51
|
Leigh N, Solomon D, Pletcher E, Labow DM, Magge DR, Sarpel U, Golas BJ. The importance of primary tumor origin in gastrointestinal malignancies undergoing cytoreductive surgery and hyperthermic intraperitoneal chemotherapy. World J Surg Oncol 2020; 18:182. [PMID: 32703239 PMCID: PMC7379772 DOI: 10.1186/s12957-020-01938-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/29/2020] [Indexed: 12/14/2022] Open
Abstract
Background Appendiceal and colorectal cancers with peritoneal carcinomatosis (PC) can derive benefit from cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC). However, its role in gastric and small bowel malignancies remains undefined. Methods We retrospectively analyzed 251 gastrointestinal adenocarcinomas with PC which underwent CRS/HIPEC at our institution from 2007 to 2017. We compared outcomes of gastric, small bowel, appendiceal, and colorectal cohorts. Results Thirty-one gastric, 8 small bowel, 91 appendiceal, and 121 colorectal cohorts were included. More gastric cancers (90%) received neoadjuvant chemotherapy than any other cohort, p = 0.002. Although colorectal had the lowest peritoneal cancer index (PCI) (9) and appendiceal the highest (16), all cohorts underwent similar rates of organ resection and complete cytoreduction. Length of stay (p = 0.005) and major perioperative morbidity (Clavien III/IV, p = 0.011) were significantly higher in gastric and small bowel. Median overall survival (OS, p < 0.001) was significantly shorter in gastric (13 months) and small bowel (9 months) than in appendiceal (33 months) and colorectal (42 months) cohorts. On multivariate analysis, complete cytoreduction and PCI score were significant predictors of OS, p < 0.05. Conclusions Primary tumor origin significantly affects outcomes after CRS/HIPEC for gastrointestinal malignancies. Though there was a survival benefit in appendiceal and colorectal, gastric and small bowel survival was comparable to systemic chemotherapy.
Collapse
Affiliation(s)
- Natasha Leigh
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA.
| | - Daniel Solomon
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Eric Pletcher
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Daniel M Labow
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Deepa R Magge
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Umut Sarpel
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| | - Benjamin J Golas
- Division of Surgical Oncology, Icahn School of Medicine at Mount Sinai St. Luke's West Hospital, 425 West 59th Street, Suite 7B, New York, NY, 10019, USA
| |
Collapse
|
52
|
Tan HL, Kim G, Charles CJ, Li RR, Jang CJ, Shabbir A, Chue KM, Tai CH, Sundar R, Goh BC, Bonney GK, Looi WD, Cheow ES, So JB, Wang L, Yong WP. Safety, pharmacokinetics and tissue penetration of PIPAC paclitaxel in a swine model. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2020; 47:1124-1131. [PMID: 32800400 DOI: 10.1016/j.ejso.2020.06.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/30/2020] [Accepted: 06/15/2020] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Peritoneal carcinomatosis is difficult to treat. Pressurized Intra-Peritoneal Aerosolised Chemotherapy (PIPAC) is a novel method of delivering chemotherapy to the peritoneal cavity, aiming for homogenous and deeper drug distribution. To date, limited chemotherapeutics have been used with promising results. Here, we evaluate the pharmacokinetics, peritoneal tissue drug concentration, penetration, and short-term safety of PIPAC using solvent-based paclitaxel in swine to guide clinical trials. MATERIALS AND METHODS PIPAC solvent-based paclitaxel was administered at 60, 30, and 15mg/m2 for 3 cohorts. Each PIPAC procedure was followed by intravenous (IV) administration of the same dose of solvent-based paclitaxel on Day 7, serving as control for pharmacokinetic comparison in the same pig. Safety and toxicity were evaluated by clinical assessment, blood counts and biochemistry. Blood samples were taken for pharmacokinetic analysis. Peritoneal biopsies were taken to measure tissue paclitaxel concentrations and distribution. RESULTS 12 Yorkshire x Landrace pigs underwent trial procedures. With PIPAC, there was linear pharmacokinetics and lower systemic exposure to paclitaxel compared to IV administration. MALDI-MSI demonstrated concentration of paclitaxel at the peritoneal surface, with estimated 2 mm penetration. PIPAC paclitaxel had favorable toxicity profile. The most significant adverse event was neutropenia which was dose dependent, with absolute neutrophil count <1.0 × 103/μL seen at the highest dose. One pig developed grade 2 hypersensitivity reaction during IV infusion and one death occurred during the PIPAC procedure, likely from anaphylaxis; these are known potential adverse events mandating standard precautions and monitoring. CONCLUSION PIPAC paclitaxel at 15mg/m2 may be considered for a Phase I study.
Collapse
Affiliation(s)
- Hon Lyn Tan
- National University Cancer Institute, Singapore, National University Health System, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Guowei Kim
- National University Cancer Institute, Singapore, National University Health System, Singapore; University Surgical Cluster, National University Health System, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher John Charles
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore, Singapore
| | - Renee R Li
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiovascular Research Institute (CVRI), National University Heart Centre Singapore, Singapore
| | - Clarisse Jm Jang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Asim Shabbir
- National University Cancer Institute, Singapore, National University Health System, Singapore; University Surgical Cluster, National University Health System, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Koy Min Chue
- University Surgical Cluster, National University Health System, Singapore
| | - Chia Hui Tai
- University Surgical Cluster, National University Health System, Singapore
| | - Raghav Sundar
- National University Cancer Institute, Singapore, National University Health System, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Boon Cher Goh
- National University Cancer Institute, Singapore, National University Health System, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, Singapore
| | - Glenn Kunnath Bonney
- University Surgical Cluster, National University Health System, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Institute for Health Innovation and Technology, National University of Singapore, Singapore
| | - Wen Donq Looi
- Bruker Daltonics, Bruker Singapore Pte Ltd, Singapore
| | - Esther Sh Cheow
- Institute for Health Innovation and Technology, National University of Singapore, Singapore
| | - Jimmy By So
- National University Cancer Institute, Singapore, National University Health System, Singapore; University Surgical Cluster, National University Health System, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| | - Lingzhi Wang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cancer Science Institute of Singapore, Singapore
| | - Wei Peng Yong
- National University Cancer Institute, Singapore, National University Health System, Singapore; Cancer Science Institute of Singapore, Singapore
| |
Collapse
|
53
|
Dranichnikov P, Graf W, Cashin PH. Readmissions after cytoreductive surgery and hyperthermic intraperitoneal chemotherapy-a national population-based study. World J Surg Oncol 2020; 18:67. [PMID: 32252768 PMCID: PMC7137266 DOI: 10.1186/s12957-020-01837-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/19/2020] [Indexed: 01/08/2023] Open
Abstract
Background Comprehensive readmission morbidity studies after cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are scarce. This study aimed to investigate readmissions and in-hospital morbidity after CRS and HIPEC. Methods The national in-hospital patient register was used to identify patients via the HIPEC ICD code JAQ10 2004–2014. Data were retrieved from the index CRS/HIPEC treatment and from all HIPEC-related readmissions within 6 months. Univariate/multivariate logistical analyses were performed to identify risk factors for reinterventions and readmissions. Results A total of 519 patients (mean age 56 years) had a mean hospital stay of 27 days. Within 6 months, 150 readmissions for adverse events were observed in 129 patients (25%) with 67 patients requiring an intervention (13%). Totally 179 patients (34%) required a reintervention during the first 6 months with 85 (16%) requiring a reoperation. Of these 179 patients, 83 patients (46%) did not undergo the intervention at the HIPEC centre. Gastric resection was the only independent risk factor for in-hospital intervention, and advanced age for readmission. Conclusion Morbidity causing HIPEC-related readmission was higher than expected with almost half of the interventions occurring outside the HIPEC centre. Gastric resection and high age are independent predictors of morbidity and readmission.
Collapse
Affiliation(s)
- Paul Dranichnikov
- Department of Surgical Sciences, Colorectal Surgery Section, Uppsala University Hospital, 1st Floor, Entrance 70, S-751 85, Uppsala, Sweden.
| | - Wilhelm Graf
- Department of Surgical Sciences, Colorectal Surgery Section, Uppsala University Hospital, 1st Floor, Entrance 70, S-751 85, Uppsala, Sweden
| | - Peter H Cashin
- Department of Surgical Sciences, Colorectal Surgery Section, Uppsala University Hospital, 1st Floor, Entrance 70, S-751 85, Uppsala, Sweden
| |
Collapse
|
54
|
Wang X, Li T. Postoperative pain pathophysiology and treatment strategies after CRS + HIPEC for peritoneal cancer. World J Surg Oncol 2020; 18:62. [PMID: 32234062 PMCID: PMC7110707 DOI: 10.1186/s12957-020-01842-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/20/2020] [Indexed: 02/08/2023] Open
Abstract
Background Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is a treatment choice for peritoneal cancer. However, patients commonly suffer from severe postoperative pain. The pathophysiology of postoperative pain is considered to be from both nociceptive and neuropathic origins. Main body The recent advances on the etiology of postoperative pain after CRS + HIPEC treatment were described, and the treatment strategy and outcomes were summarized. Conclusion Conventional analgesics could provide short-term symptomatic relief. Thoracic epidural analgesia combined with opioids administration could be an effective treatment choice. In addition, a transversus abdominis plane block could also be an alternative option, although further studies should be performed.
Collapse
Affiliation(s)
- Xiao Wang
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Yangfangdian, Haidian District, Beijing, 100038, China
| | - Tianzuo Li
- Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, No. 10 Tieyi Road, Yangfangdian, Haidian District, Beijing, 100038, China.
| |
Collapse
|
55
|
Fields AC, Lu PW, Li GZ, Welten V, Jolissaint JS, Vierra BM, Saadat LV, Larson AC, Atkinson RB, Melnitchouk N. Current practices and future steps for hyperthermic intraperitoneal chemotherapy. Curr Probl Surg 2020; 57:100727. [PMID: 32151327 DOI: 10.1016/j.cpsurg.2019.100727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 12/23/2019] [Indexed: 11/20/2022]
Affiliation(s)
- Adam C Fields
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| | - Pamela W Lu
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - George Z Li
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Vanessa Welten
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Joshua S Jolissaint
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | | | - Lily V Saadat
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Abby C Larson
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Rachel B Atkinson
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Nelya Melnitchouk
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Center for Surgery and Public Health, Brigham and Women's Hospital, Harvard Medical School, Boston, MA.
| |
Collapse
|
56
|
Lee HS, Kim J, Lee EJ, Park SJ, Mun J, Paik H, Oh SH, Park S, Ryu S, Lim W, Song G, Kim HS, Lee JC. Evaluation of a Novel Prototype for Pressurized Intraperitoneal Aerosol Chemotherapy. Cancers (Basel) 2020; 12:cancers12030633. [PMID: 32182896 PMCID: PMC7139407 DOI: 10.3390/cancers12030633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 11/16/2022] Open
Abstract
Pressurized intraperitoneal aerosol chemotherapy (PIPAC) has been suggested as an alternative option for treating peritoneal carcinomatosis (PC). Even with its clinical advantages, the current PIPAC system still suffers from limitations regarding drug distribution area and penetration depth. Thus, we evaluated the new PIPAC system using a novel prototype, and compared its performance to the results from previous studies related with the current MIP® indirectly because the system is currently not available for purchase in the market. The developed prototype includes a syringe pump, a nozzle, and controllers. Drug distribution was conducted using a methylene blue solution for performance test. For penetration depth evaluation, an ex-vivo experiment was performed with porcine tissues in a 3.5 L plastic box. Doxorubicin was sprayed using the novel prototype, and its penetration depth was investigated by confocal laser scanning microscopy. The experiment was repeated with varying nozzle levels from the bottom. The novel prototype sprays approximately 30 μm drug droplets at a flow rate of 30 mL/min with 7 bars of pressure. The average diameter of sprayed region with concentrated dye was 18.5 ± 1.2 cm, which was comparable to that of the current MIP® (about 10 cm). The depth of concentrated diffusion (DCD) did not differ among varying nozzle levels, whereas the depth of maximal diffusion (DMD) decreased with increasing distance between the prototype and the bottom (mean values, 515.3 μm at 2 cm; 437.6 μm at 4 cm; 363.2 μm at 8 cm), which was comparable to those of the current MIP® (about 350–500 μm). We developed a novel prototype that generate small droplets for drug aerosolization and that have a comparably wide sprayed area and depth of penetration to the current MIP® at a lower pressure.
Collapse
Affiliation(s)
- Hee Su Lee
- Interdisciplinary Program in Bioengineering, Seoul National University Graduate School, Seoul 08826, Republic of Korea; (H.S.L.); (J.K.)
| | - Junsik Kim
- Interdisciplinary Program in Bioengineering, Seoul National University Graduate School, Seoul 08826, Republic of Korea; (H.S.L.); (J.K.)
| | - Eun Ji Lee
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (E.J.L.); (S.J.P.); (J.M.); (H.P.); (S.H.O.)
| | - Soo Jin Park
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (E.J.L.); (S.J.P.); (J.M.); (H.P.); (S.H.O.)
| | - Jaehee Mun
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (E.J.L.); (S.J.P.); (J.M.); (H.P.); (S.H.O.)
| | - Haerin Paik
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (E.J.L.); (S.J.P.); (J.M.); (H.P.); (S.H.O.)
| | - Soo Hyun Oh
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (E.J.L.); (S.J.P.); (J.M.); (H.P.); (S.H.O.)
| | - Sunwoo Park
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (S.P.); (S.R.); (G.S.)
| | - Soomin Ryu
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (S.P.); (S.R.); (G.S.)
| | - Whasun Lim
- Department of Food and Nutrition, Kookmin University, Seoul 02707, Republic of Korea;
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea; (S.P.); (S.R.); (G.S.)
| | - Hee Seung Kim
- Department of Obstetrics and Gynecology, Seoul National University Hospital, Seoul 03080, Republic of Korea; (E.J.L.); (S.J.P.); (J.M.); (H.P.); (S.H.O.)
