1
|
Park JH, Lee SW, Choi D, Lee C, Sung YC. Harnessing the Power of IL-7 to Boost T Cell Immunity in Experimental and Clinical Immunotherapies. Immune Netw 2024; 24:e9. [PMID: 38455462 PMCID: PMC10917577 DOI: 10.4110/in.2024.24.e9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/31/2024] [Accepted: 02/01/2024] [Indexed: 03/09/2024] Open
Abstract
The cytokine IL-7 plays critical and nonredundant roles in T cell immunity so that the abundance and availability of IL-7 act as key regulatory mechanisms in T cell immunity. Importantly, IL-7 is not produced by T cells themselves but primarily by non-lymphoid lineage stromal cells and epithelial cells that are limited in their numbers. Thus, T cells depend on cell extrinsic IL-7, and the amount of in vivo IL-7 is considered a major factor in maximizing and maintaining the number of T cells in peripheral tissues. Moreover, IL-7 provides metabolic cues and promotes the survival of both naïve and memory T cells. Thus, IL-7 is also essential for the functional fitness of T cells. In this regard, there has been an extensive effort trying to increase the protein abundance of IL-7 in vivo, with the aim to augment T cell immunity and harness T cell functions in anti-tumor responses. Such approaches started under experimental animal models, but they recently culminated into clinical studies, with striking effects in re-establishing T cell immunity in immunocompromised patients, as well as boosting anti-tumor effects. Depending on the design, glycosylation, and the structure of recombinantly engineered IL-7 proteins and their mimetics, recombinant IL-7 molecules have shown dramatic differences in their stability, efficacy, cellular effects, and overall immune functions. The current review is aimed to summarize the past and present efforts in the field that led to clinical trials, and to highlight the therapeutical significance of IL-7 biology as a master regulator of T cell immunity.
Collapse
Affiliation(s)
- Jung-Hyun Park
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD 20892, USA
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Donghoon Choi
- Research Institute of NeoImmune Tech., Co, Ltd., Bio Open Innovation Center, Pohang 37666, Korea
| | - Changhyung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| | - Young Chul Sung
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37673, Korea
| |
Collapse
|
2
|
Zhao Y, Wei K, Chi H, Xia Z, Li X. IL-7: A promising adjuvant ensuring effective T cell responses and memory in combination with cancer vaccines? Front Immunol 2022; 13:1022808. [PMID: 36389666 PMCID: PMC9650235 DOI: 10.3389/fimmu.2022.1022808] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer vaccines exhibit specificity, effectiveness, and safety as an alternative immunotherapeutic strategy to struggle against malignant diseases, especially with the rapid development of mRNA cancer vaccines in recent years. However, how to maintain long-term immune memory after vaccination, especially T cells memory, to fulfill lasting surveillance against cancers, is still a challenging issue for researchers all over the world. IL-7 is critical for the development, maintenance, and proliferation of T lymphocytes, highlighting its potential role as an adjuvant in the development of cancer vaccines. Here, we summarized the IL-7/IL-7 receptor signaling in the development of T lymphocytes, the biological function of IL-7 in the maintenance and survival of T lymphocytes, the performance of IL-7 in pre-clinical and clinical trials of cancer vaccines, and the rationale to apply IL-7 as an adjuvant in cancer vaccine-based therapeutic strategy.
Collapse
Affiliation(s)
- Yue Zhao
- Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kongyuan Wei
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Hao Chi
- Clinical Medical Collage, Southwest Medical University, Luzhou, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
- *Correspondence: Zhijia Xia, ; Xiaosong Li,
| | - Xiaosong Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Zhijia Xia, ; Xiaosong Li,
| |
Collapse
|
3
|
Galarza-Muñoz G, Kennedy-Boone D, Schott G, Bradrick SS, Garcia-Blanco MA. Antisense modulation of IL7R splicing to control sIL7R expression in human CD4 + T cells. RNA (NEW YORK, N.Y.) 2022; 28:1058-1073. [PMID: 35613883 PMCID: PMC9297843 DOI: 10.1261/rna.079137.122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/06/2022] [Indexed: 06/15/2023]
Abstract
The interleukin 7 receptor (IL7R) is strongly associated with increased risk to develop multiple sclerosis (MS), an autoimmune disease of the central nervous system, and this association is likely driven by up-regulation of the soluble isoform of IL7R (sIL7R). Expression of sIL7R is determined by exclusion of the alternative exon 6 from IL7R transcripts, and our previous work revealed that the MS risk allele of the SNP rs6897932 within this exon enhances the expression of sIL7R by promoting exclusion of exon 6. sIL7R potentiates the activity of IL7, leading to enhanced expansion of T cells and increased disability in the experimental autoimmune encephalomyelitis (EAE) murine model of MS. This role in modulating T cell-driven immunity positions sIL7R as an attractive therapeutic target whose expression could be reduced for treatment of MS or increased for treatment of cancers. In this study, we identified novel antisense oligonucleotides (ASOs) that effectively control the inclusion (anti-sIL7R ASOs) or exclusion (pro-sIL7R ASOs) of this exon in a dose-dependent fashion. These ASOs provided excellent control of exon 6 splicing and sIL7R secretion in human primary CD4+ T cells. Supporting their potential for therapeutic targeting, we showed that lead anti-sIL7R ASOs correct the enhanced exon 6 exclusion imposed by the MS risk allele of rs6897932, whereas lead pro-sIL7R ASOs phenocopy it. The data presented here form the foundation for future preclinical studies that will test the therapeutic potential of these ASOs in MS and immuno-oncology.
