Skip to main content
Erschienen in: BMC Cancer 1/2020

Open Access 01.12.2020 | Research article

Neoadjuvant systemic and hyperthermic intraperitoneal chemotherapy combined with cytoreductive surgery for gastric cancer patients with limited peritoneal metastasis: a prospective cohort study

verfasst von: Pengfei Yu, Zeyao Ye, Gaiguo Dai, Yanqiang Zhang, Ling Huang, Yian Du, Xiangdong Cheng

Erschienen in: BMC Cancer | Ausgabe 1/2020

Abstract

Background

There is no currently available treatment for peritoneal metastasis of gastric cancer. This phase II study aimed to evaluate the efficacy and safety of neoadjuvant systemic chemotherapy and hyperthermic intraperitoneal chemotherapy (HIPEC) combined with cytoreductive surgery (CRS) for the treatment of these patients.

Methods

Neoadjuvant chemotherapy comprised two cycles of HIPEC and four cycles of S-1 plus paclitaxel. HIPEC was administered intraperitoneally with paclitaxel (75 mg/m2). For systemic chemotherapy, paclitaxel was administered intravenously(150 mg/m2) on day 1, and S-1 was administered orally(80 mg/m2/day)on days 1–14 of a 3-week cycle. Another two cycles of HIPEC and four cycles of S-1 plus paclitaxel were administered after second diagnostic staging laparoscopy or CRS. The primary endpoints were treatment efficiency and safety; the secondary endpoint was 3-year overall survival (OS).

Results

A total of 40 patients were enrolled and 38 patients have been analyzed. Of these, 18 (47.4%) patients received neoadjuvant systemic chemotherapy, HIPEC and CRS (conversion therapy group), while 20 patients received only chemotherapy and HIPEC (palliative chemotherapy group). Median OS was markedly improved in the conversion therapy group (21.1 months, 95% confidence interval [CI] 16.7–25.6 months) in comparison with the palliative chemotherapy group(10.8 months, 95%CI 7.3–14.2 months, p = 0.002). After neoadjuvant systemic chemotherapy and HIPEC, a second laparoscopic exploration was performed, and the prognosis of patients with low peritoneal cancer index (PCI) (PCI < 6) was significantly better than that of patients with high PCI (PCI ≥ 6)(20.1 vs.11.3 months, p = 0.006).

Conclusion

Neoadjuvant systemic chemotherapy and HIPEC combined with CRS is safe and feasible, and could potentially improve the prognosis of gastric cancer patients with limited peritoneal metastasis. However, further clinical trials are still warranted.

Trial registration

This study has been registered with ClinicalTrials.​gov as NCT02549911. Trial registration date: 15/09/2015.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
HIPEC
Hyperthermic intraperitoneal chemotherapy
CRS
Cytoreductive surgery
OS
Overall survival
PCI
Peritoneal cancer index
GC
Gastric cancer
DSL
Diagnostic staging laparoscopy
ECOG
Eastern Cooperative Oncology Group
ALT
Alanine transaminase
AST
Aspartate transaminase
ULNs
Upper limits of the normal ranges
BSA
Body surface area
CCR
Completeness of cytoreduction
TRG
Tumor regression grade
CDSC
Clavien-Dindo severity classification
MST
Median survival time

Background

Gastric cancer(GC) is the fifth most common cancer and the third leading cause of cancer-related deaths in the world [1]. Recurrence and metastasis are the main factors affecting the prognosis of GC patients, and the peritoneum represents the most common site of metastasis and postoperative recurrence [2]. Nearly 20% of patients with GC are diagnosed with peritoneal metastasis preoperatively or intraoperatively, and more than 50% of patients with stage T3 and T4 disease have peritoneal metastasis after radical resection [3]. Once peritoneal metastasis develops, symptoms such as refractory peritoneal effusion, intestinal obstruction and cachexia may occur, which are the major causes of death in patients with advanced GC [4].
Palliative chemotherapy is the main treatment strategy for peritoneal metastasis of gastric cancer, however, the response rate is low with a median survival time less than 6 months [5]. Recent retrospective studies have reported that the primary tumor and peritoneal metastasis can be well controlled in some patients by comprehensive treatment including systemic and local chemotherapy. Thus, cytoreductive surgery (CRS) of the primary tumor and peritoneal metastasis can be achieved and survival time is significantly improved [6, 7].
Hyperthermic intraperitoneal chemotherapy (HIPEC) is a highly concentrated, heated chemotherapy treatment delivered directly to the abdomen [8]. It can not only maintain the high concentration of drugs in the abdominal cavity, but also improve the anti-tumor efficacy of chemotherapy drugs through the thermo-thermal effect [9]. In recent years, HIPEC has been gradually applied in the treatment of peritoneal metastasis of gastrointestinal cancers, and achieved a good therapeutic effect [10, 11].
Therefore, this study aimed to assess the efficacy and safety of neoadjuvant systemic chemotherapy and HIPEC combined with cytoreductive surgery for the treatment of advanced GC with limited peritoneal metastasis, so as to develop a reasonable treatment strategy for these patients.

