Skip to main content
The Journal of International Medical Research logoLink to The Journal of International Medical Research
. 2019 Nov 19;47(12):5926–5936. doi: 10.1177/0300060519882545

Effect of hyperthermic intraperitoneal chemotherapy for gastric cancer patients: a meta-analysis of the randomized controlled trials

Yan-Wen Liu 1, Ying Du 1,✉,*, Bao-An Chen 1,*
PMCID: PMC7045644  PMID: 31741406

Short abstract

Objective

To determine the effectiveness and safety of hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with advanced gastric cancer and peritoneal metastases.

Methods

PubMed®, CNKI, Web of Science, VIP and WANFANG databases were searched to identify randomized controlled trials (RCTs) that examined the effect of HIPEC on survival, clinical response and adverse events. Patients with advanced gastric cancer and peritoneal metastases were divided into an experimental group and a control group. The statistical results are presented as relative ratio (RR), mean difference (MD) and 95% confidence interval (CI).

Results

Twenty-one RCTs met the inclusion criteria (n = 1674 patients). Meta-analysis showed that the 3-year survival rate was significantly higher in the HIPEC group than in the control group (RR 1.61; 95% CI 1.43, 1.82) and the complete response rate was significantly higher in the HIPEC group than in the control group (RR 2.35; 95% CI 1.67, 3.31). HIPEC was also beneficial in terms of decreased CEA (MD −1.79; 95% CI −2.22, −1.35). There was no significant difference in the rate of adverse reactions (RR 1.00; 95% CI 0.87, 1.14).

Conclusions

HIPEC had a beneficial effect on 3-year survival rate and complete response in patients with advanced gastric cancer and peritoneal metastases.

Keywords: Gastric cancer, hyperthermic intraperitoneal chemotherapy, meta-analysis

Introduction

Gastric cancer is one of the most common malignant tumours in the world and it is the second leading cause of death from malignant tumours.1 There are 21 600 new cases of gastric cancer diagnosed in the US every year.2 The National Cancer Centre of Korea predicts 221 347 new cancer cases and 82 344 cancer deaths in Korea during 2019, of which gastric cancer is one of the most common.3 In Asia, the countries with the highest incidence of gastric cancer include China, Mongolia, Japan and South Korea.4 China has the highest incidence of gastric cancer in the world.5 Early surgical treatment can improve long-term survival, while the prognosis of patients with advanced disease is poor, so early diagnosis is the key to successful treatment of gastric cancer.6 The 5-year survival rate for patients with advanced gastric cancer in Europe is only 25%.7

The peritoneum is a common metastatic site in advanced gastric cancer.8 The development of peritoneal metastasis begins with the detachment of a single tumour cell from the primary tumour, which then reaches the abdominal cavity and spreads into the peritoneal fluid. Tumour cells reaching the extracellular matrix can bind to integrins and cause degradation, which ultimately results in the invasion of the submesothelial cell layer.9 The prognosis of patients with peritoneal metastasis from a primary gastric cancer is extremely poor and the median survival time is only 4–6 months.10 There is currently no consensus on the best way to treat patients with peritoneal metastasis from a primary gastric cancer. Since the 1980s, cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) has gradually become the main treatment for peritoneal metastasis from gastrointestinal malignancies and the survival time of strictly selected patients has been significantly prolonged.11 However, this approach remains controversial because the operation is difficult, it is associated with complications and it has a high mortality rate.12 HIPEC involves the continuous heating and thermostatic infusion of a liquid containing chemotherapy drugs into the abdominal cavity to kill any residual cancer cells in the abdominal cavity that cannot be observed by the naked eye.13 In 1979, Spratt first used multiple organ resection combined with hyperthermic intraperitoneal chemotherapy to treat a 35-year-old patient with pseudomyxoma peritonei.14 To evaluate the extent of peritoneal metastasis, the concept of a peritoneal cancer index (PCI) was proposed.15 The PCI is currently the most widely used evaluation tool used to assess peritoneal metastasis, because it not only reflects the cancer burden, but it also determines to the likely prognosis of patients.16The PCI combines the distribution of the tumour cells in 13 abdominal-pelvic regions with lesion size.17 A previous study has shown a perfect linear relationship between PCI and overall survival.18

This present meta-analysis systematically evaluated the published literature in order to investigate the clinical effects of HIPEC used in patients with advanced gastric cancer and peritoneal metastasis.