- Correspondence: (H.S.K.); (J.C.L.); Tel.: +82-2-740-8573 (J.C.L.)
| | - Jung Chan Lee
- Department of Biomedical Engineering, College of Medicine and Institute of Medical and Biological Engineering, Medical Research Center, Seoul National University, Seoul 03080, Republic of Korea
- Correspondence: (H.S.K.); (J.C.L.); Tel.: +82-2-740-8573 (J.C.L.)
| |
Collapse
|
57
|
Peritoneal metastasis in gastric cancer: results from the German database. Gastric Cancer 2020; 23:11-22. [PMID: 31228044 DOI: 10.1007/s10120-019-00978-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 06/05/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Patients with peritoneal metastases of gastric cancer have a poor prognosis with a median survival of 7 months. A benefit of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) could be shown in several selected patient cohorts but remains controversial. The aim of this study was, to reflect the results of a national German HIPEC registry initiated by the German Society of General and Visceral Surgery (DGAV). METHODS The DGAV HIPEC registry StuDoQ|Peritoneum documents patients with peritoneal malignancy contributed from 52 hospitals. All consecutive documented patients from 2011 until 2016 (n = 3078) were treated with CRS and HIPEC and were analysed. A total of 315 (10%) suffered from gastric cancer and were analysed. RESULTS A complete data set of 235 patients was available for this study, including 113 male (48.1%) and 122 female (51.9%) patients with a median age of 53.4 years (SD ± 11.9). The median PCI was 8.0 (range 1-30). A complete cytoreduction was achieved in 121 patients (71.6%). Postoperative complications (Clavien-Dindo grades 3-4) occurred in 40 patients (17%). The median overall survival (OS) time was 13 months. The 5-year survival rate was 6%. According to the PCI from 0-6 (n = 74); 7-15 (n = 70) and 16-39 (n = 24) the median OS differs significantly (18 months vs. 12 months vs. 5 months; p = 0.002). CONCLUSIONS CRS and HIPEC in selected patients with gastric cancer and peritoneal spread can improve survival when they are treated in centers. An accurate staging and patient selection are of major importance to achieve long-term survival.
Collapse
|
58
|
Wang R, Song S, Harada K, Ghazanfari Amlashi F, Badgwell B, Pizzi MP, Xu Y, Zhao W, Dong X, Jin J, Wang Y, Scott A, Ma L, Huo L, Vicente D, Blum Murphy M, Shanbhag N, Tatlonghari G, Thomas I, Rogers J, Kobayashi M, Vykoukal J, Estrella JS, Roy-Chowdhuri S, Han G, Zhang S, Mao X, Song X, Zhang J, Gu J, Johnson RL, Calin GA, Peng G, Lee JS, Hanash SM, Futreal A, Wang Z, Wang L, Ajani JA. Multiplex profiling of peritoneal metastases from gastric adenocarcinoma identified novel targets and molecular subtypes that predict treatment response. Gut 2020; 69:18-31. [PMID: 31171626 PMCID: PMC6943252 DOI: 10.1136/gutjnl-2018-318070] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/14/2019] [Accepted: 04/04/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVE Peritoneal carcinomatosis (PC) occurs frequently in patients with gastric adenocarcinoma (GAC) and confers a poor prognosis. Multiplex profiling of primary GACs has been insightful but the underpinnings of PC's development/progression remain largely unknown. We characterised exome/transcriptome/immune landscapes of PC cells from patients with GAC aiming to identify novel therapeutic targets. DESIGN We performed whole-exome sequencing (WES) and whole transcriptome sequencing (RNA-seq) on 44 PC specimens (43 patients with PC) including an integrative analysis of WES, RNA-seq, immune profile, clinical and pathological phenotypes to dissect the molecular pathogenesis, identifying actionable targets and/or biomarkers and comparison with TCGA primary GACs. RESULTS We identified distinct alterations in PC versus primary GACs, such as more frequent CDH1 and TAF1 mutations, 6q loss and chr19 gain. Alterations associated with aggressive PC phenotypes emerged with increased mutations in TP53, CDH1, TAF1 and KMT2C, higher level of 'clock-like' mutational signature, increase in whole-genome doublings, chromosomal instability (particularly, copy number losses), reprogrammed microenvironment, enriched cell cycle pathways, MYC activation and impaired immune response. Integrated analysis identified two main molecular subtypes: 'mesenchymal-like' and 'epithelial-like' with discriminating response to chemotherapy (31% vs 71%). Patients with the less responsive 'mesenchymal-like' subtype had high expression of immune checkpoint T-Cell Immunoglobulin And Mucin Domain-Containing Protein 3 (TIM-3), its ligand galectin-9, V-domain Ig suppressor of T cell activation (VISTA) and transforming growth factor-β as potential therapeutic immune targets. CONCLUSIONS We have uncovered the unique mutational landscape, copy number alteration and gene expression profile of PC cells and defined PC molecular subtypes, which correlated with PC therapy resistance/response. Novel targets and immune checkpoint proteins have been identified with a potential to be translated into clinics.
Collapse
Affiliation(s)
| | - Shumei Song
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Kazuto Harada
- GI Medical Oncology, UT MDACC, Houston, Texas, USA,Gastroenterological Surgery, Kumamoto University, Kumamoto, Japan
| | | | | | | | - Yan Xu
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Wei Zhao
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | | | | | - Ying Wang
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Ailing Scott
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Lang Ma
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Longfei Huo
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | | | | | | | | | - Irene Thomas
- GI Medical Oncology, UT MDACC, Houston, Texas, USA
| | - Jane Rogers
- Pharmacy Clinical Programs, UT MDACC, Houston, TX, USA
| | | | - Jody Vykoukal
- Clinical Cancer Prevention, UT MDACC, Houston, Texas, USA
| | | | | | | | | | - Xizeng Mao
- Genomic Medicine, UT MDACC, Houston, Texas, USA
| | | | | | - Jian Gu
- Epidemiology, UT MDACC, Houston, Texas, USA
| | | | | | - Guang Peng
- Clinical Cancer Prevention, UT MDACC, Houston, Texas, USA
| | - Ju-Seog Lee
- Systems Biology, UT MDACC, Houston, Texas, USA
| | - Samir M Hanash
- Clinical Cancer Prevention, UT MDACC, Houston, Texas, USA
| | | | - Zhenning Wang
- Surgical Oncology and General Surgery, First Hospital of China Medical University, Shenyang, China
| | | | | |
Collapse
|
59
|
Conversion Surgery with HIPEC for Peritoneal Oligometastatic Gastric Cancer. Cancers (Basel) 2019; 11:cancers11111715. [PMID: 31684115 PMCID: PMC6896107 DOI: 10.3390/cancers11111715] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/22/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Peritoneal metastases (PM) of gastric cancer (GC) are characterized by a particularly poor prognosis, with median survival time of 6 months, and virtually no 5-year survival reported. Conversion therapy for GC is defined as a surgical treatment aiming at an R0 resection after systemic chemotherapy for tumours that were originally unresectable (or marginally resectable) for technical and/or oncological reasons. The aim of the present study was to evaluate early and late outcomes in GC patients with PM who underwent the cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) after neoadjuvant (conversion) chemotherapy. Thirty patients with stage IV GC underwent CRS plus HIPEC. Severe grade III/IV (Clavien-Dindo classification) complications occurred in 13 (43%) patients. The Comprehensive Complication Index (CCI) ranged from 8.7 to 100 (median, 42.4). In the multivariate survival analysis, ypT2 and P3 (according to the Japanese classification of the PM severity) were favourable and adverse prognostic factors p = 0.031 and o = 0.035, respectively. Estimated 1- and 3-year survival was 73.9% and 36.6%, respectively. The median survival was 19.3 months. Conclusion: Conversion surgery, including extended gastrectomy and multi-organ resections followed by HIPEC performed after systemic chemotherapy therapy for GC with PM is justified in downstaged patients with ypT2 and limited (less than P3) PM.
Collapse
|
60
|
Beeharry MK, Zhu ZL, Liu WT, Yao XX, Yan M, Zhu ZG. Prophylactic HIPEC with radical D2 gastrectomy improves survival and peritoneal recurrence rates for locally advanced gastric cancer: personal experience from a randomized case control study. BMC Cancer 2019; 19:932. [PMID: 31533660 PMCID: PMC6751672 DOI: 10.1186/s12885-019-6125-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 09/03/2019] [Indexed: 02/07/2023] Open
Abstract
Background To investigate the implications of prophylactic intraoperative Hyperthermic Intraperitoneal Chemotherapy (HIPEC) with D2 radical gastrectomy for locally advanced Gastric Cancer (AGC) in a randomized case control study. Method Eighty consecutive patients with locally AGC were randomly separated into 2 groups: HIPEC group (Curative Resection + intraoperative HIPEC with cisplatin 50 mg/m2 at 42.0 ± 1.0 °C for 60 min) and Control group (Curative Resection only). Intraoperative and post-operative events, clinical recovery, morbidity and the disease-free survival (DFS) rates were closely monitored. Results Faster recovery of bowel function (43 ± 5 h vs 68 ± 7, P < 0.05) and shorter postoperative stay (8d vs 14d, P < 0.05) were noted in the HIPEC group. Among the 40 HIPEC group patients, the highest intracranial temperature recorded during the procedure was 38.2 °C but the patient made an eventless recovery. Mild renal dysfunction, hyperbilirubinemia and mild liver dysfunction were recorded in the HIPEC group but their incidences were found to be statistically insignificant when compared with the control group (P > 0.05). The 3 year DFS rate analysis showed that the prophylactic HIPEC group had a higher DFS rate (93% vs 65%, P = 0.0054). The peritoneal recurrence rate was lower in the HIPEC group (3% vs 23%, P < 0.05). Conclusion Prophylactic HIPEC with radical D2 Gastrectomy improves survival and peritoneal recurrence rates for AGC with favorable post-operative recovery at low and acceptable morbidity.
Collapse
Affiliation(s)
- Maneesh Kumarsing Beeharry
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zheng-Lun Zhu
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wen-Tao Liu
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xue-Xin Yao
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Min Yan
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zheng-Gang Zhu
- Department of Surgery, Ruijin Hospital affiliated Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
61
|
Morris MC, Dhar VK, Stevenson MA, Winer LK, Lee TC, Wang J, Ahmad SA, Patel SH, Sussman JJ, Abbott DE. Adjuvant Hyperthermic Intraperitoneal Chemotherapy (HIPEC) for patients at High-Risk of Peritoneal Metastases. Surg Oncol 2019; 31:33-37. [PMID: 31518971 DOI: 10.1016/j.suronc.2019.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 08/01/2019] [Accepted: 09/04/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND Selection of patients for hyperthermic intraperitoneal chemotherapy (HIPEC) continues to evolve. We hypothesized that adjuvant HIPEC for patients at high-risk of peritoneal progression is safe and associated with favorable outcomes. METHODS The institutional database of a high-volume center was queried for patients with high-risk disease undergoing HIPEC with a peritoneal carcinomatosis index (PCI) of 0. High-risk patients were defined as those with ruptured primary tumors or locally advanced (T4) disease. RESULTS 37 patients underwent adjuvant HIPEC, with a median follow-up of 5.2 years. 54% had low-grade (LG) tumors while 46% had high-grade (HG) tumors. No patients underwent neoadjuvant chemotherapy, while eleven patients (32.4%) received adjuvant chemotherapy. There were no perioperative mortalities, and the overall complication rate was 43%. For the entire cohort, five year recurrence-free survival (RFS) and overall survival (OS) were 77% and 100%, respectively. Five year RFS and OS were 75% and 100% for LG patients and 81% and 100% for HG patients, respectively. CONCLUSIONS Adjuvant HIPEC for patients at high-risk of peritoneal progression, with PCI 0, is safe and associated with favorable long-term survival. Additional prospective investigation is needed to identify patient populations who may benefit most from HIPEC.