Collapse
Affiliation(s)
- Gaddiel Galarza-Muñoz
- Autoimmunity BioSolutions, Galveston, Texas 77550, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Department of Neurology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Debbie Kennedy-Boone
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Geraldine Schott
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Shelton S Bradrick
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas 77550, USA
| | - Mariano A Garcia-Blanco
- Autoimmunity BioSolutions, Galveston, Texas 77550, USA
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas 77550, USA
- Institute of Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas 77550, USA
| |
Collapse
|
4
|
Leilei Z, Kewen Z, Biao H, Fang H, Yigang W. The Role of Chemokine IL-7 in Tumor and Its Potential Antitumor Immunity. J Interferon Cytokine Res 2022; 42:243-250. [PMID: 35613386 DOI: 10.1089/jir.2021.0236] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interleukin-7 (IL-7) is a cytokine belonging to the chemokine family. It plays a key role in the differentiation, development, and maturation of T lymphocytes and B lymphocytes, which is pivotal to adaptive immunity. In addition to its role in lymphocyte development, recent studies have indicated the antitumor functions of IL-7 in the tumor microenvironment. In this review, we discuss the role of IL-7 in tumors and summarize its antitumor potential and clinical application in lymphoma, leukemia, breast cancer, colon cancer, and so on. Furthermore, the combinational strategies of IL-7 and other antitumor drugs have been also discussed.
Collapse
Affiliation(s)
- Zhang Leilei
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
- Zhejiang Sci-Tech University Shaoxing Academy of Biomedicine, Shaoxing, China
| | - Zhou Kewen
- Department of Immunology, University of Toronto, Bachelor of Science, Toronto, Canada
| | - Huang Biao
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
| | - Huang Fang
- Department of Pathology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Wang Yigang
- Xinyuan Institute of Medicine and Biotechnology, College of Life Sciences and Medicine, Zhejiang Sci-Tech University, Hangzhou, China
- Zhejiang Sci-Tech University Shaoxing Academy of Biomedicine, Shaoxing, China
| |
Collapse
|
5
|
Campian JL, Ghosh S, Kapoor V, Yan R, Thotala S, Jash A, Hu T, Mahadevan A, Rifai K, Page L, Lee BH, Ferrando-Martinez S, Wolfarth AA, Yang SH, Hallahan D, Chheda MG, Thotala D. Long-acting recombinant human interleukin-7, NT-I7, increases cytotoxic CD8 + T cells and enhances survival in mouse glioma models. Clin Cancer Res 2022; 28:1229-1239. [PMID: 35031547 DOI: 10.1158/1078-0432.ccr-21-0947] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 08/27/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Patients with glioblastoma (GBM) are treated with radiation therapy (RT) and temozolomide (TMZ). These treatments may cause prolonged systemic lymphopenia, which itself is associated with poor outcomes. NT-I7 is a long-acting IL-7 that expands CD4 and CD8 T cell numbers in humans and mice. We tested whether NT-I7 prevents systemic lymphopenia and improves survival in mouse models of GBM. EXPERIMENTAL DESIGN C57BL/6 mice bearing intracranial tumors (GL261 or CT2A) were treated with RT (1.8 Gy/day x 5 days), TMZ (33 mg/kg/day x 5 days), and/or NT-I7 (10 mg/kg on the final day of RT). We followed the mice for survival while serially analyzing levels of circulating T lymphocytes. We assessed regulatory T cells (Treg) and cytotoxic T lymphocytes in the tumor microenvironment, cervical lymph nodes, spleen, and thymus; and hematopoietic stem and progenitor cells (HSPCs) in the bone marrow. RESULTS GBM tumor-bearing mice treated with RT+NT-I7 increased T lymphocytes in the lymph nodes, thymus, and spleen, enhanced IFNγ production, and decreased Treg cells in the tumor which was associated with a significant increase in survival. NT-I7 also enhanced central memory and effector memory CD8 T cells in lymphoid organs and tumor. Depleting CD8 T cells abrogated the effects of NT-I7. Furthermore, NT-I7 treatment decreased progenitor cells in the bone marrow. CONCLUSION In orthotopic glioma-bearing mice, NT-I7 mitigates radiation-related lymphopenia, increases cytotoxic CD8 T lymphocytes systemically and in the tumor, and improves survival. A phase I/II trial to evaluate NT-I7 in patients with high-grade gliomas is ongoing (NCT03687957).
Collapse
Affiliation(s)
| | - Subhajit Ghosh
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Vaishali Kapoor
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Ran Yan
- Washington University in St. Louis
| | | | | | - Tong Hu
- Medicine, Washington University in St. Louis
| | - Anita Mahadevan
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Kasem Rifai
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Logan Page
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | - Byung Ha Lee
- Translational Research Division, NeoImmuneTech, Inc
| | | | | | | | - Dennis Hallahan
- Radiation Oncology, Washington University in St. Louis School of Medicine
| | | | - Dinesh Thotala
- Radiation Oncology, Washington University in St. Louis School of Medicine
| |
Collapse
|
6
|
Abstract
Clinical trials have demonstrated that an increased number of effector cells, especially tumor-specific T cells, is positively linked with patients’ prognosis. Although the discovery of checkpoint inhibitors (CPIs) has led to encouraging progress in cancer immunotherapy, the lack of either T cells or targets for CPIs is a limitation for patients with poor prognosis. Since interleukin (IL)-2 and IL-7 are cytokines that target many aspects of T-cell responses, they have been used to treat cancers. In this review, we focus on the basic biology of how these cytokines regulate T-cell response and on the clinical trials using the cytokines against cancer. Further, we introduce several recent studies that aim to improve cytokines’ biological activities and find the strategy for combination with other therapeutics.
