1
|
Lewis KA, Diggs LP, Badgwell BD. Educational Review: Updates on Therapeutic Strategies for Gastric Cancer with Peritoneal Metastasis. Ann Surg Oncol 2025:10.1245/s10434-025-17069-3. [PMID: 40016614 DOI: 10.1245/s10434-025-17069-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 02/09/2025] [Indexed: 03/01/2025]
Abstract
Gastric cancer (GC) commonly presents in advanced stages with metastatic spread to the peritoneal cavity, and outcomes associated with gastric cancer with peritoneal metastasis (GCPM) continue to carry a dismal prognosis. Persistent challenges in the detection of peritoneal metastasis (PM) have resulted in a relative paucity of high-quality data to inform management decisions. Several consensus groups have published recommendations to guide management, including most recently the National Comprehensive Cancer Network guidelines, which now include cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) as a potential treatment modality in select patients with GCPM. Multiple clinical trials have investigated the use of CRS/HIPEC and other peritoneal-directed therapies, such as intraperitoneal chemotherapy (IPC) and pressurized intraperitoneal aerosolized chemotherapy (PIPAC). As high-volume centers work to incorporate such therapies into their practice, ongoing clinical trials are aimed at understanding their efficacy. Recent findings have improved understanding of the mechanisms and pathophysiology underlying GCPM while the discovery of novel targets offers potential for drug development and therapeutic strategies to overcome treatment resistance. This review highlights recent advancements and addresses the persistent challenges in managing GCPM while also offering a comprehensive summary of current guidelines and treatment strategies.
Collapse
Affiliation(s)
- Kever A Lewis
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Laurence P Diggs
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian D Badgwell
- Division of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Gingrich A, Manguso N, Zuckerman R. Treatment of Gastric Cancer Carcinomatosis. Surg Clin North Am 2025; 105:95-107. [PMID: 39523079 DOI: 10.1016/j.suc.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Patients with gastric cancer peritoneal metastases (GCPM) have Stage IV disease. Systemic therapy is a crucial aspect of their care. Patients with GCPM should have their tumors tested for HER2 and PD-L1 expression and microsatellite instability for potential targeted therapies. If patients with synchronous GCPM have stable disease following neoadjuvant therapy, surgical intervention can be considered. Patients with positive cytology or low-volume peritoneal disease (peritoneal carcinomatosis index [PCI] < 7) may "convert" to negative cytology or resolution of peritoneal metastases following intraperitoneal therapy and may be candidates for subsequent gastrectomy.
Collapse
Affiliation(s)
- Alicia Gingrich
- Division of Surgery, Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX 77025, USA
| | - Nicholas Manguso
- Division of Surgical Oncology, Department of Surgery, University of Nevada Reno/Renown Integrated Health System, 1500 East 2nd Street, Suite 300, Reno, NV 89502, USA
| | - Randall Zuckerman
- Division of Surgical Oncology, Department of Surgery, University of Nevada Reno/Renown Integrated Health System, 1500 East 2nd Street, Suite 300, Reno, NV 89502, USA.
| |
Collapse
|
3
|
Canbay Torun B, Sobutay E, Eren Akbulut O, Saglam S, Yilmaz S, Yonemura Y, Canbay E. ASO Author Reflections: Important Predictive Factors for Prognosis in Patients with Peritoneal Metastasis of Gastric Cancer. Ann Surg Oncol 2024; 31:6028-6029. [PMID: 38862835 DOI: 10.1245/s10434-024-15613-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 05/28/2024] [Indexed: 06/13/2024]
Affiliation(s)
- Bahar Canbay Torun
- Department of General Surgery, Haseki Education and Research Hospital, Istanbul, Turkey
| | - Erman Sobutay
- Department of General Surgery, Koç Foundation American Hospital, Istanbul, Turkey
| | - Ozge Eren Akbulut
- Department of Anesthesiology, ICU, Koç Foundation American Hospital, Istanbul, Turkey
| | - Sezer Saglam
- Department of Medical Oncology, Demiroglu Bilim University, Istanbul, Turkey
| | - Serpil Yilmaz
- Department of Pathology, Koç Foundation American Hospital, Istanbul, Turkey
| | - Yutaka Yonemura
- Department of Regional Cancer Therapy, Peritoneal Surface Malignancy Center, Kishiwada, Tokushukai Hospital, Kishiwada, Japan
| | - Emel Canbay
- NPO for Peritoneal Surface Malignancies Program, Istanbul, Turkey.