Collapse
Affiliation(s)
- Mackenzie C Morris
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Vikrom K Dhar
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Megan A Stevenson
- Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Leah K Winer
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tiffany C Lee
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jiang Wang
- Department of Pathology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Syed A Ahmad
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Sameer H Patel
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Jeffrey J Sussman
- Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Daniel E Abbott
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
62
|
Abstract
Non-muscle-invasive bladder cancer (NMIBC), the most prevalent type of bladder cancer, accounts for ~75% of bladder cancer diagnoses. This disease has a 50% risk of recurrence and 20% risk of progression within 5 years, despite the use of intravesical adjuvant treatments (such as BCG or mitomycin C) that are recommended by clinical guidelines. Intravesical device-assisted therapies, such as radiofrequency-induced thermochemotherapeutic effect (RITE), conductive hyperthermic chemotherapy, and electromotive drug administration (EMDA), have shown promising efficacy. These device-assisted treatments are an attractive alternative to BCG, as issues with supply have been a problem in some countries. RITE might be an effective treatment option for some patients who have experienced BCG failure and are not candidates for radical cystectomy. Data from trials using EMDA suggest that it is effective in high-risk disease but requires further validation, and results of randomized trials are eagerly awaited for conductive hyperthermic chemotherapy. Considerable heterogeneity in patient cohorts, treatment sessions, use of maintenance regimens, and single-arm study design makes it difficult to draw solid conclusions, although randomized controlled trials have been reported for RITE and EMDA.
Collapse
|
63
|
Ortega-Deballon P, Facy O, Binquet C, Delroeux D, Rat P. CHIPOFIL: A pilot study assessing the feasibility of HIPEC without extracorporeal circuit. Pleura Peritoneum 2019; 4:20190008. [PMID: 31667330 PMCID: PMC6816282 DOI: 10.1515/pp-2019-0008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2019] [Accepted: 07/03/2019] [Indexed: 11/23/2022] Open
Abstract
Background Heated intraperitoneal chemotherapy (HIPEC) is currently performed using an external circuit including a heating device and a pump. Available devices have several drawbacks in terms of costs, technique (flow surges due to blocked tubes) and staff safety, hindering a wider use. In a previous preclinical study conducted in animals, we placed a heating wire within the abdomen to achieve and maintain hyperthermia. Our results showed this technique is safe and effective. The present pilot study was conceived as the first use of such a device in humans, aiming to confirm its safety and efficacy. Methods This was a pilot study designed to include 13 patients undergoing HIPEC. Two sets of the prototype were placed within the abdominal cavity, one in the supramesocolic and one in the inframesocolic space. The target temperature was 42–43 °C during 30–90 min according to the protocol defined for each patient. The time to set up, heat and dismantle was measured. All complications were recorded during the first postoperative year and evaluated by an independent committee. Results Nine women and four men were included. The median time to set on the device was 25 min. The target temperature was obtained in a median of 14 min and maintained uniform and homogeneously distributed within the abdomen for the scheduled duration. A permanent stirring of the viscera was performed. No thermal injury or device-related complications were observed. There were two anastomotic leaks (only one requiring reoperation), two hemoperitoneum requiring reoperation, one evisceration and one gastroparesia. Conclusions A heating cable within the peritoneal cavity can achieve safe, simple, fast and efficient HIPEC.
Collapse
Affiliation(s)
- Pablo Ortega-Deballon
- Digestive Surgical Oncology, Equipe Avenir, 14 rue Paul Gaffarel, Dijon 21079, France.,Service de Chirurgie Digestive et Cancérologique, CHU Bocage Central, 14, rue Paul Gaffarel, Dijon Cedex 21079, France
| | - Olivier Facy
- Digestive Surgical Oncology, University Hospital of Dijon, Dijon, France
| | | | - Delphine Delroeux
- Digestive Surgical Oncology, University Hospital of Besançon, Besançon, France
| | - Patrick Rat
- Digestive Surgical Oncology, University Hospital of Dijon, Dijon, France
| |
Collapse
|
64
|
Peritoneal metastases from malignant degeneration of ectopic gastric epithelium in Meckel's diverticulum: A case report. Int J Surg Case Rep 2019; 61:305-308. [PMID: 31399399 PMCID: PMC6718650 DOI: 10.1016/j.ijscr.2019.07.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/06/2019] [Indexed: 02/08/2023] Open
Abstract
Meckel’s diverticulum is an outpouching of the distal small bowel. This diverticulum may contain ectopic gastric epithelium which can become malignant. A patient with gastric cancer with peritoneal metastases had the cancer origin in a Meckel’s diverticulum. Systemic cancer chemotherapy followed by cytoreductive surgery provided 4 years of disease-free survival.
Background Meckel’s diverticulum is the most common congenital anomaly of the gastrointestinal tract, resulting from incomplete obliteration of the omphalomesenteric duct in early fetal development. The diverticulum frequently contains ectopic gastrointestinal epithelial tissue, most commonly gastric or pancreatic. Adenocarcinoma can arise from either of these epithelia but is extremely rare, with less than 50 cases reported in the world literature. Methods In a patient with peritoneal metastases from ectopic gastric mucosae in a Meckel’s diverticulum a multidisciplinary treatment plan was initiated. Results Diagnosis was evident from study of the resected specimen of small bowel in a patient with acute abdominal pain. A multiagent chemotherapy regimen using oxaliplatin, mitomycin C, doxorubicin, and capecitabine (O-Max) resulted in a clinical and radiologic response. A second surgery with extensive sampling of tissues from the abdomen and pelvis confirmed a complete response to systemic chemotherapy. At four years from onset of disease the patient has no evidence of recurrence. Conclusions Peritoneal metastases from malignant degeneration of ectopic gastric epithelium in a Meckel’s diverticulum was reported. Multidisciplinary treatments were beneficial.
Collapse
|
65
|
Abstract
PURPOSE OF REVIEW Gastric adenocarcinoma is the fifth most common and the third most lethal cancer worldwide. Surgery is the only chance of cure, but recurrence is common, even with complete resection. RECENT FINDINGS Advances in diagnosis and staging, genomic classification, surgical resection and treatment of peritoneal disease, systemic chemotherapy and chemoradiation, and targeted and immune therapies have led to the current multidisciplinary approach to gastric adenocarcinoma. Treatment of gastric cancer is rapidly evolving in an effort to combat this challenging disease.
Collapse
Affiliation(s)
- Fabian M Johnston
- Section of Gastrointestinal Surgical Oncology, Peritoneal Surface Malignancy Program, Division of Surgical Oncology, Johns Hopkins University, 600 N. Wolfe Street/Blalock 606, Baltimore, MD, 21287, USA.
| | - Michael Beckman
- Department of Surgery, Johns Hopkins Hospital, 600 N. Wolfe Street/Blalock 665, Baltimore, MD, 21287, USA
| |
Collapse
|
66
|
Brandl A, Pachmayr E, Gül-Klein S, Alberto M, Thuss-Patience P, Rau B. [Surgical treatment of peritoneal metastases of gastric cancer]. Chirurg 2019; 89:669-677. [PMID: 29616280 DOI: 10.1007/s00104-018-0625-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND Up to 17% of all patients with gastric cancer are diagnosed with the presence of peritoneal metastases, which is associated with a poor prognosis. The most promising results were shown with multimodal treatment regimens including systemic chemotherapy and cytoreductive surgery (CRS). A subsequent hyperthermic intraperitoneal chemotherapy (HIPEC).possibly has a positive effect and is currently being tested. OBJECTIVES This manuscript highlights the key role of CRS and HIPEC in patients with peritoneal metastases of gastric cancer and illustrates which patients benefit from this intensive therapy. METHODS We performed a comprehensive review of the literature to demonstrate relevant aspects in the treatment of peritoneal metastases in gastric cancer. RESULTS The use of CRS and HIPEC improves the overall survival to 11 months compared to best supportive care in selected patients. Patients who present with low volume peritoneal disease (peritoneal cancer index ≤6) have the best prognosis. This intensive treatment is associated with a relatively high morbidity (15-50%) and mortality (1-10%). Complete cytoreduction, i.e. a complete macroscopic absence of tumor tissue after resection is the most important prognostic factor. CONCLUSION The CRS and HIPEC procedures have a proven survival benefit in selected patients. Due to the relatively high morbidity and mortality, the evaluation should be performed by an experienced team including a surgical oncologist, medical oncologist and intensive care physician, to achieve the highest rate of complete cytoreduction in combination with low morbidity; however, the effect of HIPEC has to be proven and the results of the randomized GASTRIPEC trial are awaited.
Collapse
Affiliation(s)
- A Brandl
- Chirurgische Klinik, Campus Virchow-Klinikum und Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Deutschland
| | - E Pachmayr
- Chirurgische Klinik, Campus Virchow-Klinikum und Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Deutschland
| | - S Gül-Klein
- Chirurgische Klinik, Campus Virchow-Klinikum und Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Deutschland
| | - M Alberto
- Chirurgische Klinik, Campus Virchow-Klinikum und Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Deutschland
| | - P Thuss-Patience
- Medizinische Klinik mit Schwerpunkt Hämatologie, Onkologie und Tumorimmunologie, Charité - Universitätsmedizin Berlin, Berlin, Deutschland
| | - B Rau
- Chirurgische Klinik, Campus Virchow-Klinikum und Charité Campus Mitte, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Deutschland.
| |
Collapse
|
67
|
Zhu L, Chen W, Li G, Chen H, Liao W, Zhang L, Xiao X. Upregulated RACK1 attenuates gastric cancer cell growth and epithelial-mesenchymal transition via suppressing Wnt/β-catenin signaling. Onco Targets Ther 2019; 12:4795-4805. [PMID: 31417279 PMCID: PMC6592218 DOI: 10.2147/ott.s205869] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 04/29/2019] [Indexed: 12/20/2022] Open
Abstract
Purpose: As there have been few studies on the effects of the receptor for activated C kinase 1 (RACK1) on gastric cancer (GC), we aimed to explore such effects and the mechanism that may be involved. Patients and methods: Normal gastric epithelial cells and six GC cell lines were used to detect the mRNA expression of RACK1. Overexpressing RACK1 was transfected in HGC27 and MGC803 cells. The effects of overexpressing RACK1 on cell viability, migration, and invasion were determined by cell counting kit-8, wound scratch, and Transwell assay, respectively. The expressions of epithelial–mesenchymal transition (EMT) and Wnt/β-catenin signaling related genes were detected using quantitative real-time PCR or Western blot. Wnt pathway agonist LiCl was added into RACK1 overexpressing GC cells, and then cell viability, migration, and invasion were also detected. Results: RACK1 was downregulated in GC cell lines. Under the circumstance that overexpressing RACK1 was successfully transfected in the two lowest RACK1-expressing GC cells, significant inhibition of cell viability, migration, and invasion, promotion to the mRNA and protein expression of E-cadherin, as well as a decrease in the N-cadherin and Snail expressions could be observed. Overexpressing RACK1 also enhanced the protein level of phosphorylation-β-catenin/β-catenin and attenuated c-Jun protein expression. Additionally, LiCl could partially reverse the inhibitory effects of cell viability, migration and invasion by overexpressing RACK. Conclusion: We found RACK1 possibly inhibited epithelial–mesenchymal transition of GC cells through limitation of the Wnt/β-catenin pathway, thereby suppressing cell migration and invasion; RACK1 could also suppress cell growth.
Collapse
Affiliation(s)
- Lihui Zhu
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Wen Chen
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Guoqing Li
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Honghui Chen
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Wenqiu Liao
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Li Zhang
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| | - Xiaoli Xiao
- Department of Gastroenterology, The Second Hospital Affiliated to the University of South China, Hengyang, Hunan Province, People's Republic of China
| |
Collapse
|
68
|
Qin R, Yang Y, Qin W, Han J, Chen H, Zhao J, Zhao R, Li C, Gu Y, Pan Y, Wang X, Ren S, Sun Y, Gu J. The Value of Serum Immunoglobulin G Glycome in the Preoperative Discrimination of Peritoneal Metastasis from Advanced Gastric Cancer. J Cancer 2019; 10:2811-2821. [PMID: 31258789 PMCID: PMC6584920 DOI: 10.7150/jca.31380] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 05/08/2019] [Indexed: 12/16/2022] Open
Abstract
Background: Peritoneal metastasis, associated with poor prognosis in gastric cancer, is difficult to discriminate from advanced gastric cancer preoperatively. However, operative diagnosis could bring both mental and physical trauma and economic burden for patients. Consequently, a non-invasive biomarker is necessary to reduce the burden of operative diagnosis and improve survival quality of patients. This study aims to elucidate the correlation between Immunoglobulin G (IgG) N-glycome and peritoneal metastasis and find potential biomarkers in preoperative discrimination of peritoneal metastasis from advanced gastric cancer based on the comprehensive sample set. Methods: A total of 373 gastric cancer patients were enrolled and randomly sorted into training cohort (n=249) and validation cohort (n=124). The IgG N-glycome composition was analyzed by ultra-performance liquid chromatography. Results: Twenty-four glycan peaks were directly detected and 15 traits based on the same structures were evaluated between peritoneal metastasis group and advanced gastric cancer group. Several differences in IgG glycosylation were found: sialylation and fucosylation were increased in peritoneal metastasis, while neutral glycosylation, monogalacosylation and bisecting GlcNAc were decreased. Based on the significant glycomics profile, a glyco-model composed of five glycan peaks (GP6, GP9, GP11, GP21 and GP23) was established with area under the receiver operating characteristic curve (AUC) value of 0.80 (training cohort) and 0.77 (validation cohort), which showed good potential in discriminating peritoneal metastasis from advanced gastric cancer. The diagnostic performance of this model was further validated in a combined cohort (AUC=0.79). Two patients with gastric cancer were selected to perform and demonstrate the usage of the diagnostic workflow. Conclusions: Here we firstly present IgG glycome profiles in a large number of preoperative peritoneal metastasis serums. The IgG glycan was highly associated with peritoneal metastasis. These findings enhance the understanding of peritoneal metastasis. Besides, our results suggested that the newly established glyco-model could be a reliable predictor of the presence of peritoneal metastasis in patients with advanced gastric cancer.
