1
|
Li K, Zhu Q, Du S, Zhao Q, Ba D, Zeng X, Peng Q, Cai J, Zhao Y, Jin H, Qi L. EGFLAM exhibits oncogenic activity and shows promise as a prognostic biomarker and therapeutic target in glioblastoma. Int Immunopharmacol 2024; 138:112625. [PMID: 38996666 DOI: 10.1016/j.intimp.2024.112625] [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: 04/23/2024] [Revised: 06/24/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024]
Abstract
Glioblastoma (GBM) remains the most lethal primary brain tumor, characterized by dismal survival rates. Novel molecular targets are urgently required to enhance therapeutic outcomes. A combination of bioinformatics analysis and experimental validation was employed to investigate the role of EGFLAM in GBM. The Chinese Glioma Genome Atlas provided a platform for gene expression profiling, while siRNA-mediated knockdown and overexpression assays in GBM cell lines, alongside in vivo tumorigenesis models, facilitated functional validation. EGFLAM was found to be significantly overexpressed in GBM tissues, correlating with adverse prognostic factors and higher tumor grades, particularly in patients over the age of 41. Functional assays indicated that EGFLAM is vital for maintaining GBM cell proliferation, viability, and invasiveness. Knockdown of EGFLAM expression led to a marked decrease in tumorigenic capabilities. Proteomic interactions involving EGFLAM, such as with NUP205, were implicated in cell cycle regulation, providing insight into its oncogenic mechanism. In vivo studies further demonstrated that silencing EGFLAM expression could inhibit tumor growth, underscoring its therapeutic potential. The study identifies EGFLAM as a pivotal oncogenic factor in GBM, serving as both a prognostic biomarker and a viable therapeutic target. These findings lay the groundwork for future research into EGFLAM-targeted therapies, aiming to improve clinical outcomes for GBM patients.
Collapse
Affiliation(s)
- Kaishu Li
- Department of Neurosurgery, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Qihui Zhu
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Siyuan Du
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Qiuman Zhao
- Department of Neurosurgery, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Deyan Ba
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Xiangzong Zeng
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Qian Peng
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Junbin Cai
- Department of Neurosurgery, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Yubo Zhao
- Department of Neurosurgery, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China; Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China
| | - Hong Jin
- School of Clinical Medicine, Jilin Medical University, NO.5, Jilin Road, Jilin 132013, Jilin Province, PR China
| | - Ling Qi
- Institute of Digestive Diseases, the Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, PR China.
| |
Collapse
|
2
|
Ghanbari Sevari F, Mehdizadeh A, Abbasi K, Hejazian SS, Raisii M. Cytokine-induced killer cells: new insights for therapy of hematologic malignancies. Stem Cell Res Ther 2024; 15:254. [PMID: 39135188 PMCID: PMC11321231 DOI: 10.1186/s13287-024-03869-z] [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: 02/12/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND Cytokine-induced killer (CIK) cells are a novel subgroup of immune effectors, classified as one of the modified T cell-mediated arms for immunotherapy. These cells exert MHC-unrestricted cytotoxicity against both hematological and solid malignancies with low incidence of treatment-related severe complications. This study reviews the application of CIK cells in treating cases with hematologic malignancies. MAIN BODY CIK cells consist of CD3+/CD56+ natural killer (NK) T cells, CD3-/CD56+ NK cells, and CD3+/CD56- cytotoxic T cells. In this regard, the CD3+/CD56+ NK T cells are the primary effectors. Compared with the previously reported antitumor immune cells, CIK cells are characterized by improved in vitro proliferation and amplification, enhanced migration and invasive capacity to tumor region, more significant antitumor activity, and a broader antitumor spectrum. CIK cells can also induce death in tumor cells via numerous pathways and mechanisms. Hence, CIKs-based therapy has been used in various clinical trials and has shown efficacy with a very low graft versus host disease (GVHD) against several cancers, such as hematologic malignancies, even in relapsing cases, or cases not responding to other therapies. Despite the high content of T cells, CIK cells induce low alloreactivity and, thus, pose a restricted threat of GVHD induction even in MHC-mismatched transplantation cases. Phase 1 and 2 clinical trials of CIK cell therapy have also highlighted satisfactory therapeutic advantages against hematologic cancers, indicating the safety of CIK cells even in haploidentical transplantation settings. CONCLUSION CIK cells have shown promising results in the treatment of hematologic malignancies, especially in combination with other antitumor strategies. However, the existing controversies in achieving desired clinical responses underscore the importance of future studies.
Collapse
Affiliation(s)
- Faezeh Ghanbari Sevari
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Khadijeh Abbasi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Sina Hejazian
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Mortaza Raisii
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Capelli C, Cuofano C, Pavoni C, Frigerio S, Lisini D, Nava S, Quaroni M, Colombo V, Galli F, Bezukladova S, Panina-Bordignon P, Gaipa G, Comoli P, Cossu G, Martino G, Biondi A, Introna M, Golay J. Potency assays and biomarkers for cell-based advanced therapy medicinal products. Front Immunol 2023; 14:1186224. [PMID: 37359560 PMCID: PMC10288881 DOI: 10.3389/fimmu.2023.1186224] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 05/24/2023] [Indexed: 06/28/2023] Open
Abstract
Advanced Therapy Medicinal Products (ATMPs) based on somatic cells expanded in vitro, with or without genetic modification, is a rapidly growing area of drug development, even more so following the marketing approval of several such products. ATMPs are produced according to Good Manufacturing Practice (GMP) in authorized laboratories. Potency assays are a fundamental aspect of the quality control of the end cell products and ideally could become useful biomarkers of efficacy in vivo. Here we summarize the state of the art with regard to potency assays used for the assessment of the quality of the major ATMPs used clinic settings. We also review the data available on biomarkers that may substitute more complex functional potency tests and predict the efficacy in vivo of these cell-based drugs.
Collapse
Affiliation(s)
- Chiara Capelli
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Carolina Cuofano
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Chiara Pavoni
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Simona Frigerio
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daniela Lisini
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Sara Nava
- Cell Therapy Production Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Michele Quaroni
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Valentina Colombo
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Francesco Galli
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, Manchester, United Kingdom
| | - Svetlana Bezukladova
- Università Vita-Salute San Raffaele, Milan, Italy
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
| | - Paola Panina-Bordignon
- Università Vita-Salute San Raffaele, Milan, Italy
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
| | - Giuseppe Gaipa
- Laboratory of Cell and Gene Therapy Stefano Verri, ASST Monza Ospedale San Gerardo, Monza, Italy
| | - Patrizia Comoli
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Giulio Cossu
- Division of Cell Matrix Biology & Regenerative Medicine, Faculty of Biology, Medicine and Health (FBMH), University of Manchester, Manchester, United Kingdom
- Università Vita-Salute San Raffaele, Milan, Italy
| | - Gianvito Martino
- IRCCS San Raffaele Hospital, Neuroimmunology Unit, Division of Neuroscience, Milan, Italy
- Pediatric Hematology/Oncology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
| | - Andrea Biondi
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Martino Introna
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - Josée Golay
- Center of Cellular Therapy “G. Lanzani”, ASST Papa Giovanni XXIII, Bergamo, Italy
| |
Collapse
|
4
|
Zúñiga TM, Baker FL, Smith KA, Batatinha H, Lau B, Gustafson MP, Katsanis E, Simpson RJ. Acute exercise mobilizes NKT-like cells with a cytotoxic transcriptomic profile but does not augment the potency of cytokine-induced killer (CIK) cells. Front Immunol 2022; 13:938106. [PMID: 36189306 PMCID: PMC9519182 DOI: 10.3389/fimmu.2022.938106] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 07/04/2022] [Indexed: 11/17/2022] Open
Abstract
CD3+/CD56+ Natural killer (NK) cell-like T-cells (NKT-like cells) represent <5% of blood lymphocytes, display a cytotoxic phenotype, and can kill various cancers. NKT-like cells can be expanded ex vivo into cytokine-induced killer (CIK) cells, however this therapeutic cell product has had mixed results against hematological malignancies in clinical trials. The aim of this study was to determine if NKT-like cells mobilized during acute cycling exercise could be used to generate more potent anti-tumor CIK cells from healthy donors. An acute exercise bout increased NKT-like cell numbers in blood 2-fold. Single cell RNA sequencing revealed that exercise mobilized NKT-like cells have an upregulation of genes and transcriptomic programs associated with enhanced anti-tumor activity, including cytotoxicity, cytokine responsiveness, and migration. Exercise, however, did not augment the ex vivo expansion of CIK cells or alter their surface phenotypes after 21-days of culture. CIK cells expanded at rest, during exercise (at 60% and 80% VO2max) or after (1h post) were equally capable of killing leukemia, lymphoma, and multiple myeloma target cells with and without cytokine (IL-2) and antibody (OKT3) priming in vitro. We conclude that acute exercise in healthy donors mobilizes NKT-like cells with an upregulation of transcriptomic programs involved in anti-tumor activity, but does not augment the ex vivo expansion of CIK cells.
