1
|
Quan Y, He J, Zou Q, Zhang L, Sun Q, Huang H, Li W, Xie K, Wei F. Low molecular weight heparin synergistically enhances the efficacy of adoptive and anti-PD-1-based immunotherapy by increasing lymphocyte infiltration in colorectal cancer. J Immunother Cancer 2023; 11:e007080. [PMID: 37597850 PMCID: PMC10441131 DOI: 10.1136/jitc-2023-007080] [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] [Accepted: 08/04/2023] [Indexed: 08/21/2023] Open
Abstract
BACKGROUND Immunotherapy, including adoptive cell therapy (ACT) and immune checkpoint inhibitors (ICIs), has a limited effect in most patients with colorectal cancer (CRC), and the efficacy is further limited in patients with liver metastasis. Lack of antitumor lymphocyte infiltration could be a major cause, and there remains an urgent need for more potent and safer therapies for CRC. METHODS In this study, the antitumoral synergism of low molecular weight heparin (LMWH) combined with immunotherapy in the microsatellite stable (MSS) highly aggressive murine model of CRC was fully evaluated. RESULTS Dual LMWH and ACT objectively mediated the stagnation of tumor growth and inhibition of liver metastasis, neither LMWH nor ACT alone had any antitumoral activity on them. The combination of LMWH and ACT obviously increased the infiltration of intratumor CD8+ T cells, as revealed by multiplex immunohistochemistry, purified CD8+ T-cell transfer assay, and IVIM in vivo imaging. Mechanistically, evaluation of changes in the tumor microenvironment revealed that LMWH improved tumor vascular normalization and facilitated the trafficking of activated CD8+ T cells into tumors. Similarly, LMWH combined with anti-programmed cell death protein 1 (PD-1) therapy provided superior antitumor activity as compared with the single PD-1 blockade in murine CT26 tumor models. CONCLUSIONS LMWH could enhance ACT and ICIs-based immunotherapy by increasing lymphocyte infiltration into tumors, especially cytotoxic CD8+ T cells. These results indicate that combining LMWH with an immunotherapy strategy presents a promising and safe approach for CRC treatment, especially in MSS tumors.
Collapse
Affiliation(s)
- Yibo Quan
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| | - Jie He
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
- Center for Pancreatic Cancer Research and Department of Immunology, The South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| | - Qi Zou
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
- Center for Pancreatic Cancer Research and Department of Immunology, The South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| | - Liuxi Zhang
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
- Center for Pancreatic Cancer Research and Department of Immunology, The South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| | - Qihui Sun
- Center for Pancreatic Cancer Research and Department of Immunology, The South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| | - Hongli Huang
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| | - Wanglin Li
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| | - Keping Xie
- Center for Pancreatic Cancer Research and Department of Immunology, The South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| | - Fang Wei
- Guangzhou Digestive Disease Center, Guangzhou First People's Hospital and The Second Affiliated Hospital, South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
- Center for Pancreatic Cancer Research and Department of Immunology, The South China University of Technology School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
2
|
Georgilis E, Gavriatopoulou M, Tsilimigras DI, Malandrakis P, Theodosopoulos T, Ntanasis-Stathopoulos I. Optimizing Adjuvant Therapy after Surgery for Colorectal Cancer Liver Metastases: A Systematic Review. J Clin Med 2023; 12:jcm12062401. [PMID: 36983401 PMCID: PMC10051548 DOI: 10.3390/jcm12062401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 03/09/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
The liver is the most common site of colorectal cancer metastatic spread. Although metastasectomy is the gold standard for fit patients with resectable colorectal cancer liver metastases (CRLMs), their management after surgical treatment remains controversial. The objective of this systematic review was to collate the currently available data of the agents used in the adjuvant setting in order to define the most optimal therapeutic strategy. A systematic review of the literature was conducted by searching PubMed/Medline and Cochrane library databases. We included studies that evaluated the efficacy, the tolerability and the safety profile of various chemotherapeutic agents that are used as adjuvant treatment after surgical resection of CRLMs. The outcomes of interest were regression-free survival (RFS), disease-free survival (DFS), overall survival (OS) and severe toxicities. From 543 initial articles, 29 publications with 7028 patients were finally included. In general, the results of the eligible studies indicated that adjuvant therapy after resection of CRLMs led to improved RFS/DFS rates, but this benefit did not contribute to a statistically significant prolongation of OS. Moreover, the choice of the therapeutic strategy, namely systematic or regional chemotherapy or the combination of both, did not seem to have a differential impact on patient outcomes. However, these results should be interpreted with caution since the majority of the chosen studies are of low or moderate quality. In this context, further high-quality clinical trials conducted on patient sub-populations with modern therapies are required in order to reduce in-study and between-study heterogeneity and determine which patients are expected to derive the maximum benefit from adjuvant therapy after surgery for CRLMs.
Collapse
Affiliation(s)
- Emmanouil Georgilis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Panagiotis Malandrakis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Theodosios Theodosopoulos
- Second Department of Surgery, Aretaieion University Hospital, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| |
Collapse
|
3
|
Kazemi MH, Sadri M, Najafi A, Rahimi A, Baghernejadan Z, Khorramdelazad H, Falak R. Tumor-infiltrating lymphocytes for treatment of solid tumors: It takes two to tango? Front Immunol 2022; 13:1018962. [PMID: 36389779 PMCID: PMC9651159 DOI: 10.3389/fimmu.2022.1018962] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Accepted: 10/14/2022] [Indexed: 07/30/2023] Open
Abstract
Tumor-infiltrating lymphocytes (TILs), frontline soldiers of the adaptive immune system, are recruited into the tumor site to fight against tumors. However, their small number and reduced activity limit their ability to overcome the tumor. Enhancement of TILs number and activity against tumors has been of interest for a long time. A lack of knowledge about the tumor microenvironment (TME) has limited success in primary TIL therapies. Although the advent of engineered T cells has revolutionized the immunotherapy methods of hematologic cancers, the heterogeneity of solid tumors warrants the application of TILs with a wide range of specificity. Recent advances in understanding TME, immune exhaustion, and immune checkpoints have paved the way for TIL therapy regimens. Nowadays, TIL therapy has regained attention as a safe personalized immunotherapy, and currently, several clinical trials are evaluating the efficacy of TIL therapy in patients who have failed conventional immunotherapies. Gaining favorable outcomes following TIL therapy of patients with metastatic melanoma, cervical cancer, ovarian cancer, and breast cancer has raised hope in patients with refractory solid tumors, too. Nevertheless, TIL therapy procedures face several challenges, such as high cost, timely expansion, and technical challenges in selecting and activating the cells. Herein, we reviewed the recent advances in the TIL therapy of solid tumors and discussed the challenges and perspectives.