Collapse
Affiliation(s)
- Ruihuan Qin
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yupeng Yang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wenjun Qin
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jing Han
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hao Chen
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junjie Zhao
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ran Zhao
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Can Li
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yong Gu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yiqing Pan
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xuefei Wang
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shifang Ren
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yihong Sun
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jianxin Gu
- NHC Key Laboratory of Glycoconjugates Research, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
69
|
Chiu CC, Tsao CJ, Wang JJ, Yonemura Y. Can hyperthermic intraperitoneal chemotherapy effectively control gastric cancer-associated peritoneal carcinomatosis? World J Surg Proced 2019; 9:7-11. [DOI: 10.5412/wjsp.v9.i1.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 03/11/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer-associated peritoneal carcinomatosis leads to a poor prognosis and low quality of life. The current systemic chemotherapy processes cannot effectively improve survival. Hyperthermic intraperitoneal chemotherapy (HIPEC) has been used as an alternative treatment to control this disease through recurrence prevention, definitive therapeutic modality, and symptom palliation. Although HIPEC has been demonstrated to yield favorable results mainly in some Asian studies, widespread adoption of this treatment is still debatable before larger prospective randomized controlled clinical trials confirm its effectiveness.
Collapse
Affiliation(s)
- Chong-Chi Chiu
- Department of General Surgery, Chi Mei Medical Center, Liouying 73657 and Tainan 71004, Taiwan
- Department of Electrical Engineering, Southern Taiwan University of Science and Technology, Tainan 71005, Taiwan
| | - Chao-Jung Tsao
- Department of Oncology, Chi Mei Medical Center, Liouying 73657, Taiwan
| | - Jhi-Joung Wang
- Department of Medical Research, Chi Mei Medical Center, Tainan 71004, Taiwan
- AI Biomed Center, Southern Taiwan University of Science and Technology, Tainan 71005, Taiwan
| | - Yutaka Yonemura
- Peritoneal Surface Malignancy Center, Kishiwada Tokushukai Hospital, Kishiwada, Osaka 596-8522, Japan
| |
Collapse
|
70
|
Struller F, Horvath P, Solass W, Weinreich FJ, Strumberg D, Kokkalis MK, Fischer I, Meisner C, Königsrainer A, Reymond MA. Pressurized intraperitoneal aerosol chemotherapy with low-dose cisplatin and doxorubicin (PIPAC C/D) in patients with gastric cancer and peritoneal metastasis: a phase II study. Ther Adv Med Oncol 2019; 11:1758835919846402. [PMID: 31205501 PMCID: PMC6535725 DOI: 10.1177/1758835919846402] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 04/04/2019] [Indexed: 01/11/2023] Open
Abstract
Background: Efficacy of second-line systemic chemotherapy in recurrent gastric cancer with peritoneal metastasis (RGCPM) is limited. We assessed the feasibility, safety and possible efficacy of pressurized intraperitoneal aerosol chemotherapy (PIPAC) in patients with RGCPM after ⩾1 line of palliative intravenous chemotherapy. Methods: In this open-label, single-arm, monocentric phase II ICH-GCP clinical trial, patients were scheduled for three courses of PIPAC with cisplatin 7.5 mg/m2 and doxorubicin 1.5 mg/m2 (PIPAC C/D) every 6 weeks. Patients with bowel obstruction or extraperitoneal metastasis were ineligible. The primary endpoint was clinical benefit rate (CBR) by Response Evaluation Criteria in Solid Tumors based on clinical records. Secondary endpoints included overall survival (OS), median time to progression (TTP), peritoneal carcinomatosis index (PCI), histological regression and ascites volume. Safety and tolerability were assessed by Common Terminology Criteria for Adverse Events (CTCAE) version 4, quality of life (QoL) by EORTC-QLQ30 questionnaire. Results: A total of 25 patients were enrolled and available for the analysis of the primary endpoint. Of those 25 patients, 10 (40%) had a radiological complete, partial response or stable disease. Median OS [intention to treat (ITT)] was 6.7 months, median TTP was 2.7 months. Complete or major regression on histology were observed in 9/25 patients (36%, ITT) or 6/6 [100%, per protocol (PP)] patients. There were no suspected unexpected serious adverse reactions, no treatment-related deaths, no CTCAE grade 4 toxicity and three (12%) grade 3 toxicities. Changes in the QLQ-C30 scores during PIPAC C/D therapy were small and not significant. Conclusions: PIPAC C/D was well tolerated and active in patients with RGCPM. Survival was encouraging. Randomized controlled trials should now be designed in this indication.
Collapse
Affiliation(s)
- Florian Struller
- Department of General and Transplant Surgery, Tübingen, University Hospital, Hoppe-Seyler-Strasse 3, 72076 Tübingen, Germany
| | - Philipp Horvath
- Department of General and Transplant Surgery, University Hospital Tübingen, Germany
| | - Wiebke Solass
- Department of Pathology, University Hospital Tübingen, Germany
| | | | - Dirk Strumberg
- Department of Medical Oncology, Marien Hospital, Ruhr University Bochum, Germany
| | - Marios K Kokkalis
- Department of General and Transplant Surgery, University Hospital Tübingen, Germany
| | - Imma Fischer
- Institute for Clinical Epidemiology and Applied Biometrics, University Hospital Tübingen, Germany
| | - Christoph Meisner
- Institute for Clinical Epidemiology and Applied Biometrics, University Hospital Tübingen, Germany
| | - Alfred Königsrainer
- Department of General and Transplant Surgery, University Hospital Tübingen, Germany
| | - Marc A Reymond
- Department of General and Transplant Surgery, University Hospital Tübingen, Germany National Center for Pleura and Peritoneum, University Hospital, Tübingen, Germany
| |
Collapse
|
71
|
Li Y, Chen Y, Qiu C, Ma X, Lei K, Cai G, Liang X, Liu J. 17-allylamino-17-demethoxygeldanamycin impeded chemotherapy through antioxidant activation via reducing reactive oxygen species-induced cell death. J Cell Biochem 2019; 120:1560-1576. [PMID: 30378153 DOI: 10.1002/jcb.27397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 07/11/2018] [Indexed: 01/24/2023]
Abstract
Hyperthermia enhances the anticancer effects of thymidylate synthase (TYMS) inhibitors (raltitrexed, RTX) and improves the precise biochemical mechanisms partially through enhancement of intracellular drug absorption. Recent research focuses on the potential anticancer drug target Heat Shock Protein 90 (HSP90), which could increase the sensitivity of cancer cells to TYMS inhibitors; however, with different HSP90 inhibitors, several research studies finally showed a poor efficacy in preclinical or clinical research. Here, we showed that 17-allylamino-17-demethoxygeldanamycin (17-AAG, HSP90 inhibitor) affects the efficacy of chemotherapy through antioxidant activation-induced resistance. In this study, we found that RTX, alone or in combination with hyperthermia, triggers reactive oxygen species (ROS) exposure and thus induces cell death. Also, the addition of hyperthermia showed more ROS exposure and function. The pharmacologic inhibition of HSP90 reversed the effects of chemotherapeutical treatments, while the overexpression of HSP90 showed no relation with these effects, which demonstrated that dysregulation of HSP90 might have a significant impact on chemotherapeutic treatments. The addition of 17-AAG increased the activation of antioxidant with increased antioxidant enzymes, thus affecting the RTX efficacy.
Collapse
Affiliation(s)
- Yueqi Li
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Yiyang Chen
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Cen Qiu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Xiaoying Ma
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Kecheng Lei
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Guoxiang Cai
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xin Liang
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| | - Jianwen Liu
- State Key Laboratory of Bioreactor Engineering & Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
72
|
Tan HL, Chia CS, Tan GHC, Choo SP, Tai DWM, Chua CWL, Ng MCH, Soo KC, Teo MCC. Metastatic gastric cancer: Does the site of metastasis make a difference? Asia Pac J Clin Oncol 2019; 15:10-17. [PMID: 29920947 DOI: 10.1111/ajco.13025] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/14/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Metastatic gastric cancer has a poor prognosis. We aim to study how clinical features and prognosis differs between different metastatic sites, and to identify prognostic factors for overall survival. METHODS We retrospectively reviewed patients with metastatic gastric adenocarcinoma managed at a tertiary referral cancer center over a 5-year period. We divided our cohort into three groups based on the site(s) of metastasis at presentation-peritoneal metastasis only (P), distant metastasis only (D), and peritoneal and distant metastases (PD). RESULTS We studied 470 patients with 175 (37.2%), 193 (41.1%) and 102 (21.7%) patients in the P, D and PD groups, respectively. Patients with peritoneal disease (both P and PD) had higher proportions of patients experiencing chemotherapy disruption due to unplanned hospitalizations, which were also of a longer average duration. The P group had the longest overall median survival of 8.9 months compared to the PD and D groups with 7.4 and 5.5 months, respectively (P < 0.001). On multivariate Cox regression analysis, the presence of ≥1 metastatic site (hazard ratio [HR] 1.67; 95% confidence interval [CI], 1.23-2.28; P = 0.001) was significantly associated with increased overall mortality, whereas palliative systemic chemotherapy (HR 0.29; 95% CI, 0.22-0.37; P < 0.001) and palliative gastrectomy (HR 0.24; 95% CI, 0.15-0.39; P < 0.001) were significantly associated with decreased overall mortality. CONCLUSION Metastatic gastric cancer represents a heterogeneous disease, with specific disease complications and treatment outcomes unique to different metastatic sites. We can consider novel multimodality therapies for patient subgroups with isolated metastatic disease and good prognostic factors in a bid to improve long-term survival.
Collapse
Affiliation(s)
- Hwee Leong Tan
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Claramae Shulyn Chia
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Grace Hwei Ching Tan
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Su Pin Choo
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - David Wai-Meng Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Matthew Chau Hsien Ng
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | - Khee Chee Soo
- Division of Surgical Oncology, National Cancer Centre Singapore, Singapore, Singapore
| | | |
Collapse
|
73
|
Qian H, Qian K, Cai J, Yang Y, Zhu L, Liu B. Therapy for Gastric Cancer with Peritoneal Metastasis Using Injectable Albumin Hydrogel Hybridized with Paclitaxel-Loaded Red Blood Cell Membrane Nanoparticles. ACS Biomater Sci Eng 2019; 5:1100-1112. [PMID: 33405800 DOI: 10.1021/acsbiomaterials.8b01557] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The local delivery of therapeutics in a long-term sustained manner at tumor sites is attractive for the therapy of gastric cancer with peritoneal metastasis. In this manuscript, an injectable hydrogel-encapsulating paclitaxel-loaded red blood cell membrane nanoparticles (PRNP-gel) is designed on the basis of temperature-induced phase transition of polyethylene-glycol-modified bovine serum albumin (PEG-BSA). Dynamic light scattering, ζ potential, and electron microscopy were utilized to characterize the nanoparticle-hydrogel hybrid system. It was found that the PRNP had a spherical morphology with a diameter of about 133 nm and negative surface potential. The drug loading efficiency and loading content are 85% and 22%, respectively. In situ gelation occurred within 12 min when the gel precursor was incubated at 37 °C or injected subcutaneously. The in-situ-forming hydrogel showed a sustained release profile, and the cumulative release of PTX was ∼30% after 6 days. The PRNP-gel exhibited high cytocompatibility and biodegradability in vitro and in vivo. This nanoparticle-hydrogel hybrid system is applied as a drug carrier for local chemotherapy to enhance therapeutic levels at tumor site and reduce the systemic toxicity. In vivo antitumor evaluation within a subcutaneous xenograft and peritoneal dissemination model showed that the hydrogel possesses good tumor growth suppression properties after a single injection. Hence, the as-prepared injectable hydrogel system could be a promising candidate for the local delivery of chemotherapeutic drugs.