Collapse
Affiliation(s)
- Tiffany M. Zúñiga
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
| | - Forrest L. Baker
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
| | - Kyle A. Smith
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
| | - Helena Batatinha
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
| | - Branden Lau
- University of Arizona Genetics Core, The University of Arizona, Tucson, AZ, United States
| | - Michael P. Gustafson
- Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, AZ, United States
| | - Emmanuel Katsanis
- Department of Pediatrics, The University of Arizona, Tucson, AZ, United States
- The University of Arizona Cancer Center, The University of Arizona, Tucson, AZ, United States
- Department of Immunobiology, The University of Arizona, Tucson, AZ, United States
- Department of Medicine, The University of Arizona, Tucson, AZ, United States
- Department of Pathology, The University of Arizona, Tucson, AZ, United States
| | - Richard J. Simpson
- School of Nutritional Sciences and Wellness, The University of Arizona, Tucson, AZ, United States
- Department of Pediatrics, The University of Arizona, Tucson, AZ, United States
- The University of Arizona Cancer Center, The University of Arizona, Tucson, AZ, United States
- Department of Immunobiology, The University of Arizona, Tucson, AZ, United States
- *Correspondence: Richard J. Simpson,
| |
Collapse
|
5
|
Park SJ, Yoon HJ, Gu EY, Lee BS, Kim Y, Jung J, Kim J, Moon KS. A general toxicity and biodistribution study of human natural killer cells by single or repeated intravenous dose in severe combined immune deficient mice. Toxicol Res 2022; 38:545-555. [PMID: 36277368 PMCID: PMC9532477 DOI: 10.1007/s43188-022-00138-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/10/2022] [Accepted: 05/11/2022] [Indexed: 11/28/2022] Open
Abstract
AbstractNatural killer (NK) cells are a part of the innate immune system and represent the first line of defense against infections and tumors. NK cells can eliminate tumor cells without major histocompatibility restriction and are independent of the expression of tumor-associated antigens. Therefore, they are considered an emerging tool for cancer immunotherapy. However, the general toxicity and biodistribution of NK cells after transplantation remain to be understood. This study was conducted to evaluate the general toxicity and biodistribution of human NK cells after single or repeated intravenous dosing in severely combined immunodeficient (SCID) mice. There were no test item-related toxicological changes in single and repeated administration groups. The no observed adverse effect level of human NK cells was 2 × 107 cells/head for both male and female SCID mice. Results from the biodistribution study showed that human NK cells were mainly distributed in the lungs, and a small number of the cells were detected in the liver, heart, spleen, and kidney of SCID mice, in both the single and repeated dose groups. Additionally, human NK cells were completely eliminated from all organs of the mice in the single dose group on day 7, while the cells persisted in mice in the repeated dose group until day 64. In conclusion, transplantation of human NK cells in SCID mice had no toxic effects. The cells were mainly distributed in the lungs and completely disappeared from the body over time after single or repeated intravenous administration.
Collapse
Affiliation(s)
- Sang-Jin Park
- Department of Toxicological Evaluation and Research, Korea Institute of Toxicology, 141 Gaejeongro, Yuseong gu, Daejeon, Republic of Korea
| | - Hae-Jin Yoon
- Department of Toxicological Evaluation and Research, Korea Institute of Toxicology, 141 Gaejeongro, Yuseong gu, Daejeon, Republic of Korea
| | - Eun-Young Gu
- Department of Toxicological Evaluation and Research, Korea Institute of Toxicology, 141 Gaejeongro, Yuseong gu, Daejeon, Republic of Korea
| | - Byoung-Seok Lee
- Department of Toxicological Evaluation and Research, Korea Institute of Toxicology, 141 Gaejeongro, Yuseong gu, Daejeon, Republic of Korea
| | - Yongman Kim
- NKMAX Co., Ltd, SNUH Healthcare Innovation Park, Seongnam, Gyeonggi-do Republic of Korea
| | - Jaeseob Jung
- NKMAX Co., Ltd, SNUH Healthcare Innovation Park, Seongnam, Gyeonggi-do Republic of Korea
| | - Jinmoon Kim
- NKMAX Co., Ltd, SNUH Healthcare Innovation Park, Seongnam, Gyeonggi-do Republic of Korea
| | - Kyoung-Sik Moon
- Department of Toxicological Evaluation and Research, Korea Institute of Toxicology, 141 Gaejeongro, Yuseong gu, Daejeon, Republic of Korea
| |
Collapse
|
6
|
The Hematology of Tomorrow Is Here-Preclinical Models Are Not: Cell Therapy for Hematological Malignancies. Cancers (Basel) 2022; 14:cancers14030580. [PMID: 35158848 PMCID: PMC8833715 DOI: 10.3390/cancers14030580] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/13/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell therapy is revolutionizing the prospect of deadly hematological malignancies such as high-risk acute myeloid leukemia. Stem cell therapy of allogeneic source from compatible human leukocyte antigen donor has exceptional success promoting durable remissions, but the rate of relapse is currently still high and there is transplant-related mortality. This review presents the current knowledge on the clinical use of mesenchymal stromal cells to improve outcomes in hematopoietic stem cell transplants. As an alternative or adjuvant approach to prevent relapse, we summarize the status of the promising forms of cellular immunotherapy aimed at targeting not only the bulk but also the cells of origin of leukemia. Finally, we discuss the available in vivo models for disease modelling and treatment efficacy prediction in these contexts. Abstract The purpose of this review is to present the current knowledge on the clinical use of several forms of cell therapy in hematological malignancies and the preclinical models available for their study. In the context of allogeneic hematopoietic stem cell transplants, mesenchymal stromal cells are pursued to help stem cell engraftment and expansion, and control graft versus host disease. We further summarize the status of promising forms of cellular immunotherapy including CAR T cell and CAR NK cell therapy aimed at eradicating the cells of origin of leukemia, i.e., leukemia stem cells. Updates on other forms of cellular immunotherapy, such as NK cells, CIK cells and CAR CIK cells, show encouraging results in AML. The considerations in available in vivo models for disease modelling and treatment efficacy prediction are discussed, with a particular focus on their strengths and weaknesses for the study of healthy and diseased hematopoietic stem cell reconstitution, graft versus host disease and immunotherapy. Despite current limitations, cell therapy is a rapidly evolving field that holds the promise of improved cure rates, soon. As a result, we may be witnessing the birth of the hematology of tomorrow. To further support its development, improved preclinical models including humanized microenvironments in mice are urgently needed.
Collapse
|
7
|
Min XY, Liu CF, Cao B, Zhang T, Yang X, Ma N, Wang N, Li K. Human CD3 +CD56 +NKT-like cells express a range of complement receptors and C3 activation has negative effects on these cell activity and effector function. Hum Immunol 2021; 82:625-633. [PMID: 34134908 DOI: 10.1016/j.humimm.2021.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/20/2021] [Accepted: 06/02/2021] [Indexed: 11/22/2022]
Abstract
CD3+CD56+NKT-like cells are a rare population of lymphocytes that serve important roles in various types of immune-related diseases, and particularly in cancer. The complement system regulates inflammatory and immune responses by interacting with complement receptors expressed on a range of immune cells. However, whether CD3+CD56+NKT-like cells are regulated by the complement system has still not been definitively determined. In the present study, the expression of complement receptors and regulators in gated CD3+CD56+NKT-like cells isolated from human peripheral blood was assessed using PCR and flow cytometry. The results showed that human CD3+CD56+NKT-like cells expressed a range of complement receptors and regulators, such as CR3, C3aR, C5aR, C5L2, CD46 and CD55. Furthermore, the presence of complement component 3 (C3), a key component in complement activation in culture supernatant, mitigated the activity, IFN-γ production and killing function of CD3+CD56+NKT-like cells. The present study provides evidences supporting the relationship between complement activation and functional modulation of CD3+CD56+NKT-like cells, expanding our knowledge of the complement regulatory network, and also highlighting a potential target for treatment of numerous immune-related diseases, particularly NKT cell-based tumor adoptive immunotherapy.
Collapse
Affiliation(s)
- Xiao-Yun Min
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| | - Cheng-Fei Liu
- Cancer Centre, The Second Hospital of Shandong University, Ji'nan, Shandong, PR China
| | - Bo Cao
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Ting Zhang
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Xiao Yang
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Ning Ma
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Na Wang
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China
| | - Ke Li
- Core Research Laboratory, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, PR China.
| |
Collapse
|
8
|
Limongello R, Marra A, Mancusi A, Bonato S, Hoxha E, Ruggeri L, Hui S, Velardi A, Pierini A. Novel Immune Cell-Based Therapies to Eradicate High-Risk Acute Myeloid Leukemia. Front Immunol 2021; 12:695051. [PMID: 34413848 PMCID: PMC8368440 DOI: 10.3389/fimmu.2021.695051] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/06/2021] [Indexed: 12/26/2022] Open
Abstract
Adverse genetic risk acute myeloid leukemia (AML) includes a wide range of clinical-pathological entities with extremely poor outcomes; thus, novel therapeutic approaches are needed. Promising results achieved by engineered chimeric antigen receptor (CAR) T cells in other blood neoplasms have paved the way for the development of immune cell-based therapies for adverse genetic risk AML. Among these, adoptive cell immunotherapies with single/multiple CAR-T cells, CAR-natural killer (NK) cells, cytokine-induced killer cells (CIK), and NK cells are subjects of ongoing clinical trials. On the other hand, allogeneic hematopoietic stem cell transplantation (allo-HSCT) still represents the only curative option for adverse genetic risk AML patients. Unfortunately, high relapse rates (above 50%) and associated dismal outcomes (reported survival ~10–20%) even question the role of current allo-HSCT protocols and emphasize the urgency of adopting novel effective transplant strategies. We have recently demonstrated that haploidentical allo-HSCT combined with regulatory and conventional T cells adoptive immunotherapy (Treg-Tcon haplo-HSCT) is able to overcome disease-intrinsic chemoresistance, prevent leukemia-relapse, and improve survival of adverse genetic risk AML patients. In this Perspective, we briefly review the recent advancements with immune cell-based strategies against adverse genetic risk AML and discuss how such approaches could favorably impact on patients’ outcomes.
