutic approaches are required to improve the survival of patients with PM. Recent reports have highlighted several favorable outcomes of the use of simultaneous IP and systemic chemotherapy, especially the use of paclitaxel as a chemotherapeutic agent in the treatment of peritoneal seeding in GC. Additionally, laparoscopic surgery may play a role in the treatment of peritoneal seeding, while innovative strategies, such as pressurized IP aerosol chemotherapy and new materials for IP chemotherapy, present promising strategies for addressing this challenging disease.

Keywords
Chemotherapy; Gastric cancer; Paclitaxel; Peritoneum

INTRODUCTION

Although gastric cancer (GC) is considered potentially curable, this may not be true for patients with stage IV advanced GC, particularly those with peritoneal metastases (PMs). Despite recent advances in systemic chemotherapy, the prognosis of GC patients with PM remains controversial, with a median survival of 6–14 months [1, 2, 3, 4]. Among patients with stage IV GC, the peritoneum is the most common single site of distant metastasis, accounting for 14% of all GC cases in Western populations and >40% of cases in a single Korean institution [5, 6]. Interestingly, >1,800 patients were diagnosed with peritoneal seeding in Korea in 2019 [7].

In multivariate logistic regression models for tumor response to chemotherapy in stage IV GC, PM was the worst prognostic factor [8]. The recent Korean Gastric Cancer Association guidelines recommend only palliative systemic chemotherapy for stage IV GC, regardless of the metastasis site. To date, no tailored treatment strategy has been established for patients with PM [9].

In general, PM progresses from the primary tumor to the serosal exposure. Intraperitoneal (IP)-free cancer cells induce gross PM. Malignant ascites and peritoneal carcinomatosis are observed. Currently, 2 molecular models of PM in GC are available. The trans-mesothelial metastasis model suggests that IP-free cancer cells can attach to the peritoneal basement membrane between the peritoneal mesothelial cells. Conversely, the translymphatic metastasis model indicates that IP-free cancer cells can directly attach to the peritoneal lymphatic orifices, known as the lymphatic stroma [10]. Furthermore, the molecular mechanism of peritoneal carcinomatosis is complex and dynamic, and involves diverse molecules that act in a coordinated manner. Therefore, multimodal treatments, which are systemic IP chemotherapy and conversion surgery, may be applicable owing to the ineffectiveness of conventional chemotherapy (Fig. 1) [11].

Fig. 1
Mechanisms of peritoneal dissemination in gastric cancer including trans-mesothelial metastasis (A) and translymphatic metastasis (B).
PMC = peritoneal mesothelial cell; CAF = cancer associated fibroblast; TAM = tumor associated macrophage; CXCL12 = C-X-C motif chemokine ligand 12; ECM = extracellular matrix.

SPECIFIC CONSIDERATION OF IP CHEMOTHERAPY

IP paclitaxel for peritoneal dissemination

Peritoneal-plasma barrier is a 90-μm thick barrier between the peritoneum and plasma area that is composed of a monolayer of mesothelial tissue. This retards the clearance of high-molecular-weight chemotherapeutic agents from the peritoneum and prevents the exposure of small cancer nodules on the surfaces of the abdomen and pelvis. Therefore, systemic chemotherapy can pose challenges, necessitating the direct administration of chemotherapeutic drugs into the peritoneal cavity (Fig. 2) [12].

Fig. 2
Peritoneal-plasma barrier.
Anatomy of the peritoneal membrane (upper) and intraperitoneal drug transport between the tumor tissue in the peritoneum and blood vessels (lower).

IP paclitaxel is macromolecular fat-soluble, can be absorbed slowly from the peritoneal cavity, and is preserved at high concentrations in the peritoneal cavity, with minimal toxicity. Additionally, it is antiproliferative; therefore, the IP administration of chemotherapeutic drugs can be repeated without adhesions [13]. Compared to those of other known effective drugs for GC, including cisplatin, oxaliplatin, and 5-fluorouracil, paclitaxel has a higher molecular weight and area under the curve ratio, enabling it to penetrate more than 80 cell layers [14, 15, 16]. However, it has limitations regarding its ability to infiltrate tumors deeply and cover a restricted delivery area in the peritoneal cavity. Thus, we should combine it with systemic chemotherapy. Therefore, the IP administration of paclitaxel is considered a promising treatment for eliminating PM in GC because it penetrates directly into disseminated tumors (Fig. 3) [14, 15].