Materials and methods

Study methods

The methods used in this meta-analysis were in accordance with those proposed by a related meta-analysis.19

Search strategy

A systematic search of publications listed in the electronic databases (PubMed®, CNKI, Web of Science, VIP and WANFANG) between January 2004 and July 2018 was conducted using the following search terms: ‘gastric cancer’, ‘peritoneal metastasis’, ‘hyperthermic intraperitoneal chemotherapy’. There were no search or language restrictions applied. The list of articles was reviewed independently by all three authors. The reference lists of all selected articles were manually reviewed to identify any additional relevant studies.

Study selection

Studies were eligible for inclusion if they met the following criteria: (i) randomized controlled trial (RCT); (ii) patients had advanced gastric cancer with peritoneal metastasis; (iii) the experimental group received HIPEC therapy, while the control group received systemic chemotherapy only; (iv) PCI score was 0–36; (v) reported outcomes included complete response (CR) rate, 3-year survival rate, serum carcinoembryonic antigen (CEA) level and adverse reactions. The exclusion criteria were as follows: (i) not an RCT; (ii) irrelevant or repetitive research; (iii) incomplete data access; (iv) peritoneal metastatic tumour cells were derived from a variety of primary tumours. After obtaining full reports of the candidate studies, two authors (Y.W.L. & Y.D.) reviewed each article independently. Differences between the two reviewers were resolved by discussion and consensus with the supervisor (B.A.C.).

Data abtraction and quality assessment

Each study was evaluated for quality based on the following characteristics: (i) secure method used for randomization; (ii) allocation concealment; (iii) patient and observer blinding; (iv) loss to follow-up. The quality of the studies was evaluated using Jadad quality scores and classified as low quality (score <4) and high quality (score ≥4).20

For each study, the following data were extracted: article title; first author’s last name; publication year; sample size; the age of the patients; total number for CR or 3-year survival; details of the chemotherapy regimens; curative effects; adverse events; surgery plans; quality evaluation.

Statistical analyses

A meta-analysis was performed via using RevMan software (version 5.0; Cochrane Collaboration, Oxford, UK). Enumeration data are presented as relative risk (RR) with a 95% confidence interval (CI). Measurement data are expressed as the mean difference (MD) with a 95% CI. The difference was considered statistically significant when P < 0.01.

Statistical heterogeneity was determined by the P-value and I2 value in the heterogeneity test results. If homogeneity was found (P > 0.10, I2 < 50%), a fixed-effect model was employed for the meta-analysis. The source of heterogeneity was investigated if P < 0.10 and I2 > 50% and a random-effect model was used in the absence of significant clinical heterogeneity. Subgroup or descriptive analyses were used in the presence of significant clinical heterogeneity.

Results

The initial database search identified 391 articles, of which 37 were RCTs (Figure 1). Full-text assessment resulted in 21 studies that enrolled a total of 1674 patients with advanced gastric cancer who met the inclusion criteria and were included in this analysis (Table 1).2141 Patients receiving HIPEC were classified as the experimental group (n = 840) and those receiving chemotherapy alone as the control group (n = 834). The studies were stratified according to their quality into a low-quality group (Jadad score <4) and a high-quality group (Jadad score ≥4). There were three RCTs in the low-quality group.26,28,38

Figure 1.

Figure 1.

Flowchart of the literature search undertaken in this meta-analysis of the effect of hyperthermic intraperitoneal chemotherapy (HIPEC) for patients with advanced gastric cancer and peritoneal metastasis. RCT, randomized controlled trial; NRCT, nonrandomized controlled trial.

Table 1.

Basic characteristics of the 21 randomized controlled trials that were included in a meta-analysis of the effect of hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with advanced gastric cancer and peritoneal metastasis.2141