Collapse
Affiliation(s)
- Ji-Hae Kim
- Department of Life Sciences, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| | - Kun-Joo Lee
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
- Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology(POSTECH), Pohang 37673, Korea
| |
Collapse
|
7
|
Wei X, Zhang Y, Yang Z, Sha Y, Pan Y, Chen Y, Cai L. Analysis of the role of the interleukins in colon cancer. Biol Res 2020; 53:20. [PMID: 32381120 PMCID: PMC7203842 DOI: 10.1186/s40659-020-00287-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 04/28/2020] [Indexed: 01/21/2023] Open
Abstract
Background The role of interleukin family in colon cancer remained controversial. The purpose of this study was to investigate the association between interleukin family and colon cancer progression through bioinformatics methods and to validate such association in clinical patients. Methods A total of 15 differentially expressed interleukins between the colon cancer tissue and normal colon tissue were evaluated from the Cancer Genome Atlas (TCGA) database with R software and only interleukin-7 (IL-7) was significantly associated with survival. The signaling pathway associated with IL-7 was then investigated using gene enrichment analysis. In addition, subsets of TNM were analyzed in detail and univariate and multivariate COX regression analysis were conducted. Finally, we performed western blotting, immunohistochemistry, cell proliferation and cell apoptosis analysis to examine the expression of IL-7 in patients with intestinal cancer. Results The study demonstrated that IL-7 could inhibit the progression of colon cancer. In addition, IL-7 was found to be associated with overall survival (OS) and pathological stage. Further analysis of IL-7 expression with clinical data indicated that IL-7 was a key factor in inhibiting colon cancer progression. Conclusion IL-7 was a key factor in inhibiting the progression of colon cancer and was closely related to overall survival.
Collapse
Affiliation(s)
- Xiyi Wei
- Department of General Surgery, Shanghai Pudong Hospita, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, 201399, Shanghai, China.,First Clinical Medical College of Nanjing Medical University, 210029, Nanjing, China
| | - Yuan Zhang
- Department of General Surgery, Shanghai Pudong Hospita, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, 201399, Shanghai, China
| | - Zhou Yang
- Department of General Surgery, Shanghai Pudong Hospita, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, 201399, Shanghai, China
| | - Yeqin Sha
- First Clinical Medical College of Nanjing Medical University, 210029, Nanjing, China
| | - Yitong Pan
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, 211116, Nanjing, China
| | - Yusheng Chen
- Department of General Surgery, Shanghai Pudong Hospita, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, 201399, Shanghai, China.
| | - Lei Cai
- Department of General Surgery, Shanghai Pudong Hospita, Fudan University Pudong Medical Center, 2800 Gongwei Road, Huinan Town, Pudong, 201399, Shanghai, China.
| |
Collapse
|
8
|
Shi L, Xu Z, Yang Q, Huang Y, Gong Y, Wang F, Ke B. IL-7-Mediated IL-7R-JAK3/STAT5 signalling pathway contributes to chemotherapeutic sensitivity in non-small-cell lung cancer. Cell Prolif 2019; 52:e12699. [PMID: 31599032 PMCID: PMC6869130 DOI: 10.1111/cpr.12699] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Revised: 06/26/2019] [Accepted: 09/06/2019] [Indexed: 12/26/2022] Open
Abstract
Objectives The chemotherapy drug resistance is a major challenge for non‐small‐cell lung cancer (NSCLC) treatment. Combination of immunotherapy and chemotherapy has shown promise for cancer. The goal of this study was to evaluate the anti‐tumour efficacy of interleukin‐7 (IL‐7) combining cisplatin against NSCLC. Materials and Methods Cell proliferation was analysed using CCK‐8 assay, EdU proliferation assay and colony‐forming assay. Cell apoptosis was evaluated using HOECHST 33342 assay and flow cytometry. The protein expression levels were analysed by Western blot. The blocking antibody against the IL‐7 receptor and the inhibitors of STAT5 and JAK3 were used to investigate the pathway involved. A xenograft model was established to assess the anti‐tumour efficacy of IL‐7 combining cisplatin in vivo. Results Here we found IL‐7R was increased in A549/DDP cells compared with A549 cells. The block of IL‐7R reversed the inhibitory effects of IL‐7 combined with cisplatin and decreased the numbers of apoptosis cells induced by treatment of IL‐7 combined with cisplatin. The JAK3 inhibitor and STAT5 inhibitor were used to identify the pathway involved. The results showed that JAK3/STAT5 pathway was involved in enhancing role of cisplatin sensitivity of NSCLC cells by IL‐7. In vivo, cisplatin significantly inhibited tumour growth and IL‐7 combined with cisplatin achieved the best therapeutic effect. Conclusion Together, IL‐7 promoted the sensitivity of NSCLC cells to cisplatin via IL‐7R‐JAK3/STAT5 signalling pathway.