Collapse
Affiliation(s)
- Hanqing Qian
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
| | - Keyang Qian
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| | - Juan Cai
- The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China.,Department of Oncology, The First Affiliated Hospital, Yijishan Hospital of Wannan Medical College, Wuhu 241001, China
| | - Yan Yang
- Department of Oncology, The Affiliated Jiangning Hospital with Nanjing Medical School, Nanjing 211100, China
| | - Lijing Zhu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China
| | - Baorui Liu
- The Comprehensive Cancer Center of Drum Tower Hospital, Medical School of Nanjing University and Clinical Cancer Institute of Nanjing University, Nanjing 210008, China.,The Comprehensive Cancer Centre of Nanjing Drum Tower Hospital, Clinical College of Nanjing Medical University, Nanjing 210008, China
| |
Collapse
|
74
|
Garg PK, Brandl A, Rau B. Hyperthermic Intraperitoneal Chemotherapy - Fading Perspective in the Light of Modern Systemic Chemotherapy? Visc Med 2018; 34:412-416. [PMID: 30675484 PMCID: PMC6341355 DOI: 10.1159/000493493] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Peritoneal metastasis (PM) in a gastrointestinal cancer is associated with a poor prognosis. Various guidelines recommend palliative chemotherapy in these patients as cure is not achievable. However, hyperthermic intraperitoneal chemotherapy (HIPEC) in combination with cytoreductive surgery (CRS) can achieve long-term survival with low morbidity in selected patients. During the last decades, CRS and HIPEC have been performed in a number of indications, aiming for long-term survival. What is more, new drugs and treatment regimens demonstrate favorable results. METHODS The recently published important literature on PM in gastrointestinal cancer was analyzed. RESULTS Resection of isolated PM in a gastrointestinal cancer should be considered, and HIPEC is feasible. However, there is a lack of well-designed randomized controlled trials (RCTs) in gastrointestinal cancer-related PM. Many RCTs are being conducted across the world whose results will be available in 1-2 years. Systemic chemotherapy alone as an adequate management plan for isolated PM in a gastrointestinal cancer is not compatible with a high standard of care. Formulating an optimal plan combining the benefits of CRS and HIPEC coupled with systemic chemotherapy is a necessary task of the multidisciplinary team. Proper selection of the patients for CRS and HIPEC is necessary to achieve maximal oncological benefit and minimal perioperative morbidity and mortality. CONCLUSIONS A multidisciplinary approach combining the benefits of both CRS and HIPEC, and systemic chemotherapy is the way forward to manage those patients with gastrointestinal cancer and PM. Further studies are warranted to identify the selection variables for CRS and HIPEC.
Collapse
Affiliation(s)
- Pankaj Kumar Garg
- Department of Surgery, University College of Medical Sciences & Guru Teg Bahadur Hospital, University of Delhi, Delhi, India
| | - Andreas Brandl
- Department of General Surgery, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Beate Rau
- Department of General Surgery, Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
75
|
Dahdaleh FS, Turaga KK. Evolving Treatment Strategies and Outcomes in Advanced Gastric Cancer with Peritoneal Metastasis. Surg Oncol Clin N Am 2018; 27:519-537. [PMID: 29935687 DOI: 10.1016/j.soc.2018.02.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Gastric cancer (GC) has a predilection to metastasize to the peritoneum, denoting a poor prognosis. Treatment strategies available for advanced GC have significantly evolved over time and can be categorized into systemic, regional, and surgical. Although systemic therapies have been the mainstay for the treatment of advanced GC, their ability in achieving long-term survival in patients with peritoneal involvement is modest at best. This article describes advances in combined modality treatment of peritoneal metastases, specifically with an emphasis on peritoneal-directed therapies.
Collapse
Affiliation(s)
- Fadi S Dahdaleh
- Complex General Surgical Oncology, Section of General Surgery/Surgical Oncology, The University of Chicago Medicine, 5841 South Maryland Avenue, Room S214, MC 5094, Chicago, IL 60637, USA
| | - Kiran K Turaga
- The University of Chicago Medicine, Section of General Surgery/Surgical Oncology, 5841 South Maryland Avenue, Room G207, MC 5094, Chicago, IL 60637, USA.
| |
Collapse
|
76
|
Zurleni T, Gjoni E, Altomare M, Rausei S. Conversion surgery for gastric cancer patients: A review. World J Gastrointest Oncol 2018; 10:398-409. [PMID: 30487951 PMCID: PMC6247102 DOI: 10.4251/wjgo.v10.i11.398] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 09/25/2018] [Accepted: 10/07/2018] [Indexed: 02/05/2023] Open
Abstract
Gastric cancer (GC) is the third most common cancer-related cause of death worldwide. In locally advanced tumors, neoadjuvant chemotherapy has recently been introduced in most international Western guidelines. For metastatic and unresectable disease, there is still debate regarding correct management and the role of surgery. The standard approach for stage IV GC is palliative chemotherapy. Over the last decade, an increasing number of M1 patients who responded to palliative regimens of induction chemotherapy have been subsequently undergone surgery with curative intent. The objective of the present review is to analyze the literature regarding this approach, known as “conversion surgery”, which has become one of the most commonly adopted therapeutic options. It is defined as a treatment aiming at an R0 resection after chemotherapy in initially unresectable tumors. The 13 retrospective studies analyzed, with a total of 411 patients treated with conversion therapy, clearly show that even if standardization of unresectable and metastatic criteria, post-chemotherapy resectability evaluation and timing of surgery has not yet been established, an R0 surgery after induction chemotherapy with partial or complete response seems to offer superior survival results than chemotherapy alone. Additional larger sample-size randomized control trials are needed to identify subgroups of well-stratified patients who could benefit from this multimodal approach.
Collapse
Affiliation(s)
- Tommaso Zurleni
- Department of Surgery, ASST Valle Olona, Busto Arsizio 21052, Italy
| | - Elson Gjoni
- Department of Surgery, ASST Valle Olona, Busto Arsizio 21052, Italy
| | - Michele Altomare
- Department of Surgery, ASST Valle Olona, Busto Arsizio 21052, Italy
| | - Stefano Rausei
- Department of Surgery, ASST Valle Olona, Gallarate. 21013, Italy
| |
Collapse
|
77
|
Su Y, Liu M, Liang K, Liu X, Song Y, Deng Y. Evaluating the Accelerated Blood Clearance Phenomenon of PEGylated Nanoemulsions in Rats by Intraperitoneal Administration. AAPS PharmSciTech 2018; 19:3210-3218. [PMID: 30187444 DOI: 10.1208/s12249-018-1120-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/02/2018] [Indexed: 12/24/2022] Open
Abstract
The accelerated blood clearance (ABC) phenomenon is induced by repeated intravenous injection of stealth polyethylene glycol (PEG) nanocarriers and appears as the alteration of the pharmacokinetics and biodistribution of the second administration. Nevertheless, there is no any report about the ABC phenomenon induced by intraperitoneal administration of PEGylated nanocarriers. In this study, we firstly observed whether the ABC phenomenon is induced with PEGylated nanoemulsion at the dose of 0.5~100 μmol phospholipid·kg-1 by intraperitoneal/intravenous injections in rats. The PEG (molecule weight, 2000)-modified nanoemulsions PE-B and PE in which fluorescence indicator dialkylcarbocyanines (DiR) is encapsulated by PE-B were prepared for this work. The pharmacokinetics of the first injected PE via veins or peritoneal cavity features different variation trends. Moreover, the tissue distributions (in vivo or in vitro) of the first injected PE by intraperitoneal injection reveals that the PE gains access to the whole lymphatic circulatory system. Furthermore, our results demonstrate that the ABC phenomenon can be induced by intraperitoneal administration PE-B and present obvious changes with varying PE-B concentration 0.5~100 μmol phospholipid·kg-1. Moreover, an interesting point is that the ABC phenomenon induced by intraperitoneal injected PE-B can be significantly inhibited by intraperitoneal pre-injection of distilled water. For understanding this issue clear, we studied the production of anti-PEG IgM and the characteristic morphologies of immune cells. We observed that the mast cells in peritoneal cavity exhibit rapid depletion in response to the intraperitoneal pre-injection of distilled water, while the anti-PEG IgM secretes at the same level.
Collapse
|
78
|
Brandl A, Pratschke J, Rau B. Chirurgisch-onkologische Therapie primärer und sekundärer peritonealer Neoplasien. Eur Surg 2018. [DOI: 10.1007/s10353-018-0553-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
79
|
Cytoreductive surgery combined with hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with gastric cancer and peritoneal carcinomatosis. Eur J Surg Oncol 2018; 44:1805-1810. [PMID: 30087071 DOI: 10.1016/j.ejso.2018.06.036] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 05/23/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Gastric Cancer (GC) with Peritoneal Carcinomatosis (PC) has long been regarded as a terminal disease. Over the past two decades, cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) has changed the traditional concept of peritoneal metastases from being a systemic disease, to being considered a locoregional dissemination. PATIENTS AND METHODS A prospective study was performed at a high-volume Carcinomatosis Center to evaluate survival, morbi-mortality and prognostic factors for survival in a cohort of patients with GC and PC treated with CRS + HIPEC between June 2006 and December 2016. RESULTS Thirty-five patients were included in the study. Median follow-up was 54 months. Postoperative major complications (>grade IIIa) occurred in 25.7% of patients, including 2 deaths (mortality 5.7%). The median overall survival (OS) was 16 months and the 1-, 3- and 5-year OS rates were 70.8%, 21.3% and 21.3% %, respectively. The median OS for patients with PCI ≤6 was 19 months, in contrast to 12 months for the 19 patients with PCI >6. Three patients were included with only a positive cytology and their median OS was not reached. Perineural invasion was the only factor that had a negative influence in prognosis (HR 18.8) in multivariate analysis. CONCLUSION Although GC with PC still has a poor prognosis, survival has improved in selected patients with CRS + HIPEC and perioperative systemic chemotherapy. Patients with isolated positive cytology or peritoneal carcinomatosis with PCI less than 6 had encouraging survival rates.
Collapse
|
80
|
Solanki SL, Kumar PP, DeSouza A, Saklani AP. Perioperative concerns and management of pressurised intraperitoneal aerosolised chemotherapy: Report of two cases. Indian J Anaesth 2018; 62:225-228. [PMID: 29643558 PMCID: PMC5881326 DOI: 10.4103/ija.ija_623_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Pressurised intraperitoneal aerosolised chemotherapy (PIPAC) is a new, mostly supportive approach to help patients with advanced peritoneal metastasis to increase the lifespan. It carries occupational hazards to health-care workers and especially anaesthesiologist during the procedure. The aerosolised chemotherapy can also cause chemical peritonitis and organ dysfunction in the perioperative period. In this case report, we present the report of two cases and discuss the perioperative concerns and management related to PIPAC.
Collapse
Affiliation(s)
- Sohan Lal Solanki
- Department of Anesthesiology, Critical Care and Pain, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Pooja P Kumar
- Department of Anesthesiology, Critical Care and Pain, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Ashwin DeSouza
- Department of Surgical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| | - Avanish P Saklani
- Department of Surgical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, Maharashtra, India
| |
Collapse
|
81
|
Li Y, Yan X, Ren L, Li Y. miR-644a Inhibits Cellular Proliferation and Invasion via Suppression of CtBP1 in Gastric Cancer Cells. Oncol Res 2018; 26:1-8. [PMID: 27983935 PMCID: PMC7844550 DOI: 10.3727/096504016x14772410356982] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) is one of the most important mechanisms in the metastasis of various cancers, including gastric cancer (GC). In this study, we explored the putative significance of miR-644a and its role in EMT-mediated metastasis of GC. We first detected the expression of miR-644a in a cohort of 107 GC tissues using quantitative RT-PCR. The expression of miR-644a was suppressed in GC tissues and was associated with a later clinical stage and tumor metastasis. Restoring the expression of miR-644a could significantly suppress the migration and invasion of HGC-27 and SGC-7901 cells, which might be correlated to its suppressive effect on the EMT process. We also found that carboxyl-terminal-binding protein 1 (CtBP1) was a putative target gene of miR-644a in GC and might be involved in the suppressive effect. Collectively, through targeting CtBP1-mediated suppression of the EMT process, miR-644a might suppress the tumor metastasis of GC cells.
Collapse
Affiliation(s)
- Yingchao Li
- *Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, P.R. China
| | - Xiaoni Yan
- *Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, P.R. China
| | - Li Ren
- *Department of Gastroenterology, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi Province, P.R. China
| | - Yang Li
- †Department of Otolaryngology, Head and Neck Surgery, The Second Affiliated Hospital, Medical School of Xi’an Jiaotong University, Xi’an, Shaanxi, P.R. China
| |
Collapse
|
82
|
Pamela K, Matthias Z, Reinhold KR, Julia P, Peter M, Alexander P, Dietmar Ö. Cytoreductive Surgery (CRS) and Hyperthermic Intraperitoneal Chemotherapy (HIPEC): a Single-Center Experience in Austria. J Gastrointest Surg 2018; 22:884-893. [PMID: 29363016 PMCID: PMC5954007 DOI: 10.1007/s11605-017-3661-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 12/18/2017] [Indexed: 01/31/2023]
Abstract
BACKGROUND Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) can significantly influence overall and disease-free survival in selected patients suffering from peritoneal surface malignancies (PSM) of various tumor entities. Because of the extent of the therapeutic approach, the associated morbidity and mortality and the multidisciplinarity needed, implementation of a CRS + HIPEC program at an institution is often challenging. METHODS This single-center analysis included all patients (n = 60, 34 female, 26 male) with PSM from various tumor primaries [colorectal cancer (15/60; 25%), appendix neoplasia (21/60; 35%), and others (24/60; 40%)] treated with CRS + HIPEC at our institution between 2006 and 2014. Charts were reviewed for preoperative patient evaluation, procedure-specific and tumor-specific parameters, morbidity, mortality, tumor recurrence and patients' overall (OS), and disease-free survival (DFS). RESULTS In 57 of the 60 patients included in the investigation (57/60; 95%), a radical resection (CC 0/1) was achieved. Median operating time was 559 min (253-900) with a median need of packed red blood cells of 1.1 (0-7) or fresh frozen plasma of 4.4 (0-20) concentrates. Twenty (33.3%) patients experienced 24 Dindo-Clavien grade III/IV complications (24/63; 38.1%). Postoperative 30- and 90-day mortality was 0% in our study population. Five-year OS was 43%, 5-year DFS 33%. CONCLUSIONS Due to thorough preoperative patient evaluation, strict inclusion and exclusion criteria, and intense collaboration with other specialties, we were able to achieve an excellent 5-year OS of 43% with a CC score of 0/1 in 95% of our patient population. We were able to demonstrate the feasibility, efficacy, and safety of CRS + HIPEC in patients suffering from PSM at our institution.