Methods

Study design

The current prospective, single arm, open phase II clinical study was performed in Zhejiang Cancer Hospital, Hangzhou, China, following the Helsinki Declaration as well as the principles of good clinical practice. The study was approved by the ethics review committee of Zhejiang Cancer Hospital(approval number:IRB-2015-170). A written informed consent was obtained from each study participant. The trial was registered with ClinicalTrials.gov (#NCT02549911).
Inclusion criteria were as follows: (1)advanced GC with suspected peritoneal metastasis, including ascites, ovarian metastasis, or omental metastasis; (2)positive peritoneal cytology or peritoneal dissemination with a PCI ≤ 20 confirmed by diagnostic staging laparoscopy (DSL); (3)absence of other distant metastases;(4) no prior treatment (radiotherapy, chemotherapy, targeted therapy, or immunotherapy);(5) 18–75 years of age and Eastern Cooperative Oncology Group (ECOG) performance status of 0–2;(6)normal organ functions, with serum alanine transaminase (ALT) and aspartate transaminase (AST) levels less than two times the upper limits of the normal ranges (ULNs), serum total bilirubin < 1.5 × ULN, serum creatinine < 1.25 × ULN, platelet counts > 75,000/L, absolute granulocyte counts > 1500/L, hemoglobin levels > 90 g/L, and normal electrocardiogram.
Exclusion criteria were as follows: severe intraperitoneal adhesions; presence of a synchronous or metachronous malignancy; pregnant or lactating patients; severe drug hypersensitivity; mental abnormalities; serious respiratory disease; uncontrolled hypertension, diabetes, and cardiovascular disease.

Treatment

HIPEC

HIPEC was performed twice within 72 h after each diagnostic staging laparoscopy(DSL) or CRS using four drainage tubes that were placed in the abdominal cavity. First HIPEC was usually performed within 24 h of DSL or CRS, followed by a second HIPEC at an interval of 24–48 h. Approximately 3 L of heated 0.9% saline supplemented with paclitaxel (75 mg/m2) was circulated for 60 min. The heated perfusion solution was infused into the peritoneal cavity at a rate of 400–500 ml/min, which was introduced by an automatic hyperthermia perfusion device (RHL-2000B, Madain Medical Devices Co., Ltd., Jilin, China). The perfusate temperatures in the inflow and outflow tubes were obtained in real time, maintaining the perfusion solution in the peritoneum at43.0 ± 0.5 °C.After HIPEC completion, the abdominal fluid was removed as much as possible [12].

Systemic chemotherapy

Preoperative systemic chemotherapy (four cycles of paclitaxel combined with S-1) was administered three weeks after the initial HIPEC, while postoperative systemic chemotherapy (four cycles of paclitaxel combined with S-1) was administered four to six weeks after the second DSL or CRS: S-1 (80, 100 and 120 mg/day for body surface area (BSA) below 1.25 m2, between 1.25 and 1.5 m2 and above 1.5m2, respectively) was administered orally, twice a day for two consecutive weeks followed by a one-week rest, and paclitaxel (150 mg/m2) was administered intravenously on day 1.Patients with Her-2 expression received targeted therapy (intravenous trastuzumab at 8 mg/kg on day 1 of the initial cycle, and then 6 mg/kg every three weeks).

Surgical treatment

Four to six weeks after the last preoperative chemotherapy, the patient received a second laparoscopic exploration, if the tumor was well controlled, CRS was performed, including resection of the primary tumor with acceptable margins, lymphadenectomy and peritoneotomy where peritoneal surfaces were involved by tumor [13]. CRS aimed to achieve complete macroscopic cytoreduction; after resection a score estimating the completeness of cytoreduction or CCR was used and defined as: CCR-0 (no residual peritoneal tumor nodules), CCR-1 (residual tumor nodules < 2.5 mm), CCR-2 (residual tumor nodules of 2.5 mm-2.5 cm), and CCR-3 (residual tumor nodules > 2.5 cm or a confluence of unresectable tumor nodules at any site [14]. The treatment schedule is shown in Fig. 1.

Evaluation

The primary endpoints were treatment efficacy and safety. The secondary endpoint was 2-year overall survival (OS).
The characteristics of the tumor were recorded according to the Japanese gastric cancer classification (3rd edition) [15] and Union for International Cancer Control (UICC) TNM classification (7th edition) [16].Peritoneal metastasis was assessed according to the peritoneal carcinomatosis index (PCI) proposed by Jacquet and Sugarbaker [13]. Histological tumor regression grade (TRG) was assessed as previously described [17]: G1a, complete response; G1b,< 10% residual tumor/tumor bed; G2, 10–50% residual tumor/tumor bed; G3,> 50% residual tumor/tumor bed. A postoperative complication was prospectively defined as any deviation from a predetermined postoperative course within 30 days of surgery and categorized following the Clavien-Dindo severity classification (CDSC) [18]. Adverse events were evaluated using the Common Terminology Criteria for Adverse Events, Version 4.0 (CTCAE v4.0).