Collapse
Affiliation(s)
- Lin Shi
- Department of Traditional Chinese Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Zhaozhong Xu
- Department of Emergency, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Qiong Yang
- Department of Oncology, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuanyuan Huang
- Department of VIP Ward, Affiliated Cancer Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yuxin Gong
- Department of Respiratory Diseases, Zhujiang Hospital of Southern Medical University, Guangzhou, China
| | - Fang Wang
- Department of Oncology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Ke
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
9
|
Song Y, Liu Y, Hu R, Su M, Rood D, Lai L. In Vivo Antitumor Activity of a Recombinant IL7/IL15 Hybrid Cytokine in Mice. Mol Cancer Ther 2016; 15:2413-2421. [PMID: 27474151 DOI: 10.1158/1535-7163.mct-16-0111] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/15/2016] [Indexed: 11/16/2022]
Abstract
Both IL7 and IL15 have become important candidate immunomodulators for cancer treatment. However, IL7 or IL15 used alone suffers from shortcomings, such as short serum half-life and limited antitumor effect. We have cloned and expressed a recombinant (r) IL7/IL15 fusion protein in which IL7 and IL15 are linked by a flexible linker. We then compared the antitumor effect of rIL7/IL15 with the individual factors rIL7 and/or rIL15. We show here that rIL7/IL15 has a higher antitumor activity than the combination of the individual factors in both murine B16F10 melanoma and CT-26 colon cancer models. This was associated with a significant increase in tumor infiltration of T cells, DCs, and NK cells and a decrease in regulatory T cells (Tregs). In addition, rIL7/IL15-treated DCs had higher expression of costimulatory molecules CD80 and CD86. The higher antitumor activity of rIL7/IL15 is likely due to its longer in vivo half-life and different effects on immune cells. Our results suggest that rIL7/IL15 may offer a new tool to enhance antitumor immunity and treat cancer. Mol Cancer Ther; 15(10); 2413-21. ©2016 AACR.
Collapse
Affiliation(s)
- Yinhong Song
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut. Medical College, Three Gorges University, Yichang, China
| | - Yalan Liu
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Rong Hu
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Debra Rood
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, Connecticut. University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, Connecticut.
| |
Collapse
|
10
|
Trédan O, Ménétrier-Caux C, Ray-Coquard I, Garin G, Cropet C, Verronèse E, Bachelot T, Rebattu P, Heudel P, Cassier P, Chabaud S, Croughs T, Dupont P, Cadore A, Clapisson G, Delgado A, Bardin-dit-Courageot C, Rigal C, N'Kodia A, Gilles-Afchain L, Morre M, Pérol D, Blay J, Caux C. ELYPSE-7: a randomized placebo-controlled phase IIa trial with CYT107 exploring the restoration of CD4+ lymphocyte count in lymphopenic metastatic breast cancer patients. Ann Oncol 2015; 26:1353-62. [DOI: 10.1093/annonc/mdv173] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Accepted: 03/24/2015] [Indexed: 12/29/2022] Open
|
11
|
Datta J, Berk E, Cintolo JA, Xu S, Roses RE, Czerniecki BJ. Rationale for a Multimodality Strategy to Enhance the Efficacy of Dendritic Cell-Based Cancer Immunotherapy. Front Immunol 2015; 6:271. [PMID: 26082780 PMCID: PMC4451636 DOI: 10.3389/fimmu.2015.00271] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 05/15/2015] [Indexed: 02/03/2023] Open
Abstract
Dendritic cells (DC), master antigen-presenting cells that orchestrate interactions between the adaptive and innate immune arms, are increasingly utilized in cancer immunotherapy. Despite remarkable progress in our understanding of DC immunobiology, as well as several encouraging clinical applications – such as DC-based sipuleucel-T for metastatic castration-resistant prostate cancer – clinically effective DC-based immunotherapy as monotherapy for a majority of tumors remains a distant goal. The complex interplay between diverse molecular and immune processes that govern resistance to DC-based vaccination compels a multimodality approach, encompassing a growing arsenal of antitumor agents which target these distinct processes and synergistically enhance DC function. These include antibody-based targeted molecular therapies, immune checkpoint inhibitors, therapies that inhibit immunosuppressive cellular elements, conventional cytotoxic modalities, and immune potentiating adjuvants. It is likely that in the emerging era of “precision” cancer therapeutics, tangible clinical benefits will only be realized with a multifaceted – and personalized – approach combining DC-based vaccination with adjunctive strategies.
Collapse
Affiliation(s)
- Jashodeep Datta
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Erik Berk
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Jessica A Cintolo
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Shuwen Xu
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Robert E Roses
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA
| | - Brian J Czerniecki
- Division of Endocrine and Oncologic Surgery, Department of Surgery, University of Pennsylvania Perelman School of Medicine , Philadelphia, PA , USA ; Rena Rowen Breast Center, Hospital of the University of Pennsylvania , Philadelphia, PA , USA
| |
Collapse
|
12
|
Nayar S, Dasgupta P, Galustian C. Extending the lifespan and efficacies of immune cells used in adoptive transfer for cancer immunotherapies-A review. Oncoimmunology 2015; 4:e1002720. [PMID: 26155387 DOI: 10.1080/2162402x.2014.1002720] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/19/2014] [Accepted: 12/20/2014] [Indexed: 12/19/2022] Open
Abstract
Cells used in adoptive cell-transfer immunotherapies against cancer include dendritic cells (DCs), natural-killer cells, and CD8+ T-cells. These cells may have limited efficacy due to their lifespan, activity, and immunosuppressive effects of tumor cells. Therefore, increasing longevity and activity of these cells may boost their efficacy. Four cytokines that can extend immune effector-cell longevity are IL-2, IL-7, IL-21, and IL-15. This review will discuss current knowledge on effector-cell lifespans and the mechanisms by which IL-2, IL-7, IL-15, and IL-21 can extend effector-cell longevity. We will also discuss how lifespan and efficacy of these cells can be regulated to allow optimal clinical benefits.