Collapse
Affiliation(s)
- Kogler Pamela
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Anichstr. 35, 6020 Innsbruck, Austria
| | - Zitt Matthias
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Anichstr. 35, 6020 Innsbruck, Austria ,Department of Surgery, Dornbirn Hospital, Dornbirn, Austria
| | - Kafka-Ritsch Reinhold
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Anichstr. 35, 6020 Innsbruck, Austria
| | - Punter Julia
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Anichstr. 35, 6020 Innsbruck, Austria
| | - Müssigang Peter
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Anichstr. 35, 6020 Innsbruck, Austria
| | - Perathoner Alexander
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Anichstr. 35, 6020 Innsbruck, Austria
| | - Öfner Dietmar
- Department of Visceral, Transplant and Thoracic Surgery, Medical University of Innsbruck, Anichstr. 35, 6020 Innsbruck, Austria
| |
Collapse
|
83
|
Does additional HIPEC help after CRS in peritoneal disseminated gastric cancer? CURRENT ISSUES IN PHARMACY AND MEDICAL SCIENCES 2017. [DOI: 10.1515/cipms-2017-0035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
The incidence of synchronous or metachronous peritoneal metastases (PM) in patients with locally advanced gastric cancer is high, and associated with a poor prognosis. The recommended therapeutic option for these patients is systemic chemotherapy and leads to a median of 7-8 months. However, new approaches like cytoreductive surgery and hyperthermic intraperitoneal chemotherapy might help to improve the median survival in selected patients.
Indications, patient selection and the choice of the chemotherapeutic agent are described in this manuscript, as well as an overview of the most recent literature about this topic.
Collapse
|
84
|
Topal B, Demey K, Topal H, Jaekers J, Van Cutsem E, Vandecaveye V, Sagaert X, Prenen H. Cytoreductive surgery and Hyperthermic intra-operative peritoneal chemotherapy with Cisplatin for gastric peritoneal Carcinomatosis Monocentric phase-2 nonrandomized prospective clinical trial. BMC Cancer 2017; 17:771. [PMID: 29149865 PMCID: PMC5693494 DOI: 10.1186/s12885-017-3730-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 10/30/2017] [Indexed: 01/12/2023] Open
Abstract
Background Cytoreductive surgery (CRS) plus hyperthermic intra-operative peritoneal chemotherapy (HIPC) for gastric peritoneal carcinomatosis (PC) is controversial, and selection criteria for this treatment modality are lacking. Methods Thirty-two patients (F/M ratio 12/20; median (range) age 58 (32-75) years) underwent CRS + HIPC with cisplatin for PC from gastric adenocarcinoma in 2010-2014. This monocentric phase-2 nonrandomized prospective study with a power of 90% aimed to improve the 1-year overall survival (OS) rate with 40% (historical reference of 52% to 72%). Median PCI score was 8 (range 1-20), number of regions involved was 6 (range 1-11). The impact of 16 prognostic factors on survival was evaluated using univariable and multivariable Cox regression models. Follow-up was complete in all patients, and closed 2 years after patient inclusion. Results All patients had complete cytoreduction (CCR-0) and histopathological R0 resection. PCI </= 12 without PC on any small bowel region with 4 or more non-small bowel regions resulted in a median OS time of 24.7 months (15.6–29.4), and 1, 2, 5-year OS rates of 90%, 55%, 5.6%, respectively. Independent predictors of OS were PC on the small bowel combined with PC on 4 or more non-small bowel regions (p = 0.0004), number of regions involved (p = 0.0029), and overall PCI score (p = 0.0104). Conclusions CRS + HIPC with cisplatin to treat gastric PC, providing complete cytoreduction and R0 resection, should be restricted to patients with PCI of 12 or less. Patients having PC on any small bowel region with 4 or more non-small bowel regions should be refused for CRS + HIPC. Trial registration number Registration number: NCT01116791. Registration date: May 5, 2010.
Collapse
Affiliation(s)
- Baki Topal
- Department of Abdominal Surgical Oncology, University Hospitals KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Karel Demey
- Department of Abdominal Surgical Oncology, University Hospitals KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Halit Topal
- Department of Abdominal Surgical Oncology, University Hospitals KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Joris Jaekers
- Department of Abdominal Surgical Oncology, University Hospitals KU Leuven, Herestraat 49, 3000, Leuven, Belgium
| | - Eric Van Cutsem
- Department of Digestive Oncology, University Hospitals KU Leuven, Leuven, Belgium
| | | | - Xavier Sagaert
- Department of Pathology, University Hospitals KU Leuven, Leuven, Belgium
| | - Hans Prenen
- Department of Digestive Oncology, University Hospitals KU Leuven, Leuven, Belgium
| |
Collapse
|
85
|
Almerie MQ, Gossedge G, Wright KE, Jayne DG. Treatment of peritoneal carcinomatosis with photodynamic therapy: Systematic review of current evidence. Photodiagnosis Photodyn Ther 2017; 20:276-286. [PMID: 29111390 DOI: 10.1016/j.pdpdt.2017.10.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 10/11/2017] [Accepted: 10/26/2017] [Indexed: 11/28/2022]
Abstract
BACKGROUND Peritoneal carcinomatosis results when tumour cells implant and grow within the peritoneal cavity. Treatment and prognosis vary based on the primary cancer. Although therapy with intention-to-cure is offered to selective patients using cytoreductive surgery with chemotherapy, the prognosis remains poor for most of the patients. Photodynamic therapy (PDT) is a cancer-therapeutic modality where a photosensitiser is administered to patients and exerts a cytotoxic effect on cancer cells when excited by light of a specific wavelength. It has potential application in the treatment of peritoneal carcinomatosis. METHODS We systematically reviewed the evidence of using PDT to treat peritoneal carcinomatosis in both animals and humans (Medline/EMBASE searched in June 2017). RESULTS Three human and 25 animal studies were included. Phase I and II human trials using first-generation photosensitisers showed that applying PDT after surgical debulking in patients with peritoneal carcinomatosis is feasible with some clinical benefits. The low tumour-selectivity of the photosensitisers led to significant toxicities mainly capillary leak syndrome and bowel perforation. In animal studies, PDT improved survival by 15-300%, compared to control groups. PDT led to higher tumour necrosis values (categorical values 0-4 [4=highest]: PDT 3.4±1.0 vs. control 0.4±0.6, p<0.05) and reduced tumour size (residual tumour size is 10% of untreated controls, p<0.001). CONCLUSION PDT has potential in treating peritoneal carcinomatosis, but is limited by its narrow therapeutic window and possible serious side effects. Recent improvement in tumour-selectivity and light delivery systems is promising, but further development is needed before PDT can be routinely applied for peritoneal carcinomatosis.
Collapse
Affiliation(s)
- Muhammad Qutayba Almerie
- Section of Translational Anaesthesia and Surgical Sciences, Leeds Institute of Biomedical & Clinical Sciences (LIBACS), St James's University Hospital, Leeds LS9 7TF, UK.
| | - Gemma Gossedge
- Section of Translational Anaesthesia and Surgical Sciences, Leeds Institute of Biomedical & Clinical Sciences (LIBACS), St James's University Hospital, Leeds LS9 7TF, UK.
| | - Kathleen E Wright
- Section of Translational Anaesthesia and Surgical Sciences, Leeds Institute of Biomedical & Clinical Sciences (LIBACS), St James's University Hospital, Leeds LS9 7TF, UK.
| | - David G Jayne
- Section of Translational Anaesthesia and Surgical Sciences, Leeds Institute of Biomedical & Clinical Sciences (LIBACS), St James's University Hospital, Leeds LS9 7TF, UK.
| |
Collapse
|
86
|
Badgwell B, Blum M, Das P, Estrella J, Wang X, Ho L, Fournier K, Royal R, Mansfield P, Ajani J. Phase II Trial of Laparoscopic Hyperthermic Intraperitoneal Chemoperfusion for Peritoneal Carcinomatosis or Positive Peritoneal Cytology in Patients with Gastric Adenocarcinoma. Ann Surg Oncol 2017; 24:3338-3344. [PMID: 28799004 DOI: 10.1245/s10434-017-6047-4] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE The aim of this phase II study was to perform neoadjuvant hyperthermic intraperitoneal chemoperfusion (HIPEC) via a minimally invasive approach without cytoreduction for patients with gastric cancer and positive peritoneal cytology or low-volume peritoneal carcinomatosis. METHODS Patients with gastric or gastroesophageal adenocarcinoma and positive peritoneal cytology or radiologically occult peritoneal carcinomatosis after systemic chemotherapy received laparoscopic HIPEC with mitomycin C 30 mg and cisplatin 200 mg. Patients whose peritoneal disease resolved were offered gastrectomy. The primary endpoint was overall survival (OS), with secondary endpoints of HIPEC complications and gastrectomy rate. RESULTS We enrolled 19 patients (6 with positive peritoneal cytology only and 13 with peritoneal carcinomatosis) and treated them with 38 laparoscopic HIPEC procedures. Patients had received a median of 8 cycles (range 3-12) of systemic chemotherapy prior to enrollment. Fourteen patients were also treated with chemoradiotherapy before or between cycles of HIPEC. The complication rate for HIPEC was 11% (4 of 38 procedures), the 30-day mortality rate was 0%, and the median length of hospital stay after HIPEC was 3 days (range 2-6). Five patients went on to receive gastrectomy. The median follow-up was 18.9 months, the median OS from the date of diagnosis of metastatic disease was 30.2 months, and the median OS from the first laparoscopic HIPEC was 20.3 months. CONCLUSIONS Laparoscopic HIPEC was well tolerated, and an encouraging number of patients demonstrated an absence of peritoneal disease after HIPEC and were able to undergo gastrectomy. Comparative studies will be required to clarify survival benefits.