Fig. 3
Characteristics of intraperitoneal paclitaxel.

Role of laparoscopy in IP chemotherapy

For peritoneal carcinomatosis, preoperative computed tomography and positron emission tomography underestimate the tumor burden because metastatic peritoneal implants <8 mm cannot be accurately assessed. Therefore, laparoscopy can be an alternative for the assessment of the peritoneal cancer index (PCI) and histological diagnosis, making it a highly accurate diagnostic modality for peritoneal carcinomatosis [16, 17]. For the laparoscopic evaluation of peritoneal carcinomatosis, the PCI score was proposed by Sugarbaker for the laparoscopic evaluation of peritoneal carcinomatosis. This is a quantitative assessment of cancer distribution and implant size, measured by the greatest diameter (Fig. 4) [18].

Fig. 4
PCI score.
The PCI score was proposed by Jacquet and Sugarbaker [18] for the laparoscopic evaluation of peritoneal carcinomatosis. This is a quantitative assessment of cancer distribution and implant size, measured by the greatest diameter.

PCI = peritoneal cancer index.

Recently, Yonemura et al. [19, 20] proposed the significance of a comprehensive treatment for gastric peritoneal carcinomatosis in Japan that evolved with laparoscopic surgery. First, laparoscopic hyperthermic IP chemotherapy has the advantage of greater depth of drug penetration from the peritoneal surface compared to that associated with open hyperthermic IP chemotherapy. Bidirectional IP, systemic induction chemotherapy, and extensive peritoneal lavage can be administered [19]. Therefore, the authors reported the results of neoadjuvant laparoscopic hyperthermic IP chemotherapy and neoadjuvant IP/systemic chemotherapy. In this study, neoadjuvant IP and systemic chemotherapy reduced the PCI scores by approximately 50% before cytoreductive surgery [20].

Although laparoscopy is a useful tool for PM, the PCI score cannot be calculated in 70% of cases because of the incomplete visualization of the abdominal cavity and multiple tumor manifestations. The extent of tumor involvement in the small intestine appears to be more relevant than the calculation of the PCI score [21].

Recent results of IP paclitaxel chemotherapy

A meta-analysis comparing intravenous (IV) plus IP chemotherapy with IV chemotherapy alone included 5 randomized controlled trials (RCTs) and 1,072 patients. In the present study, the IP group showed better overall survival rates and the prevention of distant PM [22].

In several phase I and II trials, the clinical outcomes of IP chemotherapy with paclitaxel and systemic chemotherapy indicated an increase in the median survival time of up to 2 years, with an overall survival rate >70%. Furthermore, the overall response rate has been reported to be 40%–70% (Table 1) [23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34].

Table 1
Studies of intraperitoneal paclitaxel with systemic chemotherapy for gastric cancer with peritoneal metastasis

Prof. Ishigami at Tokyo University in Japan proposed a phase III study of IP paclitaxel plus systemic S-1 and paclitaxel compared with systemic S-1 and cisplatin (SP), namely, the PHEONIX-GC trial. In the 2018, the results of the PHEONIX-GC trial were reported. Especially, the median survival times for the IP and SP arms were approximately 18 and 15 months, respectively, and the 3-year overall survival rates were approximately 22% and 6% in the IP and SP arms, respectively. Therefore, despite a trend toward survival benefits, this trial did not demonstrate the statistical superiority of IP paclitaxel plus systemic chemotherapy. However, in a subgroup analysis of overall survival, they detected a significant interaction between the treatment effect and the amount of ascites. Analysis of the ascites volume revealed that the IP group had a more favorable response. Thus, this trial suggests possible clinical benefits of IP paclitaxel therapy [35].