Collapse
Affiliation(s)
- Mohammad Hossein Kazemi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Sadri
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Alireza Najafi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Ali Rahimi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Baghernejadan
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Reza Falak
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
4
|
Yuan J, Li J, Gao C, Jiang C, Xiang Z, Wu J. Immunotherapies catering to the unmet medical need of cold colorectal cancer. Front Immunol 2022; 13:1022190. [PMID: 36275766 PMCID: PMC9579278 DOI: 10.3389/fimmu.2022.1022190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/21/2022] [Indexed: 11/13/2022] Open
Abstract
As a common malignant tumor of gastrointestinal tract, the incidence of colorectal cancer (CRC) has gradually increased in recent years. In western developed countries, it has even become the second largest malignant tumor next to lung cancer. Immunotherapy is a hot topic in the field of cancer therapy, including immune checkpoint blockade (ICB), adoptive cell therapy (ACT), cancer vaccines and cytokines, aiming to improve the ability of the immune system to recognize, target and eliminate cancer cells. However, cold CRC, which accounts for a high proportion of CRC, is not so reactive to it. The development of immunotherapy to prevent cancer cells from forming "immune escape" pathways to the immune system in cold CRC, has been under increasing study attention. There is proof that an organic combination of radiotherapy, chemotherapy, and several immunotherapies can considerably boost the immune system's capacity to eradicate tumor cells. In this review, we summarized the role of immunotherapy in colorectal cancer. In addition, we propose a breakthrough and strategy to improve the role of immunotherapy in cold CRC based on its characteristics.
Collapse
Affiliation(s)
- Jun Yuan
- Department of Clinical Laboratory, The Yancheng Clinical College of Xuzhou Medical University, The First People’s Hospital of Yancheng, Yancheng, China
| | - Jiarui Li
- Zhejiang University School of Medicine, Hangzhou, China
| | - Ce Gao
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Chun Jiang
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Ze Xiang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Jian Wu
- Department of Clinical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
5
|
Store-Operated Ca2+ Entry Is Up-Regulated in Tumour-Infiltrating Lymphocytes from Metastatic Colorectal Cancer Patients. Cancers (Basel) 2022; 14:cancers14143312. [PMID: 35884372 PMCID: PMC9315763 DOI: 10.3390/cancers14143312] [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: 06/10/2022] [Revised: 07/03/2022] [Accepted: 07/04/2022] [Indexed: 11/29/2022] Open
Abstract
Simple Summary Store-operated Ca2+ entry (SOCE) has long been known to regulate the differentiation and effector functions of T cells as well as to be instrumental to the ability of cytotoxic T lymphocytes to target cancer cells. Currently, no information is available regarding the expression and function of SOCE in tumour-infiltrating lymphocytes (TILs) that have been expanded in vitro for adoptive cell therapy (ACT). This study provides the first evidence that SOCE is up-regulated in ex vivo-expanded TILs from metastatic colorectal cancer (mCRC) patients. The up-regulation of SOCE mainly depends on diacylglycerol kinase (DGK), which prevents the protein kinase C-dependent inhibition of Ca2+ entry in normal T cells. Of note, the pharmacological blockade of SOCE with the selective inhibitor, BTP-2, during target cell killing significantly increases cytotoxic activity at low TIL density, i.e., when TILs-mediated cancer cell death is rarer. This study, albeit preliminary, could lay the foundation to propose an alternative strategy to effect ACT. It has been shown that ex vivo-expanded TILs did not improve the disease-free survival rate in mCRC patients. Our results strongly suggest that pre-treating autologous TILs with a SOCE or DGK inhibitor before being infused into the patient could improve their cytotoxic activity against cancer cells. Abstract (1) Background: Store-operated Ca2+ entry (SOCE) drives the cytotoxic activity of cytotoxic T lymphocytes (CTLs) against cancer cells. However, SOCE can be enhanced in cancer cells due to an increase in the expression and/or function of its underlying molecular components, i.e., STIM1 and Orai1. Herein, we evaluated the SOCE expression and function in tumour-infiltrating lymphocytes (TILs) from metastatic colorectal cancer (mCRC) patients. (2) Methods: Functional studies were conducted in TILs expanded ex vivo from CRC liver metastases. Peripheral blood T cells from healthy donors (hPBTs) and mCRC patients (cPBTs) were used as controls. (3) Results: SOCE amplitude is enhanced in TILs compared to hPBTs and cPBTs, but the STIM1 protein is only up-regulated in TILs. Pharmacological manipulation showed that the increase in SOCE mainly depends on tonic modulation by diacylglycerol kinase, which prevents the protein kinase C-dependent inhibition of SOCE activity. The larger SOCE caused a stronger Ca2+ response to T-cell receptor stimulation by autologous mCRC cells. Reducing Ca2+ influx with BTP-2 during target cell killing significantly increases cytotoxic activity at low target:effector ratios. (4) Conclusions: SOCE is enhanced in ex vivo-expanded TILs deriving from mCRC patients but decreasing Ca2+ influx with BTP-2 increases cytotoxic activity at a low TIL density.
Collapse
|
6
|
Chen Y, Zheng X, Wu C. The Role of the Tumor Microenvironment and Treatment Strategies in Colorectal Cancer. Front Immunol 2021; 12:792691. [PMID: 34925375 PMCID: PMC8674693 DOI: 10.3389/fimmu.2021.792691] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) has the second highest mortality rate among all cancers worldwide. Surgery, chemotherapy, radiotherapy, molecular targeting and other treatment methods have significantly prolonged the survival of patients with CRC. Recently, the emergence of tumor immunotherapy represented by immune checkpoint inhibitors (ICIs) has brought new immunotherapy options for the treatment of advanced CRC. As the efficacy of ICIs is closely related to the tumor immune microenvironment (TME), it is necessary to clarify the relationship between the immune microenvironment of CRC and the efficacy of immunotherapy to ensure that the appropriate drugs are selected. We herein review the latest research progress in the immune microenvironment and strategies related to immunotherapy for CRC. We hope that this review helps in the selection of appropriate treatment strategies for CRC patients.
Collapse
Affiliation(s)
- Yaping Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
7
|
Sumransub N, Vantanasiri K, Prakash A, Lou E. Advances and new frontiers for immunotherapy in colorectal cancer: Setting the stage for neoadjuvant success? Mol Ther Oncolytics 2021; 22:1-12. [PMID: 34307839 PMCID: PMC8280480 DOI: 10.1016/j.omto.2021.05.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy in the metastatic setting has drastically altered the landscape of treatment for various types of malignancy, including colorectal cancer. The category of immune checkpoint inhibitors has especially emerged as a class of therapy predicated on a more comprehensive understanding of immune cell-cancer cell regulation and evolution of the tumor microenvironment over time. Strategies including adoptive cellular therapies, tumor vaccines, and antibodies have also demonstrated the ability to enhance antitumor immunity. In this article, we provide a comprehensive review of the current landscape of immunotherapeutic strategies in colorectal cancer and provide insight into how these strategies may evolve in the next decade and be adapted to more localized forms of cancers of the colon and rectum. We provide particular focus on various combination approaches under investigation for reversing cancer-induced immunosuppression, especially in mismatch repair-proficient/microsatellite-stable colorectal tumors. Finally, we summarize current understanding on a recently identified integral factor in local immune regulation, the colonic microbiome. The aim of this article is to identify current challenges and barriers to improvement and to specify opportunities for applying knowledge in the immunotherapy sphere to rational design of clinical trials intended to improve survival and related outcomes in patients treated in the neoadjuvant setting.