| |
Collapse
|
4
|
Read MD, Drake J, Hashemipour G, Powers BD, Mehta R, Sinnamon A, Pimiento JM, Dineen SP. Initial Experience Using Laparoscopic HIPEC for Gastric Cancer with Peritoneal Metastasis: Safety and Outcomes. Ann Surg Oncol 2024; 31:3750-3757. [PMID: 38430428 DOI: 10.1245/s10434-024-15102-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/12/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND Peritoneal metastases (PM) develop in approximately 20% of patients with gastric cancer (GC). For selected patients, treatment of PM with cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) has shown promising results. This report aims to describe the safety and perioperative outcomes of laparoscopic HIPEC for GC/PM. METHODS This retrospective cohort study evaluated patients who had GC and PM treated with laparoscopic HIPEC (2018-2022). The HIPEC involved cisplatin and mitomycin C (MMC) or MMC alone. The primary end point was perioperative safety. RESULTS The 22 patients in this study underwent 27 procedures. The mean age was 58 ± 13 years. All the patients were Eastern Cooperative Oncology Group (ECOG) 0 or 1 (55 and 45%, respectively). Five patients underwent a second laparoscopic HIPEC, with a median of 126 days (interquartile range [IQR], 117-166 days) between procedures. The median peritoneal carcinomatosis index (PCI) was 4 (IQR, 2-9), and the median hospital stay was 2 days (IQR, 1-3 days). No 30-day readmissions or complications occurred. Eight patients (36%) underwent gastrectomy (CRS ± HIPEC). After an average follow-up period of 11 months, 7 (32%) of the 22 patients were alive. The median overall survival was 11 months (IQR, 195-739 days) from the initial procedure and 19.3 months (IQR, 431-1204 days) from the diagnosis. CONCLUSIONS Laparoscopic HIPEC appears to be safe with minimal perioperative complications. Approximately one third of the patients undergoing initial laparoscopic HIPEC ultimately proceeded to cytoreduction and gastrectomy. Preliminary survival data from this highly selected cohort suggest that the addition of laparoscopic HIPEC to systemic chemotherapy does not compromise other treatment options. These initial results suggest that laparoscopic HIPEC may offer benefit to patients with GC and PM and aid in the selection of patients who may benefit from curative-intent resection.
Collapse
Affiliation(s)
- Meagan D Read
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Surgery, Morsani College of Medicine, Tampa, FL, USA
| | - Justin Drake
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Ghazal Hashemipour
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Benjamin D Powers
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Oncologic Sciences, Morsani College of Medicine, Tampa, FL, USA
- Department of Health Outcomes and Behavior, Moffitt Cancer Center, Tampa, FL, USA
| | - Rutika Mehta
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Andrew Sinnamon
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Oncologic Sciences, Morsani College of Medicine, Tampa, FL, USA
| | - Jose M Pimiento
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA
- Department of Oncologic Sciences, Morsani College of Medicine, Tampa, FL, USA
| | - Sean P Dineen
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA.