Collapse
Affiliation(s)
- Roberto Limongello
- Institute of Hematology and Centre of Haemato-Oncology Research (CREO), University and Hospital of Perugia, Perugia, Italy
| | - Andrea Marra
- Institute of Hematology and Centre of Haemato-Oncology Research (CREO), University and Hospital of Perugia, Perugia, Italy
| | - Antonella Mancusi
- Institute of Hematology and Centre of Haemato-Oncology Research (CREO), University and Hospital of Perugia, Perugia, Italy
| | - Samanta Bonato
- Institute of Hematology and Centre of Haemato-Oncology Research (CREO), University and Hospital of Perugia, Perugia, Italy
| | - Eni Hoxha
- Institute of Hematology and Centre of Haemato-Oncology Research (CREO), University and Hospital of Perugia, Perugia, Italy
| | - Loredana Ruggeri
- Institute of Hematology and Centre of Haemato-Oncology Research (CREO), University and Hospital of Perugia, Perugia, Italy
| | - Susanta Hui
- Department of Radiation Oncology, City of Hope Medical Center, Duarte, CA, United States.,Beckman Research Institute of City of Hope, Duarte, CA, United States
| | - Andrea Velardi
- Institute of Hematology and Centre of Haemato-Oncology Research (CREO), University and Hospital of Perugia, Perugia, Italy
| | - Antonio Pierini
- Institute of Hematology and Centre of Haemato-Oncology Research (CREO), University and Hospital of Perugia, Perugia, Italy
| |
Collapse
|
9
|
Sommaggio R, Cappuzzello E, Dalla Pietà A, Tosi A, Palmerini P, Carpanese D, Nicolè L, Rosato A. Adoptive cell therapy of triple negative breast cancer with redirected cytokine-induced killer cells. Oncoimmunology 2020; 9:1777046. [PMID: 32923140 PMCID: PMC7458615 DOI: 10.1080/2162402x.2020.1777046] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cytokine-Induced Killer (CIK) cells share several functional and phenotypical properties of both T and natural killer (NK) cells. They represent an attractive approach for cell-based immunotherapy, as they do not require antigen-specific priming for tumor cell recognition, and can be rapidly expanded in vitro. Their relevant expression of FcγRIIIa (CD16a) can be exploited in combination with clinical-grade monoclonal antibodies (mAbs) to redirect their lytic activity in an antigen-specific manner. Here, we report the efficacy of this combined approach against triple negative breast cancer (TNBC), an aggressive tumor that still requires therapeutic options. Different primitive and metastatic TNBC cancer mouse models were established in NSG mice, either by implanting patient-derived TNBC samples or injecting MDA-MB-231 cells orthotopically or intravenously. The combined treatment consisted in the repeated intratumoral or intravenous injection of CIK cells and cetuximab. Tumor growth and metastasis were monitored by bioluminescence or immunohistochemistry, and survival was recorded. CIK cells plus cetuximab significantly restrained primitive tumor growth in mice, either in patient-derived tumor xenografts or MDA-MB-231 cell line models. Moreover, this approach almost completely abolished metastasis spreading and dramatically improved survival. The antigen-specific mAb favored tumor and metastasis tissue infiltration by CIK cells, and led to an enrichment of the CD16a+ subset. Data highlight the potentiality of this novel immunotherapy strategy where a nonspecific cytotoxic cell population can be converted into tumor-specific effectors with clinical-grade antibodies, thus providing not only a therapeutic option for TNBC but also a valid alternative to more complex approaches based on chimeric antigen receptor-engineered cells. List of abbreviations ACT, Adoptive Cell Transfer; ADCC, Antibody-Dependent Cell-mediated Cytotoxicity; ADP, Adenosine diphosphate; BLI, Bioluminescence Imaging; CAR, Chimeric Antigen Receptor; CIK, Cytokine Induced Killer cells; CTX, Cetuximab; DMEM, Dulbecco’s Modified Eagle Medium; EGFR, Human Epidermal Growth Factor 1; ER, Estrogen; FBS, Fetal Bovine Serum; FFPE, Formalin-Fixed Paraffin-Embedded; GMP, Good Manufacturing Practices; GVHD, Graft Versus Host Disease; HER2, Human Epidermal Growth Factor 2; HRP, Horseradish Peroxidase; IFN-γ, Interferon-γ; IHC, Immunohistochemistry; IL-2, Interleukin-2; ISO, Irrelevant antibody; i.t., intratumoral; i.v., intravenous, mAbs, Monoclonal Antibodies; mIHC, Multiplex Fluorescence Immunohistochemistry; MHC, Major Histocompatibility Complex; NK, Natural Killer; NKG2D, Natural-Killer group 2 member D; NSG, NOD/SCID common γ chain knockout; PARP, Poly ADP-ribose polymerase; PBMCs, Peripheral Blood Mononuclear Cells; PBS, Phosphate-buffered saline; PDX, Patient-derived xenograft; PR, Progesterone; rhIFN-γ, Recombinant Human Interferon-γ; RPMI, Roswell Park Memorial Institute; STR, Short tandem Repeat; TCR, T Cell Receptor; TNBC, Triple Negative Breast Cancer; TSA, Tyramide Signal Amplification
Collapse
Affiliation(s)
| | - Elisa Cappuzzello
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Anna Tosi
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | - Pierangela Palmerini
- Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| | | | - Lorenzo Nicolè
- Department of Medicine, Surgical Pathology & Cytopathology Unit, University of Padua, Padua, Italy
| | - Antonio Rosato
- Veneto Institute of Oncology IOV - IRCCS, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, Immunology and Oncology Section, University of Padua, Padua, Italy
| |
Collapse
|
10
|
Hansrivijit P, Gale RP, Barrett J, Ciurea SO. Cellular therapy for acute myeloid Leukemia – Current status and future prospects. Blood Rev 2019; 37:100578. [DOI: 10.1016/j.blre.2019.05.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 04/23/2019] [Accepted: 05/10/2019] [Indexed: 12/31/2022]
|
11
|
Luah YH, Sundar Raj K, Koh MBC, Linn YC. A novel simplified method of generating cytomegalovirus-specific cytokine-induced killer cells of high specificity and superior potency with GMP compliance. Clin Immunol 2019; 205:83-92. [PMID: 31229666 DOI: 10.1016/j.clim.2019.06.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 04/07/2019] [Accepted: 06/19/2019] [Indexed: 12/13/2022]
Abstract
We describe a method of rendering polyclonal cytokine-induced killer cells (CIK) specific against cytomegalovirus (CMV), focusing on GMP compliance. Peripheral blood mononuclear cells (PBMNC) are stimulated with pooled CMV peptides pp65 and IE-1 for 16-24 h and the reactive T cell subset which up-regulate CD137 is further co-stimulated with anti-CD137, followed by expansion in G-Rex flasks under standard CIK culture condition. This method generates a large number CMV-specific CIK with superior potency compared to published method currently in clinical trials. The cytotoxicity as measured by chromium release assay correlates with the upregulation of CD107a upon peptide re-challenge as measured by flow cytometry. CMV-CIK at maturity consist of mainly late effector memory CD8 T cells and have a skewed TCR repertoire with preferential expansion of a few families. Such CMV-CIK retain their function after freezing and thawing. CMV-CIK thus generated is ready for clinical trial against drug-resistant CMV disease.
Collapse
Affiliation(s)
- Yen Hoon Luah
- Cell Therapy Facility, Health Sciences Authority, 11 Outram Rd, 169078, Singapore
| | | | - Mickey B C Koh
- Cell Therapy Facility, Health Sciences Authority, 11 Outram Rd, 169078, Singapore
| | - Yeh Ching Linn
- Dept of Hematology, Singapore General Hospital, Outram Rd, 169608, Singapore.
| |
Collapse
|
12
|
Lee JB, Chen B, Vasic D, Law AD, Zhang L. Cellular immunotherapy for acute myeloid leukemia: How specific should it be? Blood Rev 2019; 35:18-31. [PMID: 30826141 DOI: 10.1016/j.blre.2019.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 02/05/2019] [Accepted: 02/22/2019] [Indexed: 12/25/2022]
Abstract
Significant improvements in the survival of patients with hematological cancers following hematopoietic stem cell transplantation provide evidence supporting the potency of immune cell-mediated anti-leukemic effects. Studies focusing on immune cell-based cancer therapies have made significant breakthroughs in the last few years. Adoptive cellular therapy (ACT), and chimeric antigen receptor (CAR) T cell therapy, in particular, has significantly increased the survival of patients with B cell acute lymphoblastic leukemia and aggressive B cell lymphoma. Despite antigen-negative relapses and severe toxicities such as cytokine release syndrome after treatment, CAR-T cell therapies have been approved by the FDA in some conditions. Although a number of studies have tried to achieve similar results for acute myeloid leukemia (AML), clinical outcomes have not been as promising. In this review, we summarize recent and ongoing studies on cellular therapies for AML patients, with a focus on antigen-specific versus -nonspecific approaches.