Collapse
Affiliation(s)
- Nuttavut Sumransub
- Department of Medicine, University of Minnesota, 420 Delaware St., SE, MMC 480, Minneapolis, MN 55455, USA
| | - Kornpong Vantanasiri
- Department of Medicine, University of Minnesota, 420 Delaware St., SE, MMC 480, Minneapolis, MN 55455, USA
| | - Ajay Prakash
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Medical Center, 160 E. 34th St., New York, NY 10016, USA
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, 420 Delaware St., SE, MMC 480, Minneapolis, MN 55455, USA
| |
Collapse
|
8
|
Adoptive T-cell immunotherapy in digestive tract malignancies: Current challenges and future perspectives. Cancer Treat Rev 2021; 100:102288. [PMID: 34525422 DOI: 10.1016/j.ctrv.2021.102288] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/15/2022]
Abstract
Multiple systemic treatments are currently available for advanced cancers of the digestive tract, but none of them is curative. Adoptive T-cell immunotherapy refers to the extraction, modification and re-infusion of autologous or allogenic T lymphocytes for therapeutic purposes. A number of clinical trials have investigated either non-engineered T cells (i.e., lymphokine-activated killer cells, cytokine induced killer cells, or tumor-infiltrating lymphocytes) or engineered T cells (T cell receptor-redirected T cells or chimeric antigen receptor T cells) in patients with digestive tract malignancies over the past two decades, with variable degrees of success. While the majority of completed trials have been primarily aimed at assessing the safety of T-cell transfer strategies, a new generation of studies is being designed to formally evaluate the antitumor potential of adoptive T-cell immunotherapy in both the metastatic and adjuvant settings. In this review, we provide an overview of completed and ongoing clinical trials of passive T-cell immunotherapy in patients with cancers of the digestive tract, focusing on present obstacles and future strategies for achieving potential success.
Collapse
|
9
|
Manfredi F, Cianciotti BC, Potenza A, Tassi E, Noviello M, Biondi A, Ciceri F, Bonini C, Ruggiero E. TCR Redirected T Cells for Cancer Treatment: Achievements, Hurdles, and Goals. Front Immunol 2020; 11:1689. [PMID: 33013822 PMCID: PMC7494743 DOI: 10.3389/fimmu.2020.01689] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/24/2020] [Indexed: 12/11/2022] Open
Abstract
Adoptive T cell therapy (ACT) is a rapidly evolving therapeutic approach designed to harness T cell specificity and function to fight diseases. Based on the evidence that T lymphocytes can mediate a potent anti-tumor response, initially ACT solely relied on the isolation, in vitro expansion, and infusion of tumor-infiltrating or circulating tumor-specific T cells. Although effective in a subset of cases, in the first ACT clinical trials several patients experienced disease progression, in some cases after temporary disease control. This evidence prompted researchers to improve ACT products by taking advantage of the continuously evolving gene engineering field and by improving manufacturing protocols, to enable the generation of effective and long-term persisting tumor-specific T cell products. Despite recent advances, several challenges, including prioritization of antigen targets, identification, and optimization of tumor-specific T cell receptors, in the development of tools enabling T cells to counteract the immunosuppressive tumor microenvironment, still need to be faced. This review aims at summarizing the major achievements, hurdles and possible solutions designed to improve the ACT efficacy and safety profile in the context of liquid and solid tumors.
Collapse
Affiliation(s)
- Francesco Manfredi
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Beatrice Claudia Cianciotti
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Fondazione Centro San Raffaele, Milan, Italy
| | - Alessia Potenza
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- School of Medicine and Surgery, University of Milano – Bicocca, Milan, Italy
| | - Elena Tassi
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maddalena Noviello
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Biondi
- Clinica Pediatrica Università degli Studi di Milano Bicocca, Fondazione MBBM, Monza, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Bonini
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eliana Ruggiero
- Experimental Hematology Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
10
|
Picard E, Verschoor CP, Ma GW, Pawelec G. Relationships Between Immune Landscapes, Genetic Subtypes and Responses to Immunotherapy in Colorectal Cancer. Front Immunol 2020; 11:369. [PMID: 32210966 PMCID: PMC7068608 DOI: 10.3389/fimmu.2020.00369] [Citation(s) in RCA: 334] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is highly heterogeneous at the genetic and molecular level, which has major repercussions on the efficacy of immunotherapy. A small subset of CRCs exhibit microsatellite instability (MSI), a molecular indicator of defective DNA mismatch repair (MMR), but the majority are microsatellite-stable (MSS). The high tumor mutational burden (TMB) and neoantigen load in MSI tumors favors the infiltration of immune effector cells, and antitumor immune responses within these tumors are strong relative to their MSS counterparts. MSI has emerged as a major predictive marker for the efficacy of immune checkpoint blockade over the last few years and nivolumab or pembrolizumab targeting PD-1 has been approved for patients with MSI refractory or metastatic CRC. However, some MSS tumors show DNA polymerase epsilon (POLE) mutations that also confer a very high TMB and may also be heavily infiltrated by immune cells making them amenable to respond to immune checkpoint inhibitors (ICI). In this review we discuss the role of the different immune landscapes in CRC and their relationships with defined CRC genetic subtypes. We discuss potential reasons why immune checkpoint blockade has met with limited success for the majority of CRC patients, despite the finding that immune cell infiltration of primary non-metastatic tumors is a strong predictive, and prognostic factor for relapse and survival. We then consider in which ways CRC cells develop mechanisms to resist ICI. Finally, we address the latest advances in CRC vaccination and how a personalized neoantigen vaccine strategy might overcome the resistance of MSI and MSS tumors in patients for whom immune checkpoint blockade is not a treatment option.
Collapse
Affiliation(s)
- Emilie Picard
- Health Sciences North Research Institute, Sudbury, ON, Canada
| | | | - Grace W Ma
- Department of Surgery, Health Sciences North, Sudbury, ON, Canada
| | - Graham Pawelec
- Health Sciences North Research Institute, Sudbury, ON, Canada.,Department of Immunology, University of Tübingen, Tübingen, Germany
| |
Collapse
|
11
|
Pan QZ, Gu JM, Zhao JJ, Tang Y, Wang QJ, Zhu Q, Song MJ, Li YQ, He J, Chen SP, Weng DS, Xia JC. Retrospective analysis of the efficacy of cytokine-induced killer cell immunotherapy combined with first-line chemotherapy in patients with metastatic colorectal cancer. Clin Transl Immunology 2020; 9:e1113. [PMID: 32076550 PMCID: PMC7029432 DOI: 10.1002/cti2.1113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/14/2022] Open
Abstract
Objectives Fluoropyrimidine‐based chemotherapy regimens are the current first‐line treatment for metastatic colorectal cancer (mCRC); however, the outcome is often unsatisfactory. The present study aimed to determine the effect of combined cytokine‐induced killer (CIK) cell immunotherapy and first‐line chemotherapy in patients with mCRC. Methods This retrospective study included 252 patients with mCRC treated with first‐line chemotherapy. Among them, 126 patients received first‐line chemotherapy only (control group), while the other 126 patients, with similar demographic and clinical characteristics, received CIK cell immunotherapy combined with first‐line chemotherapy (CIK group). Overall survival (OS) and progression‐free survival (PFS) were compared between the two groups using the Kaplan–Meier method. Results The median OS for the CIK group was 54.7 versus 24.1 months for the controls, and the median PFS for the CIK group was 25.7 versus 14.6 months for the controls. Univariate and multivariate analyses indicated that CIK cell treatment was an independent prognostic factor for patients' OS and PFS. Subgroup analyses showed that CIK cell treatment significantly improved the OS and PFS of patients with metastatic colon cancer, but not those with metastatic rectal cancer. Additionally, the change in CD3+CD56+ subsets after the fourth treatment cycle might be an indicator of successful CIK cell treatment: Patients with increased CD3+CD56+ subsets had better survival than those with decreased CD3+CD56+ subsets. Conclusion Cytokine‐induced killer cell immunotherapy combined with first‐line chemotherapy could significantly improve the OS and PFS of patients with mCRC, particularly for patients with metastatic colon cancer.
