1
|
Zou JX, Chang MR, Kuznetsov NA, Kee JX, Babak MV, Ang WH. Metal-based immunogenic cell death inducers for cancer immunotherapy. Chem Sci 2025; 16:6160-6187. [PMID: 40160356 PMCID: PMC11949249 DOI: 10.1039/d4sc08495k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Immunogenic cell death (ICD) has attracted enormous attention over the past decade due to its unique characteristics in cancer cell death and its role in activating innate and adaptive immune responses against tumours. Many efforts have been dedicated to screening, identifying and discovering ICD inducers, resulting in the validation of several based on metal complexes. In this review, we provide a comprehensive summary of current metal-based ICD inducers, their molecular mechanisms for triggering ICD initiation and subsequent protective antitumour immune responses, along with considerations for validating ICD both in vitro and in vivo. We also aim to offer insights into the future development of metal complexes with enhanced ICD-inducing properties and their applications in potentiating antitumour immunity.
Collapse
Affiliation(s)
- Jiao Xia Zou
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Meng Rui Chang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Nikita A Kuznetsov
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Jia Xuan Kee
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
| | - Maria V Babak
- Drug Discovery Lab, Department of Chemistry, City University of Hong Kong 83 Tat Chee Avenue Hong Kong SAR 999077 People's Republic of China
| | - Wee Han Ang
- Department of Chemistry, National University of Singapore 4 Science Drive 2 Singapore 117544 Singapore
- NUS Graduate School - Integrative Science and Engineering Programme (ISEP), National University of Singapore 21 Lower Kent Ridge Rd Singapore 119077 Singapore
| |
Collapse
|
2
|
Kim JH, Park E, Park SY, Lim MC. A randomized, multicenter, open-label phase III trial of cytoreductive surgery and hyperthermic intraperitoneal chemotherapy in platinum-resistant recurrent ovarian cancer: a rationale and study design of the KOV-HIPEC-02 trial. Int J Gynecol Cancer 2025; 35:101630. [PMID: 39955184 DOI: 10.1016/j.ijgc.2025.101630] [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] [Received: 10/23/2024] [Revised: 01/01/2025] [Accepted: 01/05/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND This study aimed to identify the survival benefits of hyperthermic intraperitoneal chemotherapy (HIPEC) after cytoreductive surgery for platinum-resistant recurrent ovarian cancer. PRIMARY OBJECTIVES The primary objective is to evaluate the efficacy of cytoreductive surgery and HIPEC on progression-free survival in platinum-resistant recurrent ovarian cancer. STUDY HYPOTHESIS The implementation of cytoreductive surgery and HIPEC combined with doxorubicin and mitomycin C may extend progression-free survival with manageable safety in patients with platinum-resistant recurrent ovarian cancer. TRIAL DESIGN This trial (KOV-HIPEC-02) is a multicenter, open-label, 1:1 randomized, phase III trial that enrolled 140 patients with platinum-resistant recurrent ovarian cancer. The patients will receive or will not receive cytoreductive surgery and HIPEC with doxorubicin 35 mg/m2 and mitomycin 15 mg/m2. Enrolled patients will receive non-platinum compound systemic chemotherapy until disease progression. MAJOR INCLUSION/EXCLUSION CRITERIA Patients with recurrent epithelial ovarian, fallopian tube, and primary peritoneal cancer, who demonstrate resistance or refractory to platinum-based chemotherapy, are eligible for this trial. Patients exhibiting resectable intraperitoneal disease, as determined by clinical history, recent imaging findings, and laparoscopic assessment, will be enrolled. Furthermore, they must have an Eastern Cooperative Oncology Group Performance Status of 0 to 2, a life expectancy of at least 3 months, adequate organ function, and must provide informed consent to participate in the study. PRIMARY ENDPOINT Progression-free survival defined as the time from random assignment in a clinical trial to disease progression or death from any cause. SAMPLE SIZE Assuming that the enrollment period is 3 years and the follow-up period is 2 years, 140 patients will be randomized in this study design (70 patients in the HIPEC group and 70 patients in the control group). ESTIMATED DATES FOR COMPLETING ACCRUAL AND PRESENTING RESULTS The expected date of interim analysis of primary end point is early 2025, and the patient enrollment is expected to be completed in 2025. TRIAL REGISTRATION The trial is registered at ClinicalTrials.gov (NCT05316181).
Collapse
Affiliation(s)
- Ji Hyun Kim
- National Cancer Center, Gynecologic Cancer Center, Goyang, South Korea
| | - Eunyoung Park
- National Cancer Center Goyang, Biostatistics Collaboration Team, Research Core Center, South Korea
| | - Sang-Yoon Park
- National Cancer Center, Gynecologic Cancer Center, Goyang, South Korea
| | - Myong Cheol Lim
- National Cancer Center, Gynecologic Cancer Center, Goyang, South Korea; National Cancer Center, Center for Gynecologic Cancer and Center for Clinical Trials, Goyang, South Korea; National Cancer Center, Research Institute, Division of Rare and Refractory Cancer, Rare & Pediatric Cancer Branch and Immuno-oncology Branch, Goyang, South Korea.
| |
Collapse
|
3
|
Fujiwara K, Nagao S, Tan D, Hasegawa K. Intraperitoneal chemotherapy is now back for ovarian cancer. Int J Clin Oncol 2025; 30:427-433. [PMID: 39875651 PMCID: PMC11842472 DOI: 10.1007/s10147-025-02700-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 01/06/2025] [Indexed: 01/30/2025]
Abstract
The Intraperitoneal Carboplatin for Ovarian Cancer (iPocc) trial demonstrated that intraperitoneal (IP) administration of carboplatin is more effective than intravenous (IV) administration for advanced ovarian cancer, especially in cases with large residual tumors, challenging previous assumptions that IP chemotherapy is only beneficial for small residual tumors. Additionally, the iPocc trial showed that IP chemotherapy has a comparable safety profile to IV chemotherapy, with the exception of port-related toxicities. This review summarizes the principles, development, and significance of IP chemotherapy and discusses its future potential in light of recent studies. Notably, the iPocc trial, conducted under Japan's new clinical trial regulations, achieving regulatory approval based on investigator-initiated results. The iPocc regimen offers a viable treatment option for patients with advanced ovarian cancer (stages II-IV). However, bevacizumab is recommended for later-line treatments rather than combining it with IP chemotherapy until further trials support such combinations. Future studies are needed to identify biomarkers that predict response to the iPocc regimen. The trial's success underscores the dedication of patients and families who contributed to this groundbreaking research.
Collapse
Affiliation(s)
- Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan.
| | - Shoji Nagao
- Department of Obstetrics and Gynecology, Faculty of Medicine, Okayama University, Okayama, Japan
| | - David Tan
- Department of Haematology-Oncology, National University Cancer Institute, Singapore (NCIS), National University Hospital (NUH), Singapore, Singapore
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| |
Collapse
|
4
|
Zeng X, Jin X, Leng J, Zhang S, Wang Y, Chen J, Zhang S, Teng L, Hu Z, Zhou S, Zeng Z, Long J. High-dose radiation induces dendritic cells maturation by promoting immunogenic cell death in nasopharyngeal carcinoma. Front Immunol 2025; 16:1554018. [PMID: 40040692 PMCID: PMC11876370 DOI: 10.3389/fimmu.2025.1554018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Aim and background Due to the radiosensitivity and deep anatomical location of nasopharyngeal carcinoma (NPC), radiotherapy serves as the cornerstone of standardized treatment for this malignancy. Beyond its cytotoxic effects, radiotherapy can serve as an immunological adjuvant by inducing immunogenic cell death (ICD). Dendritic cells (DCs), as potent antigen-presenting cells, play a critical role in tumor immunotherapy, but their exact role in the ICD process of NPC remains unclear. The effects of high-dose radiation (≥2 Gy) on DCs and the type of immune response it elicits in NPC have not been fully elucidated. Methods An in vitro study was conducted to assess whether ICD of NPC 5-8F cells induced by high-dose radiation could regulate the immune response of DCs. Specifically, the maturation and antigen-presenting capacity of DCs were evaluated following co-culture with NPC cells exposed to high-dose radiation. Results High-dose radiation was found to induce ICD in NPC 5-8F cells, as evidenced by increased pro-inflammatory factor levels and reduced anti-inflammatory factor levels in the cell culture supernatant. Co-culture with NPC cells exposed to high-dose radiation for 15 minutes significantly enhanced the expression of surface molecules on DCs, promoting their immune sensitization. Conclusion High-dose radiation-induced apoptosis of NPC 5-8F cells is a form of ICD, which plays an important role in regulating DC immune function. These findings provide insight into the immunomodulatory effects of radiotherapy in NPC and its potential to enhance tumor immunotherapy through DC activation.
Collapse
Affiliation(s)
- Xianlin Zeng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Xianhuai Jin
- Department of Oncology, Guiyang Public Health Clinical Center, Guiyang, Guizhou, China
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Ji Leng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shuai Zhang
- Department of Interventional Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Yun Wang
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Jin Chen
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shichao Zhang
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Lijing Teng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Zuquan Hu
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
| | - Shi Zhou
- Department of Interventional Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Zhu Zeng
- School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guiyang, China
- Engineering Center of Cellular Immunotherapy of Guizhou Province, Guiyang, China
- Key Laboratory of Infectious Immunity and Antibody Engineering of Guizhou Province, Guiyang, China
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, Guizhou Medical University, Guiyang, China
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, China
| | - Jinhua Long
- Department of Oncology, School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, China
- Department of Oncology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China
- Department of Oncology, Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
5
|
Ogasawara A, Matsushita H, Tan TZ, Shintani D, Ye J, Nagao S, Demachi-Okamura A, Muraoka D, Kobayashi Y, Kakimi K, Yamaguchi R, Matsuo K, Yamamoto K, Fujiwara K, Huang RYJ, Tan DSP, Hasegawa K. Immunological impact of intraperitoneal and intravenous chemotherapy in ovarian cancer, translational analyses of the Phase 3 iPocc trial. Gynecol Oncol 2024; 191:124-131. [PMID: 39413557 DOI: 10.1016/j.ygyno.2024.09.023] [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] [Received: 07/12/2024] [Revised: 09/27/2024] [Accepted: 09/29/2024] [Indexed: 10/18/2024]
Abstract
BACKGROUND The iPocc trial, a randomized, global phase 3 study that compared intraperitoneal (IP) and intravenous (IV) carboplatin with dose-dense paclitaxel chemotherapy in epithelial ovarian cancer (EOC) patients, demonstrated improved progression-free survival in patients who received IP chemotherapy. The present study aimed to investigate the role of preexisting tumor immunity in the clinical outcomes of patients receiving IP chemotherapy. METHODS This study involved analyzing patient data from the iPocc trial, selectively of those whose tumor specimens were preserved at the time of primary surgery. A total of 116 cases ((IP; n = 59), (IV; n = 57)) were subjected to microarray analysis. Single-sample gene set enrichment analyses were performed to evaluate the tumor immune microenvironment. RESULTS Patients with enhanced tumor infiltration of T cells, natural killer (NK) cells, and cytotoxic lymphocytes in the IP group had a longer overall survival (OS) than those in the IV group, but not in the group with low infiltration. IP therapy improved the OS of patients with high expression of immune-related genes such as CD8A and FOXP3. In patients' subdivided into "immune Hot" and "immune Cold" groups based on hierarchical clustering analysis using four parameters representing "Innate immunity," "T cells," "IFNG response" and "Inhibitory molecules," IP therapy significantly improved prognosis in the "immune Hot" group, but not in the "immune Cold" group compared to that of IV therapy. CONCLUSIONS IP chemotherapy enhances the survival rates of patients with EOC with an immune-Hot phenotype in the tumor microenvironment prior to treatment. (Japan Registry of Clinical Trials number, jRCTs031180141.).
Collapse
Affiliation(s)
- Aiko Ogasawara
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan; Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Hirokazu Matsushita
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan.
| | - Tuan Zea Tan
- Genomics and Data Analytics Core, Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Daisuke Shintani
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Jieru Ye
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Shoji Nagao
- Gynecologic Oncology, Hyogo Cancer Center, Akashi, Japan
| | - Ayako Demachi-Okamura
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Daisuke Muraoka
- Division of Translational Oncoimmunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yukari Kobayashi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan
| | - Kazuhiro Kakimi
- Department of Immunotherapeutics, The University of Tokyo Hospital, Tokyo, Japan; Department of Immunology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Rui Yamaguchi
- Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Keitaro Matsuo
- Division of Cancer Epidemiology and Prevention, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kouji Yamamoto
- Department of Biostatistics, Yokohama City University School of Medicine, Yokohama, Japan
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan
| | - Ruby Yun-Ju Huang
- School of Medicine, College of Medicine, National Taiwan University, Taipei City, Taiwan; Department of Obstetrics & Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - David Shao Peng Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore; NUS Centre for Cancer Research (N2CR) National University of Singapore, Singapore, Singapore
| | - Kosei Hasegawa
- Department of Gynecologic Oncology, Saitama Medical University International Medical Center, Hidaka, Japan.
| |
Collapse
|
6
|
Thiels C, Grotz TE. The Emerging Role of Cytoreduction and Hyperthermic Intraperitoneal Chemotherapy (HIPEC) in Pancreatic Cancer. J Surg Oncol 2024; 130:1257-1262. [PMID: 39552018 DOI: 10.1002/jso.27880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 08/23/2024] [Indexed: 11/19/2024]
Abstract
Peritoneal spread is a frequent cause of metastasis in both the synchronous and metachronous settings for patients with pancreatic cancer. Recent improvements in systemic chemotherapy for pancreatic cancer may now allow for the careful selection of patients with oligometastatic disease isolated to the peritoneum who may benefit from consolidative cytoreduction and HIPEC.