Follow-up

Clinical follow-up was carried out by calling the patients and by accessing outpatient records. It was performed once every 3 months for the first 2 years, followed by once every 6 months for 2–5 years. OS was defined as the time from diagnosis of peritoneal metastasis to last follow up or death. The cutoff date for OS was December 2019.

Sample size

According to some previous studies [19, 20], the median survival time (MST) of GC patients with peritoneal metastasis was 9–10 months. After neoadjuvant systemic chemotherapy and HIPEC combined with CRS, the MST is estimated to be 18 months. Assuming a two-sided α of 0.05 and 90% statistical power, with an estimated drop-out rate of 20%, the required sample size was estimated to be 40 patients.

Statistical analysis

Statistical Package for Social Sciences (SPSS ver.19.0 SPSS Inc., Chicago, IL, USA) was employed for data analysis. Student’s t test and the chi-square test were performed for comparing continuous and discrete variables, respectively. OS was determined by the Kaplan-Meier method, and compared by the log-rank test. P < 0.05 indicated statistical significance.

Results

Patient characteristics

From September 2015 to October 2019, 40 patients were enrolled in this study. Among these, two patients declined chemotherapy and HIPEC. Therefore, a total of 38 patients were finally assessed. There were 18 males and 20 females with the median age of 52 years (range 28–72 years). There were one case with positive peritoneal lavage cytology (CY1), and 37 cases with peritoneal metastasis, including 19 cases with CY1 and 10 cases had ovarian metastasis. After the neoadjuvant treatment, PCI levels in 34 patients (89.5%) were decreased, while increasing in 4 patients. The average PCI score was significantly reduced to 5.8 ± 4.9 from 9.0 ± 5.3 in the whole cohort (p = 0.017).A total of 18 (47.4%) patients received neoadjuvant systemic chemotherapy, HIPEC and CRS (conversion therapy group), while 20 patients underwent only chemotherapy and HIPEC (palliative chemotherapy group). Tumor location, differentiation, initial PCI scores, and the levels of tumor markers were comparable in both groups (Table 1).
Table 1
Characteristics of the conversion therapy group and the palliative chemotherapy group
Variable
conversion therapy (n = 18)
palliative chemotherapy (n = 20)
p-Value
Gender
  
0.321
 Male
7
11
 Female
11
9
Median age
49.8(28 ~ 72)
53.5(35 ~ 73)
0.185
Tumor location
 Upper
1
4
 
 Middle
11
10
 
 Lower
6
6
0.417
Differentiation
  
0.758
 Well and moderately
9
11
 Poorly
9
9
Initial DSL
  
0.321
 PCI < 10
11
9
 PCI ≥ 10
7
11
Second DSL
  
<0.001
 PCI < 6
18
2
 PCI ≥ 6
0
18
Ascites
  
0.516
 Positive
8
11
 Negative
10
9
Serum CEA (ng/ml)
  
0.544
 Normal
15
18
 >5
3
2
Serum CA19–9 (U/ml)
  
0.914
 Normal
12
13
 >39
6
7
Serum CA125 (U/ml)
  
0.703
 Normal
7
9
 >35
11
11
Serum CA724 (U/ml)
 Normal
12
15
0.532
 >7
6
5
DSL diagnostic staging laparoscopy, PCI peritoneal carcinomatosis index
CEA carcinoembryonic antigen

Surgical results

Among the 18 patients who underwent conversion therapy, the second laparoscopic exploration found a PCI of 0 in 6 patients, PCI score of 1–5 in 12 patients (median PCI = 1.5) and CY1 in 3 patients. CCR-0 and CCR-1 were achieved in 15 and 3 patients, respectively. Nine patients were administered total gastrectomy, while 10 patients received combined vascular resections. Of the 20 patients who underwent palliative chemotherapy, the second laparoscopic exploration found PCI score of 5–9 in 13 patients, 10–20 in 7 patients (median PCI = 8.8) and CY1 in 14 patients. Two patients received gastrointestinal bypass, 3 patients had oophorectomy and 7 patients received biopsy of peritoneal metastasis.
A total of 6 (15.8%) patients experienced Clavien-Dindo grade II-III complications, including pneumonia (n = 3), anastomotic leakage (n = 1) and abdominal abscess (n = 2). All of these complications were successfully treated with a conservative procedure.
Histopathological assessment of the resected primary tumors showed that responses to chemotherapy were G1b in 6 patients, G2 in 11 patients and G3 in 1 patient. No patient showed a G1a response (Table 2).
Table 2
Surgical data and pathological factors of the conversion therapy group
Variable
n = 18
Operating procedure
 Total gastrectomy
9(50.0%)
 Distal gastrectomy
9(50.0%)
LN dissection
 D2
14(77.8%)
 D2+
4(22.2%)
Combined resection
 Spleen
3(16.7%)
 Ovary
7(38.9%)
 Pancreas
1(5.6%)
Operation time (min)
223(157–329)
Blood loss (ml)
150(50–800)
Postoperative complications
6(15.8%)
 pneumonia
3
 anastomotic leakage
1
 abdominal abscess
2
Residual tumor
 CCR-0
15(83.3%)
 CCR-1
3(16.7%)
yp T grade
 T1
3(16.7%)
 T2
5(27.8%)
 T3
2(11.1%)
 T4a
7(38.9%)
 T4b
1(5.6%)
yp N grade
 N0
5(27.8%)
 N1
6(33.3)
 N2
5(27.8%)
 N3
2(11.1%)
Histological response
 G1a
0
 G1b
6(33.3%)
 G2
11(61.1%)
 G3
1(5.6%)
LN Lymph node, CCR completeness of cytoreduction