- Department of Oncologic Sciences, Morsani College of Medicine, Tampa, FL, USA.
| |
Collapse
|
5
|
Chen C, Jung A, Yang A, Monroy I, Zhang Z, Chaurasiya S, Deshpande S, Priceman S, Fong Y, Park AK, Woo Y. Chimeric Antigen Receptor-T Cell and Oncolytic Viral Therapies for Gastric Cancer and Peritoneal Carcinomatosis of Gastric Origin: Path to Improving Combination Strategies. Cancers (Basel) 2023; 15:5661. [PMID: 38067366 PMCID: PMC10705752 DOI: 10.3390/cancers15235661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 02/12/2024] Open
Abstract
Precision immune oncology capitalizes on identifying and targeting tumor-specific antigens to enhance anti-tumor immunity and improve the treatment outcomes of solid tumors. Gastric cancer (GC) is a molecularly heterogeneous disease where monoclonal antibodies against human epidermal growth factor receptor 2 (HER2), vascular endothelial growth factor (VEGF), and programmed cell death 1 (PD-1) combined with systemic chemotherapy have improved survival in patients with unresectable or metastatic GC. However, intratumoral molecular heterogeneity, variable molecular target expression, and loss of target expression have limited antibody use and the durability of response. Often immunogenically "cold" and diffusely spread throughout the peritoneum, GC peritoneal carcinomatosis (PC) is a particularly challenging, treatment-refractory entity for current systemic strategies. More adaptable immunotherapeutic approaches, such as oncolytic viruses (OVs) and chimeric antigen receptor (CAR) T cells, have emerged as promising GC and GCPC treatments that circumvent these challenges. In this study, we provide an up-to-date review of the pre-clinical and clinical efficacy of CAR T cell therapy for key primary antigen targets and provide a translational overview of the types, modifications, and mechanisms for OVs used against GC and GCPC. Finally, we present a novel, summary-based discussion on the potential synergistic interplay between OVs and CAR T cells to treat GCPC.
Collapse
Affiliation(s)
- Courtney Chen
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Audrey Jung
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Annie Yang
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Isabel Monroy
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
| | - Zhifang Zhang
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Shyambabu Chaurasiya
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Supriya Deshpande
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Saul Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yuman Fong
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
| | - Anthony K. Park
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA; (I.M.); (S.P.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yanghee Woo
- Department of Surgery, City of Hope, Duarte, CA 91010, USA; (C.C.); (A.J.); (A.Y.); (Z.Z.); (S.C.); (S.D.); (Y.F.)
- Cancer Immunotherapeutics Program, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
6
|
Dai W, Chen Y, Xue Y, Wan M, Mao C, Zhang K. Progress in the Treatment of Peritoneal Metastatic Cancer and the Application of Therapeutic Nanoagents. ACS APPLIED BIO MATERIALS 2023; 6:4518-4548. [PMID: 37916787 DOI: 10.1021/acsabm.3c00662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2023]
Abstract
Peritoneal metastatic cancer is a cancer caused by the direct growth of cancer cells from the primary site through the bloodstream, lymph, or peritoneum, which is a difficult part of current clinical treatment. In the abdominal cavity of patients with metastatic peritoneal cancer, there are usually nodules of various sizes and malignant ascites. Among them, nodules of different sizes can obstruct intestinal movement and form intestinal obstruction, while malignant ascites can cause abdominal distension and discomfort, and even cause patients to have difficulty in breathing. The pathology and physiology of peritoneal metastatic cancer are complex and not fully understood. The main hypothesis is "seed" and "soil"; i.e., cells from the primary tumor are shed and implanted in the peritoneal cavity (peritoneal metastasis). In the last two decades, the main treatment modalities used clinically are cytoreductive surgery (CRS), systemic chemotherapy, intraperitoneal chemotherapy, and combined treatment, all of which help to improve patient survival and quality of life (QOL). However, the small-molecule chemotherapeutic drugs used clinically still have problems such as rapid drug metabolism and systemic toxicity. With the rapid development of nanotechnology in recent years, therapeutic nanoagents for the treatment of peritoneal metastatic cancer have been gradually developed, which has improved the therapeutic effect and reduced the systemic toxicity of small-molecule chemotherapeutic drugs to a certain extent. In addition, nanomaterials have been developed not only as therapeutic agents but also as imaging agents to guide peritoneal tumor CRS. In this review, we describe the etiology and pathological features of peritoneal metastatic cancer, discuss in detail the clinical treatments that have been used for peritoneal metastatic cancer, and analyze the advantages and disadvantages of the different clinical treatments and the QOL of the treated patients, followed by a discussion focusing on the progress, obstacles, and challenges in the use of therapeutic nanoagents in peritoneal metastatic cancer. Finally, therapeutic nanoagents and therapeutic tools that may be used in the future for the treatment of peritoneal metastatic cancer are prospected.