Collapse
Affiliation(s)
- Cornelius Thiels
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Travis E Grotz
- Division of Hepatobiliary and Pancreas Surgery, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
7
|
Roth L, Huynh-Russo L, Heeb L, Ulugöl S, Freire Dos Santos R, Breuer E, Ungethüm U, Haberecker M, Pauli C, Koelzer V, Lehmann K, Gupta A. CD8 + T-cells restrict the development of peritoneal metastasis and support the efficacy of hyperthermic intraperitoneal chemotherapy (HIPEC). Sci Rep 2024; 14:22324. [PMID: 39333597 PMCID: PMC11437079 DOI: 10.1038/s41598-024-72826-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
Multimodal therapy for peritoneal metastasis (PM) including cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) provides long-term survival in highly selected colorectal cancer patients. Mechanisms behind HIPEC are unknown and may include induction of adaptive immunity. We therefore analyzed human PM samples and explored the impact of HIPEC in experimental models. Human samples from colorectal primary tumors (n = 19) and PM lesions (n = 37) were examined for the presence of CD8 + T-cells and their association with disease free (DFS) and overall survival (OS). CD8 + T cell response after HIPEC was assessed using an in-vivo PM mouse model, tumor cell lines and patient-derived tumor organoids. Patients with high intraepithelial CD8 + T cell counts showed longer DFS and OS. In the mouse model, HIPEC controlled growth of PM and increased numbers of functional granzyme positive CD8 + T cells within tumors. Cell lines and human organoids that were treated with heated chemotherapies showed immunogenic changes, reflected by significantly higher levels of MHC-class I molecules and expression of Cancer Testis Antigens Cyclin A1 and SSX-4. Using in-vitro co-culture assays, we noticed that cancer cells treated with heated chemotherapy primed dendritic cells, which subsequently enhanced effector functions of CD8 + T cells. The presence of CD8 + T-cells within PM lesions is associated with prolonged survival of patients with PM. Data from PM mouse model and in-vitro assay show that heated chemotherapies induce immunogenic changes on cancer cells leading to induction of CD8 + T-cells mediated immunity, which seems to control growth of PM lesions in mice after HIPEC.
Collapse
Affiliation(s)
- Lilian Roth
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland
| | - Linda Huynh-Russo
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland
| | - Laura Heeb
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland
| | - Sima Ulugöl
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland
| | - Rafael Freire Dos Santos
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland
| | - Eva Breuer
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland
| | - Udo Ungethüm
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland
| | - Martina Haberecker
- Department of Pathology and Molecular Pathology, University and University Hospital of Zürich, Zürich, Switzerland
| | - Chantal Pauli
- Department of Pathology and Molecular Pathology, University and University Hospital of Zürich, Zürich, Switzerland
| | - Viktor Koelzer
- Department of Pathology and Molecular Pathology, University and University Hospital of Zürich, Zürich, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Kuno Lehmann
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland.
| | - Anurag Gupta
- Surgical Oncology Research Laboratory, Department of Surgery & Transplantation, University Hospital of Zurich, Raemistrasse 100, Zurich, CH-8091, Switzerland.
- Laboratory for Applied Cancer Research, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
8
|
Kusamura S, Delhorme JB, Taibi A, Villeneuve L, Deraco M, Dico RL, Glehen O, Moran B. The 2022 PSOGI International Consensus on HIPEC Regimens for Peritoneal Malignancies: Pseudomyxoma Peritonei. Ann Surg Oncol 2024; 31:6262-6273. [PMID: 39008204 DOI: 10.1245/s10434-024-15646-6] [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] [Received: 05/26/2023] [Accepted: 06/08/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND The combination of cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) constitutes the established standard of care for pseudomyxoma peritonei patients. However, the role of HIPEC lacks validation through randomized trials, leading to diverse proposed treatment protocols. This consensus seeks to standardize HIPEC regimens and identify research priorities for enhanced clarity. METHODS The steering committee applied the patient, intervention, comparator, and outcome method to formulate crucial clinical questions. Evaluation of evidence followed the Grading of Recommendations, Assessment, Development, and Evaluation system. Consensus on HIPEC regimens and research priorities was sought through a two-round Delphi process involving international experts. RESULTS Out of 90 eligible panelists, 71 (79%) participated in both Delphi rounds, resulting in a consensus on six out of seven questions related to HIPEC regimens. An overwhelming 84% positive consensus favored combining HIPEC with CRS, while a 70% weak positive consensus supported HIPEC after incomplete CRS. Specific HIPEC regimens also gained consensus, with 53% supporting Oxaliplatin 200 mg/m2 and 51% favoring the combination of cisplatin (CDDP) associated with mitomycin-C (MMC). High-dose MMC regimens received an 89% positive recommendation. In terms of research priorities, 61% of panelists highlighted the importance of studies comparing HIPEC regimens post CRS. The preferred regimens for such studies were the combination of CDDP/MMC and high-dose MMC. CONCLUSIONS The consensus recommends the application of HIPEC following CRS based on the available evidence. The combination of CDDP/MMC and high-dose MMC regimens are endorsed for both current clinical practice and future research efforts.
Collapse
Affiliation(s)
- Shigeki Kusamura
- Department of Surgical Oncology, PSM Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.
| | - Jean-Baptiste Delhorme
- Department of General and Digestive Surgery, Hautepierre Hospital, Strasbourg University Hospital, Strasbourg, France
| | - Abdelkader Taibi
- Department of Digestive Surgery, Dupuytren University Hospital, CNRS, XLIM, UMR 7252, Limoges, France
| | - Laurent Villeneuve
- Hospices Civils de Lyon, Hôpital Lyon Sud, Service de Recherche et d'Epidémiologie Cliniques, Pierre-Bénite, France
- CICLY, Université Claude Bernard Lyon 1, Lyon, France
| | - Marcello Deraco
- Department of Surgical Oncology, PSM Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Rea Lo Dico
- Department of General Surgery, Emergency, and New Technologies, San Camillo Forlanini Hospital, Rome, Italy
| | - Olivier Glehen
- CICLY, Université Claude Bernard Lyon 1, Lyon, France
- Department of Surgical Oncology, Hospices Civils de Lyon, Hôpital Lyon Sud, Lyon, France
| | - Brendan Moran
- Peritoneal Malignancy Unit, Basingstoke Hospital, Basingstoke, UK
| |
Collapse
|
9
|
Liu Y, Tian C, Yang Z, Huang C, Jiao K, Yang L, Duan C, Zhang Z, Li G. Effects of Chronic Heat Stress on Growth, Apoptosis, Antioxidant Enzymes, Transcriptomic Profiles, and Immune-Related Genes of Hong Kong Catfish ( Clarias fuscus). Animals (Basel) 2024; 14:1006. [PMID: 38612245 PMCID: PMC11010891 DOI: 10.3390/ani14071006] [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: 01/19/2024] [Revised: 03/14/2024] [Accepted: 03/19/2024] [Indexed: 04/14/2024] Open
Abstract
Chronic heat stress can have detrimental effects on the survival of fish. This study aimed to investigate the impact of prolonged high temperatures on the growth, antioxidant capacity, apoptosis, and transcriptome analysis of Hong Kong catfish (Clarias fuscus). By analyzing the morphological statistics of C. fuscus subjected to chronic high-temperature stress for 30, 60, and 90 days, it was observed that the growth of C. fuscus was inhibited compared to the control group. The experimental group showed a significant decrease in body weight and body length compared to the control group after 60 and 90 days of high-temperature stress (p < 0.05, p < 0.01). A biochemical analysis revealed significant alterations in the activities of three antioxidant enzymes superoxide dismutase activity (SOD); catalase activity (CAT); glutathione peroxidase activity (GPx), the malondialdehyde content (MDA), and the concentrations of serum alkaline phosphatase (ALP); Aspartate aminotransferase (AST); and alanine transaminase (ALT) in the liver. TUNEL staining indicated stronger apoptotic signals in the high-temperature-stress group compared to the control group, suggesting that chronic high-temperature-induced oxidative stress, leading to liver tissue injury and apoptosis. Transcriptome analysis identified a total of 1330 DEGs, with 835 genes being upregulated and 495 genes being downregulated compared to the control group. These genes may be associated with oxidative stress, apoptosis, and immune response. The findings elucidate the growth changes in C. fuscus under chronic high temperature and provide insights into the underlying response mechanisms to a high-temperature environment.
Collapse
Affiliation(s)
- Yong Liu
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Changxu Tian
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Zhihua Yang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Cailin Huang
- Guangxi Introduction and Breeding Center of Aquaculture, Nanning 530001, China; (C.H.); (Z.Z.)
| | - Kaizhi Jiao
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Lei Yang
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Cunyu Duan
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| | - Zhixin Zhang
- Guangxi Introduction and Breeding Center of Aquaculture, Nanning 530001, China; (C.H.); (Z.Z.)
| | - Guangli Li
- Guangdong Research Center on Reproductive Control and Breeding Technology of Indigenous Valuable Fish Species, Guangdong Provincial Engineering Laboratory for Mariculture Organism Breeding, Guangdong Provincial Key Laboratory of Aquatic Animal Disease Control and Healthy Culture, Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; (Y.L.); (C.T.); (Z.Y.); (K.J.); (L.Y.); (C.D.)
| |
Collapse
|
10
|
Breusa S, Zilio S, Catania G, Bakrin N, Kryza D, Lollo G. Localized chemotherapy approaches and advanced drug delivery strategies: a step forward in the treatment of peritoneal carcinomatosis from ovarian cancer. Front Oncol 2023; 13:1125868. [PMID: 37287910 PMCID: PMC10242058 DOI: 10.3389/fonc.2023.1125868] [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: 12/16/2022] [Accepted: 05/04/2023] [Indexed: 06/09/2023] Open
Abstract
Peritoneal carcinomatosis (PC) is a common outcome of epithelial ovarian carcinoma and is the leading cause of death for these patients. Tumor location, extent, peculiarities of the microenvironment, and the development of drug resistance are the main challenges that need to be addressed to improve therapeutic outcome. The development of new procedures such as HIPEC (Hyperthermic Intraperitoneal Chemotherapy) and PIPAC (Pressurized Intraperitoneal Aerosol Chemotherapy) have enabled locoregional delivery of chemotherapeutics, while the increasingly efficient design and development of advanced drug delivery micro and nanosystems are helping to promote tumor targeting and penetration and to reduce the side effects associated with systemic chemotherapy administration. The possibility of combining drug-loaded carriers with delivery via HIPEC and PIPAC represents a powerful tool to improve treatment efficacy, and this possibility has recently begun to be explored. This review will discuss the latest advances in the treatment of PC derived from ovarian cancer, with a focus on the potential of PIPAC and nanoparticles in terms of their application to develop new therapeutic strategies and future prospects.
Collapse
Affiliation(s)
- Silvia Breusa
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Apoptosis, Cancer and Development Laboratory- Equipe labellisée ‘La Ligue’, LabEx DEVweCAN, Institut PLAsCAN, Centre de Recherche en Cancérologie de Lyon, Institut national de santé et de la recherche médicale (INSERM) U1052-Centre National de la Recherche Scientifique - Unité Mixte de Recherche (CNRS UMR)5286, Université de Lyon, Centre Léon Bérard, Lyon, France
| | - Serena Zilio
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Sociétés d'Accélération du Transfert de Technologies (SATT) Ouest Valorisation, Rennes, France
| | - Giuseppina Catania
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
| | - Naoual Bakrin
- Department of Surgical Oncology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Lyon, France
- Centre pour l'Innovation en Cancérologie de Lyon (CICLY), Claude Bernard University Lyon 1, Lyon, France
| | - David Kryza
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
- Imthernat Plateform, Hospices Civils de Lyon, Lyon, France
| | - Giovanna Lollo
- Univ Lyon, Université Claude Bernard Lyon 1, Centre National de la Recherche Scientifique (CNRS), LAGEPP Unité Mixte de Recherche (UMR) 5007, Villeurbanne, France
| |
Collapse
|
11
|
Choi M, Shin J, Lee CE, Chung JY, Kim M, Yan X, Yang WH, Cha JH. Immunogenic cell death in cancer immunotherapy. BMB Rep 2023; 56:275-286. [PMID: 37081756 PMCID: PMC10230015 DOI: 10.5483/bmbrep.2023-0024] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 09/29/2023] Open
Abstract
Cancer immunotherapy has been acknowledged as a new paradigm for cancer treatment, with notable therapeutic effects on certain cancer types. Despite their significant potential, clinical studies over the past decade have revealed that cancer immunotherapy has low response rates in the majority of solid tumors. One of the key causes for poor responses is known to be the relatively low immunogenicity of solid tumors. Because most solid tumors are immune desert 'cold tumors' with antitumor immunity blocked from the onset of innate immunity, combination therapies that combine validated T-based therapies with approaches that can increase tumor-immunogenicity are being considered as relevant therapeutic options. This review paper focuses on immunogenic cell death (ICD) as a way of enhancing immunogenicity in tumor tissues. We will thoroughly review how ICDs such as necroptosis, pyroptosis, and ferroptosis can improve anti-tumor immunity and outline clinical trials targeting ICD. Finally, we will discuss the potential of ICD inducers. as an adjuvant for cancer immunotherapy.[BMB Reports 2023; 56(5): 275-286].
Collapse
Affiliation(s)
- Minji Choi
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Jisoo Shin
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Chae-Eun Lee
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Joo-Yoon Chung
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Minji Kim
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Xiuwen Yan
- Affiliated Cancer Institute & Hospital and Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou 910095, China, Taichung 40402, Taiwan
| | - Wen-Hao Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Jong-Ho Cha
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea, Taichung 40402, Taiwan
| |
Collapse
|
12
|
Choi M, Shin J, Lee CE, Chung JY, Kim M, Yan X, Yang WH, Cha JH. Immunogenic cell death in cancer immunotherapy. BMB Rep 2023; 56:275-286. [PMID: 37081756 PMCID: PMC10230015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 04/22/2023] Open
Abstract
Cancer immunotherapy has been acknowledged as a new paradigm for cancer treatment, with notable therapeutic effects on certain cancer types. Despite their significant potential, clinical studies over the past decade have revealed that cancer immunotherapy has low response rates in the majority of solid tumors. One of the key causes for poor responses is known to be the relatively low immunogenicity of solid tumors. Because most solid tumors are immune desert 'cold tumors' with antitumor immunity blocked from the onset of innate immunity, combination therapies that combine validated T-based therapies with approaches that can increase tumor-immunogenicity are being considered as relevant therapeutic options. This review paper focuses on immunogenic cell death (ICD) as a way of enhancing immunogenicity in tumor tissues. We will thoroughly review how ICDs such as necroptosis, pyroptosis, and ferroptosis can improve anti-tumor immunity and outline clinical trials targeting ICD. Finally, we will discuss the potential of ICD inducers. as an adjuvant for cancer immunotherapy.[BMB Reports 2023; 56(5): 275-286].