Survival outcomes

The median duration of follow-up in the whole cohort was 19.3 months (range, 5–37 months). The median survival time (MST) of the 38 patients was 15.1 months (95% confidence interval [CI] 11.9–18.4 months); the 1- and 2-year survival rates of the patients were 63.2 and 47.4%, respectively.
The median OS in the conversion therapy group was 21.1 months (95%CI 16.7–25.6 months), which was significantly better than that of the palliative chemotherapy group (10.8 months, 95%CI 7.3–14.2 months; p = 0.002) (Fig. 2). In the initial diagnostic laparoscopic exploration, patients with low PCI (PCI < 10) had better OS than those with high PCI (PCI ≥ 10), however, the difference was not statistically significant (p>0.05). After neoadjuvant systemic chemotherapy and HIPEC treatment, a second laparoscopic exploration was performed, and the prognosis of patients with low PCI (PCI < 6) was significantly better than that of patients with high PCI (PCI ≥ 6)(20.1 vs.11.3 months; p = 0.006) (Fig. 3 and Table 3).
Table 3
Univariate analysis of predictors for overall survival. (n = 38)
Variables
Overall Survival Median
p-Value
Age
  ≤ 50
16.7
0.478
 >50
13.8
Gender
 Male
12.1
0.120
 Female
17.5
Treatment
 Conversion therapy
21.1
0.002
 Palliative chemotherapy
10.8
Tumor location
 Upper
11.3
0.343
 Middle and lower
15.7
Differentiation
 Poorly
11.6
0.087
 Moderately and well
18.1
Ascites
 Positive
15.3
0.774
 Negative
14.5
Initial DSL
 PCI<10
16.6
0.391
 PCI ≥ 10
13.2
Second DSL
 PCI<6
20.1
0.006
 PCI ≥ 6
11.3
Serum CEA (ng/ml)
 Normal
15.6
0.373
 >5
12.0
Serum CA19–9 (U/ml)
 Normal
15.9
0.359
 >39
12.1
Serum CA125 (U/ml)
 Normal
10.4
0.076
 >35
17.6
DSL diagnostic staging laparoscopy, PCI peritoneal carcinomatosis index
CEA carcinoembryonic antigen
In the conversion therapy group, the 2-year OS rate in chemotherapy-treated patients with TRG of 1 was 100%, whereas that observed for counterparts with TRG of 2–3 was 50%; however, statistical significance was not achieved (p = 0.211).

Treatment toxicity

Chemotherapy combined with HIPEC was well-tolerated in most patients. A total of 10.5% (4/38) of all patients with Her-2 expression received trastuzumab treatment. Grade 3 or 4 adverse events were found in 11 of the 38 patients (28.9%) in the postoperative stage. Among these, the most common hematological toxic effects were leucopenia/neutropenia (15.8%) and thrombocytopenia (5.3%) while the most frequent non-hematological toxic effects were elevation of serum AST levels (7.9%) (Table 4).All the patients suffered from varying degrees of abdominal pain and distension from HIPEC, but these symptoms were adequately controlled by medication.
Table 4
Grade 3/4 toxic effects in the two groups (n = 38)
Toxic effects
conversion therapy (n = 18)
palliative chemotherapy (n = 20)
Hematological
 Leucopenia/neutropenia
3(16.7%)
3(15.0%)
 Thrombocytopenia
1(5.6%)
1(5.0%)
Non-hematological
 Transaminase elevation
2(11.1%)
1(5.0%)