Collapse
Affiliation(s)
- Qiu-Zhong Pan
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Jia-Mei Gu
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Jing-Jing Zhao
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Yan Tang
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Qi-Jing Wang
- Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Qian Zhu
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China
| | - Meng-Jia Song
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China
| | - Yong-Qiang Li
- Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Jia He
- Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Shi-Ping Chen
- Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - De-Sheng Weng
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| | - Jian-Chuan Xia
- State Key Laboratory of Oncology in South China Collaborative Innovation Center for Cancer Medicine Sun Yat-Sen University Cancer Center Guangzhou China.,Department of Biotherapy Sun Yat-Sen University Cancer Center Guangzhou China
| |
Collapse
|
12
|
Fan J, Shang D, Han B, Song J, Chen H, Yang JM. Adoptive Cell Transfer: Is it a Promising Immunotherapy for Colorectal Cancer? Am J Cancer Res 2018; 8:5784-5800. [PMID: 30555581 PMCID: PMC6276301 DOI: 10.7150/thno.29035] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Accepted: 10/04/2018] [Indexed: 12/12/2022] Open
Abstract
The last decade has witnessed significant advances in the adoptive cell transfer (ACT) technique, which has been appreciated as one of the most promising treatments for patients with cancer. Utilization of ACT can enhance the function of the immune system or improve the specificity and persistence of transferred cells. Various immune cells including T lymphocytes, natural killer cells, dendritic cells, and even stem cells can be used in the ACT despite their different functional mechanisms. Colorectal cancer (CRC) is among the most common malignancies and causes millions of deaths worldwide every year. In this review, we discuss the status and perspective of the ACT in the treatment of CRC.
Collapse
|
13
|
Cytokine-induced killer cells/dendritic cells-cytokine induced killer cells immunotherapy combined with chemotherapy for treatment of colorectal cancer in China: a meta-analysis of 29 trials involving 2,610 patients. Oncotarget 2018; 8:45164-45177. [PMID: 28404886 PMCID: PMC5542175 DOI: 10.18632/oncotarget.16665] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 03/17/2017] [Indexed: 12/15/2022] Open
Abstract
Purpose To systematically evaluate the efficacy and safety of Cytokine-induced killer cells/dendritic cells-cytokine induced killer cells (CIK/DC-CIK) immunotherapy in treating advanced colorectal cancer (CRC) patients. Results 29 trials including 2,610 CRC patients were evolved. Compared with chemotherapy alone, the combination of chemotherapy with CIK/DC-CIK immunotherapy significantly prolonged the overall survival rate (OS) and disease-free survival rate (DFS) (1–5 year OS, P < 0.01; 1-, 2-, 3- and 5-year DFS, P < 0.01). The combined therapy also improved patients’ overall response, disease control rate and life quality (P < 0.05). After immunotherapy, lymphocyte subsets percentages of CD3+, CD3−CD56+, CD3+CD56+ and CD16+CD56+ (P < 0.01) and cytokines levels of IL-2 and IFN-γ (P < 0.05) were increased, while CD4+, CD8+ and CD4+CD25+ and IL-6 and TNF-α did not show significant change (P > 0.05). Materials and Methods Clinical trials reporting response or safety of CIK/DC-CIK immunotherapy treating advanced CRC patients and published before September 2016 were searched in Cochrane Library, EMBASE, PubMed, Wanfang and CNKI database. Research quality and heterogeneity were evaluated before analysis. Pooled analyses were performed using random or fixed-effect models. Conclusions The combination of CIK/DC-CIK immunotherapy and chemotherapy prolong CRC patients’ survival time, enhanced patients’ immune function and alleviates the adverse effects caused by chemotherapy.
Collapse
|
14
|
Challenges in Colorectal Cancer: From Vaccines to Macrophage Repolarization. Oncoimmunology 2018. [DOI: 10.1007/978-3-319-62431-0_37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
15
|
Kim JS, Kim YG, Park EJ, Kim B, Lee HK, Hong JT, Kim Y, Han SB. Cell-based Immunotherapy for Colorectal Cancer with Cytokine-induced Killer Cells. Immune Netw 2016; 16:99-108. [PMID: 27162526 PMCID: PMC4853502 DOI: 10.4110/in.2016.16.2.99] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/11/2016] [Accepted: 02/16/2016] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer is the third leading cancer worldwide. Although incidence and mortality of colorectal cancer are gradually decreasing in the US, patients with metastatic colorectal cancer have poor prognosis with an estimated 5-year survival rate of less than 10%. Over the past decade, advances in combination chemotherapy regimens for colorectal cancer have led to significant improvement in progression-free and overall survival. However, patients with metastatic disease gain little clinical benefit from conventional therapy, which is associated with grade 3~4 toxicity with negative effects on quality of life. In previous clinical studies, cell-based immunotherapy using dendritic cell vaccines and sentinel lymph node T cell therapy showed promising therapeutic results for metastatic colorectal cancer. In our preclinical and previous clinical studies, cytokine-induced killer (CIK) cells treatment for colorectal cancer showed favorable responses without toxicities. Here, we review current treatment options for colorectal cancer and summarize available clinical studies utilizing cell-based immunotherapy. Based on these studies, we recommend the use CIK cell therapy as a promising therapeutic strategy for patients with metastatic colorectal cancer.
Collapse
Affiliation(s)
- Ji Sung Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Yong Guk Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Eun Jae Park
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Boyeong Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Hong Kyung Lee
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Youngsoo Kim
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| | - Sang-Bae Han
- College of Pharmacy, Chungbuk National University, Cheongju 28644, Korea
| |
Collapse
|
16
|
Zumwalt TJ, Goel A. Immunotherapy of Metastatic Colorectal Cancer: Prevailing Challenges and New Perspectives. CURRENT COLORECTAL CANCER REPORTS 2015; 11:125-140. [PMID: 26441489 PMCID: PMC4591512 DOI: 10.1007/s11888-015-0269-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Patients with recurring or metastatic colorectal cancer (mCRC) have strikingly low long-term survival, while conventional treatments such as chemotherapeutic intervention and radiation therapy marginally improve longevity. Although, many factors involving immunosurveillance and immunosuppression were recently validated as important for patient prognosis and care, a multitude of experimental immunotherapies designed to combat unresectable mCRC have, in few cases, successfully mobilized antitumor immune cells against malignancies, nor conclusively or consistently granted protection, complete remission, and/or stable disease from immunotherapy - of which benefit less than 10% of those receiving therapy. After decades of progress, however, new insights into the mechanisms of immunosuppression, tolerance, and mutation profiling established novel therapies that circumvent these immunological barriers. This review underlines the most exciting methods to date that manipulate immune cells to curb mCRC, including adoptive cell therapy, dendritic cell vaccines, and checkpoint inhibitor antibodies - of which hint at effective and enduring protection against disease progression and undetected micrometastases.