Collapse
Affiliation(s)
- Minji Choi
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Jisoo Shin
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Chae-Eun Lee
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Joo-Yoon Chung
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Minji Kim
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
| | - Xiuwen Yan
- Affiliated Cancer Institute & Hospital and Key Laboratory for Cell Homeostasis and Cancer Research of Guangdong Higher Education Institutes, Guangzhou Medical University, Guangzhou 910095, China, Taichung 40402, Taiwan
| | - Wen-Hao Yang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - Jong-Ho Cha
- Department of Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22212, Taiwan
- Department of Biomedical Sciences, College of Medicine, Inha University, Incheon 22212, Korea, Taichung 40402, Taiwan
| |
Collapse
|
13
|
Chen Q, Li C, Wang Q. Multifunctional Nano-Biomaterials for Cancer Therapy via Inducing Enhanced Immunogenic Cell Death. SMALL METHODS 2023; 7:e2201457. [PMID: 36703555 DOI: 10.1002/smtd.202201457] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/30/2022] [Indexed: 05/17/2023]
Abstract
Immunotherapy is considered to be one of the most promising methods to overcome cancer. Immunogenic cell death (ICD), as a special form of cell death that can trigger an antitumor immune response, has attracted increasing attention for cancer immunotherapy. Presently, ICD-mediating immunotherapy needs to overcome many hurdles including a lack of targeted delivery systems for ICD inducers, insufficient antitumor immunity, and the immunosuppressive tumor microenvironment. Recent research has demonstrated that nano-biomaterials exhibit unique biochemphysical properties at the nanoscale, providing a prospective approach to overcoming these obstacles. In this review, the authors first survey the occurrence, processes, and detection methods of ICD. Subsequently, the recent advances of nano-biomaterials applied to enhance ICD according to the key steps in the process of ICD, particularly with a focus on the mechanisms and lifting schemes are investigated. Finally, based on the achievement in the representative studies, the prospects and challenges of nanotechnology in ICD for cancer therapy are discussed to enable clinical translation.
Collapse
Affiliation(s)
- Qian Chen
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- North District of Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215008, China
| | - Chunyan Li
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Qiangbin Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine and i-Lab, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| |
Collapse
|
14
|
Sen S, Karoscik K, Maier E, Arambula JF. Immunogenic cell death-inducing metal complexes: From the benchtop to the clinic. Curr Opin Chem Biol 2023; 73:102277. [PMID: 36867977 DOI: 10.1016/j.cbpa.2023.102277] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 03/05/2023]
Abstract
The immune system presents a complex array of processes designed to maintain homeostasis in malignant cellular growth. Malignancy is the result of a breakdown in immune surveillance by cancer cells evading immune recognition. Significant efforts have been made in modulating immune checkpoint signaling cascades to bypass the resulting immune evasion and establish an anticancer effect. More recently, it was discovered that a form of regulated cell death can involve the stimulation of immune response as its downstream effect and subsequently re-establish immune surveillance. This mechanism, known as immunogenic cell death (ICD), is being exploited as a target to prevent tumor relapse and prevent cancer metastasis. It is now appreciated that metal-based compounds play a key role in ICD activation due to their unique biochemical properties and interactions within cancer cells. With fewer than 1% of known anticancer agents documented as ICD inducers, recent efforts have been made to identify novel entities capable of stimulating a more potent anticancer immune response. While the recent reviews by us or others focus primarily on either discussing the chemical library of ICD inducers or intricate detailing of biological pathways associated with ICD, this review aims to bridge these two topics as a concise summary. Furthermore, early clinical evidence and future directions of ICD are briefly summarized.
Collapse
Affiliation(s)
- Sajal Sen
- Department of Biological Engineering, Massachusetts Institute of Technology, 32 Vassar Street, Cambridge, MA 02139, USA.
| | | | - Esther Maier
- Drug Dynamics Institute, College of Pharmacy, The University of Texas at Austin, 1400 Barbara Jordan Blvd., Austin, TX 78723, USA
| | | |
Collapse
|
15
|
Nevo N, Lee Goldstein A, Bar-David S, Abu-Abeid A, Dayan D, Lahat G, Nizri E. Immunological effects of heated intraperitoneal chemotherapy can be augmented by thymosin α1. Int Immunopharmacol 2023; 116:109829. [PMID: 36758296 DOI: 10.1016/j.intimp.2023.109829] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/22/2023] [Accepted: 01/25/2023] [Indexed: 02/10/2023]
Abstract
BACKGROUND Peritoneal metastases of colorectal carcinoma origin (PM-CRC) are treated by cytoreductive surgery and heated intraperitoneal chemotherapy (HIPEC). However, the majority of patients recur, calling for novel treatments. We explored the immunogenic changes induced by HIPEC and the possibility to use thymosin α1 (Tα1) as an immune-stimulatory agent. METHODS We used an experimental murine model of PM-CRC combined with mitomycin (MMC)-based HIPEC. We determined immune cell infiltration into tumor metastases after HIPEC administration by means of immunohistochemistry, and determined immunogenic cell death signals in tumor cells by real-time polymerase chain reaction. RESULTS Mice with PM-CRC treated by HIPEC had increased overall survival (OS) compared to sham-treated mice (median OS 22.8 vs 18.9 days, respectively; P < 0.001). HIPEC induced increased infiltration of CD4+, CD8+, CD68 + and CD20 + cells into omental and visceral metastases at a magnitude of 40-100 %. We searched for potential immune signals induced by HIPEC by determining its effects on known immunogenic cell death proteins (heat-shock protein [HSP]-70, HSP-90 and calreticulin). HIPEC significantly increased HSP-90 mRNA expression (2.37 ± 1.5 vs 1-fold change, P < 0.05). The OS of Tα1 treated mice significantly improved compared to HIPEC-treated mice (16.3 ± 0.8 vs 14.1 ± 0.6 days, respectively, P = 0.02) and vs sham (11.8 ± 0.8 days, P = 0.007). CONCLUSIONS HIPEC induced immunogenic changes that led to increased immune cell infiltration. These changes were further augmented by Tα1 treatment. Future studies aimed at optimizing Tα1 treatment should focus upon the immune response it evokes.
Collapse
Affiliation(s)
- Nadav Nevo
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel; Laboratory of Surgical Oncology, Division of Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | | | - Shoshi Bar-David
- Laboratory of Surgical Oncology, Division of Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Adam Abu-Abeid
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel
| | - Danit Dayan
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel
| | - Guy Lahat
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eran Nizri
- Department of Surgery B, Weizman 6, Tel-Aviv 6423906, Israel; The Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
16
|
Alon G, Geva R, Lahat G, Nizri E. ASO Author Reflections: HIPEC Immunogenic Changes Could be Further Exploited by Immunotherapy to Treat Peritoneal Metastases from Colorectal Cancer. Ann Surg Oncol 2023; 30:2664-2665. [PMID: 36807022 DOI: 10.1245/s10434-023-13254-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/05/2023] [Indexed: 02/19/2023]
Affiliation(s)
- Gilad Alon
- Peritoneal Surface Malignancy and Melanoma Unit, Department of Surgery A, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Laboratory of Surgical Oncology, Division of Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Ravit Geva
- Institute of Oncology, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Guy Lahat
- Peritoneal Surface Malignancy and Melanoma Unit, Department of Surgery A, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Laboratory of Surgical Oncology, Division of Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Eran Nizri
- Peritoneal Surface Malignancy and Melanoma Unit, Department of Surgery A, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel. .,Laboratory of Surgical Oncology, Division of Surgery, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel. .,Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel.
| |
Collapse
|
17
|
Zhou Y, Qi M, Zhou YX, Fang SQ. Application of intraperitoneal hyperthermic perfusion chemotherapy in gastric cancer. Shijie Huaren Xiaohua Zazhi 2023; 31:1-7. [DOI: 10.11569/wcjd.v31.i1.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Gastric cancer is one of the most common malignant tumors in China. Intraperitoneal hyperthermic perfusion chemotherapy is a comprehensive therapy that combines intraperitoneal perfusion, hyperthermia, and chemotherapy. It has a good curative effect in peritoneal metastasis of gastric cancer. In recent years, with the continuous progress of technology and the deepening of research, the scope of application of intraperitoneal hyperthermic perfusion chemotherapy is more extensive. In this paper, we discuss intraperitoneal hyperthermic perfusion chemotherapy with regard to its application in gastric cancer, commonly used drugs, safety, and prospects.
Collapse
Affiliation(s)
- Yue Zhou
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese medicine, Shanghai 200437, China
| | - Mei Qi
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese medicine, Shanghai 200437, China
| | - Yu-Xuan Zhou
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese medicine, Shanghai 200437, China
| | - Sheng-Quan Fang
- Department of Gastroenterology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese medicine, Shanghai 200437, China
| |
Collapse
|
18
|
Geva R, Alon G, Nathanson M, Bar-David S, Nevo N, Aizic A, Peles-Avraham S, Lahat G, Nizri E. PD-1 Blockade Combined with Heated Intraperitoneal Chemotherapy Improves Outcome in Experimental Peritoneal Metastases from Colonic Origin in a Murine Model. Ann Surg Oncol 2023; 30:2657-2663. [PMID: 36595112 DOI: 10.1245/s10434-022-13025-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/13/2022] [Indexed: 01/04/2023]
Abstract
BACKGROUND Heated intraperitoneal chemotherapy (HIPEC) was shown to induce immunogenicity of peritoneal metastases from colorectal cancer (PM-CRC) by induction of immunogenic cell death. We aimed to explore whether the addition of a checkpoint inhibitor would augment the effect of HIPEC in an experimental murine model of PM-CRC. METHODS PM-CRC was established in C57BL mice by intraperitoneal inoculation of MC38 colon cancer cells. HIPEC was administered using the closed technique with mitomycin C (MMC). Clinical and immunological parameters were compared between animals treated with HIPEC alone and those treated with HIPEC + anti-programmed death receptor-1 (aPD-1). RESULTS MMC-based HIPEC increased the overall survival of animals compared with sham-treated animals (22.8; 95% confidence interval [CI] 21.14-24.53 vs. 18.9 days; 95% CI 17.6-20.3, p < 0.001). The extent of peritoneal disease as measured by the modified peritoneal carcinomatosis index was also reduced by HIPEC. This clinical benefit was accompanied by increased infiltration of CD8+, CD68+, and CD20+ cells into tumor metastases in HIPEC-treated animals compared with sham-treated animals. We identified heat shock protein (HSP) 90 as a potential immunogenic cell death protein whose expression is increased under HIPEC conditions (fold change: 2.37 ± 1.5 vs. 1 without HIPEC, p < 0.05). Combined HIPEC + PD-1 treatment ameliorated survival compared with HIPEC alone and sham treatment (24.66; 95% CI 20.13-29.2 vs. 19; 95% CI 15.85-22.14 and 14.33 days; 95% CI 9.6-19.04, respectively; p = 0.008). This clinical effect was accompanied by increased CD8+ tumor infiltration. CONCLUSIONS HIPEC induced the expression of immunogenic cell death signals that can support an anti-tumor immune response. This response can be further exploited by a checkpoint inhibitor.
Collapse
Affiliation(s)
- Ravit Geva
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Gilad Alon
- Department of Surgery A, Peritoneal Surface Malignancy and Melanoma Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Laboratory of Surgical Oncology, Division of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Maya Nathanson
- Department of Surgery, Assaf Harofeh Medical Center, Zerifin, Israel
| | - Shoshi Bar-David
- Department of Surgery A, Peritoneal Surface Malignancy and Melanoma Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Laboratory of Surgical Oncology, Division of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Nadav Nevo
- Department of Surgery A, Peritoneal Surface Malignancy and Melanoma Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Laboratory of Surgical Oncology, Division of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Asaf Aizic
- Institute of Pathology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Sharon Peles-Avraham
- Institute of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Guy Lahat
- Department of Surgery A, Peritoneal Surface Malignancy and Melanoma Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Laboratory of Surgical Oncology, Division of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eran Nizri
- Department of Surgery A, Peritoneal Surface Malignancy and Melanoma Unit, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel. .,Laboratory of Surgical Oncology, Division of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel. .,Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
19
|
Hu J, Wang Z, Wang X, Xie S. Side-effects of hyperthermic intraperitoneal chemotherapy in patients with gastrointestinal cancers. PeerJ 2023; 11:e15277. [PMID: 37138820 PMCID: PMC10150720 DOI: 10.7717/peerj.15277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 03/31/2023] [Indexed: 05/05/2023] Open
Abstract
Background Hyperthermic intraperitoneal chemotherapy (HIPEC) produces unwanted side-effects that are mainly caused by chemotherapeutic drugs in the treatment of gastrointestinal (GI) cancers, and these effects have not been systematically summarized. The aim of this article was to provide a comprehensive overview of the side-effects of HIPEC for GI cancers and propose practical strategies for adverse event management. Methodology PubMed, Web of Science, and the Cochrane Library were systematically searched for side-effects of HIPEC in GI cancers prior to October 20, 2022. A total of 79 articles were included in this review. Results Adverse events, such as enterocutaneous digestive fistulas, GI tract perforation, neutropenia, postoperative bleeding, ventricular tachycardia, hyperglycemia, hypocalcemia, renal impairment, encapsulating peritoneal sclerosis, scrotal ulceration, and sarcopenia were described, and their clinical management was discussed. These side-effects involve the digestive, hematopoietic, circulatory, metabolic, and urinary systems. Effective methods for adverse event management included an expert multidisciplinary team, replacing chemotherapy drugs, using Chinese medicine, and careful preoperative assessments. Conclusion The side-effects of HIPEC are frequent and can be minimized by several effective methods. This study proposes practical strategies for adverse event management of HIPEC to assist physicians in choosing the optimal treatment method.