Discussion

Peritoneal metastasis has poor prognosis, and is a major cause of death in patients with advanced gastric cancer. In the past, systemic chemotherapy was the main treatment option for these patients [21]. However, the benefit was limited as systemic chemotherapy can be affected by the plasma-peritoneal barrier. HIPEC can maintain a high concentration of drugs in the abdominal cavity and enhance the cytotoxicity of chemotherapeutic drugs against tumor cells under the hyperthermia effect [11]. It has been reported that CRS combined with chemotherapy and HIPEC has the potential to control peritoneal metastasis from GC [6, 7].
There is also some controversy over whether chemotherapy and HIPEC should be performed before CRS. Several clinical studies have demonstrated the clinical efficacy of CRS combined with HIPEC for peritoneal metastasis from GC, although it is associated with high mortality and morbidity [22, 23]. Nevertheless, the clinical significance of CRS plus HIPEC for GC is currently under investigation in the neoadjuvant setting, and this treatment model may be safer and more likely to confer a survival benefit.
Therefore, a prospective cohort study of neoadjuvant systemic and hyperthermic intraperitoneal chemotherapy combined with cytoreductive surgery was performed to determine the safety and potential survival benefits of conversion therapy for GC patients with limited peritoneal metastasis. This study found that despite the complexity of CRS following neoadjuvant chemotherapy and HIPEC, the incidence of complications was low, and there was no treatment related mortality. After neoadjuvant therapy, the primary tumor and peritoneal metastasis were well controlled (Fig. 4); thus, the scope of surgical resection and the rate of combined visceral resection were reduced. These results are consistent with previous reports [24, 25]. Therefore, CRS after neoadjuvant chemotherapy and HIPEC is safe and feasible for GC with peritoneal metastasis.
A retrospective study assessed 64 GC patients with peritoneal carcinomatosis who received HIPEC combined with systemic chemotherapy; 32 (50%) of these patients who received radical resection showed a median survival of 678 days, which was markedly prolonged compared with that of patients without operation [26]. This prospective study found that after 2 cycles of HIPEC and 4 cycles of neoadjuvant systemic chemotherapy, 18 patients (47.4%) received CRS (conversion therapy). Median OS in the conversion therapy group was 21.1 months, indicating a significant improvement compared with the palliative chemotherapy group. Therefore, after neoadjuvant chemotherapy and HIPEC, peritoneal metastasis could be well-controlled, with survival improved in some patients received complete resection of both primary tumor and peritoneal metastasis.
The evaluation of peritoneal metastasis before conversion therapy is important, and diagnostic staging laparoscopy with PCI assessment is a preferable procedure [27]. It has been proved that PCI is a predictive factor for complete cytoreduction, with the best results obtained when the PCI score is limited [24].In a Chinese study, CRS and HIPEC were administered to GC patients with peritoneal carcinomatosis and/or malignant ascites, and survival analysis indicated that patients with PCI < 20 had significantly better survival than those with PCI > 20 [28]. Another study revealed that patients with incomplete cytoreduction or PCI > 15 were not benefitting from CRS and HIPEC due to a limited survival benefit [29]. The present prospective study demonstrated that after neoadjuvant systemic chemotherapy and HIPEC, PCI scores had been improved in most patients, and the prognosis of patients with low PCI (PCI < 6) was significantly better than that of patients with high PCI (PCI ≥ 6)[20.1 vs.11.3 months, p = 0.006].Therefore, after neoadjuvant chemotherapy and HIPEC, only patients with low PCI followed by complete cytoreduction could achieve long-term survival.
The limitations of this study include a relatively small sample size, a single-center design, and short follow up. Therefore, further multi-center large sample studies are warranted to clarify the survival benefits of conversion therapy for GC patients with limited peritoneal metastasis.
Overall, neoadjuvant chemotherapy and HIPEC, combined with cytoreductive surgery, could potentially improve the survival time of GC patients with limited peritoneal metastasis. This procedure is safe and feasible, with only minor complications. However, prospective randomized clinical studies with a large sample size are warranted to validate the results of this study.

Acknowledgements

We thank all study participants and research staff who participated in this work.
The study protocol was approved by the institutional ethics review board of Zhejiang Cancer Hospital. Written informed consent was obtained from all study participants.
Not applicable.