Name Year Patient numberExperimental/control Age, yearsExperimental/control Karnofsky Performance Status score HIPEC regimens Systemic chemotherapy regimens
Zuo21 2004 46/36 53/52 (median) ≥ 70 DDP + 5-FU, 1750–2000 ml, 41–43°C, 60 min CF 100 mg, 5-FU 750 mg, THP 50-60 mg, DDP 20 mg
Cui22 2014 48/48 55/56 (mean) >60 DDP + 5-FU, 3000 ml, 41–43°C, 90 min PTX 135 mg/m2, DDP 20 mg/m2, FT207 0.8–1.0 g/day
Fan23 2017 15/15 71.9 ± 5.8/72.4 ± 5.6(mean ± SD) ND DDP + 5-FU, 4000 ml, 43°C, 60 min L-OHP 130 mg/m2, FT207 40–60 mg/time
Lu24 2016 28/20 52.4 ± 7.9/54.3 ± 6.6(mean ± SD) 50–80 DDP + 5-FU, 1500–2500ml, 41–44°C, 90 min L-OHP or TXT and FT207or CDHP and Oxo
Wang25 2017 48/48 69.3 ± 7.4/69.5 ± 7.2(mean ± SD) ND DDP + L-OHP, 3000 ml, 41–43°C, 90 min PTX 135 mg/m2, DDP 20 mg/m2, FT207 1.0 g/day
Xu26 2017 24/24 52.5 ± 1.4/51.5 ± 2.5(mean ± SD ND DDP + 5-FU, 3000 ml, 45°C, 60 min ND
Yuan27 2017 44/43 55.33 ± 4.75/56.86 ± 4.34(mean ± SD) >60 DDP, 2500 ml, 41–44°C, 60 min L-OHP 135mg/m2, CF 200 mg/m2, 5-FU 2600 mg/m2
Zhu28 2008 31/29 54/56 (median) >60 DDP, 1000 ml, 45°C, 60 min 5-FU 1000 mg/m2, L-OHP 85 mg/m2, CF 100 mg/m2
Chen29 2015 40/40 49/48 (mean) ND DDP, 41–43°C, 60 min 5-FU 500 mg, DDP 30–40 mg
Chen30 2016 30/30 ND >60 DDP + PTX, 1000 ml, 42.5–43.0°C, 60 min PTX 135 mg/m2, DDP 75 mg/m2
Hong31 2016 46/46 62.34 ± 7.37/62.43 ± 7.41(mean ± SD) >60 DDP, 3000 ml, 42–43°C, 60 min L-OHP 135mg/2, CF 200 mg/m2, 5-FU 2600 mg/m2
Hu32 2014 20/20 54.75 ± 13.63/ 58.50 ± 12.53(mean ± SD) >50 DDP, 3000 ml, 43°C, 60 min L-OHP 130 mg/m2, CAPE 2000 mg/m2
Jin33 2017 38/38 ND ND DDP + 5-FU + THP, 42–44°C, 90 min ND
Wang34 2016 50/50 62.17 ± 5.54/60.98 ± 5.02(mean ± SD) >60 5-FU, 2000 ml, 45°C, 60 min ND
Wang35 2018 32/32 46.4 ± 7.9/44.5 ± 7.3(mean ± SD) 40–70 TXT, 2000–2500 ml, 41–43°C, 60 min L-OHP 135mg/m2, 5-FU 2600 mg/m2, CF 100 mg/m2
Zhang36 2017 60/60 51.3 ± 8.7/52.6 ± 7.9(mean ± SD) >50 ND ND
Deng37 2009 44/41 52/53 (median) ND MMC + 5-FU, 3000 ml, 42–43°C, 60–90 min DDP 15 mg/m2
Liu38 2015 80/70 ND ND DDP + 5-FU, 3000 ml, 41–43°C, 90 min L-OHP 130 mg/m2, S-1 40 mg/m2
Yang39 2011 34/34 50/51 (median) >50 DDP + MMC, 6000 ml, 43.0 ± 0.5°C, 60–90 min ND
Zhang40 2013 40/40 ND ND MMC + 5-FU, 3000 ml, 45°C, 60–90min L-OHP 85mg/m2, CF 200 mg/m2, 5-FU 400 mg/m2, 5-FU 600 mg/m2
Zhang41 2007 92/120 57/57 (mean) ND DDP 100 mg, MMC 30 mg, 2000 ml, 43–45°C, 30 min DDP 10–15 mg/kg, MMC 0.1–0.15 mg/kg, ADM 0.5–1.0 mg/kg

DDP, cisplatin; 5-FU, 5-fluoruracil; CF, calcium folinate; THP, pirarubicin; PTX, paclitaxel; FT207, tegafur; L-OHP, oxaliplatin; TXT, docetaxel; CDHP, gimeracil; Oxo, oteracil; ND, not declared; CAPE, capecitabine; MMC, mitomycin; S-1, tegafur, gimeracil and oteracil potassium capsules; ADM, adriamycin.