Collapse
Affiliation(s)
- Brian Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Mariela Blum
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prajnan Das
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeannelyn Estrella
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuemei Wang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Linus Ho
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keith Fournier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Richard Royal
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Mansfield
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jaffer Ajani
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
87
|
van der Kaaij RT, Braam HJ, Boot H, Los M, Cats A, Grootscholten C, Schellens JH, Aalbers AG, Huitema AD, Knibbe CA, Boerma D, Wiezer MJ, van Ramshorst B, van Sandick JW. Treatment of Peritoneal Dissemination in Stomach Cancer Patients With Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy (HIPEC): Rationale and Design of the PERISCOPE Study. JMIR Res Protoc 2017; 6:e136. [PMID: 28705789 PMCID: PMC5532515 DOI: 10.2196/resprot.7790] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/30/2017] [Indexed: 01/16/2023] Open
Abstract
Background Patients with gastric cancer and peritoneal carcinomatosis have a very poor prognosis; median survival is 3 to 4 months. Palliative systemic chemotherapy is currently the only treatment available in the Netherlands. Intraoperative hyperthermic intraperitoneal chemotherapy (HIPEC) has an established role in the treatment of peritoneal carcinomatosis originating from colorectal cancer, appendiceal cancer, and pseudomyxoma peritonei; its role in gastric cancer is uncertain. Currently, there is no consensus on the choice of chemotherapeutic agents used in HIPEC for gastric cancer. Objective The main objectives of this study are (1) to investigate the safety, tolerability, and feasibility of gastrectomy combined with cytoreductive surgery and HIPEC after systemic chemotherapy, as a primary treatment option for patients with advanced gastric cancer with tumor positive peritoneal cytology and/or limited peritoneal carcinomatosis; and (2) to determine the maximum tolerated dose (MTD) of intraperitoneal docetaxel in combination with a fixed dose of intraperitoneal oxaliplatin. Methods The PERISCOPE study is a multicenter, open label, phase I-II dose-escalation study. The MTD of docetaxel will be studied using a 3+3 design. Patients with locally advanced (cT3-cT4) gastric adenocarcinoma are eligible for inclusion if the primary gastric tumor is considered resectable, tumor positive peritoneal cytology and/or limited peritoneal carcinomatosis is confirmed by diagnostic laparoscopy/ laparotomy, and prior systemic chemotherapy was without disease progression. At laparotomy, cytoreductive surgery (complete removal of all macroscopically visible tumor deposits) and a total or partial gastrectomy with a D2 lymph node dissection is performed. An open HIPEC technique is used with 460mg/m2 hyperthermic oxaliplatin for 30 minutes (41°C to 42°C) followed by normothermic docetaxel for 90 minutes (37°C) in a dose that will be escalated per 3 patients (0, 50, 75, 100, 125, 150 mg/m2). The primary endpoint is treatment related toxicity. Results Patient accrual is ongoing and the first results are expected in 2017. Conclusions The PERISCOPE study will determine the safety, tolerability, and feasibility of gastrectomy combined with cytoreduction and HIPEC using oxaliplatin in combination with docetaxel after systemic chemotherapy as primary treatment option for gastric cancer patients with tumor positive peritoneal cytology and/or limited peritoneal carcinomatosis. This study will provide pharmacokinetic data on the intraperitoneal administration of oxaliplatin and docetaxel, including the MTD of intraperitoneal-administered docetaxel. These data are a prerequisite for the safe conduct of future HIPEC studies in patients with gastric cancer. Trial Registration Netherlands Trial Registration (NTR): NTR4250; http://www.trialregister.nl/trialreg/admin/ rctview.asp?TC=4250 (Archived by WebCite at http://www.webcitation.org/6rWJONgkt)
Collapse
Affiliation(s)
- Rosa T van der Kaaij
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Department of Surgical Oncology, Amsterdam, Netherlands
| | - Hidde Jw Braam
- St. Antonius Hospital, Department of Surgery, Nieuwegein, Netherlands
| | - Henk Boot
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Department of Gastroenterology, Amsterdam, Netherlands
| | - Maartje Los
- St. Antonius Hospital, Department of Medical Oncology, Nieuwegein, Netherlands
| | - Annemieke Cats
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Department of Gastroenterology, Amsterdam, Netherlands
| | - Cecile Grootscholten
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Department of Medical Oncology, Amsterdam, Netherlands
| | - Jan Hm Schellens
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Department of Clinical Pharmacology, Amsterdam, Netherlands
| | - Arend Gj Aalbers
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Department of Surgical Oncology, Amsterdam, Netherlands
| | - Alwin Dr Huitema
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Department of Pharmacy and Pharmacology, Amsterdam, Netherlands.,University Medical Center Utrecht, Department of Clinical Pharmacy, Utrecht, Netherlands
| | | | - Djamila Boerma
- St. Antonius Hospital, Department of Surgery, Nieuwegein, Netherlands
| | - Marinus J Wiezer
- St. Antonius Hospital, Department of Surgery, Nieuwegein, Netherlands
| | | | - Johanna W van Sandick
- The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Department of Surgical Oncology, Amsterdam, Netherlands
| |
Collapse
|
88
|
Fugazzola P, Coccolini F, Montori G, Ceresoli M, Baggi P, Costanzo A, Tomasoni M, Gregis F, Nozza S, Ansaloni L. 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. [PMID: 28736644 DOI: 10.21037/jgo.2017.03.11] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Our experience regarding cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) was reviewed in terms of overall survival (OS) and disease-free survival (DFS) in patients with synchronous peritoneal carcinomatosis (SPC) and metachronous peritoneal carcinomatosis (MPC) from gastric cancer (GC). METHODS An analysis of prospectively collected data about patients who underwent CRS and HIPEC from July 2011 to July 2016 was carried out. Patients and tumor characteristics were taken into consideration together with pre and post-operative data. The outcomes concerned OS and DFS in both groups. RESULTS A total of 17 cases were reported. All patients of SPC group underwent neoadjuvant chemotherapy, while all patients of MPC group underwent adjuvant chemotherapy subsequently to surgery of the primary tumor. The mean follow up period was 9 months (SD±9.5). Thirteen patients (76.5%) had SPC and four (23.5%) had MPC. The mean total Peritoneal Cancer Index (PCI) was 8.5 (SD±8.4). The mean PCI was 3.75 (SD±4.9) for SPC group and 16 (SD±9.5) for MPC (P=0.003). HIPEC regimen was cisplatin plus paclitaxel for fourteen patients (82.4%) and cisplatin plus mitomycin-C (MMC) for three patients (17.6%). OS was 16 months and 6 months respectively in patients with SPC and MPC (P=0.189). DFS was 11 months and 2 months respectively in the two groups (P=0.156). Patients with SPC patients and PCI ≥12 were significantly different in terms of DFS from SPC with PCI <12 (P=0.001). Overall, twelve patients had postoperative major complications (CTCAE>2), in particular eight (61%) in SPC group while four (100%) in MPC group. Our study showed significantly better DFS for patients aged >60 years (P=0.016). CONCLUSIONS HIPEC and CRS with cisplatin and paclitaxel in patients with PC from GC showed promising results in improving the DFS and the OS, particularly for patients with PCI <12 and for those aged >60. Although a high incidence of complications was revealed, especially in MPC group.
Collapse
Affiliation(s)
- Paola Fugazzola
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Federico Coccolini
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Giulia Montori
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Marco Ceresoli
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Paolo Baggi
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Antonio Costanzo
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Matteo Tomasoni
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Francesco Gregis
- Department of Farmacology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Silvia Nozza
- Department of Farmacology, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Luca Ansaloni
- Department of General and Emergency Surgery, ASST Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
89
|
Solon JG, O'Neill M, Chang KH, Deady S, Cahill R, Moran B, Shields C, Mulsow J. An 18 year population-based study on site of origin and outcome of patients with peritoneal malignancy in Ireland. Eur J Surg Oncol 2017; 43:1924-1931. [PMID: 28583791 DOI: 10.1016/j.ejso.2017.05.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 04/15/2017] [Accepted: 05/10/2017] [Indexed: 12/17/2022] Open
Abstract
Peritoneal malignancy (PM) is predominantly metastatic spread from advanced gastrointestinal or gynaecological cancer. PM is generally considered incurable and therefore has rarely been the focus of novel therapeutic strategies. This study assessed patterns and survival outcomes for patients with PM in Ireland. The National Cancer Registry Ireland database was interrogated to identify patients diagnosed with PM during the period 1994-2012. Patient and tumour characteristics were retrieved and survival outcomes calculated. 5791 patients were diagnosed during the study period. Median age at diagnosis was 68 years; females accounted for 62%. The incidence increased annually from 228 in 1994 to 401 in 2012. Primary PM accounted for 3% of cases. Colorectal (22%), ovarian (16%) and gastric (13%) cancers accounted for the majority of cases of secondary PM. Almost 75% of patients had PM at initial presentation. Almost 40% of patients (n = 2274) underwent surgical intervention, while 44% (n = 2560) had tumour directed chemotherapy. The median survival (MS) in patients with secondary PM was 6.6 months, and did not improve significantly during the study period. Outcomes were best in patients with ovarian cancer (MS 15.9 months) and colorectal cancer (MS 14.3 months) and worst in patients with lung (MS 2.4 months) and pancreas (MS 1.9 months) cancers. This is the first population-based study from Ireland to report the incidence and outcomes for PM. PM is more common than previously reported and survival remains poor. These findings highlight the need for greater clinician awareness and the need to focus on new therapeutic approaches to improve patient outcomes.
Collapse
Affiliation(s)
- J G Solon
- National Centre for Peritoneal Malignancy, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - M O'Neill
- National Centre for Peritoneal Malignancy, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - K H Chang
- National Centre for Peritoneal Malignancy, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - S Deady
- The National Cancer Registry Ireland, Cork, Ireland
| | - R Cahill
- National Centre for Peritoneal Malignancy, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - B Moran
- National Centre for Peritoneal Malignancy, Mater Misericordiae University Hospital, Dublin 7, Ireland; Peritoneal Malignancy Institute, Basingstoke, UK
| | - C Shields
- National Centre for Peritoneal Malignancy, Mater Misericordiae University Hospital, Dublin 7, Ireland
| | - J Mulsow
- National Centre for Peritoneal Malignancy, Mater Misericordiae University Hospital, Dublin 7, Ireland.
| |
Collapse
|
90
|
Tan HL, Chia CS, Tan GHC, Choo SP, Tai DWM, Chua CWL, Ng MCH, Soo KC, Teo MCC. Gastric peritoneal carcinomatosis - a retrospective review. World J Gastrointest Oncol 2017; 9:121-128. [PMID: 28344747 PMCID: PMC5348627 DOI: 10.4251/wjgo.v9.i3.121] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 12/13/2016] [Accepted: 01/02/2017] [Indexed: 02/05/2023] Open
Abstract
AIM To characterize patients with gastric peritoneal carcinomatosis (PC) and their typical clinical and treatment course with palliative systemic chemotherapy as the current standard of care. METHODS We performed a retrospective electronic chart review of all patients with gastric adenocarcinoma with PC diagnosed at initial metastatic presentation between January 2010 and December 2014 in a single tertiary referral centre. RESULTS We studied a total of 271 patients with a median age of 63.8 years and median follow-up duration of 5.1 mo. The majority (n = 217, 80.1%) had the peritoneum as the only site of metastasis at initial presentation. Palliative systemic chemotherapy was eventually planned for 175 (64.6%) of our patients at initial presentation, of which 171 were initiated on it. Choice of first-line regime was in accordance with the National Comprehensive Cancer Network Guidelines for Gastric Cancer Treatment. These patients underwent a median of one line of chemotherapy, completing a median of six cycles in total. Chemotherapy disruption due to unplanned hospitalizations occurred in 114 (66.7%), while cessation of chemotherapy occurred in 157 (91.8%), with 42 cessations primarily attributable to PC-related complications. Patients who had initiation of systemic chemotherapy had a significantly better median overall survival than those who did not (10.9 mo vs 1.6 mo, P < 0.001). Of patients who had initiation of systemic chemotherapy, those who experienced any disruptions to chemotherapy due to unplanned hospitalizations had a significantly worse median overall survival compared to those who did not (8.7 mo vs 14.6 mo, P < 0.001). CONCLUSION Gastric PC carries a grim prognosis with a clinical course fraught with disease-related complications which may attenuate any survival benefit which palliative systemic chemotherapy may have to offer. As such, investigational use of regional therapies is warranted and required validation in patients with isolated PC to maximize their survival outcomes in the long run.
Collapse
|
91
|
Kobayashi D, Kodera Y. Intraperitoneal chemotherapy for gastric cancer with peritoneal metastasis. Gastric Cancer 2017; 20:111-121. [PMID: 27803990 DOI: 10.1007/s10120-016-0662-9] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 10/15/2016] [Indexed: 02/07/2023]
Abstract
Peritoneal metastasis is the most frequent pattern of gastric cancer recurrence or metastasis and is a definitive determinant of prognosis. However, an effective means of treating peritoneal disease has not yet been established. Systemic chemotherapy has only a limited effect on peritoneal metastasis, although some progress has been shown in terms of median survival time, especially among patients with a minimal or moderate disease burden. Clinical research related to intraperitoneal administration of anticancer drugs is currently underway. An advantage of intraperitoneal chemotherapy is the ability to achieve high concentrations of anticancer drugs in the peritoneal cavity and the direct exposure of peritoneal deposits and free cancer cells to those drugs. In addition, pharmacokinetic studies with taxanes have shown that these high intraperitoneal drug concentrations are sustained for a considerable length of time, allowing prolonged exposure. As taxanes are the most appropriate drugs for intraperitoneal administration, the development of repeated intraperitoneal chemotherapy using taxanes for gastric cancer peritoneal metastasis-either alone or in combination with systemic chemotherapy-has taken place over the past decade, mostly in Japan. Several phase II trials and a phase III trial have recently demonstrated the efficacy of this therapy, including median survival times of 14.4-24.6 months and one-year overall survival rates of 67-91%. These results may lead to the approval of intraperitoneal taxanes, especially paclitaxel, for official insurance coverage in the near future.
Collapse
Affiliation(s)
- Daisuke Kobayashi
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Yasuhiro Kodera
- Department of Gastroenterological Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
92
|
Kono K, Yong WP, Okayama H, Shabbir A, Momma T, Ohki S, Takenoshita S, So J. Intraperitoneal chemotherapy for gastric cancer with peritoneal disease: experience from Singapore and Japan. Gastric Cancer 2017; 20:122-127. [PMID: 27766496 DOI: 10.1007/s10120-016-0660-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 10/10/2016] [Indexed: 02/07/2023]
Abstract
Among advanced gastric cancer cases, peritoneal dissemination is a life-threatening mode of metastasis, and any strategy to control peritoneal metastasis will significantly improve treatment outcomes. Since intraperitoneal administration of anticancer drugs can induce an extremely high concentration of drugs in the peritoneal cavity, intraperitoneal chemotherapy would appear to be a reasonable and promising strategy to control the peritoneal dissemination. However, it has been reported in the past that intraperitoneal administration of mitomycin C or cisplatin resulted in no significant clinical effects against peritoneal metastasis of gastric cancer. In contrast, intraperitoneal paclitaxel is expected to remain inside the peritoneal cavity due to its large molecular weight and fat solubility, leading to a high concentration of the drug in the peritoneal cavity. In fact, promising results in several phase II clinical trials using intraperitoneal paclitaxel have been reported, including a median survival time of 16.2-24.6 months and a 1-year overall survival rate of 69-78 %. Thereafter, a phase III randomized control study (PHOENIX-GC trial) with intraperitoneal paclitaxel plus systemic S-1 and intravenous paclitaxel in comparison to systemic S-1 plus cisplatin was conducted in Japan. Moreover, a phase II clinical trial of combination chemotherapy of intraperitoneal paclitaxel with systemic capecitabine plus oxaliplatin is currently ongoing in Singapore. In this review, based on clinical experience from Singapore and Japan, the clinical significance of intraperitoneal chemotherapy for gastric cancer with peritoneal disease is discussed.
Collapse
Affiliation(s)
- Koji Kono
- Department of Organ Regulatory Surgery, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan.
- Department of Surgery, National University of Singapore, Singapore, Singapore.