Collapse
Affiliation(s)
- Sandeep Nayar
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| | - Prokar Dasgupta
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| | - Christine Galustian
- MRC Centre for Transplantation; Kings College London; Guys Hospital ; London, UK
| |
Collapse
|
13
|
Yuan CH, Yang XQ, Zhu CL, Liu SP, Wang BC, Wang FB. Interleukin-7 enhances the in vivo anti-tumor activity of tumor-reactive CD8+ T cells with induction of IFN-gamma in a murine breast cancer model. Asian Pac J Cancer Prev 2014; 15:265-71. [PMID: 24528037 DOI: 10.7314/apjcp.2014.15.1.265] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Interleukin-7 (IL-7) is a potent anti-apoptotic cytokine that enhances immune effector cell functions and is essential for lymphocyte survival. While it known to induce differentiation and proliferation in some haematological malignancies, including certain types of leukaemias and lymphomas, little is known about its role in solid tumours, including breast cancer. In the current study, we investigated whether IL-7 could enhance the in vivo antitumor activity of tumor-reactive CD8+ T cells with induction of IFN-γ in a murine breast cancer model. Human IL-7 cDNA was constructed into the eukaryotic expression plasmid pcDNA3.1, and then the recombinational pcDNA3.1-IL-7 was intratumorally injected in the TM40D BALB/C mouse graft model. Serum and intracellular IFN-γ levels were measured by ELISA and flow cytometry, respectively. CD8+ T cell-mediated cytotoxicity was analyzed using the MTT method. Our results showed that IL-7 administration significantly inhibited tumor growth from day 15 after direct intratumoral injection of pcDNA3.1-IL-7. The anti-tumor effect correlated with a marked increase in the level of IFN-γ and breast cancer cells-specific CTL cytotoxicity. In vitro cytotoxicity assays showed that IL-7-treatment could augment cytolytic activity of CD8+ T cells from tumor bearing mice, while anti-IFN-γ blocked the function of CD8+ T cells, suggesting that IFN-γ mediated the cytolytic activity of CD8+ T cells. Furthermore, in vivo neutralization of CD8+ T lymphocytes by CD8 antibodies reversed the antitumor benefit of IL-7. Thus, we demonstrated that IL-7 exerts anti-tumor activity mainly through activating CD8+ T cells and stimulating them to secrete IFN-γ in a murine breast tumor model. Based on these results, our study points to a potential novel way to treat breast cancer and may have important implications for clinical immunotherapy.
Collapse
Affiliation(s)
- Chun-Hui Yuan
- Department of Laboratory Medicine and Center for Gene Diagnosis, Zhongnan Hospital of Wuhan University, Wuhan, China E-mail :
| | | | | | | | | | | |
Collapse
|
14
|
Zarogoulidis P, Lampaki S, Yarmus L, Kioumis I, Pitsiou G, Katsikogiannis N, Hohenforst-Schmidt W, Li Q, Huang H, Sakkas A, Organtzis J, Sakkas L, Mpoukovinas I, Tsakiridis K, Lazaridis G, Syrigos K, Zarogoulidis K. Interleukin-7 and interleukin-15 for cancer. J Cancer 2014; 5:765-73. [PMID: 25368677 PMCID: PMC4216801 DOI: 10.7150/jca.10471] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 10/06/2014] [Indexed: 11/30/2022] Open
Abstract
Interleukin 7 and 15 are considered powerful pro-inflammatory cytokines, they have the ability to destabilize chromosomes and induce tumorigenesis. Additionally, they can control malignancy proliferation by influencing the tumor microenvironment and immune system. Immunotherapy has been proposed as a treatment modality for malignancy for over a decade; the exact mechanisms of action and pathways are still under investigation. Interleukin 7 and 15 have been extensively investigated in hematological malignancies since their mode of action influences the stimulation of the immune system in a more direct way than other malignancies such as lung, melanoma, and breast, renal and colorectal cancer.
Collapse
Affiliation(s)
- Paul Zarogoulidis
- 1. Pulmonary Department-Oncology Unit, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sofia Lampaki
- 1. Pulmonary Department-Oncology Unit, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Lonny Yarmus
- 2. Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, U.S.A
| | - Ioannis Kioumis
- 1. Pulmonary Department-Oncology Unit, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Georgia Pitsiou
- 1. Pulmonary Department-Oncology Unit, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Nikolaos Katsikogiannis
- 3. Surgery Department (NHS), University General Hospital of Alexandroupolis, Alexandroupolis, Greece
| | | | - Qiang Li
- 5. Department of Respiratory Diseases, Changhai Hospital/First Affiliated Hospital of the Second Military Medical University, Shanghai, China
| | - Haidong Huang
- 5. Department of Respiratory Diseases, Changhai Hospital/First Affiliated Hospital of the Second Military Medical University, Shanghai, China
| | - Antonios Sakkas
- 6. Pathology Department, ``G. Papanikolaou`` General Hospital, Thessaloniki, Greece
| | - John Organtzis
- 1. Pulmonary Department-Oncology Unit, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Leonidas Sakkas
- 6. Pathology Department, ``G. Papanikolaou`` General Hospital, Thessaloniki, Greece
| | - Ioannis Mpoukovinas
- 7. Oncology Department, ``BioMedicine`` Private Hospital, Thessaloniki, Greece
| | - Kosmas Tsakiridis
- 8. Cardiothoracic Surgery Department, ``Saint Luke`` Private Hospital, Thessaloniki, Greece
| | - George Lazaridis
- 9. Oncology Department, ``G. Papageorgiou`` University Hospital, Thessaloniki, Greece
| | - Konstantinos Syrigos
- 10. Oncology Department, ``Sotiria`` Hospital, University of Athens, Athens, Greece
| | - Konstantinos Zarogoulidis
- 1. Pulmonary Department-Oncology Unit, ``G. Papanikolaou`` General Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
15
|
Gou HF, Huang J, Shi HS, Chen XC, Wang YS. Chemo-immunotherapy with oxaliplatin and interleukin-7 inhibits colon cancer metastasis in mice. PLoS One 2014; 9:e85789. [PMID: 24465710 PMCID: PMC3897491 DOI: 10.1371/journal.pone.0085789] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 12/05/2013] [Indexed: 02/05/2023] Open
Abstract
Combination of immunotherapy and chemotherapy has shown promise for cancer. Interleukin-7 (IL-7) can potentially enhance immune responses against tumor, while oxaliplatin (OXP), a platinum-based drug, can promote a favorable immune microenvironment and stimulate anticancer immune responses. We evaluated the anti-tumor activity of IL-7 combining OXP against a murine colon carcinoma in vitro and in vivo and studied the tumor immune microenvironment to investigate whether the combined treatment affects on the local immune cell populations. Utilizing lung and abdomen metastasis models by inoculation of CT26 mice colon cancer cells, we evaluated the anti-tumor efficacy of combining IL-7 and OXP in mice models. Tumor immune microenvironment was evaluated by flow cytometric analysis and immunohistochemical staining. Our study showed that the in vivo administration of IL-7 combined with OXP markedly inhibited the growth of tumors in lung and abdomen metastasis models of colon cancer. IL-7 alone had no effect on tumor growth in mice and IL-7 did not alter cell sensitivity to OXP in culture. The antitumor effect of combining IL-7 and OXP correlated with a marked increase in the number of tumor-infiltrating activated CD8+ T cells and a marked decrease in the number of regulatory T (Treg) cells in spleen. Our data suggest that OXP plus IL-7 treatment inhibits tumor cell growth by immunoregulation rather than direct cytotoxicity. Our findings justify further evaluation of combining IL-7 and chemotherapy as a novel experimental cancer therapy.