Collapse
Affiliation(s)
- Wenjun Dai
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Yidan Chen
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Yunxin Xue
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Mimi Wan
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ke Zhang
- Department of Radiation Oncology, Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| |
Collapse
|
7
|
Green BL, Blumenthaler AN, Gamble LA, McDonald JD, Robinson K, Connolly M, Epstein M, Hernandez JM, Blakely AM, Badgwell BD, Davis JL. Cytoreduction and HIPEC for Gastric Carcinomatosis: Multi-institutional Analysis of Two Phase II Clinical Trials. Ann Surg Oncol 2023; 30:1852-1860. [PMID: 36348206 PMCID: PMC10683488 DOI: 10.1245/s10434-022-12761-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 10/24/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION There are no approved locoregional therapies for peritoneal carcinomatosis from gastric adenocarcinoma (GA). Cytoreductive surgery with hyperthermic intraperitoneal chemotherapy (CRS-HIPEC) represents a potential treatment for advanced GA with isolated peritoneal metastasis. PATIENTS AND METHODS Two separate single-institution phase II, single-arm studies evaluating CRS-HIPEC using cisplatin with mitomycin C (NIH: NCT03092518, MDACC: NCT02891447) in patients with GA and confirmed peritoneal metastasis were analyzed. The primary endpoint of each trial was overall survival (OS). Clinical, pathologic, and treatment variables were analyzed for association with outcomes. RESULTS Over 4 years, 41 patients with peritoneal carcinomatosis from GA underwent CRS-HIPEC. All patients had synchronous peritoneal metastasis and received systemic chemotherapy as front-line therapy. A total of 23 patients also received laparoscopic HIPEC prior to open CRS-HIPEC. The majority (63%, n = 26) were male, and median PCI score at CRS-HIPEC was 2. Median OS was 24.9 months from diagnosis and 14.4 months from CRS-HIPEC. Three-year OS was 25% from diagnosis and 22% from CRS-HIPEC. Median RFS was 7.4 months. The rate of 30-day Clavien-Dindo grade ≥ 3 complications was 32%; specifically, the rate of anastomotic leak was 22%. Multivariable analysis identified the number of pathologically positive lymph nodes as an independent predictor of postoperative OS. CONCLUSIONS In patients with gastric adenocarcinoma and isolated peritoneal metastasis treated with CRS-HIPEC, 3-year OS was 22% from CRS-HIPEC, and complications were common. The number of pathologic lymph node metastases was inversely correlated with overall survival. Further investigation of CRS-HIPEC for GA should include patient selection based on response to systemic chemotherapy or incorporate novel intraperitoneal treatment strategies.