In 2017, Korean gastrointestinal surgeons planned to establish a multicenter trial that included a Perioperative Intraperitoneal and Systemic Chemotherapy for Gastric Cancer (PIPS-GC) trial. First, they gathered and reported multicenter retrospective data on IP and systemic chemotherapy with paclitaxel. Eighty-two patients from 2015 to 2018 were included in the analysis. The mean number of IP chemotherapy cycles was 6.6. Regarding the type of palliative surgery, 51 cases were synchronous and the mean PCI was 22. Postoperative complications were reported in 24 patients, and IP catheter-related and port-related complications were observed in 15 patients [36].

Two recent clinical trials on IP chemotherapy have been conducted in South Korea. Paclitaxel was used in both trials as an IP chemotherapy regimen. The PIPS-GC trial is a multicenter study that primarily included phase I and II trials. Twelve hospitals participated in that study [27]. However, the IPLUS study was a single-center trial involving a surgeon, oncologist, pathologist, and radiologist participating in this study. The IPLUS study group reported the results of a phase I study. Two cases of dose-limiting toxicity occurred with 80 mg; therefore, they decided to administer 60 mg, which is the recommended phase II dose. Of the 6 patients, 3 showed a partial response, 2 showed stable disease, and 1 showed progression. Conversion surgery was possible in three patients. The median overall survival period was 16.6 months and progression free survival was 9.6 months [32].

Nakashima et al. [37] reported the results of a retrospective study comparing IP paclitaxel with non-IP systemic chemotherapy and showed better survival outcomes in the IP paclitaxel group.

Conversion surgery for peritoneal dissemination

Conversion therapy for stage IV GC has recently attracted considerable attention. It is defined as a surgical treatment with no residual curative resection after chemotherapy for tumors that were originally unresectable or marginally resectable for technical and/or oncological reasons [38]. In 2016, Yoshida et al. [39] proposed a classification system for stage IV GC based on resectability and curability. This system is largely determined by macroscopic peritoneal dissemination. Therefore, the efficacy of IP and systemic chemotherapy in cases of macroscopic peritoneal dissemination suggests that achieving complete disappearance of lesions through IP chemotherapy might enable the prospect of R0 surgery, even in patients diagnosed with PM.

Ishigami et al. [40] reported the results of IP paclitaxel chemotherapy. In 2017, the authors reported conversion surgery after IP and systemic chemotherapy for PM. In total, 100 patients were enrolled, 64 of whom underwent conversion gastrectomy. The median survival time was 30 months, and better survival was noted in the surgical cases, especially in R0 cases.

In 2017, Yonemura et al. [20] proposed neoadjuvant laparoscopic hyperthermic intraperitoneal chemoperfusion (NLHIPEC) and neoadjuvant intraperitoneal and systemic chemotherapy (NIPS). In this study, NLHIPEC and NIPS reduced the PCI score in approximately 50% of the patients before cytoreductive surgery.

Recently, Kim et al. [41] reported the results of minimally invasive surgery combined with IP and systemic chemotherapy for with GC patients with PM in Korea. In total, 26 patients were enrolled, and five of them underwent conversion surgery, including laparoscopic gastrectomy.

Other modalities of IP chemotherapy

Pressurized intraperitoneal aerosol chemotherapy (PIPAC) has emerged as an alternative treatment for peritoneal carcinomatosis. Fig. 5 illustrates the PIPAC procedure using a laparoscopic CO2 insufflator and a high-pressure injector [42].

Fig. 5
Pressurized intraperitoneal aerosol chemotherapy systm.

In 2016, Nadiradze and colleagues [43] published the first report of PIPAC for gastric peritoneal carcinomatosis. Sixty PIPAC procedures were performed in 24 patients and tumor responses were observed in 50% of them. Subsequently, several studies have reported the application of PIPAC in GC-associated PM. Two studies focused exclusively on GC patients, whereas the others included heterogeneous populations [43, 44]. A single study emphasized the role of PIPAC in the neoadjuvant setting [45]. All studies revealed that PIPAC is feasible and has minimal perioperative morbidity even after repeated applications [46].

Thereafter, early clinical trials of PIPAC therapy using albumin-bound paclitaxel have been reported, which may result in superior efficacy in the treatment of PM compared to that associated with standard IP paclitaxel chemotherapy [47, 48]. Additionally, the retrospective results of PIPAC with taxanes have been reported by an Indian group. Thus, the role of PIPAC in combination with docetaxel and paclitaxel, either alone or in combination with other drugs, should be investigated in future prospective studies [49].