Collapse
Affiliation(s)
- Hong-Feng Gou
- Department of Abdominal Cancer, Cancer Center, the State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Juan Huang
- Department of Abdominal Cancer, Cancer Center, the State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hua-Shan Shi
- Department of Head and Neck Cancer, Cancer Center, the State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Xin-chuan Chen
- Department of Hematology, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Yong-Sheng Wang
- Department of Thoracic Cancer, Cancer Center, the State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
- * E-mail:
| |
Collapse
|
16
|
Morre M, Beq S. Interleukin-7 and immune reconstitution in cancer patients: a new paradigm for dramatically increasing overall survival. Target Oncol 2012; 7:55-68. [PMID: 22383042 PMCID: PMC3304058 DOI: 10.1007/s11523-012-0210-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 01/16/2012] [Indexed: 12/27/2022]
Abstract
Although great effort is being expended in the development of cancer immunotherapies, it is surprising that global lymphopenia and its various dimensions are not being systematically assessed in cancer patients. The incident pathologies associated with various immunosuppressed conditions such as those found in HIV infection have taught us that measuring various T cell populations including CD4 provides the clinician with a reliable measure for gauging the risk of cancer and opportunistic infections. Importantly, recent data emphasize the key link between lymphocyte T cell counts and overall survival in cancer patients receiving chemotherapy. Treatment of immunocompromised patients with interleukin-7 (IL-7), a critical growth and homeostatic factor for T cells, has been shown to produce a compelling profile of T cell reconstitution. The clinical results of this investigational therapy confirm data obtained from numerous preclinical studies and demonstrate the long-term stability of this immune reconstitution, not only on CD4 but also on CD8 T cells, involving recent thymic emigrants as well as naive, memory, and central memory T cells. Furthermore, IL-7 therapy also contributes to restoration of a broadened diversity of the T cell repertoire as well as to migration of these cells to lymph nodes and tissues. All these properties support the initiation of new clinical studies aimed at reconstituting the immune system of cancer patients before or immediately after chemotherapy in order to demonstrate a potentially profound increase in overall survival.
Collapse
Affiliation(s)
- Michel Morre
- Cytheris S.A., 175 rue Jean Jacques Rousseau, 92130, Issy-Les-Moulineaux, France.
| | | |
Collapse
|
17
|
Mouse embryonic stem cell-derived thymic epithelial cell progenitors enhance T-cell reconstitution after allogeneic bone marrow transplantation. Blood 2011; 118:3410-8. [DOI: 10.1182/blood-2011-03-340794] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Abstract
We have reported that mouse embryonic stem cells (mESCs) can be selectively induced in vitro to differentiate into thymic epithelial cell progenitors (TEPs). When placed in vivo, these mESC-derived TEPs differentiate into cortical and medullary thymic epithelial cells, reconstitute the normal thymic architecture, and enhance thymocyte regeneration after syngeneic BM transplantation (BMT). Here, we show that transplantation of mESC-derived TEPs results in the efficient establishment of thymocyte chimerism and subsequent generation of naive T cells in both young and old recipients of allo-geneic BM transplant. GVHD was not induced, whereas graft-versus-tumor activity was significantly enhanced. Importantly, the reconstituted immune system was tolerant to host, mESC, and BM transplant donor antigens. Therefore, ESC-derived TEPs may offer a new approach for the rapid and durable correction of T-cell immune deficiency after BMT, and the induction of tolerance to ESC-derived tissue and organ transplants. In addition, ESC-derived TEPs may also have use as a means to reverse age-dependent thymic involution, thereby enhancing immune function and decreasing infection rates in the elderly.