Collapse
Affiliation(s)
- Benjamin L Green
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alisa N Blumenthaler
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lauren A Gamble
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - James D McDonald
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kristen Robinson
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maureen Connolly
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Monica Epstein
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan M Hernandez
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Andrew M Blakely
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Brian D Badgwell
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jeremy L Davis
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
Zhang J, Li L, Yin J, Zhang X, Zheng Y, Feng R. Study on the thermal stability of nab-paclitaxel during hyperthermic intraperitoneal chemotherapy. BMC Pharmacol Toxicol 2023; 24:13. [PMID: 36859304 PMCID: PMC9979565 DOI: 10.1186/s40360-023-00653-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 02/14/2023] [Indexed: 03/03/2023] Open
Abstract
BACKGROUND Albumin-bound paclitaxel (nab-paclitaxel), as a special targeted preparation of paclitaxel, has the advantages of good curative effect and less side effects in anti-tumor therapy. The existence of the plasma-peritoneal barrier and insufficient blood supply make intravenous drugs hard to reach the peritoneum, while hyperthermic intraperitoneal chemotherapy can solve the difficulty. And compared with systemic medications, HIPEC can also give higher concentrations of chemotherapy drugs in the abdominal cavity, while ensuring lower systemic toxicity. However, at present, there is no relevant report on the clinical study of nab-paclitaxel during intraperitoneal hyperthermic chemotherapy, and its stability under special temperature conditions has not been reported either. METHODS In this study, We examined three batches of albumin-bound paclitaxel dissolved in saline at different temperatures (25 °C, 37 °C, 41 °C, 42 °C and 43 °C) for the changes of human serum albumin content, human serum albumin polymer content, related substance content, in-vitro release rate, paclitaxel binding rate and paclitaxel content at different temperatures. RESULTS Our results demonstrated that the indicators including human serum albumin content, human serum albumin polymer content, in-vitro release rate, paclitaxel binding rate and paclitaxel content were stable to the several temperatures, except that Taxane (0.1%) and other individual impurities in the determination of related substance content fluctuated comparatively widely with the change of temperature. In addition, only Taxane (0.1%) and 7-Epitaxol (1%) were detected. CONCLUSIONS Overall, albumin-bound paclitaxel is relatively stable to different temperatures (25 °C, 37 °C, 41 °C, 42 °C and 43 °C). This study will lay a foundation for further studies on the albumin-bound paclitaxel during hyperthermic intraperitoneal chemotherapy.
Collapse
Affiliation(s)
- Jingjing Zhang
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, P. R. China
| | - Luya Li
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, P. R. China
| | - Jintuo Yin
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, P. R. China
| | - Xidong Zhang
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, P. R. China
| | - Ying Zheng
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, P. R. China.
| | - Rui Feng
- Department of Pharmacy, The Fourth Hospital of Hebei Medical University, Shijiazhuang, 050011, P. R. China.
| |
Collapse
|
9
|
Mariani A, Zaanan A, Glehen O, Karoui M. Outcomes of a Phase II Study of Intraperitoneal Paclitaxel Plus Systemic Capecitabine and Oxaliplatin (XELOX) for Gastric Cancer with Peritoneal Metastases. Ann Surg Oncol 2023; 30:816-817. [PMID: 36383333 DOI: 10.1245/s10434-022-12837-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Accepted: 10/28/2022] [Indexed: 11/17/2022]
Affiliation(s)
- Antoine Mariani
- Department of Digestive Surgery, Assistance Publique Hôpitaux de Paris, Georges Pompidou University Hospital, Université Paris Cité, Paris, France.
| | - Aziz Zaanan
- Department of Digestive Oncology, Assistance Publique Hôpitaux de Paris, Georges Pompidou University Hospital, Université Paris Cité, Paris, France
| | - Olivier Glehen
- Surgical Oncology Department, Hôpital Lyon Sud, Hospices Civils de Lyon, Pierre Bénite, France
| | - Mehdi Karoui
- Department of Digestive Surgery, Assistance Publique Hôpitaux de Paris, Georges Pompidou University Hospital, Université Paris Cité, Paris, France
| |
Collapse
|
10
|
Lianos GD, Bali CD, Schizas D, Mitsis M, Galli F, Rausei S. Management of positive peritoneal cytology gastric cancer patients. G Chir 2022; 42:e06. [DOI: 10.1097/ia9.0000000000000005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Prognosis of patients with stage IV gastric cancer remains up to date dramatically poor. A subgroup of these patients has only positive peritoneal cytology in the peritoneal lavage and represents a target issue of the recent ongoing research. The administration of neoadjuvant chemotherapy, either systemic or intraperitoneal or hyperthermic intraoperative peritoneal chemotherapy, with a variety of combinations of drugs has assisted some of these patients to become peritoneal cytology negative, with a significant implication in their prognosis. Staging laparoscopy is widely used to detect occult peritoneal disease and has, therefore, reduced the number of unnecessary laparotomies. However, recent reports point out a low sensitivity of staging laparoscopy and cytological exam in detecting viable cancer cells and focus on the need of more accurate methods of examining the peritoneal fluid by immunohistochemistry or molecular assays. This review focuses on the latest scientific evidence regarding the optimal management of positive peritoneal cytology gastric cancer patients.