The meta-analysis of the effect of treatment on 3-year survival included 13 RCTs (n = 1142 patients).2128,3741 The results of the heterogeneity analysis showed that P > 0.5 and I2 < 50%, so there was no heterogeneity between the two study groups. The meta-analysis using a fixed-effect model showed that the 3-year survival rate was significantly higher in the HIPEC group than in the control group (RR 1.61; 95% CI 1.43, 1.82; P < 0.00001) (Figure 2).

Figure 2.

Figure 2.

Meta-analysis of the effect of hyperthermic intraperitoneal chemotherapy (HIPEC) on 3-year survival in patients with advanced gastric cancer and peritoneal metastasis.2128,3741

The meta-analysis of the effect of treatment on CR rate included nine RCTs.24,2936 The meta-analysis using a fixed-effect model showed that the CR rate was significantly higher in the HIPEC group than in the control group (RR 2.35; 95% CI 1.67, 3.31; P < 0.00001) (Figure 3).

Figure 3.

Figure 3.

Meta-analysis of the effect of hyperthermic intraperitoneal chemotherapy (HIPEC) on clinical response in patients with advanced gastric cancer and peritoneal metastasis.24,2936

Adverse events included bone marrow suppression, anastomotic leak, bowel fistula, gastrointestinal reactions, kidney damage and liver disfunction. The meta-analysis of the effect of treatment on adverse events included nine RCTs.23,25,26,3540 The meta-analysis using a fixed-effect model showed that there was no difference in the rate of adverse events between the two groups (RR 1.00; 95% CI 0.87, 1.14; P = 0.95) (Figure 4).

Figure 4.

Figure 4.

Meta-analysis of the effect of hyperthermic intraperitoneal chemotherapy (HIPEC) on adverse events in patients with advanced gastric cancer and peritoneal metastasis.23,25,26,3540

The meta-analysis of the effect of treatment on CEA levels included three RCTs.25,32,35 There was an acceptable level of statistical heterogeneity between the three studies. The meta-analysis showed that the extent of the decrease in CEA level in the HIPEC group was better than the control group (MD –1.79; 95% CI −2.22, −1.35; P < 0.00001) (Figure 5).

Figure 5.

Figure 5.

Meta-analysis of the effect of hyperthermic intraperitoneal chemotherapy (HIPEC) on carcinoembryonic antigen levels in patients with advanced gastric cancer and peritoneal metastasis.25,32,35

Discussion

Although the clinical treatment of gastric cancer has been greatly improved, the prognosis of gastric cancer patients with peritoneal metastasis remains unsatisfactory.42 At present, peritoneal perfusion chemotherapy, combined with systemic chemotherapy or surgery, is used to improve the prognosis of patients.43 The combination of cytoreductive surgery and HIPEC is considered to be a promising comprehensive treatment strategy for gastric cancer peritoneal metastases and it has shown good initial results, but there is an urgent need to evaluate this strategy further in RCTs.44 The optimal therapeutic dose of HIPEC remains to be elucidated and many potential drug regimens for perfusion exist. Studies show that the perfusion of paclitaxel into the abdominal cavity is safe and effective, and it provides significant pharmacological advantages compared with intravenous chemotherapy.45,46 For example, intraperitoneal perfusion chemotherapy results in higher bioavailability of the drug.47 The bioavailability of intraperitoneal chemotherapy for docetaxel is two-times that of intravenous chemotherapy.48

There have been several RCTs that have investigated combined intraperitoneal chemotherapy and the results showed that the combined treatment group had a better curative effect than single operation or chemotherapy.49,50 This current meta-analysis demonstrated that the 3-year survival rate, the CR rate and the reductions in CEA level following treatment were all significantly more favourable in the HIPEC group compared with the control group; and there was no difference in the occurrence of adverse events between the two groups. However, it should be noted that there were only three studies that examined the CEA levels after treatment.25,32,35 In addition, the start and stop times of intervention for patients in each treatment centre were different. A large multi-centre RCT should be undertaken to improve the homogeneity of the data.

In conclusion, this current meta-analysis demonstrated that HIPEC resulted in a higher 3-year survival rate, a higher CR rate and greater reductions in CEA level following treatment than systemic chemotherapy alone in patients with advanced gastric cancer and peritoneal metastases. There was no difference in the occurrence of adverse events between the two treatment groups. As effective treatment of advanced gastric cancer is the key to improving the prognosis of the patient, choosing the most appropriate and effective treatment is likely to have a considerable impact on the patient’s long-term outcomes.