- Department of Advanced Cancer Immunotherapy, Fukushima Medical University, Fukushima, Japan.
| | - Wei-Peng Yong
- Department of Hematology-Oncology, National University of Singapore, Singapore, Singapore
| | - Hirokazu Okayama
- Department of Organ Regulatory Surgery, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Asim Shabbir
- Department of Surgery, National University of Singapore, Singapore, Singapore
| | - Tomoyuki Momma
- Department of Organ Regulatory Surgery, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Shinji Ohki
- Department of Organ Regulatory Surgery, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Seiichi Takenoshita
- Department of Organ Regulatory Surgery, Fukushima Medical University, 1 Hikarigaoka, Fukushima, 960-1295, Japan
| | - Jimmy So
- Department of Surgery, National University of Singapore, Singapore, Singapore
| |
Collapse
|
93
|
Madani A, Thomassen I, van Gestel YRBM, van der Bilt JDW, Haak HR, de Hingh IHJT, Lemmens VEPP. Peritoneal Metastases from Gastroenteropancreatic Neuroendocrine Tumors: Incidence, Risk Factors and Prognosis. Ann Surg Oncol 2017; 24:2199-2205. [DOI: 10.1245/s10434-016-5734-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Indexed: 12/27/2022]
|
94
|
Abstract
OBJECTIVE The purpose of this article is to discuss the role of the radiologist in the treatment of peritoneal cancer, with focus placed on advanced treatment options and selection of patients with resectable disease for whom complete cytoreduction can be achieved. CONCLUSION Peritoneal cancers traditionally have been associated with significant morbidity and universal mortality; however, the management of such cancers has evolved substantially. Advanced treatment options, including cytoreductive surgery and intraperitoneal chemotherapy, are associated with significantly improved long-term patient survival. To ensure that patients benefit from aggressive multimodality treatments, the radiologist plays a pivotal role in the multidisciplinary team to ensure careful patient selection, identifying individuals with resectable disease for whom complete cytoreduction can be achieved.
Collapse
|
95
|
Ellison LM, Man Y, Stojadinovic A, Xin H, Avital I. Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in treatment of gastric cancer with peritoneal carcinomatosis. Chin J Cancer Res 2017; 29:86-92. [PMID: 28373757 PMCID: PMC5348479 DOI: 10.21147/j.issn.1000-9604.2017.01.10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Although gastric cancer with peritoneal carcinomatosis is associated with poor prognosis and is generally treated with palliative systemic therapy, recent studies have shown that cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) may prove to be an efficacious treatment option. In addition to reviewing the natural history of gastric cancer with peritoneal carcinomatosis, this mini-review examines literature on the efficacy of CRS and HIPEC as compared to chemotherapy and surgical options. Both randomized and non-randomized studies were summarized with the emphasis focused on overall survival. In summary, CRS and HIPEC are indeed a promising treatment option for gastric cancer with peritoneal carcinomatosis and large randomized clinical trials are warranted.
Collapse
Affiliation(s)
- Lynne M Ellison
- Bon Secours Cancer Institute, Bon Secours Health System, Richmond, VA 23226, USA
| | - Yangao Man
- Bon Secours Cancer Institute, Bon Secours Health System, Richmond, VA 23226, USA
| | | | - Hongwu Xin
- Laboratory of Oncology, the First People's Hospital of Jingzhou City, the First Hospital and Clinical Medical School of Yangtze University, Jingzhou 434008, China; Laboratory of Oncology, Center for Molecular Medicine, Medical School, Yangtze University, Jingzhou 434023, China
| | - Itzhak Avital
- Bon Secours Cancer Institute, Bon Secours Health System, Richmond, VA 23226, USA
| |
Collapse
|
96
|
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. [PMID: 27678344 PMCID: PMC5039790 DOI: 10.1186/s12957-016-1008-0] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Accepted: 09/17/2016] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a novel drug delivery system able to induce regression of peritoneal metastasis (PM) in the salvage situation. The aim of this study was to determine the clinical characteristics, tumor histology, and extent of disease of the patients having undergone cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) after "neoadjuvant" PIPAC. METHODS This study was performed at a single institution, tertiary center. In a prospective registry, retrospective analysis was done. PIPAC indication was restricted to patients in the salvage situation who were not eligible for cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC). RESULTS Nine-hundred sixty-one PIPAC sessions were successfully performed in 406 patients: 21 patients (5.2 %) were scheduled for CRS and HIPEC. Twelve of these patients had a low PCI (mean 5.8 ± 5.6). The remaining nine patients showed an advanced peritoneal disease (mean PCI 14.3 ± 5.3) at initial laparoscopy. After repeated PIPAC (mean number of cycles 3.5 ± 0.9), radiological tumor regression was observed in 7/9 patients and major histological regression was observed in 8/9 patients, so that secondary CRS and HIPEC became possible. CONCLUSIONS PIPAC might be used as a neoadjuvant therapy before CRS and HIPEC in order to improve the outcome of CRS and HIPEC, to select patients with chemosensitive, biologically favorable tumors, to extent the indications of CRS and HIPEC in the presence of diffuse small bowel involvement, and to reduce the extent of cytoreductive surgery.
Collapse
Affiliation(s)
- Ramy Girshally
- Therapy center for peritoneal diseases, Elisabethengruppe, Herne, Germany
- Ruhr-University Bochum, Bochum, Germany
| | - Cedric Demtröder
- Therapy center for peritoneal diseases, Elisabethengruppe, Herne, Germany
- Ruhr-University Bochum, Bochum, Germany
| | - Nurettin Albayrak
- Therapy center for peritoneal diseases, Elisabethengruppe, Herne, Germany
| | - Jürgen Zieren
- Therapy center for peritoneal diseases, Elisabethengruppe, Herne, Germany
- Ruhr-University Bochum, Bochum, Germany
| | - Clemens Tempfer
- Therapy center for peritoneal diseases, Elisabethengruppe, Herne, Germany
- Ruhr-University Bochum, Bochum, Germany
| | - Marc A. Reymond
- Department of Surgery, University of Tübingen, Hoppe-Seyler Str. 3, T2076 Tübingen, Germany
| |
Collapse
|
97
|
Khomyakov V, Ryabov A, Ivanov A, Bolotina L, Utkina A, Volchenko N, Kaprin A. 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. [PMID: 30911619 PMCID: PMC6386494 DOI: 10.1515/pp-2016-0017] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 09/02/2016] [Indexed: 01/11/2023] Open
Abstract
Background: Peritoneal metastasis (PM) develop in more than 50 % of gastric cancer (GC) patients. Median survival without treatment is not more than 3-7 months, and 8-12 months after modern combination chemotherapy. Innovative therapeutic approaches are urgently needed. Methods: Phase-2, open label prospective clinical trial assessing safety and efficacy of bidirectional chemotherapy for treating peritoneal metastasis of gastric cancer (PMGC). Treatment protocol included initial staging laparoscopy or laparotomy, 3-4 courses of systemic chemotherapy (XELOX) followed by Pressurized IntraPeritoneal Aerosol Chemotherapy (PIPAC) procedures every 6 weeks until progression of disease or death. Primary endpoints were overall survival and histological peritoneal regression grading score after rebiopsy. Results: 31 patients were included (9 men, 22 women, mean age 52 years), 24 with synchronous PM at diagnosis, 7 with metachronous PM after previous chemotherapy. Mean PCI was 13.8 (min-max 6-34). XELOX was administered in all patients and combined with 56 PIPAC procedures. Complete and partial pathological response was found in 60 % of the 15 patients eligible for tumor response assessment (4 and 5 patients, respectively). Median survival was 13 months. Conclusions: Bidirectional chemotherapy combining XELOX with PIPAC with cisplatin and doxororubicin is well tolerated, can induce objective tumor regression and is associated with a promising survival in PMGC.
Collapse
Affiliation(s)
- Vladimir Khomyakov
- Moscow Research Oncological Institute n.a. P.A. Herzen, Thoracoabdominal, 2-nd Botkinsky proesd, 3, Moscow 125284, Russian Federation
| | - Andrey Ryabov
- Moscow Research Oncological Institute n.a. P.A. Herzen, Thoracoabdominal, Moscow, Russian Federation
| | - Andrey Ivanov
- Moscow Research Oncological Institute n.a. P.A. Herzen, Thoracoabdominal, Moscow, Russian Federation
| | - Larisa Bolotina
- Moscow Research Oncological Institute n.a. P.A. Herzen, Chemotherapy
| | - Anna Utkina
- Moscow Research Oncological Institute n.a. P.A. Herzen, Thoracoabdominal, Moscow, Russian Federation
| | | | - Andrey Kaprin
- Moscow Research Oncological Institute n.a. P.A. Herzen, Director General
| |
Collapse
|
98
|
Wu Z, Li Z, Ji J. Morbidity and mortality of cytoreductive surgery with hyperthermic intraperitoneal chemotherapy in advanced gastric cancer. Transl Gastroenterol Hepatol 2016; 1:63. [PMID: 28138629 DOI: 10.21037/tgh.2016.07.03] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 07/14/2016] [Indexed: 12/29/2022] Open
Abstract
Gastric cancer (GC) is one of the leading cancer causes of death worldwide with high incidence of mortality. With limited available data from the previous literature, cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS-HIPEC) seems to provide substantial survival benefits to the advanced GC patients, especially in those with peritoneal carcinomatosis (PC). However, it is also considered as a high morbid approach. This review summarizes the current evidence regarding the perioperative safety of CRS-HIPEC in advanced GC patients. According to the currently available evidence, CRS-HIPEC causes surgery-related morbidity including abscess, fistula, and anastomotic leak, and chemotherapy-related morbidities such as leucopenia, anemia, thrombopenia, and heart, liver or renal toxicity. The incidence of the morbidity and mortality approximate 20% and 4.8% respectively, which are comparable to a major gastrointestinal surgery. Repeated evidence demonstrates that incidence of morbidity or mortality is significantly influenced by the institutional experience. Centers undertaking this treatment strategy must aim to minimize morbidity and mortality by learning from the experienced units and carefully selecting candidate patients. Patients with advanced age or greater disease burden seem to be at greater risks and thus application of CRS-HIPEC on them must be based on an extensive evaluation and multi-disciplinary team (MDT) discussion.
Collapse
Affiliation(s)
- Zhouqiao Wu
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Ziyu Li
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jiafu Ji
- Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
99
|
Cytoreductive surgery plus hyperthermic intraperitoneal chemotherapy with lobaplatin and docetaxel to treat synchronous peritoneal carcinomatosis from gastric cancer: Results from a Chinese center. Eur J Surg Oncol 2016; 42:1024-34. [DOI: 10.1016/j.ejso.2016.04.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 04/05/2016] [Accepted: 04/18/2016] [Indexed: 12/29/2022] Open
|
100
|
Tu Y, Tian Y, Fang Z, Ruan Q, Tang H, Zhang X, Wu Y, Ding Y, Cui S. Cytoreductive surgery combined with hyperthermic intraperitoneal chemoperfusion for the treatment of gastric cancer: A single-centre retrospective study. Int J Hyperthermia 2016; 32:587-94. [PMID: 27362668 DOI: 10.1080/02656736.2016.1190987] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AIM Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemoperfusion (HIPEC) is the treatment regime most likely to achieve prolonged survival in patients with peritoneal carcinomatosis from gastroenteric cancer. To date, few publications have focused on the treatment of patients with gastric cancer alone. Several controversies remain unsolved, including the safety and effectiveness of the CRS-HIPEC combination regime, particularly in cases where HIPEC is used as adjuvant treatment after CRS. Therefore, in the current study, we aimed to evaluate the safety and effectiveness of CRS combined with HIPEC in patients with gastric cancer. METHOD Data from 231 patients with a median age of 55.1 years treated with the CRS-HIPEC combination regime between January 2009 and December 2014 were retrospectively reviewed. All patients underwent the combination therapy (mean of 2.4 cycles per patient, range, 1 to 4 cycles). RESULTS Median overall survival was 37.0 months, with 1-, 2- and 3-year survival rates recorded as 83.4%, 68.5%, and 38.7%, respectively. The serum levels of carcinoembryonic antigen (CEA) and carbohydrate antigen 199 (CA199) were significantly decreased after combination therapy in the completeness of cytoreduction (CCR)-0 and CCR-1 groups, while no significant changes observed in marker levels were observed in the CC ≥2 group. The post-operative morbidity and mortality rates were 6.9% and 0.9%, respectively. Multivariate analysis revealed low TNM tumour stage, ascites condition and CCR score as independent predictors for better survival. CONCLUSION In view of the acceptable morbidity and mortality rates we propose that CRS combined with HIPEC presents an effective and safe treatment modality for patients with gastric cancer, especially in cases where optimal cytoreduction is achieved before the HIPEC procedure.
Collapse
Affiliation(s)
- Yinuo Tu
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| | - Yunhong Tian
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| | - Zhiyuan Fang
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| | - Qiang Ruan
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| | - Hongsheng Tang
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| | - Xiangliang Zhang
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| | - Yinbing Wu
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| | - Yan Ding
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| | - Shuzhong Cui
- a Cancer Centre of Guangzhou Medical University , Guangzhou , Guangdong Province , China
| |
Collapse
|