Nab-paclitaxel is an albumin-bound nanoparticle formulation of paclitaxel specifically designed to overcome the limitations of conventional paclitaxel formulations, including barriers to effective drug delivery of highly lipophilic agents [50]. Nab-paclitaxel has fewer side effects, increased tumor cell cytotoxicity, and higher overall response rates than those associated with equal doses of solvent-based paclitaxel for many solid malignancies [51]. Recently, a phase I study protocol for IP cisplatin and nab-paclitaxel administered via the PIPAC for the treatment of gastric peritoneal carcinomatosis was published [52].

CONCLUSIONS

The IP administration of anticancer drugs can induce extremely high drug concentrations in the peritoneal cavity, and IP chemotherapy appears to be a reasonable and promising strategy for controlling peritoneal dissemination. Paclitaxel administered into the peritoneal cavity is a chemotherapeutic agent that shows unusually prolonged retention within the peritoneal space. Therefore, the use of this drug to treat patients with PM was investigated.

Laparoscopy is a highly accurate diagnostic modality for detecting peritoneal carcinomatosis in patients with advanced GC. Additionally, laparoscopic surgery may play a role in the treatment of peritoneal seeding, including staging, IP catheter insertion, and palliative surgery.

Favorable outcomes of simultaneous IP paclitaxel and systemic chemotherapy have been reported in several cases involving the peritoneal dissemination of GC. Promising results from several phase II clinical trials and RCTs using IP paclitaxel have shown inconclusive but suggestive outcomes. Additionally, conversion surgery after repeated IP paclitaxel treatment has been reported in GC patients with PM.

In future studies, further therapeutic forms, such as PIPAC, and new materials, such as nanoparticle albumin-bound paclitaxel for IP chemotherapy, are anticipated to achieve better outcomes in GC patients with PM.

Notes

Funding:The author has no financial support to disclose.

Conflict of Interest:No potential conflict of interest relevant to this article was reported.