Collapse
Affiliation(s)
- Timothy J Zumwalt
- Center for Gastrointestinal Research; Center for Epigenetics, Cancer Prevention and Cancer Genomics, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas, USA
| | - Ajay Goel
- Center for Gastrointestinal Research; Center for Epigenetics, Cancer Prevention and Cancer Genomics, Baylor Research Institute and Sammons Cancer Center, Baylor University Medical Center, Dallas, Texas, USA
| |
Collapse
|
17
|
Maker AV, Ito H, Mo Q, Weisenberg E, Qin LX, Turcotte S, Maithel S, Shia J, Blumgart L, Fong Y, Jarnagin WR, DeMatteo RP, D'Angelica MI. Genetic evidence that intratumoral T-cell proliferation and activation are associated with recurrence and survival in patients with resected colorectal liver metastases. Cancer Immunol Res 2015; 3:380-8. [PMID: 25600439 DOI: 10.1158/2326-6066.cir-14-0212] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 01/06/2015] [Indexed: 01/03/2023]
Abstract
Though immune responses correlate with prognosis in primary colorectal cancer, the role of tumor immunity in metastatic disease is less clear. We hypothesized that patient survival and tumor recurrence correlate with transcriptional evidence of lymphocyte proliferation/activation in resected colorectal cancer liver metastases (CRLM). Microarray gene analysis was performed on liver tumor specimens from 96 patients who underwent resection for CRLM. A Cox proportional hazards model identified genes associated with overall survival (OS) and recurrence-free survival (RFS). Conventional gene ontology (GO) enrichment analysis ranked biologically relevant processes. Survival probabilities of prioritized processes were assessed. Protein expression was validated with immunohistochemistry in an independent set of patients. GO analysis identified and ranked unique biologic processes that correlated with survival. Genes that specifically functioned in the biologic process of "T-cell proliferation" were significant predictors of OS (P = 0.01), and both "T-cell proliferation" and "activation" were highly associated with RFS (P ≤ 0.01). Analysis of genes in these GO categories identified increased TNFSF14/LIGHT expression to be most associated with improved OS and RFS (P ≤ 0.0006). Immunohistochemistry of an independent validation set of CRLM confirmed that both increased tumor-infiltrating lymphocytes (TIL) and higher LIGHT expression on TILs were associated with improved OS and RFS. Differential expression of genes involved in T-cell proliferation/activation was associated with survival outcomes in a large number of surgical patients who underwent resection of CRLM. These biologic functions determined by GO analysis of the tumor microenvironment have identified specific immune-related genes that may be involved in an antitumor immune response.
Collapse
Affiliation(s)
- Ajay V Maker
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York. Department of Surgery, Division of Surgical Oncology, University of Illinois at Chicago, Chicago, Illinois.
| | - Hiromichi Ito
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Qianxing Mo
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Elliot Weisenberg
- Department of Pathology, University of Illinois at Chicago, Chicago, Illinois
| | - Li-Xuan Qin
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Simon Turcotte
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Shishir Maithel
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jinru Shia
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Leslie Blumgart
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuman Fong
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - William R Jarnagin
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ronald P DeMatteo
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael I D'Angelica
- Department of Surgery, Hepatopancreatobiliary Service, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
18
|
Di Caro G, Castino GF, Bergomas F, Cortese N, Chiriva-Internati M, Grizzi F, Marchesi F. Immune-based therapies in pancreatic and colorectal cancers and biomarkers of responsiveness. Expert Rev Anticancer Ther 2014; 14:1219-1228. [PMID: 25222571 DOI: 10.1586/14737140.2014.947277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Immune-based strategies are the most promising treatments to improve cancer disease control. Early clinical trials are ongoing to test the safety and feasibility of immune-based therapies for gastrointestinal cancers. However, to date, immunotherapy has been only an experimental option for these diseases and a better understanding of their molecular, cellular, structural and clinical dissimilarities is crucial in the generation of tailored immunotherapeutic treatments. In this review, we will summarize the key mechanisms that regulate the action of immune system in cancer and the different immune-based approaches aimed at improving disease control in patients with advanced disease. We will then move on to discussing the current immunotherapeutic approaches in two types of gastrointestinal (colo-rectal and pancreatic) cancers, whose immune microenvironment has been lately object of intense analyses and has emerged as an important determinant of clinical outcome.
Collapse
Affiliation(s)
- Giuseppe Di Caro
- Humanitas Clinical and Research Center, Via Manzoni 56, Rozzano 20089, Italy
| | | | | | | | | | | | | |
Collapse
|
19
|
Atreya I, Neurath MF. Immune cells in colorectal cancer: prognostic relevance and therapeutic strategies. Expert Rev Anticancer Ther 2014; 8:561-72. [DOI: 10.1586/14737140.8.4.561] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
20
|
Toomey PG, Vohra NA, Ghansah T, Sarnaik AA, Pilon-Thomas SA. Immunotherapy for gastrointestinal malignancies. Cancer Control 2013; 20:32-42. [PMID: 23302905 DOI: 10.1177/107327481302000106] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Gastrointestinal (GI) cancers are the most common human tumors encountered worldwide. The majority of GI cancers are unresectable at the time of diagnosis, and in the subset of patients undergoing resection, few are cured. There is only a modest improvement in survival with the addition of modalities such as chemotherapy and radiation therapy. Due to an increasing global cancer burden, it is imperative to integrate alternative strategies to improve outcomes. It is well known that cancers possess diverse strategies to evade immune detection and destruction. This has led to the incorporation of various immunotherapeutic strategies, which enable reprogramming of the immune system to allow effective recognition and killing of GI tumors. METHODS A review was conducted of the results of published clinical trials employing immunotherapy for esophageal, gastroesophageal, gastric, hepatocellular, pancreatic, and colorectal cancers. RESULTS Monoclonal antibody therapy has come to the forefront in the past decade for the treatment of colorectal cancer. Immunotherapeutic successes in solid cancers such as melanoma and prostate cancer have led to the active investigation of immunotherapy for GI malignancies, with some promising results. CONCLUSIONS To date, monoclonal antibody therapy is the only immunotherapy approved by the US Food and Drug Administration for GI cancers. Initial trials validating new immunotherapeutic approaches, including vaccination-based and adoptive cell therapy strategies, for GI malignancies have demonstrated safety and the induction of antitumor immune responses. Therefore, immunotherapy is at the forefront of neoadjuvant as well as adjuvant therapies for the treatment and eradication of GI malignancies.