Collapse
Affiliation(s)
- Georgios D. Lianos
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | - Christina D. Bali
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | - Dimitrios Schizas
- First Department of Surgery, National and Kapodistrian University of Athens, Laikon General Hospital, Athens, Greece
| | - Michail Mitsis
- Department of Surgery, University Hospital of Ioannina, Ioannina, Greece
| | - Federica Galli
- Department of Surgery, ASST Valle Olona, Gallarate, Italy
| | - Stefano Rausei
- Department of Surgery, ASST Valle Olona, Gallarate, Italy
| |
Collapse
|
11
|
Current Trends in Cytoreductive Surgery (CRS) and Hyperthermic Intraperitoneal Chemotherapy (HIPEC) for Peritoneal Disease from Appendiceal and Colorectal Malignancies. J Clin Med 2022; 11:jcm11102840. [PMID: 35628966 PMCID: PMC9143396 DOI: 10.3390/jcm11102840] [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: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/12/2022] [Indexed: 12/10/2022] Open
Abstract
Peritoneal carcinomatosis (PC) is a poor prognostic factor for all malignancies. This extent of metastatic disease progression remains difficult to treat with systemic therapies due to poor peritoneal vascularization resulting in limited drug delivery and penetration into tissues. Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) are surgical interventions that directly target peritoneal tumors and have improved outcomes for PC resulting from appendiceal and colorectal cancer (CRC). Despite these radical therapies, long-term survival remains infrequent, and recurrence is common. The reasons for these outcomes are multifactorial and signal the need for the continued development of novel therapeutics, techniques, and approaches to improve outcomes for these patients. Here, we review landmark historical studies that serve as the foundation for current recommendations, recent discoveries, clinical trials, active research, and areas of future interest in CRS/HIPEC to treat PC originating from appendiceal and colorectal malignancies.
Collapse
|
12
|
Mazurek M, Szlendak M, Forma A, Baj J, Maciejewski R, Roviello G, Marano L, Roviello F, Polom K, Sitarz R. Hyperthermic Intraperitoneal Chemotherapy in the Management of Gastric Cancer: A Narrative Review. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 19:681. [PMID: 35055500 PMCID: PMC8776178 DOI: 10.3390/ijerph19020681] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/02/2022] [Accepted: 01/04/2022] [Indexed: 02/05/2023]
Abstract
Gastric cancer (GC) patients with peritoneal metastasis tend to achieve poor clinical outcomes. Until recently, the treatment options were limited mainly to either palliative chemotherapy or radiation therapy in exceptional cases. Currently, these patients benefit from multimodal treatment, such as cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC). Despite good overall results, this treatment modality is still widely debated. The following study is designed to assess the papers about the possible application and utility of HIPEC in GC. A search in the PubMed, Web of Science, and Scopus databases was performed to assess the papers devoted to the role of HIPEC in GC treatment; a literature search was performed until March 21st; and, finally, 50 studies with a total number of 3946 patients were analyzed. According to the most recent data, it seems to be reasonable to limit the duration of HIPEC to the shortest effective time. Moreover, the drugs used in HIPEC need to have equal concentrations and the same solvent. Perioperative chemotherapy needs to be reported in detail and, furthermore, the term "morbidity" should be defined more clearly by the authors.