Collapse
Affiliation(s)
- Jiyun Hu
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenxing Wang
- Department of Hepatobiliary Surgery, Haikou People’s Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| | - Xinrun Wang
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Shucai Xie
- Department of Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Department of Hepatobiliary Surgery, Haikou People’s Hospital/Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, Hainan, China
| |
Collapse
|
20
|
Thymosin alpha 1 as an adjuvant to hyperthermic intraperitoneal chemotherapy in an experimental model of peritoneal metastases from colonic carcinoma. Int Immunopharmacol 2022; 111:109166. [PMID: 35994852 DOI: 10.1016/j.intimp.2022.109166] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/07/2022] [Accepted: 08/12/2022] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Heated intraperitoneal chemotherapy (HIPEC) is currently implemented in the treatment of peritoneal metastases from colorectal carcinoma (PM-CRC) origin. However, recurrence is common and the effectiveness of HIPEC has been questioned. The aim of this study was to evaluate the use of thymosin alpha 1 (Tα1), an immunomodulatory molecule, as an adjuvant to HIPEC treatment. METHODS We developed an experimental model of HIPEC by the induction of PM-CRC in C57BL mice and intra-abdominal perfusion of mitomycin C (MMC). Mice were treated with Tα1 at 0.6 mg/kg for 5 days after HIPEC. Clinical and immunological parameters were compared between HIPEC and HIPEC + Tα1 groups. RESULTS Treatment with Tα1 increased overall survival of mice compared to HIPEC treatment alone and sham-treated animals (16.1 ± 0.8 vs. 14.1 ± 0.6 and 11.8 ± 0.8, respectively, p = 0.02). Tα1 had no direct anti-tumor effect, as seen by lack of inhibition of tumor cell proliferation. Tα1 treatment induced a T helper (Th) 1 immune response in tumor metastases as evidenced by a significant increase of the Th1-specific markers IFN-γ and T-bet (1.21 ± 0.3 vs. 0.52 ± 0.08, p < 0.05; 0.88 ± 0.04 vs. 0.64 ± 0.14, p < 0.05, respectively). This Th1 skew was accompanied by increased CD8+ infiltration into omental and visceral metastases by Tα1 treatment compared to sham and HIPEC-treated animals (21.24 ± 2.16 vs. 10.45 ± 0.89 and 7.7 ± 1.3, p < 0.001; 14.12 ± 1.54 vs. 12.12 ± 0.01 and 6.64 ± 0.87, p < 0.01, respectively). CONCLUSIONS Tα1 augments the effect of HIPEC by the induction of a Th1 anti-tumor immune response. Further experiments should evaluate Tα1 and other novel immunomodulators in order to exploit the immunological opportunities created by HIPEC.
Collapse
|
21
|
Fiorentini C, Sarti D, Guadagni S, Fiorentini G. Immune response and locoregional treatments for peritoneal carcinomatosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2022; 371:97-116. [PMID: 35965002 DOI: 10.1016/bs.ircmb.2022.04.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Peritoneal Carcinomatosis (PC) is considered as a terminal disease with short survival. It is treated with palliative therapies, consisting of repeated drainages and sometimes instillation of chemotherapy. Since the nineties, surgery has been combined with more effective systemic chemotherapy, intraperitoneal chemotherapy and hyperthermic intraperitoneal chemotherapy (HIPEC) for the treatment of PC. This combination therapy significantly increases the overall survival of selected PC patients. The understanding of how intraperitoneal chemotherapy and HIPEC can cure patients is still unclear. Experts hypothesized that the efficacy is obtained by the ability of high peritoneal drug exposure and hyperthermia to directly kill cancer cells. Several studies indicate that cancer cells death directly influences the response of the immune system. For this reason, the protective effect of intraperitoneal chemotherapy and HIPEC could be mediated by its ability to kill cancer cells in an immuno-genic way, causing an efficient anticancer immune response. In this review, we investigate the role of the innate peritoneal or locoregional therapy-induced immune response in PC therapy.
Collapse
Affiliation(s)
- Caterina Fiorentini
- Department of Prevention and Sport Medicine, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Donatella Sarti
- Oncology Department, S. Maria Della Misericordia Hospital, ASUR1, Urbino, Italy
| | - Stefano Guadagni
- Department of Applied Clinical Sciences and Biotechnology, University of L'Aquila, L'Aquila, Italy
| | - Giammaria Fiorentini
- Department of Onco-Hematology, Azienda Ospedaliera "Ospedali Riuniti Marche Nord", Pesaro, Italy.
| |
Collapse
|
22
|
Keeping Cell Death Alive: An Introduction into the French Cell Death Research Network. Biomolecules 2022; 12:biom12070901. [PMID: 35883457 PMCID: PMC9313292 DOI: 10.3390/biom12070901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Since the Nobel Prize award more than twenty years ago for discovering the core apoptotic pathway in C. elegans, apoptosis and various other forms of regulated cell death have been thoroughly characterized by researchers around the world. Although many aspects of regulated cell death still remain to be elucidated in specific cell subtypes and disease conditions, many predicted that research into cell death was inexorably reaching a plateau. However, this was not the case since the last decade saw a multitude of cell death modalities being described, while harnessing their therapeutic potential reached clinical use in certain cases. In line with keeping research into cell death alive, francophone researchers from several institutions in France and Belgium established the French Cell Death Research Network (FCDRN). The research conducted by FCDRN is at the leading edge of emerging topics such as non-apoptotic functions of apoptotic effectors, paracrine effects of cell death, novel canonical and non-canonical mechanisms to induce apoptosis in cell death-resistant cancer cells or regulated forms of necrosis and the associated immunogenic response. Collectively, these various lines of research all emerged from the study of apoptosis and in the next few years will increase the mechanistic knowledge into regulated cell death and how to harness it for therapy.
Collapse
|
23
|
Li L, Liu Z, Quan J, Lu J, Zhao G, Sun J. Dietary nanoselenium supplementation for heat-stressed rainbow trout: effects on organizational structure, lipid changes, and biochemical parameters as well as heat-shock-protein- and selenoprotein-related gene expression. FISH PHYSIOLOGY AND BIOCHEMISTRY 2022; 48:707-722. [PMID: 35597860 DOI: 10.1007/s10695-022-01084-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 05/11/2022] [Indexed: 06/15/2023]
Abstract
Nanoselenium (nano-Se) shows unique protective effects against environmental heat stress in rainbow trout as a selenium source additive and free radical scavenger. Accordingly, we investigated the effects of supplementation with different levels of nano-Se (0, 5, and 10 mg/kg) and before and after heat stress (24°C) for different treatment times on the dynamic changes of rainbow trout liver tissue structure, lipid changes, biochemical properties, and gene expression. The results showed that, under heat stress, the fish supplementation of 5 mg/kg nano-Se significantly increased liver glutathione peroxidase (GPx) activity and upregulated expression levels of HSP70b, HSP90a1, GPx1a, and Trx mRNAs, while liver alanine aminotransferase (ALT), aspartate aminotransferase (AST), superoxide dismutase (SOD), and malondialdehyde (MDA) levels as well as tissue structure damage and lipid accumulation were decreased. Combining the trends for the above indicators indicated that stress began to increase significantly at 8 h. It can be concluded that supplementation with 5 mg/kg nano-Se effectively alleviates stress damage in rainbow trout. Furthermore, stress at 24°C for 8 h can be thought of as a critical time point for the study of heat stress in rainbow trout, with significant changes in response but no serious damage. Thus, these results provide a reference for the addition of nano-Se to rainbow trout feed and provide theoretical and practical guidance for enhancing the resistance of rainbow trout to heat stress.
Collapse
Affiliation(s)
- Lanlan Li
- College of Animal Science & Technology, Gansu Agricultural University, Lanzhou, 730070, People's Republic of China
| | - Zhe Liu
- College of Animal Science & Technology, Gansu Agricultural University, Lanzhou, 730070, People's Republic of China.
| | - Jinqiang Quan
- College of Animal Science & Technology, Gansu Agricultural University, Lanzhou, 730070, People's Republic of China
| | - Junhao Lu
- College of Animal Science & Technology, Gansu Agricultural University, Lanzhou, 730070, People's Republic of China
| | - Guiyan Zhao
- College of Animal Science & Technology, Gansu Agricultural University, Lanzhou, 730070, People's Republic of China
| | - Jun Sun
- College of Animal Science & Technology, Gansu Agricultural University, Lanzhou, 730070, People's Republic of China
| |
Collapse
|
24
|
Xu M, Lu JH, Zhong YZ, Jiang J, Shen YZ, Su JY, Lin SY. Immunogenic Cell Death-Relevant Damage-Associated Molecular Patterns and Sensing Receptors in Triple-Negative Breast Cancer Molecular Subtypes and Implications for Immunotherapy. Front Oncol 2022; 12:870914. [PMID: 35444934 PMCID: PMC9013947 DOI: 10.3389/fonc.2022.870914] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 03/08/2022] [Indexed: 01/23/2023] Open
Abstract
Objectives Triple-negative breast cancer (TNBC) is defined as a highly aggressive type of breast cancer which lacks specific biomarkers and drug targets. Damage-associated molecular pattern (DAMP)-induced immunogenic cell death (ICD) may influence the outcome of immunotherapy for TNBC patients. This study aims to develop a DAMPs gene signature to classify TNBC patients and to further predict their prognosis and immunotherapy outcome. Methods We identified the DAMPs-associated subtypes of 330 TNBCs using K-means analysis. Differences in immune status, genomic alterations, and predicted immunotherapy outcome were compared among each subtype. Results A total of 330 TNBCs were divided into three subtypes according to DAMPs gene expression: the nuclear DAMPs subtype, featuring the upregulation of nuclear DAMPs; the inflammatory DAMPs subtype, characterized by the gene set enrichment of the adaptive immune system and cytokine signaling in the immune system; and the DAMPs-suppressed subtype, having the lowest level of ICD-associated DAMPs. Among them, the inflammatory subtype patients had the most favorable survival, while the DAMPs-suppressed subtype was associated with the worst prognosis. The DAMPs subtyping system was successfully validated in the TCGA cohort. Furthermore, we systemically revealed the genomic alterations among the three DAMPs subtypes. The inflammatory DAMPs subtype was predicted to have the highest response rate to immunotherapy, suggesting that the constructed DAMPs clustering had potential for immunotherapy efficacy prediction. Conclusion We established a novel ICD-associated DAMPs subtyping system in TNBC, and DAMPs expression might be a valuable biomarker for immunotherapy strategies. Our work could be helpful to the development of new immunomodulators and may contribute to the development of precision immunotherapy for TNBC.
Collapse
Affiliation(s)
- Ming Xu
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China.,Department of Traditional Chinese Medicine, The First People's Hospital of Tongxiang, Zhejiang, China
| | - Jin-Hua Lu
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Ya-Zhen Zhong
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Jing Jiang
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Yue-Zhong Shen
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Jing-Yang Su
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| | - Sheng-You Lin
- Department of Oncology, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, Zhejiang, China
| |
Collapse
|
25
|
Dellinger TH, Han ES, Raoof M, Lee B, Wu X, Cho H, He TF, Lee P, Razavi M, Liang WS, Schmolze D, Priceman SJ, Lee S, Lin WC, Lin JF, Kebria M, Hakim A, Ruel N, Stewart DB, Wang EW, Paz BI, Wakabayashi MT, Cristea MC, Rodriguez-Rodriguez L. Hyperthermic Intraperitoneal Chemotherapy-Induced Molecular Changes in Humans Validate Preclinical Data in Ovarian Cancer. JCO Precis Oncol 2022; 6:e2100239. [PMID: 35357903 PMCID: PMC8984280 DOI: 10.1200/po.21.00239] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Hyperthermic intraperitoneal chemotherapy (HIPEC) confers a survival benefit in epithelial ovarian cancer (EOC) and in preclinical models. However, the molecular changes induced by HIPEC have not been corroborated in humans.