Collapse
Affiliation(s)
- Paul G Toomey
- Department of Surgery, USF Health Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | | | | | | | | |
Collapse
|
21
|
Yang JY, Li X, Gao L, Teng ZH, Liu WC. Co-transfection of dendritic cells with AFP and IL-2 genes enhances the induction of tumor antigen-specific antitumor immunity. Exp Ther Med 2012; 4:655-660. [PMID: 23170121 PMCID: PMC3501441 DOI: 10.3892/etm.2012.635] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2012] [Accepted: 06/28/2012] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) are highly efficient, specialized antigen-presenting cells and DCs transfected with tumor-related antigens are regarded as promising vaccines in cancer immunotherapy. The aim of the present study was to investigate whether DCs co-transfected with the α-fetoprotein (AFP) and human interleukin-2 (IL-2) genes were able to induce stronger therapeutic antitumor immunity in transfected DCs. In this study, DCs from hepatocellular carcinoma (HCC) patients were co-transfected with the IL-2 gene and/or the AFP gene. The reverse transcription-PCR (RT-PCR) data revealed that the DCs transfected with the adenovirus AdAFP/IL-2 expressed AFP and IL-2. The DCs co-transfected with IL-2 and AFP (AFP/IL-2-DCs) enhanced the cytotoxicities of cytotoxic T lymphocytes (CTLs) and increased the production of IL-2 and interferon-γ significantly compared with their AFP-DC, green fluorescent protein (GFP)-DC, DC or phosphate-buffered saline (PBS) counterparts. In vivo data suggested that immunization with AFP-DCs enhances antigen-specific antitumor efficacy more potently than immunization with IL-2-DCs or AFP-DCs. These findings provide a potential strategy to improve the efficacy of DC-based tumor vaccines.
Collapse
Affiliation(s)
- Jing-Yue Yang
- Department of Clinical Oncology, State Key Discipline of Cell Biology, Xijing Hospital
| | | | | | | | | |
Collapse
|
22
|
Ellebaek E, Andersen MH, Svane IM, Straten PT. Immunotherapy for metastatic colorectal cancer: present status and new options. Scand J Gastroenterol 2012; 47:315-24. [PMID: 22214467 DOI: 10.3109/00365521.2012.640831] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although no immunotherapeutic treatment is approved for colorectal cancer (CRC) patients, promising results from clinical trials suggest that several immunotherapeutic strategies may prove efficacious and applicable to this group of patients. This review describes the immunogenicity of CRC and presents the most interesting strategies investigated so far: cancer vaccination including antigen-defined vaccination and dendritic cell vaccination, chemo-immunotherapy, and adoptive cell transfer. Future treatment options as well as the possibility of combining existing therapies will be discussed along with the challenges presented by tumor escape mechanisms.
Collapse
Affiliation(s)
- Eva Ellebaek
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital Herlev, Herlev, Denmark.
| | | | | | | |
Collapse
|
23
|
Abstract
Cancer immunotherapy consists of approaches that modify the host immune system, and/or the utilization of components of the immune system, as cancer treatment. During the past 25 years, 17 immunologic products have received regulatory approval based on anticancer activity as single agents and/or in combination with chemotherapy. These include the nonspecific immune stimulants BCG and levamisole; the cytokines interferon-α and interleukin-2; the monoclonal antibodies rituximab, ofatumumab, alemtuzumab, trastuzumab, bevacizumab, cetuximab, and panitumumab; the radiolabeled antibodies Y-90 ibritumomab tiuxetan and I-131 tositumomab; the immunotoxins denileukin diftitox and gemtuzumab ozogamicin; nonmyeloablative allogeneic transplants with donor lymphocyte infusions; and the anti-prostate cancer cell-based therapy sipuleucel-T. All but two of these products are still regularly used to treat various B- and T-cell malignancies, and numerous solid tumors, including breast, lung, colorectal, prostate, melanoma, kidney, glioblastoma, bladder, and head and neck. Positive randomized trials have recently been reported for idiotype vaccines in lymphoma and a peptide vaccine in melanoma. The anti-CTLA-4 monoclonal antibody ipilumumab, which blocks regulatory T-cells, is expected to receive regulatory approval in the near future, based on a randomized trial in melanoma. As the fourth modality of cancer treatment, biotherapy/immunotherapy is an increasingly important component of the anticancer armamentarium.
Collapse
Affiliation(s)
- Robert O Dillman
- Hoag Cancer Institute of Hoag Hospital , Newport Beach, California 92658, USA.
| |
Collapse
|
24
|
Nguyen LT, Yen PH, Nie J, Liadis N, Ghazarian D, Al-Habeeb A, Easson A, Leong W, Lipa J, McCready D, Reedijk M, Hogg D, Joshua AM, Quirt I, Messner H, Shaw P, Crump M, Sharon E, Ohashi PS. Expansion and characterization of human melanoma tumor-infiltrating lymphocytes (TILs). PLoS One 2010; 5:e13940. [PMID: 21085676 PMCID: PMC2978109 DOI: 10.1371/journal.pone.0013940] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Accepted: 10/19/2010] [Indexed: 12/26/2022] Open
Abstract
Background Various immunotherapeutic strategies for cancer are aimed at augmenting the T cell response against tumor cells. Adoptive cell therapy (ACT), where T cells are manipulated ex vivo and subsequently re-infused in an autologous manner, has been performed using T cells from various sources. Some of the highest clinical response rates for metastatic melanoma have been reported in trials using tumor-infiltrating lymphocytes (TILs). These protocols still have room for improvement and furthermore are currently only performed at a limited number of institutions. The goal of this work was to develop TILs as a therapeutic product at our institution. Principal Findings TILs from 40 melanoma tissue specimens were expanded and characterized. Under optimized culture conditions, 72% of specimens yielded rapidly proliferating TILs as defined as at least one culture reaching ≥3×107 TILs within 4 weeks. Flow cytometric analyses showed that cultures were predominantly CD3+ T cells, with highly variable CD4+:CD8+ T cell ratios. In total, 148 independent bulk TIL cultures were assayed for tumor reactivity. Thirty-four percent (50/148) exhibited tumor reactivity based on IFN-γ production and/or cytotoxic activity. Thirteen percent (19/148) showed specific cytotoxic activity but not IFN-γ production and only 1% (2/148) showed specific IFN-γ production but not cytotoxic activity. Further expansion of TILs using a 14-day “rapid expansion protocol” (REP) is required to induce a 500- to 2000-fold expansion of TILs in order to generate sufficient numbers of cells for current ACT protocols. Thirty-eight consecutive test REPs were performed with an average 1865-fold expansion (+/− 1034-fold) after 14 days. Conclusions TILs generally expanded efficiently and tumor reactivity could be detected in vitro. These preclinical data from melanoma TILs lay the groundwork for clinical trials of ACT.
Collapse
Affiliation(s)
- Linh T. Nguyen
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Pei Hua Yen
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Jessica Nie
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Nicole Liadis
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
| | - Danny Ghazarian
- Department of Pathology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Ayman Al-Habeeb
- Department of Pathology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Alexandra Easson
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Wey Leong
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Joan Lipa
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - David McCready
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Michael Reedijk
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - David Hogg
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Anthony M. Joshua
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Ian Quirt
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Hans Messner
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Patricia Shaw
- Department of Pathology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Michael Crump
- Department of Medical Oncology/Hematology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Eran Sharon
- Department of Surgical Oncology, Princess Margaret Hospital, University Health Network, Toronto, Canada
| | - Pamela S. Ohashi
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, University Health Network, Toronto, Canada
- Departments of Medical Biophysics and Immunology, University of Toronto, Toronto, Canada
- * E-mail:
| |
Collapse
|
25
|
Peng RQ, Chen YB, Ding Y, Zhang R, Zhang X, Yu XJ, Zhou ZW, Zeng YX, Zhang XS. Expression of calreticulin is associated with infiltration of T-cells in stage IIIB colon cancer. World J Gastroenterol 2010; 16:2428-34. [PMID: 20480531 PMCID: PMC2874150 DOI: 10.3748/wjg.v16.i19.2428] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the correlation between expression of calreticulin and infiltration of lymphocytes in stage IIIB colon cancer.