Collapse
Affiliation(s)
- Jong Bok Lee
- Toronto General Research Institute, University Health Network, 2-207 101 College St., Toronto, Ontario M5G 1L7, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada.
| | - Branson Chen
- Toronto General Research Institute, University Health Network, 2-207 101 College St., Toronto, Ontario M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| | - Daniel Vasic
- Toronto General Research Institute, University Health Network, 2-207 101 College St., Toronto, Ontario M5G 1L7, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| | - Arjun D Law
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, 6-711 700 University Ave., Toronto, Ontario M5G 1Z5, Canada.
| | - Li Zhang
- Toronto General Research Institute, University Health Network, 2-207 101 College St., Toronto, Ontario M5G 1L7, Canada; Department of Immunology, University of Toronto, Toronto, Ontario, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
13
|
Shirjang S, Alizadeh N, Mansoori B, Mahmoodpoor A, Kafil HS, Hojjat-Farsangi M, Yousefi M. Promising immunotherapy: Highlighting cytokine-induced killer cells. J Cell Biochem 2018; 120:8863-8883. [PMID: 30556298 DOI: 10.1002/jcb.28250] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 10/22/2018] [Indexed: 12/20/2022]
Abstract
For many years, cancer therapy has appeared to be a challenging issue for researchers and physicians. By the introduction of novel methods in immunotherapy, the prospect of cancer therapy even more explained than before. Cytokine-induced killer (CIK) cell-based immunotherapy demonstrated to have potentiality in improving clinical outcomes and relieving major side effects of standard treatment options. In addition, given the distinctive features such as high safety, low toxicity effects on healthy cells, numerous clinical trials conducted on CIK cells. Due to the shortcomings that observed in CIK cell immunotherapy alone, arising a tendency to make modifications (combined modality therapy or combination therapy) including the addition of various types of cytokines, genetic engineering, combination with immune checkpoints, and so on. In this review, we have tried to bring forth the latest immunotherapy methods and their overview. We have discussed the combination therapies with CIK cells and the conducted clinical trials. This helps the future studies to use integrated therapies with CIK cells as a promising treatment of many types of cancers.
Collapse
Affiliation(s)
- Solmaz Shirjang
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazila Alizadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Mansoori
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Combined Treatment with Autologous CIK Cells, Radiotherapy and Chemotherapy in Advanced Cervical Cancer. Pathol Oncol Res 2018; 25:691-696. [DOI: 10.1007/s12253-018-0541-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/06/2018] [Indexed: 01/05/2023]
|
15
|
Xiao X, Ye X, Xu C, Huang J. Successful alternative treatment for relapsed adult acute lymphoblastic leukemia with dendritic cells-cytokine-induced killer cells combined with a rituximab-based regimen. Onco Targets Ther 2018; 11:7555-7558. [PMID: 30464503 PMCID: PMC6214580 DOI: 10.2147/ott.s177503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Objective Acute lymphoblastic leukemia (ALL) is a malignant disease characterized by the accumulation of lymphoblasts, and a poor prognosis for adults with ALL is closely associated with disease recurrence. Thus far, treatment approaches have been limited, particularly in patients who are unable to tolerate chemotherapy. In this study, we report an effective treatment for such patients. Materials and methods A 52-year-old man diagnosed with Ph-negative B-precursor ALL went into remission after inductive treatment. Unfortunately, when he subsequently relapsed, severe complications drove him to refuse intensive chemotherapy. Instead, he received a cycle of dendritic cells-cytokine-induced killer cells (DC-CIK) before chemotherapy. Result The patient tolerated rituximab in combination with a vincristine, daunorubicin, l-asparaginase, and prednisone regimen without complications, and was in remission after DC-CIK infusion. After consolidation chemotherapy, including rituximab followed by eight cycles of DC-CIK, the patient has been free of leukemia for 2 years since the relapse. Conclusion This case of relapsed ALL was successfully treated with DC-CIK combined with a rituximab regimen.
Collapse
Affiliation(s)
- XiaoFang Xiao
- Department of Hematology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China,
| | - XingNong Ye
- Department of Hematology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China,
| | - Cheng Xu
- Department of Hematology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China,
| | - Jian Huang
- Department of Hematology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China,
| |
Collapse
|
16
|
Meng M, Li L, Li R, Wang W, Chen Y, Xie Y, Han R, Zhu K, Huang W, Yang L, Li S, Shi J, Tan W, Gao H, Zhao Y, Yang L, Tan J, Hou Z. A dynamic transcriptomic atlas of cytokine-induced killer cells. J Biol Chem 2018; 293:19600-19612. [PMID: 30333226 PMCID: PMC6314136 DOI: 10.1074/jbc.ra118.003280] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 10/11/2018] [Indexed: 12/31/2022] Open
Abstract
Several clinical immunotherapy trials with cytokine-induced killer (CIK) cells have been reported. However, molecular evidence of cell expansion, acquisition of tumor cytotoxicity, and safety of CIK cells is required before putting them to clinical use. Here, we performed dynamic transcriptomic analyses of CIKs generated from primary peripheral blood mononuclear cells exposed to interferon-γ, OKT3, and interleukin-2. CIK mRNAs were extracted and sequenced at days 0, 1, 7, and 14 and subjected to bioinformatics analyses. Using weighted correlation network analysis (WGCNA), we identified two major gene modules that mediate immune cell activation and mitosis. We found that activation and cytotoxicity of CIK cells likely rely on cluster of differentiation 8 (CD8) and its protein partner LCK proto-oncogene, Src family tyrosine kinase (LCK). A time-course series analysis revealed that CIK cells have relatively low immunogenicity because of decreased expression of some self-antigens. Importantly, we identified several crucial activating receptors and auxiliary adhesion receptors expressed on CIK cells that may function as tumor sensors. Interestingly, cytotoxicity-associated genes, including those encoding PRF1, GZMB, FASL, and several cytokines, were up-regulated in mature CIK cells. Most immune-checkpoint molecules and inflammatory tumor-promoting factors were down-regulated in the CIK cells, suggesting efficacy and safety in future clinical trials. Notably, insulin-like growth factor 1 (IGF-1) was highly expressed in CIK cells and may promote cytotoxicity, although it also could facilitate tumorigenesis. The transcriptomic atlas of CIK cells presented here may inform efforts to improve CIK-associated tumor cytotoxicity and safety in clinical trials.
Collapse
Affiliation(s)
- Mingyao Meng
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Lin Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Ruhong Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Wenju Wang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Yang Chen
- the Ministry of Education (MOE) Key Laboratory of Bioinformatics, Bioinformatics Division and Center for Synthetic and Systems Biology, BNRist, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Yanhua Xie
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Rui Han
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Kai Zhu
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Wenwen Huang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Lili Yang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Shuo Li
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Jianlin Shi
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Weiwei Tan
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Hui Gao
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Yiyi Zhao
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Li Yang
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China.,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Jing Tan
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China, .,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| | - Zongliu Hou
- From the Yan'an Affiliated Hospital of Kunming Medical University, Kunming 650051, Yunnan, China, .,the Key Laboratory of Tumor Immunological Prevention and Treatment of Yunnan Province, Kunming 650051, Yunnan, China, and
| |
Collapse
|
17
|
Introna M, Correnti F. Innovative Clinical Perspectives for CIK Cells in Cancer Patients. Int J Mol Sci 2018; 19:ijms19020358. [PMID: 29370095 PMCID: PMC5855580 DOI: 10.3390/ijms19020358] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 01/19/2018] [Accepted: 01/22/2018] [Indexed: 12/18/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are T lymphocytes that have acquired, in vitro, following extensive manipulation by Interferon gamma (IFN-γ), OKT3 and Interleukin 2 (IL-2) addition, the expression of several Natural Killer (NK) cell-surface markers. CIK cells have a dual "nature", due to the presence of functional TCR as well as NK molecules, even if the antitumoral activity can be traced back only to the NK-like structures (DNAM-1, NKG2D, NKp30 and CD56). In addition to antineoplastic activity in vitro and in several in-vivo models, CIK cells show very limited, if any, GvHD toxicity as well as a strong intratumoral homing. For all such reasons, CIK cells have been proposed and tested in many clinical trials in cancer patients both in autologous and allogeneic combinations, up to haploidentical mismatching. Indeed, genetic modification of CIK cells as well as the possibility of combining them with specific monoclonal antibodies will further expand the possibility of their clinical utilization.