Declaration of conflicting interest

The authors declare that there are no conflicts of interest.

Funding

This work was supported by the National Natural Science Foundation of China (no. 81773284).

References

  • 1.Song Z, Wu Y, Yang J, et al. Progress in the treatment of advanced gastric cancer. Tumour Biol 2017; 39: 1010428317714626. [DOI] [PubMed] [Google Scholar]
  • 2.American Cancer Society®. Cancer Facts and Figures 2013. Atlanta: American Cancer Society®, http://www.cancer.org/research/cancer-facts-statistics/all-cancer-facts-figures/cancer-facts-figures-2013.html
  • 3.Jung KW, Won YJ, Kong HJ, et al. Prediction of cancer incidence and mortality in Korea, 2019. Cancer Res Treat 2019; 51: 431–437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Ferlay J, Shin HR, Bray F, et al. Estimates of worldwide burden of cancer in 2008: GLOBOCAN 2008. Int J Cancer 2010; 127: 2893–2917. [DOI] [PubMed] [Google Scholar]
  • 5.Ang TL, Fock KM. Clinical epidemiology of gastric cancer. Singapore Med J 2014; 55: 621–628. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Yuasa N, Nimura Y. Survival after surgical treatment of early gastric cancer, surgical techniques, and long-term survival. Langenbecks Arch Surg 2005; 390: 286–293. [DOI] [PubMed] [Google Scholar]
  • 7.den Hoed CM, Kuipers EJ. Gastric Cancer: How Can We Reduce the Incidence of this Disease? Curr Gastroenterol Rep 2016; 18: 34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Japanese Gastric Cancer Association. Japanese gastric cancer treatment guidelines 2014 (ver. 4). Gastric Cancer 2017; 20: 1–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Ceelen WP, Bracke ME. Peritoneal minimal residual disease in colorectal cancer: mechanisms, prevention, and treatment. Lancet Oncol 2009; 10; 72–79. [DOI] [PubMed] [Google Scholar]
  • 10.Saito H, Kono Y, Murakami Y, et al. Gross Appearance and Curability Are Predictive Factors of a Better Prognosis After Gastrectomy in Gastric Cancer Patients with Metastasis to the Adjacent Peritoneum of the Stomach. Yonago Acta Med 2017; 60: 174–178. [PMC free article] [PubMed] [Google Scholar]
  • 11.Seshadri RA, Glehen O. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in gastric cancer. World J Gastroenterol 2016; 22: 1114–1130. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Morano WF, Khalili M, Chi DS, et al. Clinical studies in CRS and HIPEC: Trials, tribulations, and future directions – A systematic review. J Surg Oncol 2018; 117: 245–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Shiu MH, Fortner JG. Intraperitoneal hyperthermic treatment of implanted peritoneal cancer in rats. Cancer Res 1980; 40: 4081–4084. [PubMed] [Google Scholar]
  • 14.Spratt JS, Adcock RA, Muskovin M, et al. Clinical delivery system for intraperitoneal hyperthermic chemotherapy. Cancer Res 1980; 40: 256–260. [PubMed] [Google Scholar]
  • 15.Jacquet P, Sugarbaker PH. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. Cancer Treat Res 1996; 82: 359–374. [DOI] [PubMed] [Google Scholar]
  • 16.Ng JL, Ong WS, Chia CS, et al. Prognostic Relevance of the Peritoneal Surface Disease Severity Score Compared to the Peritoneal Cancer Index for Colorectal Peritoneal Carcinomatosis Int J Surg Oncol 2016; 2016: 2495131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Garcia JR, Villasboas-Rosciolesi D, Soler M, et al. Peritoneal Cancer Index by (18)F-FDG PET/TC pre and post-hyperthermic intraperitoneal chemotherapy. Report of a case. Rev Esp Med Nucl Imagen Mol 2016; 35: 329–331. [DOI] [PubMed] [Google Scholar]
  • 18.Faron M, Macovei R, Goéré D, et al. Linear Relationship of Peritoneal Cancer Index and Survival in Patients with Peritoneal Metastases from Colorectal Cancer. Ann Surg Oncol 2016; 23: 114–119. [DOI] [PubMed] [Google Scholar]