Collapse
Affiliation(s)
- Thanh H Dellinger
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Ernest S Han
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Mustafa Raoof
- Division of Surgical Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Byrne Lee
- Department of Surgery, Stanford University, Stanford, CA
| | - Xiwei Wu
- Integrative Genomics Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Hyejin Cho
- Integrative Genomics Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Ting-Fang He
- Immuno-oncology Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Peter Lee
- Immuno-oncology Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Marianne Razavi
- Women's Cancer Center, City of Hope National Medical Center, Duarte, CA
| | | | - Daniel Schmolze
- Department of Pathology, City of Hope National Medical Center, Duarte, CA
| | - Saul J Priceman
- Hematology & Hematopoietic Cell Transplantation and Immuno-Oncology, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Stephen Lee
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Wei-Chien Lin
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Jeff F Lin
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Mehdi Kebria
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Amy Hakim
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Nora Ruel
- Biostatistics Core, City of Hope National Medical Center Beckman Research Institute, Duarte, CA
| | - Daphne B Stewart
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| | - Edward W Wang
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| | - Benjamin I Paz
- Department of Surgery, Stanford University, Stanford, CA
| | - Mark T Wakabayashi
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| | - Mihaela C Cristea
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA
| | - Lorna Rodriguez-Rodriguez
- Division of Gynecologic Oncology, Department of Surgery, City of Hope National Medical Center, Duarte, CA
| |
Collapse
|
26
|
Batista TP, Carneiro VCG, Tancredi R, Badiglian Filho L, Rangel RLC, Lopes A, Sarmento BJQ, Leão CS. A phase 2 trial of short-course Hyperthermic IntraPeritoneal Chemotherapy (HIPEC) at interval cytoreductive surgery (iCRS) for advanced ovarian cancer. Rev Col Bras Cir 2022; 49:e20223135. [PMID: 35239855 PMCID: PMC10578806 DOI: 10.1590/0100-6991e-20223135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/06/2021] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE to report the final analysis of a phase 2 trial assessing the efficacy and safety of short-course hyperthermic intraperitoneal chemotherapy (HIPEC) for patients with advanced epithelial ovarian cancer (EOC). METHODS this was an open-label, multicenter, single-arm trial of HIPEC in patients with advanced EOC who underwent interval cytoreductive surgery (iCRS) after neoadjuvant chemotherapy (NACT). HIPEC was performed as a concentration-based regimen of platinum-based chemotherapy for 30 minutes. Primary endpoint was the rate of disease progression occurring at nine months following iCRS plus HIPEC (PD9). Secondary endpoints were postoperative complications, time to start adjuvant chemotherapy, length of hospital and ICU stay, quality of life (QoL) over treatment, and ultimately 2-year progression-free survival (PFS) and overall survival (OS). Analysis was by intention-to-treat with final database lock for survival outcomes on February 23, 2021. RESULTS fifteen patients with stage III EOC were enrolled between February 2015 and July 2019, in four centers. The intention to treat PD9 was 6.7%. With a median follow-up of 33 months (IQR, 24.3-46.5), the median PFS was 18.1 months and corresponding 2-year rates of PFS and OS was 33.3% and 93.3%, respectively. Three patients (20%) experienced graded III complications. Median length of hospital and ICU stay was 5 (IQR, 4-6.5) and 1 (IQR, 1-1) days, respectively. Time to restart systemic chemotherapy was 39 (IQR, 35-49.3) days and no significant difference over time in QoL was observed. CONCLUSIONS we demonstrate preliminary efficacy and safety of short-course HIPEC in patient with advanced EOC.
Collapse
Affiliation(s)
- Thales Paulo Batista
- - IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Department of Surgery/Oncology - Recife - PE - Brasil
- - UFPE - Universidade Federal de Pernambuco, Center of Medical Science - Recife - PE - Brasil
| | - Vandré Cabral Gomes Carneiro
- - IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Department of Surgery/Oncology - Recife - PE - Brasil
- - HCP - Hospital de Câncer de Pernambuco, Department of Gynecology - Recife - PE - Brasil
| | - Rodrigo Tancredi
- - IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Department of Clinical Oncology - Recife - PE - Brasil
- - HCP - Hospital de Câncer de Pernambuco, Department of Clinical Oncology - Recife - PE - Brasil
| | | | | | - André Lopes
- - IBCC - Instituto Brasileiro de Controle do Câncer, Department of Gynecology - São Paulo - SP - Brasil
| | - Bruno José Queiroz Sarmento
- - IHBDF - Instituto Hospital de Base do Distrito Federal, Serviço de Oncologia Cirúrgica - Brasília - DF - Brasil
| | - Cristiano Souza Leão
- - IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Departamento de Cirurgia - Recife - PE - Brasil
| |
Collapse
|
27
|
Peritoneal Metastasis: Current Status and Treatment Options. Cancers (Basel) 2021; 14:cancers14010060. [PMID: 35008221 PMCID: PMC8750973 DOI: 10.3390/cancers14010060] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/16/2021] [Accepted: 12/20/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Surgical and locoregional treatments of peritoneal metastasis, e.g., from colorectal cancer, has gained increasing acceptance after the publication of excellent patient outcomes from many groups around the world. Apart from systemic chemotherapy and surgical removal of the tumor, locoregional therapies such as HIPEC or PIPAC may improve tumor control. Understanding the molecular characteristics of peritoneal metastasis is crucial to evolve future therapeutic strategies for peritoneal metastasis. This includes the genetic background of PM, which is often different from other sites of metastasis, and promotes peritoneal dissemination and the growth of tumor cells. Growing knowledge and insight into the physiology of the peritoneal tumor microenvironment and the specific role of the immune system in this compartment may provide a critical step to move locoregional therapy to the next level. This review summarizes the current knowledge and highlights the molecular characteristics of peritoneal metastasis. Abstract Peritoneal metastasis (PM) originating from gastrointestinal cancer was considered a terminal disease until recently. The advent of better systemic treatment, a better understanding of prognostic factors, and finally, the advent of novel loco-regional therapies, has opened the door for the multimodal treatment of PM. These strategies, including radical surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) showed surprisingly good results, leading to the prolonged survival of patients with peritoneal metastasis. This has triggered a significant body of research, leading to the molecular characterization of PM, which may further help in the development of novel treatments. This review summarizes current evidence on peritoneal metastasis and explores potential novel mechanisms and therapeutic approaches to treat patients with peritoneal metastasis.
Collapse
|
28
|
Lin YX, Wang Y, Ding J, Jiang A, Wang J, Yu M, Blake S, Liu S, Bieberich CJ, Farokhzad OC, Mei L, Wang H, Shi J. Reactivation of the tumor suppressor PTEN by mRNA nanoparticles enhances antitumor immunity in preclinical models. Sci Transl Med 2021; 13:13/599/eaba9772. [PMID: 34162754 DOI: 10.1126/scitranslmed.aba9772] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 12/29/2020] [Accepted: 05/17/2021] [Indexed: 12/12/2022]
Abstract
Increasing clinical evidence has demonstrated that the deletion or mutation of tumor suppressor genes such as the gene-encoding phosphatase and tensin homolog deleted on chromosome 10 (PTEN) in cancer cells may correlate with an immunosuppressive tumor microenvironment (TME) and poor response or resistance to immune checkpoint blockade (ICB) therapy. It is largely unknown whether the restoration of functional PTEN may modulate the TME and improve the tumor's sensitivity to ICB therapy. Here, we demonstrate that mRNA delivery by polymeric nanoparticles can effectively induce expression of PTEN in Pten-mutated melanoma cells and Pten-null prostate cancer cells, which in turn induces autophagy and triggers cell death-associated immune activation via release of damage-associated molecular patterns. In vivo results illustrated that PTEN mRNA nanoparticles can reverse the immunosuppressive TME by promoting CD8+ T cell infiltration of the tumor tissue, enhancing the expression of proinflammatory cytokines, such as interleukin-12, tumor necrosis factor-α, and interferon-γ, and reducing regulatory T cells and myeloid-derived suppressor cells. The combination of PTEN mRNA nanoparticles with an immune checkpoint inhibitor, anti-programmed death-1 antibody, results in a highly potent antitumor effect in a subcutaneous model of Pten-mutated melanoma and an orthotopic model of Pten-null prostate cancer. Moreover, the combinatorial treatment elicits immunological memory in the Pten-null prostate cancer model.
Collapse
Affiliation(s)
- Yao-Xin Lin
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yi Wang
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,CAS Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianxun Ding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Aiping Jiang
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jie Wang
- CAS Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China.,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Mian Yu
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Guangzhou, Guangdong 510006, China
| | - Sara Blake
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.,Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA
| | - Shuaishuai Liu
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21250 USA
| | - Charles J Bieberich
- Department of Biological Sciences, University of Maryland Baltimore County, Baltimore, MD 21250 USA.,University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD 21201, USA
| | - Omid C Farokhzad
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Lin Mei
- School of Pharmaceutical Sciences (Shenzhen), Sun Yat-Sen University, Guangzhou, Guangdong 510006, China. .,Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Hao Wang
- CAS Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), Beijing 100190, China. .,Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jinjun Shi
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
29
|
Affiliation(s)
- Thanh H Dellinger
- Department of Surgery, Division of Gynecologic Oncology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA 91010, United States of America
| | - Ernest S Han
- Department of Surgery, Division of Gynecologic Oncology, City of Hope Comprehensive Cancer Center, 1500 E. Duarte Road, Duarte, CA 91010, United States of America.
| |
Collapse
|
30
|
Grześkowiak BF, Maziukiewicz D, Kozłowska A, Kertmen A, Coy E, Mrówczyński R. Polyamidoamine Dendrimers Decorated Multifunctional Polydopamine Nanoparticles for Targeted Chemo- and Photothermal Therapy of Liver Cancer Model. Int J Mol Sci 2021; 22:E738. [PMID: 33451063 PMCID: PMC7828497 DOI: 10.3390/ijms22020738] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/07/2021] [Accepted: 01/11/2021] [Indexed: 12/23/2022] Open
Abstract
The development of multifunctional drug delivery systems combining two or more nanoparticle-mediated therapies for efficient cancer treatment is highly desired. To face this challenge, a photothermally active polydopamine (PDA) nanoparticle-based platform was designed for the loading of chemotherapeutic drug and targeting of cancer cells. PDA spheres were first functionalized with polyamidoamine (PAMAM) dendrimers followed by the conjugation with polyethylene glycol (PEG) moieties and folic acid (FA) targeting ligand. The anticancer drug doxorubicin (DOX) was then absorbed on the particle surface. We performed the physico-chemical characterization of this versatile material and we assessed further its possible application in chemo- and photothermal therapy using liver cancer cell model. These nanoparticles exhibited high near-infrared photothermal conversion efficacy and allowed for loading of the drug, which upon release in specifically targeted cancer cells suppressed their growth. Using cell proliferation, membrane damage, apoptosis, and oxidative stress assays we demonstrated high performance of this nanosystem in cancer cell death induction, providing a novel promising approach for cancer therapy.
Collapse
Affiliation(s)
- Bartosz F. Grześkowiak
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Wszechnicy Piastowskiej 3, PL-61614 Poznań, Poland; (D.M.); (A.K.); (A.K.); (E.C.)
| | - Damian Maziukiewicz
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Wszechnicy Piastowskiej 3, PL-61614 Poznań, Poland; (D.M.); (A.K.); (A.K.); (E.C.)
- Faculty of Physics, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 2, PL-61614 Poznań, Poland
| | - Agata Kozłowska
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Wszechnicy Piastowskiej 3, PL-61614 Poznań, Poland; (D.M.); (A.K.); (A.K.); (E.C.)
| | - Ahmet Kertmen
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Wszechnicy Piastowskiej 3, PL-61614 Poznań, Poland; (D.M.); (A.K.); (A.K.); (E.C.)
- Faculty of Physics, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 2, PL-61614 Poznań, Poland
| | - Emerson Coy
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Wszechnicy Piastowskiej 3, PL-61614 Poznań, Poland; (D.M.); (A.K.); (A.K.); (E.C.)
| | - Radosław Mrówczyński
- NanoBioMedical Centre, Adam Mickiewicz University in Poznań, Wszechnicy Piastowskiej 3, PL-61614 Poznań, Poland; (D.M.); (A.K.); (A.K.); (E.C.)
- Faculty of Chemistry, Adam Mickiewicz University in Poznań, Uniwersytetu Poznańskiego 8, PL-61614 Poznań, Poland
| |
Collapse
|
31
|
Critical Analysis of Stage IV Epithelial Ovarian Cancer Patients after Treatment with Neoadjuvant Chemotherapy followed by Cytoreductive Surgery and Hyperthermic Intraperitoneal Chemotherapy (CRS/HIPEC). Int J Surg Oncol 2020; 2020:1467403. [PMID: 33381312 PMCID: PMC7759396 DOI: 10.1155/2020/1467403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/14/2020] [Accepted: 11/26/2020] [Indexed: 12/02/2022] Open
Abstract
Background Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC) after neoadjuvant chemotherapy (NACT) showed promise as initial treatment for stage IIIC (SIII) epithelial ovarian cancer (EOC); however, stage IV (SIV) outcomes are rarely reported. We assessed our experience and outcomes treating newly diagnosed SIV EOC with NACT plus CRS/HIPEC compared to SIII patients. Methods Advanced EOC from 2015–2018 managed with NACT (carboplatin/paclitaxel) due to unresectable disease or poor performance status followed by interval CRS/HIPEC were reviewed. Perioperative factors were assessed. Overall survival (OS) and progression-free survival (PFS) were analyzed by stage. Results Twenty-seven FIGO stage IIIC (n = 12) and IV (n = 15) patients were reviewed. Median NACT cycles were 3 and 4, respectively. Post-NACT omental caking, ascites, and pleural effusions decreased/resolved in 91%, 91%, and 100% of SIII and 85%, 92%, and 71% of SIV. SIII/SIV median PCI was 21 and 20 obtaining 92% and 100% complete cytoreduction (≤0.25 cm), respectively. Median organ resections were 6 and 7, respectively. Grade III/IV surgical complications were 0% SIII and 23% SIV, without hospital mortality. Median time to adjuvant chemotherapy was 53 and 74 days, respectively (p=0.007). SIII OS at 1 and 2 years was 100% and 83% and 87% and 76% in SIV (p=0.269). SIII 1-year PFS was 54%; median PFS: 12 months. SIV 1- and 2- year PFS was 47% and 23%; median PFS: 12 months (p=0.944). Conclusion Outcomes in select initially diagnosed and unresectable SIV EOC are similar to SIII after NACT plus CRS/HIPEC. SIV EOC may benefit from CRS/HIPEC, and further studies should explore this treatment approach.
Collapse
|
32
|
Abstract
Immunogenic cell death (ICD) is a type of cancer cell death triggered by certain chemotherapeutic drugs, oncolytic viruses, physicochemical therapies, photodynamic therapy, and radiotherapy. It involves the activation of the immune system against cancer in immunocompetent hosts. ICD comprises the release of damage-associated molecular patterns (DAMPs) from dying tumor cells that result in the activation of tumor-specific immune responses, thus eliciting long-term efficacy of anticancer drugs by combining direct cancer cell killing and antitumor immunity. Remarkably, subcutaneous injection of dying tumor cells undergoing ICD has been shown to provoke anticancer vaccine effects in vivo. DAMPs include the cell surface exposure of calreticulin (CRT) and heat-shock proteins (HSP70 and HSP90), extracellular release of adenosine triphosphate (ATP), high-mobility group box-1 (HMGB1), type I IFNs and members of the IL-1 cytokine family. In this review, we discuss the cell death modalities connected to ICD, the DAMPs exposed during ICD, and the mechanism by which they activate the immune system. Finally, we discuss the therapeutic potential and challenges of harnessing ICD in cancer immunotherapy.