Collapse
Affiliation(s)
- Martino Introna
- USS Center of Cell Therapy "G. Lanzani", USC Ematologia, ASST Papa Giovanni XXIII Bergamo, 24124 Bergamo, Italy.
| | - Fabio Correnti
- USS Center of Cell Therapy "G. Lanzani", USC Ematologia, ASST Papa Giovanni XXIII Bergamo, 24124 Bergamo, Italy.
| |
Collapse
|
18
|
Introna M. CIK as therapeutic agents against tumors. J Autoimmun 2017; 85:32-44. [DOI: 10.1016/j.jaut.2017.06.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 06/19/2017] [Accepted: 06/19/2017] [Indexed: 01/26/2023]
|
19
|
Wang Y, Lv B, Li K, Zhang A, Liu H. Adjuvant immunotherapy of dendritic cells and cytokine-induced killer cells is safe and enhances chemotherapy efficacy for multiple myeloma in China: a meta-analysis of clinical trials. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:3245-3256. [PMID: 29180849 PMCID: PMC5695269 DOI: 10.2147/dddt.s146959] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Objective The aim of this study was to systematically evaluate the efficacy and safety of the combination of dendritic cells and cytokine-induced killer cells (DC–CIK) adjuvant immunotherapy and chemotherapy in the treatment of multiple myeloma (MM). Methods Clinical trials were gathered by searching Web of Science, PubMed, Embase, Cochrane Library, Wanfang, and CNKI database. Outcome measurements including therapeutic efficacy, prognosis, immune function, and adverse events were extracted and evaluated. Results A total of 12 trials including 594 MM patients were involved in this study for statistical analysis. Results indicated that compared to chemotherapy alone, the combination of DC–CIK immunotherapy with chemotherapy significantly improved patients’ overall response rate (ORR, odds ratio [OR] =2.77, 95% confidence interval [CI] =1.88–4.10, P<0.00001), disease control rate (DCR, OR =2.90, CI =1.72–4.90, P<0.0001), and life quality (P<0.00001). The combined therapy showed advantages over chemotherapy alone in prognostic indicators including percentage of tumor cells (P=0.04), serum levels of β2-microglobin (P<0.0001), M protein (P<0.00001), and creatinine (P<0.0001), and 24 h urine light chains (P<0.00001). After combined treatment, CD4+ lymphocyte subsets’ percentages, CD4+/CD8+ ratio, and cytokines levels of AgNOR, IFN-γ, IL-2, and IL-12 were significantly increased (P<0.05), whereas CD8+ and CD4+CD25+ percentages and IL-4, IL-6, IL-10, and TGF-β levels were obviously decreased (P<0.01), indicating a recovered immune condition. Conclusion Adjuvant DC–CIK immunotherapy enhances the efficacy of chemotherapy for MM and improves prognosis probably by reconstructing immune function.
Collapse
Affiliation(s)
- Yan Wang
- Department of Clinical Laboratory
| | | | - Ke Li
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong Province, China
| | - Anqi Zhang
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong Province, China
| | - Hong Liu
- Department of Central Laboratory, Liaocheng People's Hospital, Liaocheng Clinical School of Taishan Medical University, Liaocheng, Shandong Province, China
| |
Collapse
|
20
|
Li DP, Li W, Feng J, Chen K, Tao M. Adjuvant chemotherapy with sequential cytokine-induced killer (CIK) cells in stage IB non-small cell lung cancer. Oncol Res 2017; 22:67-74. [PMID: 25706393 PMCID: PMC7838450 DOI: 10.3727/096504014x14024160459168] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
For non-small cell lung cancer (NSCLC) patients at stage IB, adjuvant chemotherapy does not improve survival. Evidence suggests that dendritic cell (DC)-activated cytokine-induced killer (DC-CIK) cell therapy in addition to chemotherapy improves survival for stage I-IIIA NSCLC patients after surgery, but there are not enough data to confirm this benefit specifically for those at stage IB. Herein, we retrospectively evaluated the efficacy and safety of this therapy administered to stage IB NSCLC patients. Sixty-six patients were treated with four-cycle adjuvant chemotherapy initiated 3 weeks after surgical resection. In addition, 28 of these patients underwent DC-CIK therapy on a trimonthly basis (average 3.1 times, range 1-6) beginning 1 month after chemotherapy. The disease-free survival (DFS) rates of the two groups were statistically similar, although patients who received DC-CIK therapy showed slightly higher 1- and 2-year DFS rates (100.0% and 96.4%, respectively, compared with 81.6% and 76.3%). More importantly, patients in the DC-CIK therapy group had significantly longer overall survival (p=0.018). For patients who received treatment after recurrence, the DC-CIK therapy group had longer progression-free survival compared with the chemotherapy-only group. In addition, patients given DC-CIK therapy experienced less fatigue and appetite loss. The rate of adverse side effects was similar between the two groups. In conclusion, for these stage IB NSCLC patients, DC-CIK therapy significantly improved 2-year DFS rates compared with those who received chemotherapy only. DC-CIK therapy also benefited patients' quality of life, and adverse events were acceptable.
Collapse
Affiliation(s)
- Da-Peng Li
- Department of Oncology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, People's Republic of China
| | | | | | | | | |
Collapse
|
21
|
Luo H, Gong L, Zhu B, Huang Y, Tang C, Yu S, Yang Z, Zhou X. Therapeutic outcomes of autologous CIK cells as a maintenance therapy in the treatment of lung cancer patients: A retrospective study. Biomed Pharmacother 2016; 84:987-993. [PMID: 27764762 DOI: 10.1016/j.biopha.2016.10.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2016] [Revised: 10/03/2016] [Accepted: 10/09/2016] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Few clinical studies have confirmed the role of cytokine-induced killer cells (CIKs) in the maintenance therapy of advanced lung cancer patients. We investigate effectiveness and tolerability of CIKs as a maintenance therapy in the treatment of advanced lung cancer patients. METHODS 70 patients with advanced lung cancer (stage IIIB to IV) admitted to the First Affiliated Hospital of Third Military Medical University in Chongqing from Nov. 2011 to Jan. 2015 and treated with CIKs were enrolled as a CIKs group (T group), and another 70 advanced lung cancer patients treated with optimal supportive care during the same period were enrolled as a control group(C group). The changes of immune system, response rate, disease control rate, overall survival, and side effects were compared between the two groups. Furthermore, the factors that might influence the efficacy of CIKs therapy were evaluated. RESULTS Compared with the healthy people, the ratios of CD3+, CD4+ and CD8+ T cells significantly decreased (P<0.05) in lung cancer patients. After CIKs treatment, the ratios of CD3+ and CD4+ T cells and CD4+/CD8+ significantly increased (P<0.05). The response rate (RR) and disease control rate (DCR) were 34.3% and 80.0% in the CIKs group, which were significantly higher than those in the control group (11.4% and 54.3%, both P<0.05). Besides, the median PFS was significantly improved in the CIKs group than that in control group (6 months vs. 4 months, P<0.05). Although median OS was 28 months in CIKs group while 22 months in control group, no significant difference was observed (P>0.05). However, The 2-year, 3-year survival rates were 56.8% and 21.6% in the CIK group, respectively, which were significantly improved compared to that in the control group (both P<0.05). KPS score significantly increased in the CIKs group (P=0.001). 6 patients suffered from transient fever or chills in the process of CIKs transfusion, and no other side effect was observed. Furthermore, we also found that TNM stage, tumor size, metastasis in vital organs and KPS score were all factors associated with efficacy of CIKs treatment. CONCLUSION CIKs treatment, as a maintenance therapy, is safe and effective for advanced lung cancer patients, and can also improve the immune imbalance, RR, DCR, PFS, OS and quality of life of the lung cancer patients.
Collapse
Affiliation(s)
- Hu Luo
- Departments of Respiratory Medicine, The First Affiliated Hospital of Third Military Medical University, Chongqing 400038, PR China
| | - Liang Gong
- Departments of Respiratory Medicine, The First Affiliated Hospital of Third Military Medical University, Chongqing 400038, PR China
| | - Bingjing Zhu
- Departments of Respiratory Medicine, The First Affiliated Hospital of Third Military Medical University, Chongqing 400038, PR China
| | - Ying Huang
- Departments of Respiratory Medicine, The First Affiliated Hospital of Third Military Medical University, Chongqing 400038, PR China
| | - Chunlan Tang
- Departments of Respiratory Medicine, The First Affiliated Hospital of Third Military Medical University, Chongqing 400038, PR China
| | - Shicang Yu
- Center of Biotherapy, The First Affiliated Hospital of Third Military Medical University, Chongqing 400038, PR China
| | - Zhi Yang
- Center of Biotherapy, The First Affiliated Hospital of Third Military Medical University, Chongqing 400038, PR China
| | - Xiangdong Zhou
- Departments of Respiratory Medicine, The First Affiliated Hospital of Third Military Medical University, Chongqing 400038, PR China.
| |
Collapse
|
22
|
Allogeneic hematopoietic stem cell transplantation following donor CIK cell infusion: A phase I study in patients with relapsed/refractory hematologic malignancies. Leuk Res 2016; 48:6-10. [DOI: 10.1016/j.leukres.2016.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 01/16/2023]
|
23
|
Chan WC, Linn YC. A comparison between cytokine- and bead-stimulated polyclonal T cells: the superiority of each and their possible complementary role. Cytotechnology 2016; 68:735-48. [PMID: 25481728 PMCID: PMC4960124 DOI: 10.1007/s10616-014-9825-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 11/17/2014] [Indexed: 01/13/2023] Open
Abstract
Cytokine-induced killer (CIK) cells and T cells expanded by co-stimulation with beads presenting anti-CD3 and -CD28 antibodies are both polyclonal T cells under intensive laboratory and clinical studies, but there has not been any direct comparison between both. We compared the expansion, memory T cell subsets and cytotoxicity for T cells expanded in parallel by the two methods. Bead-stimulated T cells showed superior expansion as compared to CIK cells on D14 of culture. Bead-stimulated T cells consisted of a significantly higher CD4(+) subset and significantly lower CD8(+) subset as compared to CIK cells, as well as a higher proportion of less terminally differentiated T cells and a higher proportion of homing molecules. On the other hand, CIK cells exhibited significantly superior cytotoxicity against two myelomonocytic leukemia cell lines (THP-1 and U937) and two RCC cell lines (786.0 and CaKi-2). The cytotoxicity on D14 against THP-1 was 58.1 % for CIK cells and 8.3 % for bead-stimulated T cells at E:T of 10:1 (p < 0.01). Cytotoxicity correlated positively with the proportion of the CD8 subset in the culture and was independent of NKG2D recognition of susceptible targets. Polyclonal T cells expanded by different methods exhibit different characteristics which may define the specific role of each in different clinical scenario. We postulate that the more potent CIK cells may offer short term benefit while bead-stimulated T cells may offer a more sustained immune response.