References

    1. Koizumi W, Narahara H, Hara T, Takagane A, Akiya T, Takagi M, et al. S-1 plus cisplatin versus S-1 alone for first-line treatment of advanced gastric cancer (SPIRITS trial): a phase III trial. Lancet Oncol 2008;9:215–221.
    1. Kang YK, Kang WK, Shin DB, Chen J, Xiong J, Wang J, et al. Capecitabine/cisplatin versus 5-fluorouracil/cisplatin as first-line therapy in patients with advanced gastric cancer: a randomised phase III noninferiority trial. Ann Oncol 2009;20:666–673.
    1. Wilke H, Muro K, Van Cutsem E, Oh SC, Bodoky G, Shimada Y, et al. Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 2014;15:1224–1235.
    1. Bang YJ, Van Cutsem E, Feyereislova A, Chung HC, Shen L, Sawaki A, et al. Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 2010;376:687–697.
    1. Thomassen I, van Gestel YR, van Ramshorst B, Luyer MD, Bosscha K, Nienhuijs SW, et al. Peritoneal carcinomatosis of gastric origin: a population-based study on incidence, survival and risk factors. Int J Cancer 2014;134:622–628.
    1. Lee CM, Choi IK, Kim JH, Park DW, Kim JS, Park SH. Is noncurative gastrectomy always a beneficial strategy for stage IV gastric cancer? Ann Surg Treat Res 2017;92:23–27.
    1. Information Committee of the Korean Gastric Cancer Association. Korean Gastric Cancer Association-led nationwide survey on surgically treated gastric cancers in 2019. J Gastric Cancer 2021;21:221–235.
    1. Chau I, Norman AR, Cunningham D, Waters JS, Oates J, Ross PJ. Multivariate prognostic factor analysis in locally advanced and metastatic esophago-gastric cancer--pooled analysis from three multicenter, randomized, controlled trials using individual patient data. J Clin Oncol 2004;22:2395–2403.
    1. Kim TH, Kim IH, Kang SJ, Choi M, Kim BH, Eom BW, et al. Korean Practice Guidelines for Gastric Cancer 2022: an evidence-based, multidisciplinary approach. J Gastric Cancer 2023;23:3–106.
    1. Sun F, Feng M, Guan W. Mechanisms of peritoneal dissemination in gastric cancer. Oncol Lett 2017;14:6991–6998.
    1. Kanda M, Kodera Y. Molecular mechanisms of peritoneal dissemination in gastric cancer. World J Gastroenterol 2016;22:6829–6840.
    1. Sugarbaker PH, Van der Speeten K, Stuart OA. Pharmacologic rationale for treatments of peritoneal surface malignancy from colorectal cancer. World J Gastrointest Oncol 2010;2:19–30.
    1. Kamei T, Kitayama J, Yamaguchi H, Soma D, Emoto S, Konno T, et al. Spatial distribution of intraperitoneally administrated paclitaxel nanoparticles solubilized with poly (2-methacryloxyethyl phosphorylcholine-co n-butyl methacrylate) in peritoneal metastatic nodules. Cancer Sci 2011;102:200–205.
    1. Yonemura Y, Endou Y, Bando E, Kuno K, Kawamura T, Kimura M, et al. Effect of intraperitoneal administration of docetaxel on peritoneal dissemination of gastric cancer. Cancer Lett 2004;210:189–196.
    1. Soma D, Kitayama J, Ishigami H, Kaisaki S, Nagawa H. Different tissue distribution of paclitaxel with intravenous and intraperitoneal administration. J Surg Res 2009;155:142–146.
    1. Fujimura T, Kinami S, Ninomiya I, Kitagawa H, Fushida S, Nishimura G, et al. Diagnostic laparoscopy, serum CA125, and peritoneal metastasis in gastric cancer. Endoscopy 2002;34:569–574.
    1. Ramos RF, Scalon FM, Scalon MM, Dias DI. Staging laparoscopy in gastric cancer to detect peritoneal metastases: a systematic review and meta-analysis. Eur J Surg Oncol 2016;42:1315–1321.
    1. Jacquet P, Sugarbaker PH. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. Cancer Treat Res 1996;82:359–374.
    1. Yonemura Y, Canbay E, Li Y, Coccolini F, Glehen O, Sugarbaker PH, et al. A comprehensive treatment for peritoneal metastases from gastric cancer with curative intent. Eur J Surg Oncol 2016;42:1123–1131.
    1. Yonemura Y, Ishibashi H, Hirano M, Mizumoto A, Takeshita K, Noguchi K, et al. Effects of neoadjuvant laparoscopic hyperthermic intraperitoneal chemotherapy and neoadjuvant intraperitoneal/systemic chemotherapy on peritoneal metastases from gastric cancer. Ann Surg Oncol 2017;24:478–485.