Collapse
Affiliation(s)
- Asma Ahmed
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowUK
| | - Stephen W.G. Tait
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowUK
| |
Collapse
|
33
|
Zhu L, Xu Z, Wu Y, Liu P, Qian J, Yu S, Xia B, Lai J, Ma S, Wu Z. Prophylactic chemotherapeutic hyperthermic intraperitoneal perfusion reduces peritoneal metastasis in gastric cancer: a retrospective clinical study. BMC Cancer 2020; 20:827. [PMID: 32867714 PMCID: PMC7461269 DOI: 10.1186/s12885-020-07339-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Peritoneal metastasis is the most frequent failure in gastric cancer. This study evaluated the role of prophylactic chemotherapeutic hyperthermic intraperitoneal perfusion (CHIP) in patients after D2 dissection. METHODS Gastric cancer patients after D2 dissection were enrolled in this study. Patients received either chemotherapy (IV group) or CHIP (CHIP group). Sites of recurrence or metastasis, disease-free survival (DFS), overall survival (OS) and adverse events were evaluated. RESULTS Twenty-two patients received CHIP treatment, and 21 patients received chemotherapy alone. The median DFS time was 24.5 and 36.5 months in the IV group and CHIP group (P = 0.044), respectively. The median OS time was 33.1 months in the IV group and not reached in the CHIP group (P = 0.037). We also found that CHIP could reduce the total recurrence/metastasis rate, especially that of peritoneal metastasis. In the subgroup analysis, DFS and OS were both superior in deficient mismatch repair (dMMR) patients than in proficient MMR (pMMR) patients. CONCLUSION This hypothesis-generating study indicates that CHIP might be feasible for gastric cancer patients after D2 resection.
Collapse
Affiliation(s)
- Lucheng Zhu
- Department of Radiotherapy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou Cancer Hospital, Hangzhou, 310002, P.R. China
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, P.R. China
| | - Zhizheng Xu
- Department of oncology, Changxing people's hospital, Huzhou, 313100, P.R. China
| | - Yajun Wu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, P.R. China
| | - Pengyuan Liu
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, P.R. China
| | - Jianing Qian
- The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, 310053, P.R. China
| | - Shuhuan Yu
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, P.R. China
| | - Bing Xia
- Department of Radiotherapy, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou Cancer Hospital, Hangzhou, 310002, P.R. China
- Department of Oncology, Jiande Second People's Hospital, Zhejiang, 311604, P.R. China
| | - Jianjun Lai
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, P.R. China
| | - Shenglin Ma
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, P.R. China.
| | - Zhibing Wu
- Department of Oncology, Zhejiang Hospital, Hangzhou, Zhejiang, 310013, P.R. China.
| |
Collapse
|
34
|
Ye J, Chen L, Zuo J, Peng J, Chen C, Cai S, Song W, He Y, Yuan Y. A precise temperature control during hyperthermic intraperitoneal chemotherapy promises an early return of bowel function. Cancer Biol Ther 2020; 21:726-732. [PMID: 32644887 PMCID: PMC7515524 DOI: 10.1080/15384047.2020.1775444] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Introduction The hyperthermic intraperitoneal chemotherapy (HIPEC) has been widely applied in clinical practice for peritoneal carcinomatosis (PC). The temperature is one of the important elements affecting the efficacy of HIPEC, and it can become fluctuant by several factors. This study is aimed to explore the role of a stable perfusion temperature in promoting bowel recovery of PC patients due to gastrointestinal malignancies. Methods Between January 2012 and July 2013, 59 PC patients undergoing cytoreductive surgery and three-cycle HIPEC were included. Patients having stable perfusion temperature for all cycles were assigned into the study group, with the rest having unstable temperature into the control group. Time of flatus and defecation passage and initiation time of enteral nutrition were compared between both groups to detect the significance in bowel function recovery, with visual analogue scale (VAS) pain intensity and overall survival (OS) compared meanwhile. Results In sum, 33 (55.9%) patients obtained stable temperature during HIPEC, and the rest of 26 (44.1%) developed fluctuant perfusion temperature. Average time of flatus (2.3 ± 1.2 vs 3.9 ± 2.2 days, P =.002), defecation passage (5.2 ± 2.1 vs 7.1 ± 2.9 days, P =.004) and enteral nutrition initiation (4.3 ± 1.5 vs 6.7 ± 2.3 days, P <.001) were much shorter in the study group than the control group. Additionally, the VAS score (4.5 ± 2.3 vs 6.3 ± 1.3, P <.001) and 5-year OS rate (17.8% vs 11.1%, P=.135) were both improved, with significance observed in postoperative pain control. Conclusions During HIPEC, a precise temperature control could promise an early recovery of bowel function and reduce postoperative pain, without survival significance found based on the current cohort.
Collapse
Affiliation(s)
- Jinning Ye
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Liuhua Chen
- Department of Biliary-Pancreatic Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Jidong Zuo
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Jianjun Peng
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Chuangqi Chen
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Shirong Cai
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Wu Song
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Yulong He
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| | - Yujie Yuan
- Center of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R.China.,Center of Gastric Cancer, the First Affiliated Hospital, Sun Yat-Sen University , Guangzhou, P.R. China
| |
Collapse
|
35
|
de Bree E, Michelakis D. An overview and update of hyperthermic intraperitoneal chemotherapy in ovarian cancer. Expert Opin Pharmacother 2020; 21:1479-1492. [PMID: 32486865 DOI: 10.1080/14656566.2020.1766024] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Despite, the strong rationale and evidence of the benefit of postoperative intraperitoneal chemotherapy in advanced ovarian cancer, it has not been widely adopted, mainly due to its high morbidity and logistical difficulties. Intraoperative hyperthermic intraperitoneal chemotherapy (HIPEC) is a more tolerable and technically feasible method of intraperitoneal chemotherapy, whereas other potential advantages include homogenous drug distribution, application before tumor regrowth and combination with hyperthermia, which is directly cytotoxic and enhances the efficacy of many drugs. AREAS COVERED In this review, the authors explain the rationale and indications for cytoreductive surgery (CRS) and HIPEC in advanced ovarian cancer. Data of major clinical studies, meta-analyses, and recent randomized trials are discussed. EXPERT OPINION After many encouraging clinical studies and meta-analyses, a recent randomized study demonstrated survival benefit for HIPEC during interval CRS in primary ovarian cancer, without increased morbidity, whereas another implied its benefit in recurrent ovarian cancer. Results of recently completed and numerous ongoing randomized studies will further determine the benefit of HIPEC in ovarian cancer at different time points. Patient selection and appraisal of the best protocols are crucial. The field of gynecological oncology will most likely evolve to include HIPEC eventually as a routine treatment for ovarian cancer.
Collapse
Affiliation(s)
- Eelco de Bree
- Department of Surgical Oncology, Medical School of Crete University Hospital , Heraklion, Greece
| | - Dimosthenis Michelakis
- Department of Surgical Oncology, Medical School of Crete University Hospital , Heraklion, Greece
| |
Collapse
|
36
|
Ting WH, Hsiao CH, Chen HH, Wei MC, Lin HH, Hsiao SM. Comparisons of Clinical Outcomes in Women with Advanced Ovarian Cancer Treated with Frontline Intraperitoneal versus Dose-Dense Platinum/Paclitaxel Chemotherapy without Bevacizumab. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17103603. [PMID: 32443934 PMCID: PMC7277334 DOI: 10.3390/ijerph17103603] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/18/2020] [Accepted: 05/19/2020] [Indexed: 06/11/2023]
Abstract
Background: We aimed to compare the clinical outcomes between intraperitoneal chemotherapy and dose-dense chemotherapy for the frontline treatment of advanced ovarian, fallopian tube and primary peritoneal cancer in women not receiving bevacizumab. Methods: All consecutive women with stage II~IV cancer treated with either frontline intraperitoneal or dose-dense platinum/paclitaxel chemotherapy and not receiving bevacizumab between March 2006 and June 2019 were reviewed. Results: A total of 50 women (intraperitoneal group, n = 22; dose-dense group, n = 28) were reviewed. Median progression-free survival (32.6 months versus 14.2 months; adjusted hazard ratio = 0.38; 95% CI = 0.16 to 0.90, p = 0.03) and overall survival (not reached versus 30.7 months; adjusted hazard ratio = 0.23, 95% CI = 0.07 to 0.79, p = 0.02) were significantly higher in the intraperitoneal group than in the dose-dense group. A multivariable Cox proportional-hazards model also indicated that the number of frontline chemotherapy cycles (adjusted hazard ratio = 0.66, 95% CI 0.47 to 0.94, p = 0.02) was a predictor of better overall survival. Nausea/vomiting and nephrotoxicity occurred more frequently in the intraperitoneal group (p = 0.02 and <0.0001, respectively). Conclusions: Intraperitoneal chemotherapy seems to be superior in progression free survival and overall survival to dose-dense chemotherapy in the frontline treatment of women with optimally resected advanced ovarian, fallopian tube or primary peritoneal cancer and not receiving bevacizumab.
Collapse
Affiliation(s)
- Wan-Hua Ting
- Department of Obstetrics and Gynecology, Far Eastern Memorial Hospital, New Taipei 220409, Taiwan; (W.-H.T.); (H.-H.C.); (M.-C.W.); (H.-H.L.)
| | - Chi-Huang Hsiao
- Division of Medical Oncology and Hematology, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei 220409, Taiwan;
| | - Hui-Hua Chen
- Department of Obstetrics and Gynecology, Far Eastern Memorial Hospital, New Taipei 220409, Taiwan; (W.-H.T.); (H.-H.C.); (M.-C.W.); (H.-H.L.)
| | - Ming-Chow Wei
- Department of Obstetrics and Gynecology, Far Eastern Memorial Hospital, New Taipei 220409, Taiwan; (W.-H.T.); (H.-H.C.); (M.-C.W.); (H.-H.L.)
| | - Ho-Hsiung Lin
- Department of Obstetrics and Gynecology, Far Eastern Memorial Hospital, New Taipei 220409, Taiwan; (W.-H.T.); (H.-H.C.); (M.-C.W.); (H.-H.L.)
- Department of Obstetrics and Gynecology, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Sheng-Mou Hsiao
- Department of Obstetrics and Gynecology, Far Eastern Memorial Hospital, New Taipei 220409, Taiwan; (W.-H.T.); (H.-H.C.); (M.-C.W.); (H.-H.L.)
- Department of Obstetrics and Gynecology, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei 100225, Taiwan
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan
| |
Collapse
|
37
|
Wang Y, Li C, Pan C, Liu E, Zhao X, Ling Q. Alterations to transcriptomic profile, histopathology, and oxidative stress in liver of pikeperch (Sander lucioperca) under heat stress. FISH & SHELLFISH IMMUNOLOGY 2019; 95:659-669. [PMID: 31706008 DOI: 10.1016/j.fsi.2019.11.014] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/29/2019] [Accepted: 11/05/2019] [Indexed: 05/27/2023]
Abstract
Pikeperch (Sander lucioperca) is an economically important cool-water fish. In recent years, its cultivation has become threatened by higher temperatures in summer. We previously investigated the effects of heat stress on pikeperch liver under different temperatures, but the molecular mechanism of the heat-stress response is still unknown. This study applied consistent heat stress (29 °C, 0-48 h) to pikeperch juveniles, and a transcriptomic profile of pikeperch liver under heat stress (29 °C, 0 h) was performed by RNA-Seq. The antioxidant status, changes in liver histology, and antioxidant gene expression at different time points were examined. We identified 403 differentially expressed genes (DEGs), many of which were enriched in KEGG pathways, including protein processing in endoplasmic reticulum (ER), insulin signaling, and immune-related pathways. Among these, the most significant heat-stress-related pathway was protein processing in ER, indicating that this pathway is critical for the heat-stress response. After consistent heat stress at 29 °C, the total antioxidant capacity (T-AOC), the activities of total superoxide dismutase (T-SOD) and catalase (CAT), and the mRNA expression of manganese SOD (Mn-SOD), CAT, and glutathione peroxidase 1 and 7 (GPx1 and GPx7) in the treated groups showed the same trend of first increasing and then decreasing. Levels of malondialdehyde (MDA) content did not show significant differences between samples at 0 h and 3 h, but significantly increased by 6 h, and thereafter decreased. The liver tissue was normal at 0 h (29 °C); however, it suffered histological damage with increased duration of the heat stress. Above all, heat stress at 29 °C seemed to cause oxidative damage and dysfunction in pikeperch liver between 3 h and 48 h. The present results indicate that pikeperch have the capacity to defend against heat stress and maintain relative balance of oxidation-reduction reactions mainly through activating the antioxidant system, protein processing in ER, the insulin-signaling pathway, and immune-related pathways.