Collapse
Affiliation(s)
- Weng-Chee Chan
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | - Yeh-Ching Linn
- Department of Haematology, Singapore General Hospital, Academia, Level 3, 20, College Road, Singapore, 169856, Singapore.
| |
Collapse
|
24
|
Lan XP, Chen YG, Wang Z, Yuan CW, Wang GG, Lu GL, Mao SW, Jin XB, Xia QH. Immunotherapy of DC-CIK cells enhances the efficacy of chemotherapy for solid cancer: a meta-analysis of randomized controlled trials in Chinese patients. J Zhejiang Univ Sci B 2016; 16:743-56. [PMID: 26365116 DOI: 10.1631/jzus.b1500003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Professional antigen-presenting dendritic cells (DCs) and cytokine-induced killer (CIK) cells, components of anti-cancer therapy, have shown clinical benefits and potential to overcome chemotherapeutic resistance. To evaluate whether DC-CIK cell-based therapy improves the clinical efficacy of chemotherapy, we reviewed the literature on DC-CIK cells and meta-analyzed randomized controlled trials (RCTs). METHODS We searched several databases and selected studies using predefined criteria. RCTs that applied chemotherapy with and without DC-CIK cells separately in two groups were included. Odds ratio (OR) and mean difference (MD) were reported to measure the pooled effect. RESULTS Twelve reported RCTs (826 patients), which were all performed on Chinese patients, were included. Combination therapy exhibited better data than chemotherapy: 1-year overall survival (OS) (OR=0.22, P<0.01), 2-year OS (OR=0.28, P<0.01), 3-year OS (OR=0.41, P<0.01), 1-year disease-free survival (DFS) (OR=0.16, P<0.05), 3-year DFS (OR=0.32, P<0.01), objective response rate (ORR) (OR=0.54, P<0.01), and disease control rate (DCR) (OR=0.46, P<0.01). Moreover, the levels of CD3(+) T-lymphocytes (MD=-11.65, P<0.05) and CD4(+) T-lymphocytes (MD=-8.18, P<0.01) of the combination group were higher. CONCLUSIONS Immunotherapy of DC-CIK cells may enhance the efficacy of chemotherapy on solid cancer and induces no specific side effect. Further RCTs with no publishing bias should be designed to confirm the immunotherapeutic effects of DC-CIK cells.
Collapse
Affiliation(s)
- Xiao-peng Lan
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| | - You-gen Chen
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| | - Zheng Wang
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| | - Chuan-wei Yuan
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| | - Gang-gang Wang
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| | - Guo-liang Lu
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| | - Shao-wei Mao
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| | - Xun-bo Jin
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| | - Qing-hua Xia
- Minimally Invasive Urology Center, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250014, China
| |
Collapse
|
25
|
Samudio I, Rezvani K, Shaim H, Hofs E, Ngom M, Bu L, Liu G, Lee JTC, Imren S, Lam V, Poon GFT, Ghaedi M, Takei F, Humphries K, Jia W, Krystal G. UV-inactivated HSV-1 potently activates NK cell killing of leukemic cells. Blood 2016; 127:2575-86. [PMID: 26941401 PMCID: PMC4892253 DOI: 10.1182/blood-2015-04-639088] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 02/26/2016] [Indexed: 11/20/2022] Open
Abstract
Herein we demonstrate that oncolytic herpes simplex virus-1 (HSV-1) potently activates human peripheral blood mononuclear cells (PBMCs) to lyse leukemic cell lines and primary acute myeloid leukemia samples, but not healthy allogeneic lymphocytes. Intriguingly, we found that UV light-inactivated HSV-1 (UV-HSV-1) is equally effective in promoting PBMC cytolysis of leukemic cells and is 1000- to 10 000-fold more potent at stimulating innate antileukemic responses than UV-inactivated cytomegalovirus, vesicular stomatitis virus, reovirus, or adenovirus. Mechanistically, UV-HSV-1 stimulates PBMC cytolysis of leukemic cells, partly via Toll-like receptor-2/protein kinase C/nuclear factor-κB signaling, and potently stimulates expression of CD69, degranulation, migration, and cytokine production in natural killer (NK) cells, suggesting that surface components of UV-HSV-1 directly activate NK cells. Importantly, UV-HSV-1 synergizes with interleukin-15 (IL-15) and IL-2 in inducing activation and cytolytic activity of NK cells. Additionally, UV-HSV-1 stimulates glycolysis and fatty acid oxidation-dependent oxygen consumption in NK cells, but only glycolysis is required for their enhanced antileukemic activity. Last, we demonstrate that T cell-depleted human PBMCs exposed to UV-HSV-1 provide a survival benefit in a murine xenograft model of human acute myeloid leukemia (AML). Taken together, our results support the preclinical development of UV-HSV-1 as an adjuvant, alone or in combination with IL-15, for allogeneic donor mononuclear cell infusions to treat AML.
Collapse
Affiliation(s)
- Ismael Samudio
- Programa de Investigacion e Innovacion en Leucemia Aguda y Cronica, Bogotá, Colombia; Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Hila Shaim
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX; and
| | - Elyse Hofs
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Mor Ngom
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Luke Bu
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Guoyu Liu
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Jason T C Lee
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Suzan Imren
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Vivian Lam
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Grace F T Poon
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Maryam Ghaedi
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Fumio Takei
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - Keith Humphries
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| | - William Jia
- Brain Research Centre, University of British Columbia, Vancouver, Canada
| | - Gerald Krystal
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, Canada
| |
Collapse
|
26
|
Poh SL, Linn YC. Immune checkpoint inhibitors enhance cytotoxicity of cytokine-induced killer cells against human myeloid leukaemic blasts. Cancer Immunol Immunother 2016; 65:525-36. [PMID: 26961084 PMCID: PMC11029729 DOI: 10.1007/s00262-016-1815-8] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Accepted: 02/22/2016] [Indexed: 01/10/2023]
Abstract
We studied whether blockade of inhibitory receptors on cytokine-induced killer (CIK) cells by immune checkpoint inhibitors could increase its anti-tumour potency against haematological malignancies. CIK cultures were generated from seven normal donors and nine patients with acute myeloid leukaemia (AML), acute lymphoblastic leukaemia (ALL) or multiple myeloma (MM). The inhibitory receptors B and T lymphocyte attenuator, CD200 receptor, lymphocyte activation gene-3 (LAG-3) and T cell immunoglobulin and mucin-domain-containing-3 (TIM-3) were present at variable percentages in most CIK cultures, while cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed death-1 (PD-1) and killer cell immunoglobulin-like receptors (KIR2DL1/2/3) were expressed at low level in most cultures. Without blockade, myeloid leukaemia cells were susceptible to autologous and allogeneic CIK-mediated cytotoxicity. Blockade of KIR, LAG-3, PD-1 and TIM-3 but not CTLA-4 resulted in remarkable increase in killing against these targets, even in those with poor baseline cytotoxicity. ALL and MM targets were resistant to CIK-mediated cytotoxicity, and blockade of receptors did not increase cytotoxicity to a meaningful extent. Combination of inhibitors against two receptors did not further increase cytotoxicity. Interestingly, potentiation of CIK killing by blocking antibodies was not predicted by expression of receptors on CIK and their respective ligands on the targets. Compared to un-activated T and NK cells, blockade potentiated the cytotoxicity of CIK cells to a greater degree and at a lower E:T ratio, but without significant increase in cytotoxicity against normal white cell. Our findings provide the basis for clinical trial combining autologous CIK cells with checkpoint inhibitors for patients with AML.
Collapse
MESH Headings
- Acute Disease
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/pharmacology
- Antigens, Surface/immunology
- Antigens, Surface/metabolism
- CTLA-4 Antigen/immunology
- CTLA-4 Antigen/metabolism
- Cell Line, Tumor
- Cytokine-Induced Killer Cells/drug effects
- Cytokine-Induced Killer Cells/immunology
- Cytokine-Induced Killer Cells/metabolism
- Cytotoxicity Tests, Immunologic/methods
- Cytotoxicity, Immunologic/drug effects
- Cytotoxicity, Immunologic/immunology
- Hepatitis A Virus Cellular Receptor 2
- Humans
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/pathology
- Membrane Proteins/immunology
- Membrane Proteins/metabolism
- Neoplastic Stem Cells/immunology
- Orexin Receptors
- Programmed Cell Death 1 Receptor/immunology
- Programmed Cell Death 1 Receptor/metabolism
- Receptors, Cell Surface/immunology
- Receptors, Cell Surface/metabolism
- Receptors, KIR/immunology
- Receptors, KIR/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Su Li Poh
- Department of Haematology, Singapore General Hospital, Level 3, Academia, 20, College Road, Singapore, 169856, Singapore
| | - Yeh Ching Linn
- Department of Haematology, Singapore General Hospital, Level 3, Academia, 20, College Road, Singapore, 169856, Singapore.
| |
Collapse
|
27
|
Zhang B, Zhao W, Li H, Chen Y, Tian H, Li L, Zhang L, Gao C, Zheng J. Immunoreceptor TIGIT inhibits the cytotoxicity of human cytokine-induced killer cells by interacting with CD155. Cancer Immunol Immunother 2016; 65:305-14. [PMID: 26842126 PMCID: PMC11029225 DOI: 10.1007/s00262-016-1799-4] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/19/2016] [Indexed: 12/22/2022]
Abstract
T cell Ig and ITIM domain (TIGIT) is a newly identified inhibitory receptor expressed on T and natural killer (NK) cells. Cytokine-induced killer (CIK) cells express CD3 and CD56 molecules, and share functional properties with both NK and T cells. However, it remains unknown whether TIGIT is expressed in CIK cells. Here, we show that TIGIT is expressed by CIK cells and interacts with CD155. By blocking TIGIT using an anti-TIGIT functional antibody, we demonstrate that CIK cells display increased proliferation; higher cytotoxic targeting of tumor cells expressing CD155; and higher expression of interferon-γ (IFN-γ), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α). Furthermore, increases in IFN-γ and cytotoxicity by blockade of TIGIT were reduced by blocking DNAX accessory molecule-1 (DNAM-1) signaling, implying that TIGIT exerts immunosuppressive effects by competing with DNAM-1 for the same ligand, CD155. Our results provide evidence that blockade of TIGIT may be a novel strategy to improve the cytotoxic activity of CIK cells.