Collapse
|
18
|
Silva A, Laranjeira AB, Martins LR, Cardoso BA, Demengeot J, Yunes JA, Seddon B, Barata JT. IL-7 Contributes to the Progression of Human T-cell Acute Lymphoblastic Leukemias. Cancer Res 2011; 71:4780-9. [DOI: 10.1158/0008-5472.can-10-3606] [Citation(s) in RCA: 104] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
19
|
Lai L, Jin J, Goldschneider I. In vivo antitumor activity of a recombinant IL-7/HGFbeta hybrid cytokine in mice. Cancer Res 2010; 71:61-7. [PMID: 21084268 DOI: 10.1158/0008-5472.can-10-3198] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The immune cytokine interleukin (IL)-7 and the β-chain of hepatocyte growth factor (HGF) aggregate to form a naturally occurring heterodimer that stimulates the growth of common lymphoid progenitors and immature B and T lymphoid cells. We have cloned and expressed the heterodimer as a single-chain hybrid cytokine [recombinant (r) IL-7/HGFβ], which stimulates short-term hematopoietic stem cells as well as lymphoid precursors. Inasmuch as IL-7 and HGF are known to have antitumor and protumor activities, respectively, we determined here whether either of these activities is exhibited by rIL-7/HGFβ. We show that the in vivo administration of rIL-7/HGFβ markedly inhibits the growth of newly initiated and established tumors and the formation of pulmonary metastases in murine models of colon cancer and melanoma. The antitumor effect of rIL-7/HGFβ correlated with a marked increase in the number of tumor-infiltrating CD4(+) and CD8(+) T cells and activated dendritic cells. A major role for these immune cells in tumor suppression was indicated by the inability of rIL-7/HGFβ to inhibit the growth of tumor cells in vitro and in congenitally athymic mice. Analysis of interferon-γ-secreting T cells showed that the immune response was tumor specific. Our findings justify further evaluation of rIL-7/HGFβ as a novel experimental cancer therapy.
Collapse
Affiliation(s)
- Laijun Lai
- Department of Immunology, School of Medicine, University of Connecticut Health Center, Farmington, Connecticut 06030, USA.
| | | | | |
Collapse
|
20
|
Sportès C, Gress RE, Mackall CL. Perspective on potential clinical applications of recombinant human interleukin-7. Ann N Y Acad Sci 2010; 1182:28-38. [PMID: 20074272 DOI: 10.1111/j.1749-6632.2009.05075.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Interleukin-7 has critical and nonredundant roles in T cell development, hematopoiesis, and postdevelopmental immune functions as a prototypic homeostatic cytokine. Based on a large body of preclinical evidence, it may have multiple therapeutic applications in immunodeficiency states, either physiologic (immuno-senescence), pathologic (HIV) or iatrogenic (postchemotherapy and posthematopoietic stem cell transplant) and may have roles in immune reconstitution or enhancement of immunotherapy. Early clinical development trials in humans show that, within a short time, rhIL-7 administration results in a marked preferential expansion of both naive and memory CD4 and CD8 T cell pools with a tendency toward enhanced CD8 expansion. As a result, lymphopenic or normal older hosts develop an expanded circulating T cell pool with a profile that resembles that seen earlier in life with increased T cell repertoire diversity. These results, along with a favorable toxicity profile, open a wide perspective of potential future clinical applications.
Collapse
Affiliation(s)
- Claude Sportès
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, DHHS, Bethesda, Maryland 20892-1104, USA
| | | | | |
Collapse
|
21
|
Sportès C, Babb RR, Krumlauf MC, Hakim FT, Steinberg SM, Chow CK, Brown MR, Fleisher TA, Noel P, Maric I, Stetler-Stevenson M, Engel J, Buffet R, Morre M, Amato RJ, Pecora A, Mackall CL, Gress RE. Phase I study of recombinant human interleukin-7 administration in subjects with refractory malignancy. Clin Cancer Res 2010; 16:727-35. [PMID: 20068111 DOI: 10.1158/1078-0432.ccr-09-1303] [Citation(s) in RCA: 162] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Interleukin-7 (IL-7) has critical and nonredundant roles in T-cell development, hematopoiesis, and postdevelopmental immune functions as a prototypic homeostatic cytokine. Based on a large body of preclinical evidence, it may have multiple therapeutic applications in immunodeficiency states, either physiologic (immunosenescence), pathologic (HIV), or iatrogenic (postchemotherapy and posthematopoietic stem cell transplant), and may have roles in immune reconstitution or enhancement of immunotherapy. We report here on the toxicity and biological activity of recombinant human IL-7 (rhIL-7) in humans. DESIGN Subjects with incurable malignancy received rhIL-7 subcutaneously every other day for 2 weeks in a phase I interpatient dose escalation study (3, 10, 30, and 60 microg/kg/dose). The objectives were safety and dose-limiting toxicity determination, identification of a range of biologically active doses, and characterization of biological and, possibly, antitumor effects. RESULTS Mild to moderate constitutional symptoms, reversible spleen and lymph node enlargement, and marked increase in peripheral CD3(+), CD4(+), and CD8(+) lymphocytes were seen in a dose-dependent and age-independent manner in all subjects receiving >or=10 microg/kg/dose, resulting in a rejuvenated circulating T-cell profile, resembling that seen earlier in life. In some subjects, rhIL-7 induced in the bone marrow a marked, transient polyclonal proliferation of pre-B cells showing a spectrum of maturation as well as an increase in circulating transitional B cells. CONCLUSION This study shows the potent biological activity of rhIL-7 in humans over a well-tolerated dose range and allows further exploration of its possible therapeutic applications.