METHODS: Sixty-eight pathologically-confirmed specimens were obtained from stage IIIB (T3N1M0) colon cancer patients who underwent radical resection between January 1999 and May 2002 at the Cancer Center of Sun Yat-Sen University, Guangzhou, China. Immunohistochemical analysis was performed to show infiltration of lymphocytes and expression of calreticulin in colon cancer. Association between calreticulin expression, infiltration of lymphocytes, and 5-year survival rate of patients was assessed.
RESULTS: The expression level of calreticulin was lower in cancer nest than in its adjacent normal epithelium since 61.8% (42/68) of the samples were stained with calreticulin in colon cancer. The expression of calreticulin in colon cancer was associated with the infiltration of CD45RO+ cells rather than with that of CD3+ cells. In addition, the stronger expression of calreticulin and the higher infiltration of CD3+ and CD45RO+ cells in colon cancer were associated with the higher 5-year survival rate of patients.
CONCLUSION: Expression of calreticulin is associated with infiltration of T-cells, which implies that a low expression level of molecular marker may represent a new mechanism underlying immune escape in colon cancer.
Collapse
|
26
|
Lei XY, Xu YM, Wang T, Xie QS, Jia LT, Wang LF, Jin BQ, Yan Z, Yao LB, Yang AG. Knockdown of human bid gene expression enhances survival of CD8+ T cells. Immunol Lett 2009; 122:30-36. [PMID: 19046991 DOI: 10.1016/j.imlet.2008.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2008] [Revised: 10/26/2008] [Accepted: 10/30/2008] [Indexed: 12/27/2022]
Abstract
Tumor cells have developed immune evasion mechanisms such as considerably heterogenous FasL expression on their surface via which they could induce apoptosis of tumor-specific cytotoxic T lymphocytes (CTLs) in the immune system. Meanwhile, the competition of normal immune cells with tumor cells results in relative growth factors shortage for growth and proliferation of nontumor cells, which improves a susceptibility to early apoptosis of CTL. In an attempt to develop strategies for prolonging the survival of adoptively transferred T cells in a hostile pro-apoptotic tumor microenvironment, we used synthetic siRNA and vector-based shRNA to suppress the expression of Bid in human uterocervical carcinoma HeLa cells, followed by the further achievement of Bid gene silencing in human primary cells-CD8(+) lymphocytes via retrovirus-delivered siRNAs. Our results indicated that Bid knockdown HeLa cells are partially resistant to Fas antibody- or serum deprivation-induced apoptosis. Additionally, the blockade of Bid expression in CD8(+) lymphocytes resulted in a less susceptiveness to Fas antibody-induced apoptosis and a survival advantage following recombinant human interleukin-2 (rhIL-2) withdrawal or under lower rhIL-2 concentrations compared with control lymphocytes. These data suggest that knockdown of Bid might serve as an approach to enhancing the survival and tumoricidal activity of T lymphocytes in adoptive immunotherapy.
Collapse
Affiliation(s)
- Xiao-Ying Lei
- Department of Biochemistry and Molecular Biology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Power DG, Healey-Bird BR, Kemeny NE. Regional Chemotherapy for Liver-Limited Metastatic Colorectal Cancer. Clin Colorectal Cancer 2008; 7:247-59. [DOI: 10.3816/ccc.2008.n.032] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
28
|
Shimbo T, Kawachi M, Saga K, Fujita H, Yamazaki T, Tamai K, Kaneda Y. Development of a transferrin receptor-targeting HVJ-E vector. Biochem Biophys Res Commun 2007; 364:423-8. [DOI: 10.1016/j.bbrc.2007.09.135] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2007] [Accepted: 09/28/2007] [Indexed: 11/25/2022]
|
29
|
Kawano H, Komaba S, Kanamori T, Kaneda Y. A new therapy for highly effective tumor eradication using HVJ-E combined with chemotherapy. BMC Med 2007; 5:28. [PMID: 17883878 PMCID: PMC2039728 DOI: 10.1186/1741-7015-5-28] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Accepted: 09/21/2007] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Inactivated HVJ (hemagglutinating virus of Japan; Sendai virus) particles (HVJ envelope vector; HVJ-E can incorporate and deliver plasmid DNA, siRNA, antibody and peptide and anti-cancer drugs to cells both in vitro and in vivo. We attempted to eradicate tumors derived from mouse colon cancer cells, CT26, by combining bleomycin (BLM)-incorporated HVJ-E (HVJ-E/BLM) with cisplatin (CDDP) administration. METHODS CT-26 tumor mass was intradermally established in Balb/c mice. HVJ-E/BLM was directly injected into the tumor mass with or without intraperitoneal administration of CDDP. The anti-tumor effect was evaluated by measuring tumor size and cytotoxic T cell activity against CT26. Re-challenge of tumor cells to treated mice was performed 10 days or 8 months after the initial tumor inoculation. RESULTS We found that three intratumoral injections of HVJ-E/BLM along with a single intraperitoneal administration of CDDP eradicated CT26 tumors with more than 75% efficiency. When tumor cells were intradermally re-injected on day 10 after the initial tumor inoculation, tumors on both sides disappeared in most of the mice that received the combination therapy of HVJ-E/BLM and CDDP. Eight months after the initial tumor eradication, surviving mice were re-challenged with CT26 cells. The re-challenged tumors were rejected in all of the surviving mice treated with the combination therapy. Cytotoxic T lymphocytes specific for CT26 were generated in these surviving mice. CONCLUSION Combination therapy consisting of HVJ-E and chemotherapy completely eradicated the tumor, and generated anti-tumor immunity. The combination therapy could therefore be a promising new strategy for cancer therapy.
Collapse
Affiliation(s)
- Hirokazu Kawano
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
- GenomIdea Inc., 7-7-15 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Shintarou Komaba
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
- GenomIdea Inc., 7-7-15 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Toshihide Kanamori
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
- GenomIdea Inc., 7-7-15 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Yasufumi Kaneda
- Division of Gene Therapy Science, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
30
|
Abstract
BACKGROUND Advances in immunology and molecular biology have shown that colorectal cancer is potentially immunogenic and that host immune responses influence survival. However, immune surveillance and activation is frequently ineffective in preventing and/or controlling tumour growth. AIM To discuss potential ways in which colorectal cancer induces immune suppression, its effect upon prognosis and avenues for therapeutic development. METHOD A literature review was undertaken for evidence of colorectal cancer-induced immune suppression using PubMed and Medline searches. Further studies were identified from the reference lists of identified papers. RESULTS Immune suppression occurs at a molecular and cellular level and can result in a shift from cellular to humoral immunity. Several mechanisms for immune suppression have been described affecting innate and adaptive immunity with suppression linked to poorer clinical outcome. CONCLUSIONS Colorectal cancer causes direct inhibition of the host's immune response with a detrimental effect upon prognosis. Immunotherapy offers a therapeutic strategy to counteract these effects with promising results seen particularly in precancerous conditions and early tumours. This review strongly suggests that immunotherapy should be incorporated into adjuvant therapeutic trials for stage 2 tumours and be considered as adjuvant treatment in conjunction with standard chemotherapy regimes for advanced disease.