Collapse
Affiliation(s)
- Baofu Zhang
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Weina Zhao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Huizhong Li
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Yuanyuan Chen
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Hui Tian
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Liantao Li
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Longzhen Zhang
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China
| | - Chao Gao
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China.
| | - Junnian Zheng
- Cancer Center, The Affiliated Hospital of Xuzhou Medical College, 89 West Huai-hai Road, Xuzhou, 221006, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, 221002, Jiangsu, China.
| |
Collapse
|
28
|
Selection and expansion of natural killer cells for NK cell-based immunotherapy. Cancer Immunol Immunother 2016; 65:477-84. [PMID: 26810567 PMCID: PMC4826432 DOI: 10.1007/s00262-016-1792-y] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 01/01/2016] [Indexed: 01/19/2023]
Abstract
Natural killer (NK) cells have been used in several clinical trials as adaptive immunotherapy. The low numbers of these cells in peripheral blood mononuclear cells (PBMC) have resulted in various approaches to preferentially expand primary NK cells from PBMC. While some clinical trials have used the addition of interleukin 2 (IL-2) to co-stimulate the expansion of purified NK cells from allogeneic donors, recent studies have shown promising results in achieving in vitro expansion of NK cells to large numbers for adoptive immunotherapy. NK cell expansion requires multiple cell signals for survival, proliferation and activation. Thus, expansion strategies have been focused either to substitute these factors using autologous feeder cells or to use genetically modified allogeneic feeder cells. Recent developments in the clinical use of genetically modified NK cell lines with chimeric antigen receptors, the development of expansion protocols for the clinical use of NK cell from human embryonic stem cells and induced pluripotent stem cells are challenging improvements for NK cell-based immunotherapy. Transfer of several of these protocols to clinical-grade production of NK cells necessitates adaptation of good manufacturing practice conditions, and the development of freezing conditions to establish NK cell stocks will require some effort and, however, should enhance the therapeutic options of NK cells in clinical medicine.
Collapse
|
29
|
Pfirrmann V, Oelsner S, Rettinger E, Huenecke S, Bonig H, Merker M, Wels WS, Cinatl J, Schubert R, Klingebiel T, Bader P. Cytomegalovirus-specific cytokine-induced killer cells: concurrent targeting of leukemia and cytomegalovirus. Cytotherapy 2015; 17:1139-51. [PMID: 26072027 DOI: 10.1016/j.jcyt.2015.04.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 01/10/2023]
Abstract
BACKGROUND AIMS Human cytomegalovirus (CMV) infection and reactivation is a leading complication of allogeneic hematopoietic stem cell transplantation (HSCT). In addition to drug treatment, the adoptive transfer of virus-specific T cells to restore cellular immunity has become a standard therapy after allogeneic HSCT. We recently demonstrated potent anti-leukemic activity of interleukin (IL)-15-activated cytokine-induced killer (CIK) cells. With the use of the same expansion protocol, we asked whether concurrent CMV antigen-pulsing might generate CIK cells with anti-leukemic and anti-CMV activity. METHODS CIK cells expanded in the presence of interferon-γ, IL-2, IL-15 and anti-CD3 antibody were pulsed once with CMV(pp65) peptide pool. CMV-specific CIK (CIK(pp65)) and conventional CIK cells were phenotypically and functionally characterized according to their cytokine secretion pattern, degranulation capacity and T-cell receptor (TCR)-mediated and NKG2D-mediated cytotoxicity. RESULTS We demonstrated that among CIK cells generated from CMV-seropositive donors, a single stimulation with CMV(pp65) protein co-expanded cytotoxic CMV-specific cells without sacrificing anti-tumor reactivity. Cells generated in this fashion lysed CMV(pp65)-loaded target cells and CMV-infected fibroblasts but also leukemic cells. Meanwhile, the alloreactive potential of CIK(pp65) cells remained low. Interestingly, CMV reactivity was TCR-mediated and CMV-specific cells could be found in CD3(+)CD8(+)CD56(+/-) cytotoxic T-cell subpopulations. CONCLUSIONS We provide an efficient method to generate CIK(pp65) cells that may represent a useful cell therapy approach for preemptive immunotherapy in patients who have both an apparent risk of CMV and impending leukemic relapse after allogeneic stem cell transplantation.
Collapse
Affiliation(s)
- Verena Pfirrmann
- Division of Stem Cell Transplantation and Immunology, Department of Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany.
| | - Sarah Oelsner
- Division of Stem Cell Transplantation and Immunology, Department of Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany; Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Eva Rettinger
- Division of Stem Cell Transplantation and Immunology, Department of Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Sabine Huenecke
- Division of Stem Cell Transplantation and Immunology, Department of Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology and German Red Cross Blood Donor Service, University Hospital Frankfurt, Goethe University, Baden-Wuerttemberg-Hessen, Frankfurt/Main, Germany
| | - Michael Merker
- Division of Stem Cell Transplantation and Immunology, Department of Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Winfried S Wels
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt/Main, Germany
| | - Jindrich Cinatl
- Institute for Experimental Cancer Research in Pediatrics, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Ralf Schubert
- Division of Allergology, Pneumology and Cystic Fibrosis, Department of Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Thomas Klingebiel
- Division of Stem Cell Transplantation and Immunology, Department of Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany
| | - Peter Bader
- Division of Stem Cell Transplantation and Immunology, Department of Children and Adolescents, University Hospital Frankfurt, Goethe University, Frankfurt/Main, Germany.
| |
Collapse
|
30
|
Zhang Q, Liu XY, Zhang T, Zhang XF, Zhao L, Long F, Liu ZK, Wang EH. The dual-functional capability of cytokine-induced killer cells and application in tumor immunology. Hum Immunol 2015; 76:385-91. [DOI: 10.1016/j.humimm.2014.09.021] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 09/27/2014] [Accepted: 09/27/2014] [Indexed: 01/18/2023]
|
31
|
Schmeel LC, Schmeel FC, Coch C, Schmidt-Wolf IGH. Cytokine-induced killer (CIK) cells in cancer immunotherapy: report of the international registry on CIK cells (IRCC). J Cancer Res Clin Oncol 2015; 141:839-49. [PMID: 25381063 DOI: 10.1007/s00432-014-1864-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/21/2014] [Indexed: 01/05/2023]
Abstract
PURPOSE Cytokine-induced killer (CIK) cells represent an exceptional T cell population uniting a T cell and natural killer cell like phenotype in their terminally differentiated CD3(+)CD56(+) subset, which features non-MHC-restricted tumor-killing activity. CIK cells are expandable from peripheral blood mononuclear cells and mature following the addition of certain cytokines. CIK cells have provided encouraging results in initial clinical studies and revealed synergistic antitumor effects when combined with standard therapeutic procedures. METHODS Therefore, we established the international registry on CIK cells in order to collect and evaluate data about clinical trials using CIK cells for the treatment of cancer patients. Moreover, our registry is expected to set new standards on the reporting of results from clinical trials using CIK cells. Clinical responses, overall survival (OS), adverse reactions and immunologic effects were analyzed in 45 studies present in our database. These studies investigated 22 different tumor entities altogether enrolling 2,729 patients. RESULTS A mean response rate of 39 % and significantly increased OS, accompanied by an improved quality of life, were reported. Interestingly, side effects of CIK cell treatment were minor. Mild fevers, chills, headache and fatigue were, however, seen regularly after CIK cell infusion. Moreover, CIK cells revealed numerous immunologic effects such as changes in T cell subsets, tumor markers, cytokine secretion and HBV viral load. CONCLUSION Due to their easy availability and potent antitumor activity, CIK cells emerged as a promising immunotherapy approach in oncology and may gain major importance on the prognosis of cancer.