Collapse
Affiliation(s)
- Yunfeng Wang
- School of Biology and Basic Medical Sciences, Soochow University, 199 Renai Road, Suzhou, Jiangsu, PR China
| | - Caijuan Li
- School of Biology and Basic Medical Sciences, Soochow University, 199 Renai Road, Suzhou, Jiangsu, PR China
| | - Chenglong Pan
- School of Biology and Basic Medical Sciences, Soochow University, 199 Renai Road, Suzhou, Jiangsu, PR China
| | - Enguang Liu
- School of Biology and Basic Medical Sciences, Soochow University, 199 Renai Road, Suzhou, Jiangsu, PR China
| | - Xuqian Zhao
- School of Biology and Basic Medical Sciences, Soochow University, 199 Renai Road, Suzhou, Jiangsu, PR China
| | - Qufei Ling
- School of Biology and Basic Medical Sciences, Soochow University, 199 Renai Road, Suzhou, Jiangsu, PR China.
| |
Collapse
|
38
|
Shrestha B, Tang L, Romero G. Nanoparticles‐Mediated Combination Therapies for Cancer Treatment. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900076] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Binita Shrestha
- Department of Biomedical Engineering University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| | - Liang Tang
- Department of Biomedical Engineering University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| | - Gabriela Romero
- Department of Chemical Engineering University of Texas at San Antonio One UTSA Circle San Antonio TX 78249 USA
| |
Collapse
|
39
|
Mahanty A, Purohit GK, Mohanty S, Mohanty BP. Heat stress-induced alterations in the expression of genes associated with gonadal integrity of the teleost Puntius sophore. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:1409-1417. [PMID: 31144086 DOI: 10.1007/s10695-019-00643-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 04/22/2019] [Indexed: 06/09/2023]
Abstract
Temperature plays an important role on reproductive physiology of vertebrates including mammals, fish, and birds. It has varying effects on fish reproduction depending on the species; higher temperatures favor the spring-spawning species, while lower temperatures stimulate reproduction in autumn spawners. To evaluate the impact of high temperature on the reproductive physiology of minnow Puntius sophore, we carried out expression analysis of selected genes associated with gamete quality (hsp60, hsp70, hsp90, hsf1, vtg), pleuripotency (sox2, oct4, nanog), and sex determination (dmrt1) in gonads (ovary and testis) of P. sophore, heat stressed for different time periods (36 °C/7 days or 60 days) using real-time quantitative polymerase chain reaction (RT-qPCR). Expression of most of the hsp, vtg, and pleuripotency marker genes sox-2, oct-4, and nanog genes was downregulated in both ovary and testis of heat-stressed fish. The expression of dmrt-1 was upregulated in testis but downregulated in ovary of the heat-stressed fish which could be a male favoring effect of high temperature in P. sophore. This study suggests that the reproductive physiology and health of the nutrient dense P. sophore would be negatively affected by high temperature stress.
Collapse
Affiliation(s)
- Arabinda Mahanty
- Fishery Resource and Environmental Management Division, Biochemistry Laboratory, ICAR - Central Inland Fisheries Research Institute, Barrackpore, Kolkata, 700120, India
- Crop Protection Division, ICAR-National Rice Research Institute, Cuttack, 753 006, India
| | - Gopal Krishna Purohit
- School of Biotechnology, KIIT Deemed University, Bhubaneswar, 751024, India
- Santaan Fertility Centre and Research Institute, KIIT-TBI, KIIT Deemed University, Bhubaneswar, 751024, India
| | - Sasmita Mohanty
- School of Biotechnology, KIIT Deemed University, Bhubaneswar, 751024, India.
- Department of Biotechnology, Ramadevi Women's University, Bhubaneswar, India.
| | - Bimal Prasanna Mohanty
- Fishery Resource and Environmental Management Division, Biochemistry Laboratory, ICAR - Central Inland Fisheries Research Institute, Barrackpore, Kolkata, 700120, India.
| |
Collapse
|
40
|
Farolfi A, Gurioli G, Fugazzola P, Burgio SL, Casanova C, Ravaglia G, Altavilla A, Costantini M, Amadori A, Framarini M, Ansaloni L, De Giorgi U. Immune System and DNA Repair Defects in Ovarian Cancer: Implications for Locoregional Approaches. Int J Mol Sci 2019; 20:E2569. [PMID: 31130614 PMCID: PMC6566239 DOI: 10.3390/ijms20102569] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/07/2019] [Accepted: 05/23/2019] [Indexed: 01/26/2023] Open
Abstract
In the last few years, substantial progress has been made in the treatment of ovarian cancer, with increased knowledge about the biology of the disease. Ovarian cancer is a neoplasm strongly linked to defects in DNA repair mechanisms, where deficiency in the homologous recombination (HR) system results in a better response of ovarian cancers to therapy, whether platinum-based chemotherapy, anthracyclines, or poly (ADP-ribose) polymerase (PARP) inhibitors. More recently, it has been demonstrated that different ovarian cancer histotypes may have different immunogenicity. Interestingly, defects in HR systems are associated more frequently with higher tumor infiltrating lymphocytes, providing a rationale for developing combination therapy with immune-modulating agents and PARP inhibitors. Again, locoregional therapies combining heat shock and chemotherapy delivery have been shown to induce an anticancer immune response in vitro. Thus, the potential for locoregional therapeutic approaches that may impact the immune system, perhaps in combination with immune-modulating agents or PARP inhibitors, needs to be further explored. With this premise, we reviewed the main biological and clinical data demonstrating a strict interplay between the immune system, DNA repair mechanisms, and intraperitoneal therapies in ovarian cancer, with a focus on potential future therapeutic implications.
Collapse
Affiliation(s)
- Alberto Farolfi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy.
| | - Giorgia Gurioli
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy.
| | - Paola Fugazzola
- General and Emergency Surgery, Maurizio Bufalini Hospital, Cesena 47521, Italy.
| | - Salvatore Luca Burgio
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy.
| | - Claudia Casanova
- Oncology Department, Santa Maria delle Croci Hospital, Ravenna 48121, Italy.
| | - Giorgia Ravaglia
- Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy.
| | - Amelia Altavilla
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy.
| | | | - Andrea Amadori
- Department of Gynecological, Morgagni-Pierantoni Hospital, Forlì 47121, Italy.
| | - Massimo Framarini
- Department of General Surgery, Morgagni-Pierantoni Hospital, Forlì 47121, Italy.
| | - Luca Ansaloni
- General and Emergency Surgery, Maurizio Bufalini Hospital, Cesena 47521, Italy.
| | - Ugo De Giorgi
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola 47014, Italy.
| |
Collapse
|
41
|
Zhu L, Xu Y, Shan Y, Zheng R, Wu Z, Ma S. Intraperitoneal perfusion chemotherapy and whole abdominal hyperthermia using external radiofrequency following radical D2 resection for treatment of advanced gastric cancer. Int J Hyperthermia 2019; 36:403-407. [PMID: 30829551 DOI: 10.1080/02656736.2019.1579372] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Revised: 01/26/2019] [Accepted: 02/02/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND The peritoneum is the most frequent site of disease recurrence in gastric cancer, and the prognosis remains poor. This study assessed the role of adjuvant intraperitoneal (IP) chemotherapy with whole abdominal hyperthermia using external radiofrequency in gastric cancer patients after D2 dissection. METHODS Patients with gastric cancer who underwent gastrectomy with D2 regional lymph node dissection were enrolled in the study. Patients received IP chemotherapy with whole abdominal hyperthermia. Preheated normal saline containing 75 mg/m2 of cisplatin was delivered into the abdominal cavity through a Tenckhoff catheter at McBurney's point. Regional hyperthermia was performed using two sets of orthogonal radiofrequency waves immediately after all saline was irrigated into the abdominal cavity. For each patient, recurrent or metastatic sites and adverse events were evaluated. RESULTS A total of 22 patients were finally included. All patients tolerated hyperthermia well. Only two patients experienced grade 1 superficial thermal injury. The most frequent grade 3/4 adverse events were myelosuppression, nausea/vomiting, trichomadesis and liver dysfunction. We also found IP chemotherapy with whole abdominal hyperthermia could reduce the total recurrent/metastatic rate, especially peritoneal metastasis (4.5%). CONCLUSIONS This hypothesis-generating study indicated that IP chemotherapy with whole abdominal hyperthermia might be feasible for gastric cancer patients after D2 resection.
Collapse
Affiliation(s)
- Lucheng Zhu
- a Department of Radiotherapy , Hangzhou Cancer Hospital , Hangzhou , PR China
- b Department of Oncology, Affiliated Hangzhou First People's Hospital , Zhejiang University School of Medicine , Hangzhou , PR China
| | - Yasi Xu
- b Department of Oncology, Affiliated Hangzhou First People's Hospital , Zhejiang University School of Medicine , Hangzhou , PR China
| | - Yuqiang Shan
- c Department of Gastrointestinal Surgery, Affiliated Hangzhou First People's Hospital , Zhejiang University School of Medicine , Hangzhou , PR China
| | - Ruzhen Zheng
- a Department of Radiotherapy , Hangzhou Cancer Hospital , Hangzhou , PR China
| | - Zhibing Wu
- b Department of Oncology, Affiliated Hangzhou First People's Hospital , Zhejiang University School of Medicine , Hangzhou , PR China
| | - Shenglin Ma
- b Department of Oncology, Affiliated Hangzhou First People's Hospital , Zhejiang University School of Medicine , Hangzhou , PR China
| |
Collapse
|
42
|
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
|
43
|
Sarhan M, Land WG, Tonnus W, Hugo CP, Linkermann A. Origin and Consequences of Necroinflammation. Physiol Rev 2018; 98:727-780. [PMID: 29465288 DOI: 10.1152/physrev.00041.2016] [Citation(s) in RCA: 141] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
When cells undergo necrotic cell death in either physiological or pathophysiological settings in vivo, they release highly immunogenic intracellular molecules and organelles into the interstitium and thereby represent the strongest known trigger of the immune system. With our increasing understanding of necrosis as a regulated and genetically determined process (RN, regulated necrosis), necrosis and necroinflammation can be pharmacologically prevented. This review discusses our current knowledge about signaling pathways of necrotic cell death as the origin of necroinflammation. Multiple pathways of RN such as necroptosis, ferroptosis, and pyroptosis have been evolutionary conserved most likely because of their differences in immunogenicity. As the consequence of necrosis, however, all necrotic cells release damage associated molecular patterns (DAMPs) that have been extensively investigated over the last two decades. Analysis of necroinflammation allows characterizing specific signatures for each particular pathway of cell death. While all RN-pathways share the release of DAMPs in general, most of them actively regulate the immune system by the additional expression and/or maturation of either pro- or anti-inflammatory cytokines/chemokines. In addition, DAMPs have been demonstrated to modulate the process of regeneration. For the purpose of better understanding of necroinflammation, we introduce a novel classification of DAMPs in this review to help detect the relative contribution of each RN-pathway to certain physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Maysa Sarhan
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Walter G Land
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Wulf Tonnus
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Christian P Hugo
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| | - Andreas Linkermann
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University Vienna , Vienna , Austria ; INSERM UMR_S 1109, Laboratory of Excellence Transplantex, University of Strasbourg , Strasbourg , France ; German Academy of Transplantation Medicine, Munich , Germany ; and Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden , Dresden , Germany
| |
Collapse
|
44
|
Uehara Y, Temma K, Kobayashi Y, Irie N, Yamaguchi T. Reduction of Thermotolerance by Heat Shock Protein 90 Inhibitors in Murine Erythroleukemia Cells. Biol Pharm Bull 2018; 41:1393-1400. [PMID: 30175776 DOI: 10.1248/bpb.b18-00190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cells induce heat shock proteins (HSPs) against various stress. However, murine erythroleukemia (MEL) cells do not express HSP72, a heat-inducible member of HSP70 family. So, it is of interest to examine how MEL cells respond to heat stress (44°C, 30 min). Heat stress-induced apoptosis was suppressed by pretreatment of heat shock (44°C, 10 min). Such suppressive effects were maximal at 6 h after heat shock and remained up to 12 h. Interestingly, such effects of heat shock were abrogated by specific inhibitors of HSP90 such as 17-allylamino-17-demethoxygeldanamycin (17-AAG) and novobiocin. From flow cytometric analysis, it was found that MEL cells arrest in G2 phase at 6 h after heat shock, but restore original cell cycle at 12 h. High expression level of HSP90 was maintained before and after heat shock. Phosphorylation of HSP90α was observed in apoptotic cells induced by heat stress, but inhibited by pretreatment of heat shock. Such inhibition was abrogated by 17-AAG. Moreover, c-Jun NH2-terminal kinase (JNK) was activated in heat stress-induced apoptotic cells. Taken together, these results suggest that HSP90α in MEL cells plays an important role in the thermotolerance, i.e., suppression of heat stress-induced apoptosis by heat shock.
Collapse
Affiliation(s)
- Yousuke Uehara
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Kazunari Temma
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Yuuya Kobayashi
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Nobuyuki Irie
- Department of Chemistry, Faculty of Science, Fukuoka University
| | - Takeo Yamaguchi
- Department of Chemistry, Faculty of Science, Fukuoka University
| |
Collapse
|
45
|
Batista TP, Sarmento BJQ, Loureiro JF, Petruzziello A, Lopes A, Santos CC, Quadros CDA, Akaishi EH, Cordeiro EZ, Coimbra FJF, Laporte GA, Castro LS, Batista RMSS, Aguiar S, Costa WL, Ferreira FO. A proposal of Brazilian Society of Surgical Oncology (BSSO/SBCO) for standardizing cytoreductive surgery (CRS) plus hyperthermic intraperitoneal chemotherapy (HIPEC) procedures in Brazil: pseudomixoma peritonei, appendiceal tumors and malignant peritoneal mesothelioma. ACTA ACUST UNITED AC 2018; 44:530-544. [PMID: 29019584 DOI: 10.1590/0100-69912017005016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 06/08/2017] [Indexed: 12/13/2022]
Abstract
Cytoreductive surgery plus hypertermic intraperitoneal chemotherapy has emerged as a major comprehensive treatment of peritoneal malignancies and is currently the standard of care for appendiceal epithelial neoplasms and pseudomyxoma peritonei syndrome as well as malignant peritoneal mesothelioma. Unfortunately, there are some worldwide variations of the cytoreductive surgery and hypertermic intraperitoneal chemotherapy techniques since no single technique has so far demonstrated its superiority over the others. Therefore, standardization of practices might enhance better comparisons between outcomes. In these settings, the Brazilian Society of Surgical Oncology considered it important to present a proposal for standardizing cytoreductive surgery plus hypertermic intraperitoneal chemotherapy procedures in Brazil, with a special focus on producing homogeneous data for the developing Brazilian register for peritoneal surface malignancies.