Collapse
Affiliation(s)
- Marek Mazurek
- Department of Surgical Oncology, Voivodship Hospital in Siedlce, 08-110 Siedlce, Poland;
| | - Małgorzata Szlendak
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (M.S.); (J.B.); (R.M.)
- Department of Oncology, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Alicja Forma
- Department of Forensic Medicine, Medical University of Lublin, 20-090 Lublin, Poland;
| | - Jacek Baj
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (M.S.); (J.B.); (R.M.)
| | - Ryszard Maciejewski
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (M.S.); (J.B.); (R.M.)
| | | | - Luigi Marano
- Department of General Surgery and Surgical Oncology, University of Siena, 53100 Siena, Italy; (L.M.); (F.R.)
| | - Franco Roviello
- Department of General Surgery and Surgical Oncology, University of Siena, 53100 Siena, Italy; (L.M.); (F.R.)
| | - Karol Polom
- Department of Surgical Oncology, Medical University of Gdansk, 80-070 Gdansk, Poland;
| | - Robert Sitarz
- Department of Human Anatomy, Medical University of Lublin, 20-090 Lublin, Poland; (M.S.); (J.B.); (R.M.)
- Department of Surgical Oncology, St. John’s Cancer Center, 20-090 Lublin, Poland
| |
Collapse
|
13
|
Wu MF, Wang LJ, Ye YF, Liu CH, Lu HW, Yao TT, Zhang BZ, Chen Q, Li JB, Peng YP, Zhou H, Lin ZQ, Li J. Efficacy of neoadjuvant hyperthermic intraperitoneal chemotherapy in advanced high-grade serous ovarian cancer (the NHIPEC trial): study protocol for a randomised controlled trial. BMJ Open 2021; 11:e046415. [PMID: 34916306 PMCID: PMC8679100 DOI: 10.1136/bmjopen-2020-046415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy (NACT) is an important treatment option for patients with ovarian cancer. Although intravenous NACT can improve optimal resection rates and decrease surgical morbidity and mortality, these advantages do not translate into a survival benefit. Ovarian carcinoma is mainly confined to the peritoneal cavity, which makes it a potential target for hyperthermic intraperitoneal chemotherapy (HIPEC). Our previous study showed that HIPEC could be used in the neoadjuvant setting, which was named neoadjuvant HIPEC (NHIPEC). Since hyperthermia is an excellent chemosensitiser, we hypothesised that the combination of NHIPEC and intravenous NACT could show superior efficacy to intravenous NACT alone. METHODS This study is a single-centre, open-label, randomised (1:1 allocation ratio) phase 2 trial. A total of 80 patients will be randomly assigned into an experimental group (NHIPEC+intravenous NACT) or a control group (intravenous NACT). Patients in the experimental group will receive NHIPEC following laparoscopic evaluation, and four tubes will be placed via the laparoscopic ports, which will be used to administer NHIPEC. Then, perfusion with docetaxel (60-75 mg/m2) will be performed (43°C for 60 min, Day 0) followed by cisplatin (75 mg/m2, Day 1) infusion (43°C for 60 min) 24 hours later. After NHIPEC, two cycles of intravenous NACT will be given. Patients in the control group will receive three cycles of intravenous NACT. The primary endpoint is the proportion of patients who achieve a Chemotherapy Response Score (CRS) of 3 according to the CRS system. The secondary endpoints include progression-free survival, overall survival and the rates of complete resection and NHIPEC-related adverse events. ETHICS APPROVAL AND DISSEMINATION This study was approved by the Ethics Committee of Sun Yat-sen Memorial Hospital (approval number: 2020-ky-050). Results will be submitted to peer-reviewed journals and presented at national and international conferences. TRIAL REGISTRATION NUMBER ChiCTR2000038173.