    1. von Breitenbuch P, Boerner T, Jeiter T, Piso P, Schlitt HJ. Laparoscopy as a useful selection tool for patients with prior surgery and peritoneal metastases suitable for multimodality treatment strategies. Surg Endosc 2018;32:2288–2294.
    1. Yang S, Feng R, Pan ZC, Jiang T, Xu Q, Chen Q. A comparison of intravenous plus intraperitoneal chemotherapy with intravenous chemotherapy alone for the treatment of gastric cancer: a meta-analysis. Sci Rep 2015;5:12538.
    1. Ishigami H, Kitayama J, Kaisaki S, Hidemura A, Kato M, Otani K, et al. Phase II study of weekly intravenous and intraperitoneal paclitaxel combined with S-1 for advanced gastric cancer with peritoneal metastasis. Ann Oncol 2010;21:67–70.
    1. Imano M, Yasuda A, Itoh T, Satou T, Peng YF, Kato H, et al. Phase II study of single intraperitoneal chemotherapy followed by systemic chemotherapy for gastric cancer with peritoneal metastasis. J Gastrointest Surg 2012;16:2190–2196.
    1. Yamaguchi H, Kitayama J, Ishigami H, Emoto S, Yamashita H, Watanabe T. A phase 2 trial of intravenous and intraperitoneal paclitaxel combined with S-1 for treatment of gastric cancer with macroscopic peritoneal metastasis. Cancer 2013;119:3354–3358.
    1. Saito S, Yamaguchi H, Ohzawa H, Miyato H, Kanamaru R, Kurashina K, et al. Intraperitoneal administration of paclitaxel combined with S-1 plus oxaliplatin as induction therapy for patients with advanced gastric cancer with peritoneal metastases. Ann Surg Oncol 2021;28:3863–3870.
    1. Kim DW, Seo WJ, Youn SI, Jee YS, Jang YJ, Kim JH, et al. Intraperitoneal paclitaxel combined with S-1 plus oxaliplatin for advanced gastric cancer with peritoneal metastasis: a phase I study. J Gastric Cancer 2021;21:418–425.
    1. Shinkai M, Imano M, Hiraki Y, Momose K, Kato H, Shiraishi O, et al. Efficacy of conversion surgery after a single intraperitoneal administration of paclitaxel and systemic chemotherapy for gastric cancer with peritoneal metastasis. Langenbecks Arch Surg 2022;407:975–983.
    1. Shi M, Yang Z, Lu S, Liu W, Ni Z, Yao X, et al. Oxaliplatin plus S-1 with intraperitoneal paclitaxel for the treatment of Chinese advanced gastric cancer with peritoneal metastases. BMC Cancer 2021;21:1344.
    1. Yang ZY, Yuan F, Lu S, Xu W, Wu JW, Xi WQ, et al. Efficacy and safety of conversion therapy by intraperitoneal and intravenous paclitaxel plus oral S-1 in gastric cancer patients with peritoneal metastasis: a prospective phase II study. Front Oncol 2022;12:905922
    1. Chia DK, Sundar R, Kim G, Ang JJ, Lum JH, Nga ME, et al. Outcomes of a phase II study of intraperitoneal paclitaxel plus systemic capecitabine and oxaliplatin (XELOX) for gastric cancer with peritoneal metastases. Ann Surg Oncol 2022;29:8597–8605.
    1. Kang SH, Min SH, Kim JW, Lee E, Park SW, Lee S, et al. Safety and efficacy of intraperitoneal paclitaxel plus intravenous fluorouracil, leucovorin, and oxaliplatin (FOLFOX) for gastric cancer with peritoneal metastasis. Ann Surg Oncol 2022;29:5084–5091.
    1. Tu L, Zhang W, Ni L, Xu Z, Yang K, Gou H, et al. Study of SOX combined with intraperitoneal high-dose paclitaxel in gastric cancer with synchronous peritoneal metastasis: a phase II single-arm clinical trial. Cancer Med 2023;12:4161–4169.
    1. Kobayashi D, Kodera Y, Fukushima R, Morita M, Fushida S, Yamashita N, et al. Phase II study of intraperitoneal administration of paclitaxel combined with S-1 and cisplatin for gastric cancer with peritoneal metastasis. Ann Surg Oncol 2024;31:735–743.
    1. Ishigami H, Fujiwara Y, Fukushima R, Nashimoto A, Yabusaki H, Imano M, et al. Phase III trial comparing intraperitoneal and intravenous paclitaxel plus S-1 versus cisplatin plus S-1 in patients with gastric cancer with peritoneal metastasis: PHOENIX-GC trial. J Clin Oncol 2018;36:1922–1929.
    1. Kim DW, Jee YS, Kim CH, Kim JJ, Park S, Choi SI, et al. Multicenter retrospective analysis of intraperitoneal paclitaxel and systemic chemotherapy for advanced gastric cancer with peritoneal metastasis. J Gastric Cancer 2020;20:50–59.