  • 19.Chen J, Gong TT, Wu QJ. Parity and gastric cancer risk: a systematic review and dose-response meta-analysis of prospective cohort studies. Sci Rep 2016; 6: 18766. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Sun J, Song Y, Wang Z, et al. Benefits of hyperthermic intraperitoneal chemotherapy for patients with serosal invasion in gastric cancer: a meta-analysis of the randomized controlled trials. BMC Cancer 2012; 12: 526. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Zuo Y, Xu M, Shen D, et al. Postoperative intraperitioneal hyperthermic chemoperfusion combined with intravenous chemotherapy for 82 advanced gastric cancer patients. Zhonghua Zhong Liu Za Zhi 2004; 26: 247–249 [Article in Chinese, English abstract]. [PubMed] [Google Scholar]
  • 22.Cui HB, Ge HE, Bai XY, et al. Effect of neoadjuvant chemotherapy combined with hyperthermic intraperitoneal perfusion chemotherapy on advanced gastric cancer. Exp Ther Med 2014; 7: 1083–1088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Fan DS, Zhou G. Effect of intraperitoneal hyperthermic perfusion chemotherapy combined with radical resection on advanced gastric cancer. Jiangsu Med J 2017; 43: 1726–1727. [Google Scholar]
  • 24.Lu C, Li L, Luo Z, et al. Clinical efficacy of type-B ultrasound-guided intraperitoneal hyperthermic chemoperfusion combined with systemic chemotherapy in advanced gastric cancer patients with malignant ascites. Neoplasma 2016; 63: 299–303. [DOI] [PubMed] [Google Scholar]
  • 25.Wang QL. Clinical effect of adjuvant chemotherapy combined with hyperthermic intraperitoneal perfusion chemotherapy on advanced gastric cancer. Journal of Military Surgeon in Southwest China 2017; 19: 462–465. [Google Scholar]
  • 26.Xu Q. Clinical evaluation of efficacy and safety of postoperative hyperthermic intraperitoneal chemotherapy for advanced gastric cancer. Journal of Clinical Medical 2017; 4: 417–418. [Google Scholar]
  • 27.Yuan H, Liu ZZ, Zeng HG. Effect of intraperitoneal hyperthermic perfusion combined with systemic intravenous chemotherapy for advanced gastric cancer. Chin J Curr Adv Gen Surg 2017; 20: 822–824. [Google Scholar]
  • 28.Zhu M, Zou ZY, Qian XP. Clinical evaluation of hyperthermic intraperitoneal chemotherapy in postoperative patients with advanced gastric carcinoma. Journal of Clinical Medicine in Practice 2008; 12: 33–36. [Google Scholar]
  • 29.Chen XC. Clinical observation of cisplatin hyperthermia combined with intravenous chemotherapy in the treatment of advanced gastric cancer. Mod Diagn Treat 2015; 26: 5648–5649. [Google Scholar]
  • 30.Chen M, Jia L, Su Z. Paclitaxel combined with cisplatin intraperitoneal hyperthermic perfusion chemotherapy for malignant ascites of gastric cancer clinical research. J Mod Med Health 2016; 32: 3453–3455. [Google Scholar]
  • 31.Hong YN, Pang HX, Zhu JF, et al. Clinical effect of hyperthermic intraperitoneal chemotherapy combined with intravenous chemotherapy in elderly gastric carcinoma patients complicated with malignant ascites. Jilin Med 2016; 37: 2645–2647. [Google Scholar]
  • 32.Hu M, Zhan GF, Lei J, et al. Clinical study of hyperthermic intraperitoneal chemotherapy on unresectable gastric carcinoma with malignant ascites. J Dig Oncol (Electronic Version) 2014; 6: 19–22. [Google Scholar]
  • 33.Jin CY, Jing LL. Clinical efficacy of intraperitoneal hyperthermic perfusion chemotherapy combined with intravenous chemotherapy in the treatment of malignant ascites of gastric cancer. Guide of China Medicine 2017; 15: 160. [Google Scholar]
  • 34.Wang BL. Exploring clinical effect of gastric cancer intraperitoneal hyperthermic perfusion chemotherapy combined with intravenous chemotherapy treating malignant ascites. World Latest Medicine Information (Electronic Version) 2016; 16: 8. [Google Scholar]
  • 35.Wang WF, Jiang ZH, Dong XN. Clinical study of intraperitoneal hyperthermic perfusion combined with intravenous chemotherapy for malignant ascites in patients with gastric cancer. Journal of Navy Medicine 2018; 39: 188–190. [Google Scholar]