Collapse
Affiliation(s)
- Miao-Fang Wu
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Li-Juan Wang
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Yan-Fang Ye
- Clinical Research Design Division, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Chang-Hao Liu
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Huai-Wu Lu
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Ting-Ting Yao
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Bing-Zhong Zhang
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Qing Chen
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Ji-Bin Li
- Department of Clinical Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yong-Pai Peng
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Hui Zhou
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Zhong-Qiu Lin
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Jing Li
- Department of Gynecologic Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, China
- Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Laparoscopic Heated Intraperitoneal Chemotherapy in the Treatment of Carcinomatosis of Gastric Adenocarcinoma Origin. J Clin Med 2021; 10:jcm10204757. [PMID: 34682880 PMCID: PMC8539356 DOI: 10.3390/jcm10204757] [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: 09/16/2021] [Revised: 10/09/2021] [Accepted: 10/15/2021] [Indexed: 11/30/2022] Open
Abstract
The use of heated intraperitoneal chemotherapy (HIPEC) in conjunction with cytoreductive surgery has been gaining increasing traction in treating gastric adenocarcinoma with metastasis to the peritoneum in recent years. The addition of laparoscopic HIPEC (LS-HIPEC) to these treatment algorithms has increased the flexibility and adaptability of HIPEC integrating into treatment sequencing, allowing for iterative protocols of LS-HIPEC prior to cytoreduction as neoadjuvant treatment, as well as in the palliation of patients with unresectable disease and uncontrolled ascites. As the use of HIPEC in gastric adenocarcinoma continues to be refined, LS-HIPEC algorithms should continue to be considered and utilized both in curative treatment algorithms as well as in patients in the palliative setting. Given that LS-HIPEC remains a relatively nascent treatment modality, we advocate for its use in the setting of a clinical trial when feasible.
Collapse
|
15
|
Li Z, Wang Y, Ying X, Zhang L, Gao X, Jia Y, Zhang L, Wu A, Su X, Ji J. Prognostic and predictive value of mismatch repair deficiency in gastric and gastroesophageal junction adenocarcinoma patients receiving neoadjuvant or adjuvant chemotherapy. J Surg Oncol 2021; 124:1356-1364. [PMID: 34515995 DOI: 10.1002/jso.26669] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 08/09/2021] [Accepted: 08/29/2021] [Indexed: 12/30/2022]
Abstract
BACKGROUND AND OBJECTIVES Evidence is inconclusive regarding the prognostic significance of deficient DNA mismatch repair (dMMR) in gastric and gastroesophageal junction (GEJ) adenocarcinoma patients receiving chemotherapy. We aim to explore such associations with a large cohort. METHODS We retrospectively identified a consecutive cohort of patients who had histology proven gastric or GEJ adenocarcinoma and received neoadjuvant chemotherapy plus surgery or upfront surgery plus adjuvant chemotherapy. MMR status was assessed by immunohistochemistry staining on surgical specimen. The association of MMR status with tumor regression grade (TRG), overall survival (OS), and disease-free survival (DFS) were analyzed. RESULTS In total, 1568 patients received neoadjuvant or adjuvant chemotherapy, of which 128 (8.2%) had dMMR tumors. No significant difference was found in the frequencies of TRG categories between proficient MMR (pMMR) and dMMR tumors (p = .62). Among patients receiving neoadjuvant chemotherapy, dMMR status was associated with better OS (log-rank p = .044) and DFS (log-rank p = .022) in the univariate analysis; this association became nonsignificant after adjusting for pathologic stages and other prognostic factors. Similar results were found for patients receiving adjuvant chemotherapy. CONCLUSIONS dMMR status was not significantly associated with OS and DFS among gastric and GEJ adenocarcinoma patients with neoadjuvant and adjuvant platinum and fluorouracil-based chemotherapy.
Collapse
Affiliation(s)
- Ziyu Li
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yinkui Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangji Ying
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Li Zhang
- Department of Pathology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangyu Gao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Yongning Jia
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Lianhai Zhang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Aiwen Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiangqian Su
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jiafu Ji
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Surgery, Peking University Cancer Hospital & Institute, Beijing, China
| |
Collapse
|