Collapse
Affiliation(s)
- C Evans
- Institution Colorectal Surgery Unit & Division of Oncology, St George's Hospital, Blackshaw Road, London, UK
| | | | | |
Collapse
|
31
|
Mima H, Yamamoto S, Ito M, Tomoshige R, Tabata Y, Tamai K, Kaneda Y. Targeted chemotherapy against intraperitoneally disseminated colon carcinoma using a cationized gelatin–conjugated HVJ envelope vector. Mol Cancer Ther 2006; 5:1021-8. [PMID: 16648574 DOI: 10.1158/1535-7163.mct-05-0352] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The hemagglutinating virus of Japan envelope (HVJ-E; Sendai virus) vector derived from inactivated HVJ particles can be used to deliver DNA, proteins, and drugs into cells both in vitro and in vivo. HVJ-E is capable of delivering bleomycin, an anticancer drug, to various cancer cell lines, thereby producing 300-fold greater cytotoxicity than administration of bleomycin alone. In a mouse model of peritoneally disseminated colon cancer, we injected HVJ-E containing the luciferase gene into the peritoneum. Unexpectedly, luciferase gene expression was not observed within the tumor deposits or any organs. However, when combined with cationized gelatin (CG), CG-HVJ-E produced a high level of luciferase gene expression primarily within the tumor deposits. Forty-eight hours after introducing colon cancer cells into the peritoneum of experimental mice, CG-HVJ-E with or without bleomycin was injected into the abdominal cavity. Following six injections of bleomycin-incorporated CG-HVJ-E, complete responses were observed in 40% of the mice examined. All of the mice that received either empty CG-HVJ-E or bleomycin alone died within 40 days of having cancer cells introduced into the peritoneum. When the mice with complete responses were rechallenged with colon cancer cells from the same cell line, no tumors developed. Thus, CG-HVJ-E may suppress peritoneal dissemination of cancer.
Collapse
Affiliation(s)
- Hidetoshi Mima
- Division of Gene Therapy Science, Graduate School of Medicine, Osaka University, 2-2 Yamada-oka, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Tian JQ, Wang ZP, Rodriguez R, Fu JS, Lu JZ, Ma BL. In vitro enhanced cytotoxicity of tumor-infiltrating lymphocytes transfected with tumor necrosis factor-related apoptosis-inducing ligand and/or interleukin-2 gene in human renal cell carcinoma. Urology 2006; 67:1093-8. [PMID: 16635513 DOI: 10.1016/j.urology.2005.11.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 10/24/2005] [Accepted: 11/10/2005] [Indexed: 11/19/2022]
Abstract
OBJECTIVES To investigate whether tumor-infiltrating lymphocytes (TILs) transfected with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) and interleukin-2 (IL-2) genes are capable of improving the potency and efficacy of propagation and cytotoxicity against renal cell carcinoma (RCC) cells in vitro. METHODS A mammal expression vector system was constructed. TILs were transfected by liposome-mediated gene transfection. The degree of cytokine mRNA expression was evaluated with Northern blot. Protein expression was determined with Western blot and enzyme-linked immunosorbent assay. Cytotoxicity of TILs against autologous RCC cells and the human RCC cell line (786-0) were examined by chromium release assay. Flow cytometric analyses were performed to determine the apoptosis of tumor cells. RESULTS A high level of expression of the human TRAIL and IL-2 stable transfected TILs was observed. The mean IL-2 production was 22.6 +/- 5.2, 507.7 +/- 52.4, and 549.0 +/- 74.0 ng/10(6) cells/24 hours in the TIL/parental, TIL/IL-2, and TIL/TRAIL+IL-2 genes, respectively. The mean cytotoxicity (effector/target ratio 20:1) of TIL/parental, TIL/IL-2, TIL/TRAIL, and TIL/TRAIL+IL-2 against autologous RCC cells in the percentage of cytolysis was 21.2% +/- 4.8%, 32.1% +/- 5.5%, 63.5% +/- 6.6%, and 78.1% +/- 9.63%, respectively. These four groups showed cytotoxic activity against allogeneic 786-0 RCC cells; the corresponding values were 9.8% +/- 3.5%, 12.3% +/- 3.4%, 24.1% +/- 4.9%, and 30.4% +/- 6.2%. The number of apoptotic cells was significantly greater for autologous RCC cells than for 786-0 cells after TIL/TRAIL and TIL/TRAIL+IL-2 treatment. CONCLUSIONS TIL/TRAIL+IL-2 and TIL/IL-2 were expanded by autocrine IL-2. TIL/TRAIL+IL-2 and TIL/TRAIL showed significant cytotoxicity that was induced by TRAIL. TILs, including parental TILs and transfected TILs, demonstrated a potent cytotoxicity against RCC cells with remarkable selectivity. Autologous RCC cells seemed more sensitive than allogeneic RCC cells.
Collapse
Affiliation(s)
- Jun-Qiang Tian
- Lanzhou University School of Life Science, Lanzhou, GanSu, China
| | | | | | | | | | | |
Collapse
|
33
|
Wang P, Chen Z, Huang WX, Liu LM. Current preventive treatment for recurrence after curative hepatectomy for liver metastases of colorectal carcinoma: A literature review of randomized control trials. World J Gastroenterol 2005; 11:3817-22. [PMID: 15991275 PMCID: PMC4504878 DOI: 10.3748/wjg.v11.i25.3817] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
To review the preventive approaches for recurrence after curative resection of hepatic metastases from colorectal carcinoma, we have summarized all available publications reporting randomized control trials (RCTs) covered in PubMed. The treatment approaches presented above include adjuvant intrahepatic arterial infusion chemotherapy, systemic chemotherapy, neoadjuvant chemotherapy, and immunotherapy. Although no standard treatment has been established, several approaches present promising results, which are both effective and tolerable in post-hepatectomy patients. Intrahepatic arterial infusion chemotherapy should be regarded as effective and tolerable and it increases overall survival (OS) and disease-free survival (DFS) of patients, while 5-fluorouracil-based systemic chemotherapy has not shown any significant survival benefit. Fortunately chemotherapy combined with hepatic arterial infusion and intravenous infusion has shown OS and DFS benefit in many researches. Few neoadjuvant RCT studies have been conducted to evaluate its effect on prolonging survivals although many retrospective studies and case reports are published in which unresectable colorectal liver metastases are downstaged and made resectable with neoadjuvant chemotherapy. Liver resection supplemented with immunotherapy is associated with optimal results; however, it is also questioned by others. In conclusion, several adjuvant approaches have been studied for their efficacy on recurrence after hepatectomy for liver metastases from colorectal cancer (CRC), but multi-centric RCT is still needed for further evaluation on their efficacy and systemic or local toxicities. In addition, new adjuvant treatment should be investigated to provide more effective and tolerable methods for the patients with resectable hepatic metastases from CRC.
Collapse
Affiliation(s)
- Peng Wang
- Department of Liver Neoplasms, Cancer Hospital of Fudan University, 270 Dong An Road, Shanghai 200032, China.
| | | | | | | |
Collapse
|