Competing interests

The authors declare that they have no competing interests.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRef Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, et al. Cancer incidence and mortality worldwide: sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 2015;136:E359–86.CrossRef
2.
Zurück zum Zitat Ji ZH, Peng KW, Yu Y, Li XB, Yonemura Y, Liu Y, et al. Current status and future prospects of clinical trials on CRS + HIPEC for gastric cancer peritoneal metastases. Int J Hyperth. 2017;33(5):562–70.CrossRef Ji ZH, Peng KW, Yu Y, Li XB, Yonemura Y, Liu Y, et al. Current status and future prospects of clinical trials on CRS + HIPEC for gastric cancer peritoneal metastases. Int J Hyperth. 2017;33(5):562–70.CrossRef
3.
Zurück zum Zitat Yonemura Y, Bandou E, Kawamura T, Endou Y, Sasaki T. Quantitative prognostic indicators of peritoneal dissemination of gastric cancer. Eur J Surg Oncol. 2006;32(6):602–6.CrossRef Yonemura Y, Bandou E, Kawamura T, Endou Y, Sasaki T. Quantitative prognostic indicators of peritoneal dissemination of gastric cancer. Eur J Surg Oncol. 2006;32(6):602–6.CrossRef
4.
Zurück zum Zitat Geng X, Liu H, Lin T, Hu Y, Chen H, Zhao L, et al. Survival benefit of gastrectomy for gastric cancer with peritoneal carcinomatosis: a propensity score-matched analysis. Cancer Med. 2016;5(10):2781–91.CrossRef Geng X, Liu H, Lin T, Hu Y, Chen H, Zhao L, et al. Survival benefit of gastrectomy for gastric cancer with peritoneal carcinomatosis: a propensity score-matched analysis. Cancer Med. 2016;5(10):2781–91.CrossRef
5.
Zurück zum Zitat 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(3):622–8.CrossRef 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(3):622–8.CrossRef
6.
Zurück zum Zitat Mielko J, Rawicz-Pruszyński K, Skórzewska M, Ciseł B, Pikuła A, Kwietniewska M, et al. Conversion Surgery with HIPEC for Peritoneal Oligometastatic Gastric Cancer. Cancers (Basel). 2019; 11(11):pii: E1715. Mielko J, Rawicz-Pruszyński K, Skórzewska M, Ciseł B, Pikuła A, Kwietniewska M, et al. Conversion Surgery with HIPEC for Peritoneal Oligometastatic Gastric Cancer. Cancers (Basel). 2019; 11(11):pii: E1715.
7.
Zurück zum Zitat Rau B, Brandl A, Piso P, Pelz J, Busch P, Demtröder C, et al. Peritoneum surface oncology group and members of the StuDoQ|peritoneum registry of the German Society for General and Visceral Surgery (DGAV). Peritoneal metastasis in gastric cancer: results from the German database. Gastric Cancer. 2020;23(1):11–22.CrossRef Rau B, Brandl A, Piso P, Pelz J, Busch P, Demtröder C, et al. Peritoneum surface oncology group and members of the StuDoQ|peritoneum registry of the German Society for General and Visceral Surgery (DGAV). Peritoneal metastasis in gastric cancer: results from the German database. Gastric Cancer. 2020;23(1):11–22.CrossRef
8.
Zurück zum Zitat Dodson RM, Kuncewitch M, Votanopoulos KI, Shen P, Levine EA. Techniques for Cytoreductive surgery with Hyperthermic Intraperitoneal chemotherapy. Ann Surg Oncol. 2018;25(8):2152–8.CrossRef Dodson RM, Kuncewitch M, Votanopoulos KI, Shen P, Levine EA. Techniques for Cytoreductive surgery with Hyperthermic Intraperitoneal chemotherapy. Ann Surg Oncol. 2018;25(8):2152–8.CrossRef
9.
Zurück zum Zitat Cai Z, Cai Z, He T, Zhao Z, Yin Y, Shen C, et al. Comparative effectiveness of hyperthermic intraperitoneal chemotherapy for gastric cancer: a systematic review and network meta-analysis protocol. Medicine (Baltimore). 2018;97(33):e11949.CrossRef Cai Z, Cai Z, He T, Zhao Z, Yin Y, Shen C, et al. Comparative effectiveness of hyperthermic intraperitoneal chemotherapy for gastric cancer: a systematic review and network meta-analysis protocol. Medicine (Baltimore). 2018;97(33):e11949.CrossRef
10.
Zurück zum Zitat Eng OS, Turaga KK. Cytoreduction and hyperthermic intraperitoneal chemotherapy in metastatic colorectal cancer. J Surg Oncol. 2019;119(5):613–5.CrossRef Eng OS, Turaga KK. Cytoreduction and hyperthermic intraperitoneal chemotherapy in metastatic colorectal cancer. J Surg Oncol. 2019;119(5):613–5.CrossRef
11.
Zurück zum Zitat Seshadri RA, Glehen O. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in gastric cancer. World J Gastroenterol. 2016;22(3):1114–30.CrossRef Seshadri RA, Glehen O. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in gastric cancer. World J Gastroenterol. 2016;22(3):1114–30.CrossRef
12.
Zurück zum Zitat Wu XJ, Yuan P, Li ZY, Bu ZD, Zhang LH, Wu AW, et al. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy improves the survival of gastric cancer patients with ovarian metastasis and peritoneal dissemination. Tumour Biol. 2013;34(1):463–9.CrossRef Wu XJ, Yuan P, Li ZY, Bu ZD, Zhang LH, Wu AW, et al. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy improves the survival of gastric cancer patients with ovarian metastasis and peritoneal dissemination. Tumour Biol. 2013;34(1):463–9.CrossRef
13.
Zurück zum Zitat Jacquet P, Sugarbaker PH. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. Cancer Treat Res. 1996;82:359–74.CrossRef Jacquet P, Sugarbaker PH. Clinical research methodologies in diagnosis and staging of patients with peritoneal carcinomatosis. Cancer Treat Res. 1996;82:359–74.CrossRef
14.
Zurück zum Zitat Glehen O, Osinsky D, Cotte E, Kwiatkowski F, Freyer G, Isaac S, et al. Intraperitoneal chemohyperthermia using a closed abdominal procedure and cytoreduction surgery for the treatment of peritoneal carcinomatosis: morbidity and mortality analysis of 216 consecutive procedures. Ann Surg Oncol. 2003;10:863–9.CrossRef Glehen O, Osinsky D, Cotte E, Kwiatkowski F, Freyer G, Isaac S, et al. Intraperitoneal chemohyperthermia using a closed abdominal procedure and cytoreduction surgery for the treatment of peritoneal carcinomatosis: morbidity and mortality analysis of 216 consecutive procedures. Ann Surg Oncol. 2003;10:863–9.CrossRef
15.