    1. Nakashima T, Arigami T, Uenosono Y, Matsushita D, Shimonosono M, Tsuruda Y, et al. Clinical significance of intraperitoneal paclitaxel combined with systemic chemotherapy for gastric cancer with peritoneal metastasis. Int J Clin Oncol 2023;28:1371–1377.
    1. Terashima M. Conversion therapy for gastric cancer: who can make conversion as successful as Goromaru? Gastric Cancer 2016;19:685–686.
    1. Yoshida K, Yamaguchi K, Okumura N, Tanahashi T, Kodera Y. Is conversion therapy possible in stage IV gastric cancer: the proposal of new biological categories of classification. Gastric Cancer 2016;19:329–338.
    1. Ishigami H, Yamaguchi H, Yamashita H, Asakage M, Kitayama J. Surgery after intraperitoneal and systemic chemotherapy for gastric cancer with peritoneal metastasis or positive peritoneal cytology findings. Gastric Cancer 2017;20:128–134.
    1. Kim S, Lee CM, Lee D, Kim JH, Park S, Park SH. Can we reboot the role of intraperitoneal chemotherapy in the treatment for gastric cancer with peritoneal carcinomatosis?: a retrospective cohort study regarding minimally invasive surgery conjoined with intraperitoneal plus systemic chemotherapy. Cancers (Basel) 2022;14:2334.
    1. Solass W, Kerb R, Mürdter T, Giger-Pabst U, Strumberg D, Tempfer C, et al. Intraperitoneal chemotherapy of peritoneal carcinomatosis using pressurized aerosol as an alternative to liquid solution: first evidence for efficacy. Ann Surg Oncol 2014;21:553–559.
    1. Nadiradze G, Giger-Pabst U, Zieren J, Strumberg D, Solass W, Reymond MA. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) with low-dose cisplatin and doxorubicin in gastric peritoneal metastasis. J Gastrointest Surg 2016;20:367–373.
    1. Khomyakov V, Ryabov A, Ivanov A, Bolotina L, Utkina A, Volchenko N, et al. Bidirectional chemotherapy in gastric cancer with peritoneal metastasis combining intravenous XELOX with intraperitoneal chemotherapy with low-dose cisplatin and Doxorubicin administered as a pressurized aerosol: an open-label, phase-2 study (PIPAC-GA2). Pleura Peritoneum 2016;1:159–166.
    1. Girshally R, Demtröder C, Albayrak N, Zieren J, Tempfer C, Reymond MA. Pressurized intraperitoneal aerosol chemotherapy (PIPAC) as a neoadjuvant therapy before cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. World J Surg Oncol 2016;14:253.
    1. Garg PK, Jara M, Alberto M, Rau B. The role of pressurized intraperitoneal aerosol chemotherapy in the management of gastric cancer: a systematic review. Pleura Peritoneum 2019;4:20180127
    1. Van De Sande L, Graversen M, Hubner M, Pocard M, Reymond M, Vaira M, et al. Intraperitoneal aerosolization of albumin-stabilized paclitaxel nanoparticles (Abraxane™) for peritoneal carcinomatosis - a phase I first-in-human study. Pleura Peritoneum 2018;3:20180112
    1. Ceelen W, Sandra L, de Sande LV, Graversen M, Mortensen MB, Vermeulen A, et al. Phase I study of intraperitoneal aerosolized nanoparticle albumin based paclitaxel (NAB-PTX) for unresectable peritoneal metastases. EBioMedicine 2022;82:104151
    1. Mehta S, Kammar P, Patel A, Goswami G, Shaikh S, Sukumar V, et al. Feasibility and safety of taxane-PIPAC in patients with peritoneal malignancies-a retrospective bi-institutional study. Indian J Surg Oncol 2022;14:1–9.
    1. Schettini F, Giuliano M, De Placido S, Arpino G. Nab-paclitaxel for the treatment of triple-negative breast cancer: rationale, clinical data and future perspectives. Cancer Treat Rev 2016;50:129–141.
    1. Cullis J, Siolas D, Avanzi A, Barui S, Maitra A, Bar-Sagi D. Macropinocytosis of nab-paclitaxel drives macrophage activation in pancreatic cancer. Cancer Immunol Res 2017;5:182–190.
    1. Lang N, Diciola A, Labidi-Galy I, Ris F, Di Marco M, Mach N, et al. Nab-PIPAC: a phase IB study protocol of intraperitoneal cisplatin and nab-paclitaxel administered by pressurised intraperitoneal aerosol chemotherapy (PIPAC) in the treatment of advanced malignancies confined to the peritoneal cavity. BMJ Open 2023;13:e067691

Metrics
Share
Figures

1 / 5

Tables

1 / 1

ORCID IDs
PERMALINK