Collapse
Affiliation(s)
- Leonard Christopher Schmeel
- Department of Internal Medicine III, Center for Integrated Oncology (CIO), University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Straße 25, 53105, Bonn, Germany
| | | | | | | |
Collapse
|
32
|
Yang XY, Zeng H, Chen FP. Cytokine-induced killer cells: A novel immunotherapy strategy for leukemia. Oncol Lett 2014; 9:535-541. [PMID: 25621022 PMCID: PMC4301482 DOI: 10.3892/ol.2014.2780] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 11/12/2014] [Indexed: 12/14/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are NK-like T cells derived from peripheral blood mononuclear cells that are co-stimulated and expanded using cytokines for 14–21 days in vitro. CIK cells are a heterogeneous subset of highly-efficient cytotoxic T effector cells that mediate major histocompatibility complex-unrestricted cytotoxicity against a broad array of tumor cells. These effector cells are generated from patients with leukemia or healthy donors who demonstrate similar cytotoxic activity against leukemia blasts. Allogeneic CIK cells retain the ability to produce the graft versus tumor response and generate minimal graft versus host disease. In addition, CIK cells possess no cytotoxicity against normal hematopoietic stem cells in vivo. Leukemia recurrence remains a formidable obstacle, but adoptive immunotherapy offers promise for the eradication of minimal residual disease and prevention of leukemia relapse following hematopoietic stem cell transplantation. CIK cell infusion started a novel generation of adoptive immunotherapy and exhibits particular potential applications in the area of hematological malignancy. In the present study, the previous strategies of leukemia immunotherapy using CIK cells are reviewed and the future directions of development are discussed.
Collapse
Affiliation(s)
- Xin-Yu Yang
- Depatment of Haematology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Hui Zeng
- Department of Hematology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, P.R. China
| | - Fang-Ping Chen
- Depatment of Haematology, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
33
|
Schmeel FC, Schmeel LC, Gast SM, Schmidt-Wolf IGH. Adoptive immunotherapy strategies with cytokine-induced killer (CIK) cells in the treatment of hematological malignancies. Int J Mol Sci 2014; 15:14632-48. [PMID: 25196601 PMCID: PMC4159872 DOI: 10.3390/ijms150814632] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/14/2014] [Accepted: 08/11/2014] [Indexed: 12/21/2022] Open
Abstract
Cytokine-induced killer (CIK) cells are a heterogeneous population of immune effector cells that feature a mixed T- and Natural killer (NK) cell-like phenotype in their terminally-differentiated CD3+CD56+ subset. The easy availability, high proliferation rate and widely major histocompatibility complex (MHC)-unrestricted antitumor activity of CIK cells contribute to their particularly advantageous profile, making them an attractive approach for adoptive immunotherapy. CIK cells have shown considerable cytotoxicity against both solid tumors and hematological malignancies in vitro and in animal studies. Recently, initial clinical experiences demonstrated the feasibility and efficacy of CIK cell immunotherapy in cancer patients, even at advanced disease stages. Likewise, the clinical application of CIK cells in combination with standard therapeutic procedures revealed synergistic antitumor effects. In this report, we will focus our consideration on CIK cells in the treatment of hematological malignancies. We will give insight into the latest advances and future perspectives and outline the most prominent results obtained in 17 clinical studies. Overall, CIK cells demonstrated a crucial impact on the treatment of patients with hematological malignancies, as evidenced by complete remissions, prolonged survival durations and improved quality of life. However, up to now, the optimal application schedule eventually favoring their integration into clinical practice has still to be developed.
Collapse
Affiliation(s)
- Frederic Carsten Schmeel
- Medizinische Klinik und Poliklinik III, Center for Integrated Oncology (CIO), University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Straße 25, 53105 Bonn, Germany.
| | - Leonard Christopher Schmeel
- Medizinische Klinik und Poliklinik III, Center for Integrated Oncology (CIO), University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Straße 25, 53105 Bonn, Germany.
| | - Sanna-Marie Gast
- Medizinische Klinik und Poliklinik III, Center for Integrated Oncology (CIO), University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Straße 25, 53105 Bonn, Germany.
| | - Ingo G H Schmidt-Wolf
- Medizinische Klinik und Poliklinik III, Center for Integrated Oncology (CIO), University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, Sigmund-Freud-Straße 25, 53105 Bonn, Germany.
| |
Collapse
|
34
|
Pan K, Guan XX, Li YQ, Zhao JJ, Li JJ, Qiu HJ, Weng DS, Wang QJ, Liu Q, Huang LX, He J, Chen SP, Ke ML, Zeng YX, Xia JC. Clinical activity of adjuvant cytokine-induced killer cell immunotherapy in patients with post-mastectomy triple-negative breast cancer. Clin Cancer Res 2014; 20:3003-11. [PMID: 24668644 DOI: 10.1158/1078-0432.ccr-14-0082] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
PURPOSE Triple-negative breast cancer (TNBC) is a high risk form of this disease, even after surgery, due to the absence of targets for hormone treatment and anti-Her-2 therapy. Chemotherapy is the main therapeutic strategy for such patients with breast cancer, although the outcome is often unsatisfactory. Thus, the development of combination adjuvant therapies is essential for improved prognosis in patients with TNBC. In this study, we investigated the efficacy of a sequential combination of cytokine-induced killer cell (CIK) infusion and chemotherapy for patients with post-mastectomy TNBC. EXPERIMENTAL DESIGN From 2008 to 2012, 90 patients with post-mastectomy TNBC were included in this retrospective study: 45 cases received chemotherapy alone or with sequential radiotherapy; a further 45 cases received chemotherapy with/without radiotherapy and sequential CIK infusion. RESULTS Survival analysis showed significantly higher disease-free survival (DFS) and overall survival (OS) rates in the CIK treatment group compared with the control group (P = 0.0382, P = 0.0046, respectively; log-rank test). Multivariate survival analysis showed that CIK adjuvant treatment was an independent prognostic factor for OS of patients with TNBC. In subgroup analyses, CIK adjuvant treatment significantly increased the DFS rate of patients with pathologic grade 3, and significantly increased the OS rate of patients in N1, N2, N3, IIB, III TNM (tumor-node-metastasis) stages, and with pathologic grade 3. CONCLUSIONS These data indicate that adjuvant CIK treatment combined with chemotherapy is an effective therapeutic strategy to prevent disease recurrence and prolong survival of patients with TNBC, particularly those with lymph node metastasis, advanced TNM stage, and poor pathologic grade. Clin Cancer Res; 20(11); 3003-11. ©2014 AACR.
Collapse
Affiliation(s)
- Ke Pan
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR ChinaAuthors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Xun-Xing Guan
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yong-Qiang Li
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jing-Jing Zhao
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jian-Jun Li
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Hui-Juan Qiu
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - De-Sheng Weng
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Qi-Jing Wang
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Qing Liu
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Li-Xi Huang
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jia He
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Shi-Ping Chen
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Miao-La Ke
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yi-Xin Zeng
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR ChinaAuthors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Jian-Chuan Xia
- Authors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR ChinaAuthors' Affiliations: State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine; Departments of Biotherapy; and Radiation Oncology, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| |
Collapse
|
35
|
Tao L, Huang G, Shi S, Chen L. Bevacizumab improves the antitumor efficacy of adoptive cytokine-induced killer cells therapy in non-small cell lung cancer models. Med Oncol 2013; 31:777. [DOI: 10.1007/s12032-013-0777-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 11/16/2013] [Indexed: 01/08/2023]
|
36
|
Abstract
Cytokine Induced Killer (CIK) cells are in vitro activated human CD8 T cells which have maintained several characteristics of T-EMRA cells and additionally acquired non specific anti tumoral cytotoxicity and CD56 overexpression, thus representing a cell population with double T and NK phenotype. Due to their in vivo intratumoral homing and lack of Graft versus Host (GVH) reactivity, CIK cells have been extensively used in cancer patients either in autologous or allogeneic contexts. Here we summarise CIK main biological features as well as their most prominent clinical results.
Collapse
|
37
|
Abstract
Drug-delivery carriers have the potential to not only treat but also diagnose many diseases; however, they still lack the complexity of natural-particulate systems. Cell-based therapies using tumor-targeting T cells and tumor-homing mesenchymal stem cells have given researchers a means to exploit the characteristics exhibited by innate-biological entities. Similarly, immune evasion by pathogens has inspired the development of natural polymers to cloak drug carriers. The 'marker-of-self' CD47 protein, which is found ubiquitously on mammalian cell surfaces, has been used for evading phagocyte clearance of drug carriers. This review will focus on the recent progress of drug carriers co-opting the tricks that cells in nature use to hide safely under the radar of the body's innate immune system.
Collapse
|
38
|
Linn YC. Adoptive immunotherapy with polyclonal T cells and natural killer cells for hematological malignancies: current status and future prospects. Int J Hematol Oncol 2013. [DOI: 10.2217/ijh.13.31] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
SUMMARY Adoptive cellular therapy with polyclonal T cells and natural killer cells are immunotherapeutic modalities being studied in solid tumors and hematological malignancies to treat disease and prevent relapse. These include unexpanded polyclonal T cells, short-term activation by cytokine into lymphokine-activated killer cells, longer term expansion by cytokine stimulation giving rise to cytokine-induced killer cells or expansion under costimulation with beads expressing anti-CD3 and anti-CD28. Similarly natural killer cells can be given with or without activation and expansion. Here we review the published work and clinical trials involving each cell type in the autologous, matched allogeneic, haploidentical and nontransplant settings, comparing and contrasting each cell type and discussing their potential applications.
Collapse
Affiliation(s)
- Yeh-Ching Linn
- Department of Hematology, Singapore General Hospital, Outram Road, Singapore 169608
| |
Collapse
|