Zurück zum Zitat Japanese Gastric Cancer Association. Japanese classification of gastric carcinoma: 3rd English edition. Gastric Cancer. 2011;14:101–12.CrossRef Japanese Gastric Cancer Association. Japanese classification of gastric carcinoma: 3rd English edition. Gastric Cancer. 2011;14:101–12.CrossRef
16.
Zurück zum Zitat Sobin LH, Gospodarowicz MK, Wittekind C. TNM classification of malignant tumours. 7th ed. Hoboken: Wiley-Blackwell; 2010. Sobin LH, Gospodarowicz MK, Wittekind C. TNM classification of malignant tumours. 7th ed. Hoboken: Wiley-Blackwell; 2010.
17.
Zurück zum Zitat Becker K, Mueller JD, Schulmacher C, Ott K, Fink U, Busch R, et al. Histomorphology and grading of regression in gastric carcinoma treated with neoadjuvant chemotherapy. Cancer. 2003;98:1521–30.CrossRef Becker K, Mueller JD, Schulmacher C, Ott K, Fink U, Busch R, et al. Histomorphology and grading of regression in gastric carcinoma treated with neoadjuvant chemotherapy. Cancer. 2003;98:1521–30.CrossRef
18.
Zurück zum Zitat Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240(2):205–13.CrossRef Dindo D, Demartines N, Clavien PA. Classification of surgical complications: a new proposal with evaluation in a cohort of 6336 patients and results of a survey. Ann Surg. 2004;240(2):205–13.CrossRef
19.
Zurück zum Zitat Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomark Prev. 2014;23(5):700–13.CrossRef Karimi P, Islami F, Anandasabapathy S, Freedman ND, Kamangar F. Gastric cancer: descriptive epidemiology, risk factors, screening, and prevention. Cancer Epidemiol Biomark Prev. 2014;23(5):700–13.CrossRef
20.
Zurück zum Zitat Bozzetti F, Yu W, Baratti D, Kusamura S, Deraco M. Locoregional treatment of peritoneal carcinomatosis from gastric cancer. J Surg Oncol. 2008;98(4):273–6.CrossRef Bozzetti F, Yu W, Baratti D, Kusamura S, Deraco M. Locoregional treatment of peritoneal carcinomatosis from gastric cancer. J Surg Oncol. 2008;98(4):273–6.CrossRef
21.
Zurück zum Zitat Kitayama J, Ishigami H, Yamaguchi H, Sakuma Y, Horie H, Hosoya Y, et al. Treatment of patients with peritoneal metastases from gastric cancer. Ann Gastroenterol Surg 2018;2(2):116–123. Kitayama J, Ishigami H, Yamaguchi H, Sakuma Y, Horie H, Hosoya Y, et al. Treatment of patients with peritoneal metastases from gastric cancer. Ann Gastroenterol Surg 2018;2(2):116–123.
22.
Zurück zum Zitat Yang XJ, Huang CQ, Suo T, Mei LJ, Yang GL, Cheng FL, 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(6):1575–81.CrossRef Yang XJ, Huang CQ, Suo T, Mei LJ, Yang GL, Cheng FL, 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(6):1575–81.CrossRef
23.
Zurück zum Zitat Rudloff U, Langan RC, Mullinax JE, Beane JD, Steinberg SM, Beresnev T, 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;110:275–84.CrossRef Rudloff U, Langan RC, Mullinax JE, Beane JD, Steinberg SM, Beresnev T, 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;110:275–84.CrossRef
24.
Zurück zum Zitat Takeshita K, Liu Y, Ishibashi H, Yonemura Y. Laparoscopic Hyperthermic Intraperitoneal chemotherapy for peritoneal Carcinomatosisfrom gastric Cancer: its beneficial effects on reduction and exact evaluation of the peritoneal Cancer index. Am Surg. 2017;83(11):1315–20.CrossRef Takeshita K, Liu Y, Ishibashi H, Yonemura Y. Laparoscopic Hyperthermic Intraperitoneal chemotherapy for peritoneal Carcinomatosisfrom gastric Cancer: its beneficial effects on reduction and exact evaluation of the peritoneal Cancer index. Am Surg. 2017;83(11):1315–20.CrossRef
25.
Zurück zum Zitat Yonemura Y, Endou Y, Sasaki T, Hirano M, Mizumoto A, Matsuda T, et al. Surgical treatment for peritoneal carcinomatosis from gastric cancer. Eur J Surg Oncol. 2010;36(12):1131–8.CrossRef Yonemura Y, Endou Y, Sasaki T, Hirano M, Mizumoto A, Matsuda T, et al. Surgical treatment for peritoneal carcinomatosis from gastric cancer. Eur J Surg Oncol. 2010;36(12):1131–8.CrossRef
26.
Zurück zum Zitat Wang D, Xing Y, Guo Y, Zhang Y, Chen Y, Suo J. Study of introperitoneal hyperthermic perfusion chemotherapy combined with systemic neoadjuvent chemotherapy in treatment of gastric cancer patients with peritoneal carcinomatosis. Zhonghua Wei Chang Wai Ke Za Zhi. 2016;19(5):540–4.PubMed Wang D, Xing Y, Guo Y, Zhang Y, Chen Y, Suo J. Study of introperitoneal hyperthermic perfusion chemotherapy combined with systemic neoadjuvent chemotherapy in treatment of gastric cancer patients with peritoneal carcinomatosis. Zhonghua Wei Chang Wai Ke Za Zhi. 2016;19(5):540–4.PubMed
27.
Zurück zum Zitat 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(8):1123–31.CrossRef 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(8):1123–31.CrossRef
28.
Zurück zum Zitat Yang XJ, Li Y, Yonemura Y. Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy to treat gastric cancer with ascites and/or peritoneal carcinomatosis: results from a Chinese center. J Surg Oncol. 2010;101(6):457–64.CrossRef Yang XJ, Li Y, Yonemura Y. Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy to treat gastric cancer with ascites and/or peritoneal carcinomatosis: results from a Chinese center. J Surg Oncol. 2010;101(6):457–64.CrossRef
29.
Zurück zum Zitat Chan DL, Sjoquist KM, Goldstein D, Price TJ, Martin AJ, Bang YJ, et al. The effect of anti-angiogenic agents on overall survival in metastatic oesophago-gastric cancer: a systematic review and metaanalysis. PLoS One. 2017;12(2):e0172307.CrossRef Chan DL, Sjoquist KM, Goldstein D, Price TJ, Martin AJ, Bang YJ, et al. The effect of anti-angiogenic agents on overall survival in metastatic oesophago-gastric cancer: a systematic review and metaanalysis. PLoS One. 2017;12(2):e0172307.CrossRef
Metadaten
Titel
Neoadjuvant systemic and hyperthermic intraperitoneal chemotherapy combined with cytoreductive surgery for gastric cancer patients with limited peritoneal metastasis: a prospective cohort study
verfasst von
Pengfei Yu
Zeyao Ye
Gaiguo Dai
Yanqiang Zhang
Ling Huang
Yian Du
Xiangdong Cheng
Publikationsdatum
01.12.2020
Verlag
BioMed Central
Erschienen in
BMC Cancer / Ausgabe 1/2020
Elektronische ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-020-07601-x