Collapse
Affiliation(s)
- Thales Paulo Batista
- - Medicina Integral Professor Fernando Figueira Institute, Department of Surgery / Oncology, Recife, PE, Brazil.,- University of Pernambuco, Department of Surgery, Recife, PE, Brazil
| | | | | | - Andrea Petruzziello
- - Marcelino Champagnat Hospital, Department of Surgical Oncology, Curitiba, PR, Brazil.,- AC Camargo Cancer Center, Department of Abdominal Surgery, São Paulo, SP, Brazil
| | - Ademar Lopes
- - AC Camargo Cancer Center, Department of Pelvic Surgery, São Paulo, SP, Brazil
| | | | | | - Eduardo Hiroshi Akaishi
- - Hospital das Clínicas, University of São Paulo, Department of Surgical Oncology, São Paulo, SP, Brazil
| | | | | | - Gustavo Andreazza Laporte
- - Santa Casa de Misericórdia de Porto Alegre, Department of Surgical Oncology, Porto Alegre, RS, Brazil
| | - Leonaldson Santos Castro
- - Complexo Hospitalar de Niterói, Service of Surgical Oncology, Niterói, RJ, Brazil.,- Nacional Cancer Institute, Service of Abdomino-Pelvic Surgery, Rio de Janeiro, RJ, Brazil
| | | | - Samuel Aguiar
- - AC Camargo Cancer Center, Department of Pelvic Surgery, São Paulo, SP, Brazil
| | - Wilson Luiz Costa
- - AC Camargo Cancer Center, Department of Abdominal Surgery, São Paulo, SP, Brazil
| | | | | |
Collapse
|
46
|
Garg AD, Agostinis P. Cell death and immunity in cancer: From danger signals to mimicry of pathogen defense responses. Immunol Rev 2018; 280:126-148. [PMID: 29027218 DOI: 10.1111/imr.12574] [Citation(s) in RCA: 308] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The immunogenicity of cancer cells is an emerging determinant of anti-cancer immunotherapy. Beyond developing immunostimulatory regimens like dendritic cell-based vaccines, immune-checkpoint blockers, and adoptive T-cell transfer, investigators are beginning to focus on the immunobiology of dying cancer cells and its relevance for the success of anticancer immunotherapies. It is currently accepted that cancer cells may die in response to anti-cancer therapies through regulated cell death programs, which may either repress or increase their immunogenic potential. In particular, the induction of immunogenic cancer cell death (ICD), which is hallmarked by the emission of damage-associated molecular patterns (DAMPs); molecules analogous to pathogen-associated molecular patterns (PAMPs) acting as danger signals/alarmins, is of great relevance in cancer therapy. These ICD-associated danger signals favor immunomodulatory responses that lead to tumor-associated antigens (TAAs)-directed T-cell immunity, which paves way for the removal of residual, treatment-resistant cancer cells. It is also emerging that cancer cells succumbing to ICD can orchestrate "altered-self mimicry" i.e. mimicry of pathogen defense responses, on the levels of nucleic acids and/or chemokines (resulting in type I interferon/IFN responses or pathogen response-like neutrophil activity). In this review, we exhaustively describe the main molecular, immunological, preclinical, and clinical aspects of immunosuppressive cell death or ICD (with respect to apoptosis, necrosis and necroptosis). We also provide an extensive historical background of these fields, with special attention to the self/non-self and danger models, which have shaped the field of cell death immunology.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Batista TP, Carneiro VCG, Tancredi R, Teles ALB, Badiglian-Filho L, Leão CS. Neoadjuvant chemotherapy followed by fast-track cytoreductive surgery plus short-course hyperthermic intraperitoneal chemotherapy (HIPEC) in advanced ovarian cancer: preliminary results of a promising all-in-one approach. Cancer Manag Res 2017; 9:869-878. [PMID: 29263704 PMCID: PMC5732565 DOI: 10.2147/cmar.s153327] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Purpose Hyperthermic intraperitoneal chemotherapy (HIPEC) has been considered a promising treatment option for advanced or recurrent ovarian cancer, but there is no clear evidence based on randomized controlled trials to advocate this approach as a standard therapy. In this study, we aim to present the early outcomes and insights after an interim analysis of a pioneering clinical trial in Brazil. Methods This study was a cross-sectional analysis of early data from our ongoing clinical trial – an open-label, double-center, single-arm trial on the safety and efficacy of using HIPEC for advanced ovarian cancer (ClinicalTrials.gov: NCT02249013). A fast-track recovery strategy was also applied to improve patient outcomes. Results Nine patients with stage IIIB (n=1) or IIIC (n=8) epithelial malignancies were enrolled until February 2017. The median (range) serum CA125 level at diagnosis was 692 (223.7–6550) U/mL. The median number of preoperative cycles of intravenous (i.v.) chemotherapy was 3 (2–4), resulting in peritoneal cancer index scores of 9 (3–18) at the time of HIPEC. Time of restarting i.v. chemotherapy was 37 (33–50) days with all patients completing 6 cycles as planned. The median operation time was 395 (235–760) minutes, the length of hospital stay was 4 (3–10) days, and all the patients left the ICU on the morning after the procedure. Two patients experienced no postoperative complications, whereas 91% of the complications were minor G1/G2 events. Preliminary assessment also suggested no impairment of the patient’s quality of life. Conclusion Our comprehensive protocol might represent a promising all-in-one approach for advanced ovarian cancer. The patient recruitment for this trial is ongoing.
Collapse
Affiliation(s)
- Thales Paulo Batista
- Department of Surgery/Oncology, IMIP - Instituto de Medicina Integral Professor Fernando Figueira.,Department of Surgery, UFPE - Universidade Federal de Pernambuco
| | - Vandré Cabral G Carneiro
- Department of Surgery/Oncology, IMIP - Instituto de Medicina Integral Professor Fernando Figueira.,Department of Gynecology, HCP - Hospital de Câncer de Pernambuco
| | - Rodrigo Tancredi
- Department of Clinical Oncology, IMIP - Instituto de Medicina Integral Professor Fernando Figueira.,Department of Clinical Oncology, HCP - Hospital de Câncer de Pernambuco
| | - Ana Ligia Bezerra Teles
- Department of Anaesthesiology, IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Recife
| | | | - Cristiano Souza Leão
- Department of Surgery, IMIP - Instituto de Medicina Integral Professor Fernando Figueira, Recife, Brazil
| |
Collapse
|
48
|
Mahanty A, Mohanty S, Mohanty BP. Dietary supplementation of curcumin augments heat stress tolerance through upregulation of nrf-2-mediated antioxidative enzymes and hsps in Puntius sophore. FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:1131-1141. [PMID: 28315162 DOI: 10.1007/s10695-017-0358-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 02/23/2017] [Indexed: 06/06/2023]
Abstract
Heat stress is one of the major environmental concerns in global warming regime and rising temperature has resulted in mass mortalities of animals including fishes. Therefore, strategies for high temperature stress tolerance and ameliorating the effects of heat stress are being looked for. In an earlier study, we reported that Nrf-2 (nuclear factor E2-related factor 2) mediated upregulation of antioxidative enzymes and heat shock proteins (Hsps) provide survivability to fish under heat stress. In this study, we have evaluated the ameliorative potential of dietary curcumin, a potential Nrf-2 inducer in heat stressed cyprinid Puntius sophore. Fishes were fed with diet supplemented with 0.5, 1.0, and 1.5% curcumin at the rate 2% of body weight daily in three separate groups (n = 40 in each group) for 60 days. Fishes fed with basal diet (without curcumin) served as the control (n = 40). Critical thermal maxima (CTmax) was determined for all the groups (n = 10, in duplicates) after the feeding trial. Significant increase in the CTmax was observed in the group fed with 1.5% curcumin- supplemented fishes whereas it remained similar in groups fed with 0.5%, and 1% curcumin-supplemented diet, as compared to control. To understand the molecular mechanism of elevated thermotolerance in the 1.5% curcumin supplemented group, fishes were given a sub-lethal heat shock treatment (36 °C) for 6 h and expression analysis of nrf-2, keap-1, sod, catalase, gpx, and hsp27, hsp60, hsp70, hsp90, and hsp110 was carried out using RT-PCR. In the gill, expression of nrf-2, sod, catalase, gpx, and hsp60, hsp70, hsp90, and hsp110 was found to be elevated in the 1.5% curcumin-fed heat-shocked group compared to control and the basal diet-fed, heat-shocked fishes. Similarly, in the liver, upregulation in expression of nrf-2, sod, catalase, and hsp70 and hsp110 was observed in 1.5% curcumin supplemented and heat shocked group. Thus, this study showed that supplementation of curcumin augments tolerance to high temperature stress in P. sophore that could be attributed to nrf-2-induced upregulation of antioxidative enzymes sod, catalase, gpx, and the hsps.
Collapse
Affiliation(s)
- Arabinda Mahanty
- Fishery Resource and Environmental Management Division, Biochemistry Laboratory, ICAR - Central Inland Fisheries Research Institute, Barrackpore, Kolkata, 700120, India
- School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | - Sasmita Mohanty
- School of Biotechnology, KIIT University, Bhubaneswar, 751024, India
| | - Bimal P Mohanty
- Fishery Resource and Environmental Management Division, Biochemistry Laboratory, ICAR - Central Inland Fisheries Research Institute, Barrackpore, Kolkata, 700120, India.
| |
Collapse
|
49
|
Mahanty A, Purohit GK, Yadav RP, Mohanty S, Mohanty BP. hsp90 and hsp47 appear to play an important role in minnow Puntius sophore for surviving in the hot spring run-off aquatic ecosystem. FISH PHYSIOLOGY AND BIOCHEMISTRY 2017; 43:89-102. [PMID: 27522494 DOI: 10.1007/s10695-016-0270-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 08/01/2016] [Indexed: 06/06/2023]
Abstract
Changes in the expression of a number of hsp genes in minnow Puntius sophore collected from a hot spring run-off (Atri hot spring in Odisha, India; 20o09'N 85°18'E, 36-38 °C) were investigated to study the upper thermal acclimation response under heat stress, using same species from aquaculture ponds (water temperature 27 °C) as control. Expression of hsp genes was analyzed in both groups using RT-qPCR, which showed up-regulation of hsp90 (2.1-fold) and hsp47 (2.5-fold) in hot spring run-off fishes, whereas there was no alteration in expression of other hsps. As the fish inhabit the hot spring run-off area for very long duration, they could have adapted to the environment. To test this hypothesis, fishes collected from hot spring run-off were divided into two groups; one was heat-shocked at 41 °C/24 h, and the other was acclimatized at 27 °C/24 h. Up-regulation of all the hsps (except hsp78) was observed in the heat-shocked fishes, whereas expression of all hsps was found to be down-regulated to the basal level in fishes maintained at 27 °C/24 h. Pathway analysis showed that the expressions of all the hsps except hsp90 are regulated by the transcription factor heat shock factor 1 (Hsf1). This study showed that hsp90 and hsp47 play an important role in Puntius sophore for surviving in the high-temperature environment of the hot spring run-off. Additionally, we show that plasticity in hsp gene expression is not lost in the hot spring run-off population.
Collapse
Affiliation(s)
- Arabinda Mahanty
- Biochemistry Laboratory, Proteomics Unit, Fishery Resource and Environmental Management Division, ICAR- Central Inland Fisheries Research Institute, Barrackpore, Kolkata, 700120, India
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India
| | | | - Ravi Prakash Yadav
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India
| | - Sasmita Mohanty
- KIIT School of Biotechnology, KIIT University, Bhubaneswar, Odisha, 751024, India
| | - Bimal Prasanna Mohanty
- Biochemistry Laboratory, Proteomics Unit, Fishery Resource and Environmental Management Division, ICAR- Central Inland Fisheries Research Institute, Barrackpore, Kolkata, 700120, India.
| |
Collapse
|
50
|
Land WG, Agostinis P, Gasser S, Garg AD, Linkermann A. Transplantation and Damage-Associated Molecular Patterns (DAMPs). Am J Transplant 2016; 16:3338-3361. [PMID: 27421829 DOI: 10.1111/ajt.13963] [Citation(s) in RCA: 113] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 06/24/2016] [Accepted: 07/10/2016] [Indexed: 01/25/2023]
Abstract
Upon solid organ transplantation and during cancer immunotherapy, cellular stress responses result in the release of damage-associated molecular patterns (DAMPs). The various cellular stresses have been characterized in detail over the last decades, but a unifying classification based on clinically important aspects is lacking. Here, we provide an in-depth review of the most recent literature along with a unifying concept of the danger/injury model, suggest a classification of DAMPs, and review the recently elaborated mechanisms that result in the emission of such factors. We further point out the differences in DAMP responses including the release following a heat shock pattern, endoplasmic reticulum stress, DNA damage-mediated DAMP release, and discuss the diverse pathways of regulated necrosis in this respect. The understanding of various forms of DAMPs and the consequences of their different release patterns are prerequisite to associate serum markers of cellular stresses with clinical outcomes.
Collapse
Affiliation(s)
- W G Land
- German Academy of Transplantation Medicine, Munich, Germany.,Laboratoire d'ImmunoRhumatologie Moléculaire, INSERM UMR_S1109, Plateforme GENOMAX, Faculté de Médecine, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Université de Strasbourg, Strasbourg, France.,LabexTRANSPLANTEX, Faculté de Médecine, Université de Strasbourg, Strasbourg, France
| | - P Agostinis
- Cell Death Research and Therapy (CDRT) Lab, Department of Cellular and Molecular Medicine, KU Leuven, University of Leuven, Leuven, Belgium
| | - S Gasser
- Immunology Programme and Department of Microbiology and Immunology, Centre for Life Sciences, National University of Singapore, Singapore, Singapore
| | - A D Garg
- Cell Death Research and Therapy (CDRT) Lab, Department of Cellular and Molecular Medicine, KU Leuven, University of Leuven, Leuven, Belgium
| | - A Linkermann
- Cluster of Excellence EXC306, Inflammation at Interfaces, Schleswig-Holstein, Germany.,Clinic for Nephrology and Hypertension, Christian-Albrechts-University, Kiel, Germany
| |
Collapse
|