  • 36.Zhang TP. clinical efficacy and safety of intraperitoneal hyperthermic infusion Chemotherapy combined with intravenous Chemotherapy in the treatment of malignant ascites. Mod Diagn Treat 2017; 28: 2164–2166. [Google Scholar]
  • 37.Deng HJ, Wei ZG, Zhen L, et al. Clinical application of perioperative continuous hyperthermic peritoneal perfusion chemotherapy for gastric cancer. Nan Fang Yi Ke Da Xue Xue Bao 2009; 29: 295–297 [Article in Chinese, English abstract]. [PubMed] [Google Scholar]
  • 38.Liu T. Clinical observation of perioperative hyperthermic intraperitoneal chemotherapy combined with adjuvant chemotherapy for advanced gastric cancer. China Continuing Medical Education 2015; 7: 82–83. [Google Scholar]
  • 39.Yang XJ, Huang CQ, Suo T, et al. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy improves survival of patients with peritoneal carcinomatosis from gastric cancer: final results of a phase III randomized clinical trial. Ann Surg Oncol 2011; 18: 1575–1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Zhang GS. Effect of perioperative hyperthermic peritoneal perfusion on advanced gastric cancer. Chin J Curr Adv Gen Surg 2013; 16: 896–898. [Google Scholar]
  • 41.Zhang GY, Chen XC, Pan K, et al. Application of hyperthermic intraoperative intraperitoneal chemotherapy in patients with gastric cancer. Chin J Gastrointest Surg 2007; 10: 362–364. [PubMed] [Google Scholar]
  • 42.Hartgrink HH, Jansen EP, van Grieken NC, et al. Gastric cancer. Lancet 2009; 374: 477–490. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Glockzin G, Zeman F, Croner RS, et al. Perioperative Systemic Chemotherapy, Cytoreductive Surgery, and Hyperthermic Intraperitoneal Chemotherapy in Patients With Colorectal Peritoneal Metastasis: Results of the Prospective Multicenter Phase 2 COMBATAC Trial. Clin Colorectal Cancer 2018; 17: 285–296. [DOI] [PubMed] [Google Scholar]
  • 44.Ji ZH, Peng KW, Yu Y, et al. Current status and future prospects of clinical trials on CRS + HIPEC for gastric cancer peritoneal metastases. Int J Hyperthermia 2017; 33: 562–570. [DOI] [PubMed] [Google Scholar]
  • 45.Fugazzola P, Coccolini F, Montori G, et al. Overall and disease-free survival in patients treated with CRS + HIPEC with cisplatin and paclitaxel for gastric cancer with peritoneal carcinomatosis. J Gastrointest Oncol 2017; 8: 572–582. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ohguri T, Imada H, Narisada H, et al. Systemic chemotherapy using paclitaxel and carboplatin plus regional hyperthermia and hyperbaric oxygen treatment for non-small cell lung cancer with multiple pulmonary metastases: preliminary results. Int J Hyperthermia 2009; 25: 160–167. [DOI] [PubMed] [Google Scholar]
  • 47.Bouquet W, Ceelen W, Adriaens E, et al. In vivo toxicity and bioavailability of Taxol and a paclitaxel/beta-cyclodextrin formulation in a rat model during HIPEC. Ann Surg Oncol 2010; 17: 2510–2517. [DOI] [PubMed] [Google Scholar]
  • 48.Morgan RJ, Jr, Doroshow JH, Synold T, et al. Phase I trial of intraperitoneal docetaxel in the treatment of advanced malignancies primarily confined to the peritoneal cavity: dose-limiting toxicity and pharmacokinetics. Clin Cancer Res 2003; 9: 5896–5901. [PubMed] [Google Scholar]
  • 49.Verwaal VJ, van Ruth S, de Bree E, et al. Randomized trial of cytoreduction and hyperthermic intraperitoneal chemotherapy versus systemic chemotherapy and palliative surgery in patients with peritoneal carcinomatosis of colorectal cancer. J Clin Oncol 2003; 21: 3737–3743. [DOI] [PubMed] [Google Scholar]
  • 50.Rudloff U, Langan RC, Mullinax JE, et al. Impact of maximal cytoreductive surgery plus regional heated intraperitoneal chemotherapy (HIPEC) on outcome of patients with peritoneal carcinomatosis of gastric origin: results of the GYMSSA trial. J Surg Oncol 2014; 110:275–284. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from The Journal of International Medical Research are provided here courtesy of SAGE Publications

RESOURCES