Weitere Artikel der Ausgabe 1/2020

BMC Cancer 1/2020 Zur Ausgabe

Blutdrucksenkung könnte Uterusmyome verhindern

Frauen mit unbehandelter oder neu auftretender Hypertonie haben ein deutlich erhöhtes Risiko für Uterusmyome. Eine Therapie mit Antihypertensiva geht hingegen mit einer verringerten Inzidenz der gutartigen Tumoren einher.

Alphablocker schützt vor Miktionsproblemen nach der Biopsie

16.05.2024 alpha-1-Rezeptorantagonisten Nachrichten

Nach einer Prostatabiopsie treten häufig Probleme beim Wasserlassen auf. Ob sich das durch den periinterventionellen Einsatz von Alphablockern verhindern lässt, haben australische Mediziner im Zuge einer Metaanalyse untersucht.

Antikörper-Wirkstoff-Konjugat hält solide Tumoren in Schach

16.05.2024 Zielgerichtete Therapie Nachrichten

Trastuzumab deruxtecan scheint auch jenseits von Lungenkrebs gut gegen solide Tumoren mit HER2-Mutationen zu wirken. Dafür sprechen die Daten einer offenen Pan-Tumor-Studie.

Mammakarzinom: Senken Statine das krebsbedingte Sterberisiko?

15.05.2024 Mammakarzinom Nachrichten

Frauen mit lokalem oder metastasiertem Brustkrebs, die Statine einnehmen, haben eine niedrigere krebsspezifische Mortalität als Patientinnen, die dies nicht tun, legen neue Daten aus den USA nahe.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.