Collapse
Affiliation(s)
- Claude Sportès
- Experimental Transplantation and Immunology Branch, National Cancer Institute, NIH, Bethesda, Maryland 20892-1203, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Intratumoral IL-7 delivery by mesenchymal stromal cells potentiates IFNgamma-transduced tumor cell immunotherapy of experimental glioma. J Neuroimmunol 2009; 218:140-4. [PMID: 19914721 DOI: 10.1016/j.jneuroim.2009.10.017] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Revised: 10/23/2009] [Accepted: 10/23/2009] [Indexed: 12/24/2022]
Abstract
The present study reports regression of pre-established experimental rat gliomas as a result of combining peripheral immunization using interferon gamma (IFNgamma) transduced autologous tumor cells with local intratumoral delivery of interleukin 7 (IL-7) by mesenchymal stromal cells. IL-7 alone significantly decreased the tumor area and this effect was enhanced with IFNgamma immunization. A higher density of intratumoral T-cells was observed in animals receiving combined therapies compared to rats receiving either cytokine alone suggesting that the therapeutic effect is dependent on a T-cell response.
Collapse
|
23
|
Nguyen XD, Müller-Berghaus J, Kälsch T, Schadendorf D, Borggrefe M, Klüter H. Differentiation of monocyte-derived dendritic cells under the influence of platelets. Cytotherapy 2009; 10:720-9. [PMID: 18985478 DOI: 10.1080/14653240802378912] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Monocytapheresis has been established to collect a sufficient number of monocytes (MO) for differentiation to dendritic cells (DC) as a cancer vaccine. Platelets (Plt) are invariably found as a contaminant in the final monocytapheresis product. The aim of this study was to investigate DC differentiation under the influence of Plt with regard to their function and phenotype. METHODS MO were isolated and co-cultured with autologous Plt at different MO:Plt ratios (1:1.7, 1:5, 1:15, 1:45 and 1:135) in the presence of interleukin-4 (IL-4) and granulocyte-macrophage colony-stimulating factor (GM-CSF). IL-12p70 release after ligation of CD40L was determined in the supernatant by enzyme-linked immunosorbent assay (ELISA). For T-cell stimulation, tetanus toxoid was added to immature DC and maturation was induced by adding cytokines (IL-1beta, IL-6, tumor necrosis factor-alpha and prostaglandin E(2)). Stimulated T cells were analyzed for activation and proliferation as well as for intracellular cytokines by flow cytometry. RESULTS All DC cultures were strongly positive for CD83. At a contaminating concentration of 5 Plt/MO, matured DC showed the highest expression of HLA-DR, CD80 and CD86, inducing a strong T-cell proliferation with high production of IL-4 and interferon-gamma. The highest level of IL-12p70 production was observed by the same DC group. DISCUSSION Plt did not negatively influence DC maturation but enhanced the expression of co-stimulatory molecules and the release of IL-12. Functionally this was reflected by a strong T-cell response that involved T-helper 1 (Th1)- as well as Th2-biased T cells. Our findings show that controlling the Plt concentration may provide important advantages for the generation of DC for use in immunotherapy.
Collapse
Affiliation(s)
- X D Nguyen
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, Red-Cross Blood Donation Service of Baden-Wurttemberg-Hessen, Germany.
| | | | | | | | | | | |
Collapse
|
24
|
Li B, Simmons A, Du T, Lin C, Moskalenko M, Gonzalez-Edick M, VanRoey M, Jooss K. Allogeneic GM-CSF-secreting tumor cell immunotherapies generate potent anti-tumor responses comparable to autologous tumor cell immunotherapies. Clin Immunol 2009; 133:184-97. [PMID: 19664962 DOI: 10.1016/j.clim.2009.07.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2009] [Revised: 06/26/2009] [Accepted: 07/09/2009] [Indexed: 11/28/2022]
Abstract
Clinical studies of cell-based immunotherapies have included both patient-specific (autologous) and non-patient-specific (allogeneic) approaches. Major concerns in using allogeneic immunotherapies are that the induced immune responses may be predominantly directed against the allogeneic HLA molecules of the cellular immunotherapy and not against its potential tumor antigens and that only the allogeneic responses will be enhanced when the immunotherapies are combined with immune checkpoint regulators in an effort to enhance overall immunotherapy potency. To evaluate these possibilities, studies were performed using the GM-CSF-secreting B16F1 cell line as autologous immunotherapy (Auto) and the same cell line modified to over-express the MHC molecule K(d) to generate an immunotherapy that expresses an allogeneic component (Allo) when injected into C57/Bl6 mice. The goal was to compare the specific anti-tumor immune responses induced by these two immunotherapies, which share an identical antigen repertoire, with the exception of the allogeneic MHC class I molecule expressed by the Allo cells, and have identical GM-CSF-secretion levels. Both immunotherapies provided similar therapeutic benefit to tumor-bearing animals with a trend towards a more pronounced tumor growth delay in animals injected with the Allo immunotherapy. This correlated with a significant increase in the number of activated DCs and T-cells in the DLN of Allo-treated animals. In addition, persistent infiltration of effector CD8(+) T-cells was detected in the tumors of animals treated with the Allo immunotherapy, which correlated with a trend towards a greater antigen-specific T-cell response in these animals. When combined with the immune checkpoint regulator anti-PD-1, tumor-specific and allogeneic immune responses were equally enhanced. Thus, the ability of an allogeneic tumor cell immunotherapy to induce a therapeutic anti-tumor immune response is comparable, if not superior, to an autologous tumor cell immunotherapy and its anti-tumor potency can be enhanced when combined with immunomodulatory compounds.
Collapse
Affiliation(s)
- Betty Li
- Cell Genesys Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Interleukin-7 (IL-7) is required for the development and survival of T cells and plays a critical role in modulating T-cell homeostasis. This review will address current understanding of IL-7 biology, review recent clinical experiences and discuss potential future clinical applications of IL-7, or IL-7 blockade, in the setting of disease.
Collapse
Affiliation(s)
- C M Capitini
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | |
Collapse
|
26
|
Schroten-Loef C, de Ridder CMA, Reneman S, Crezee M, Dalgleish A, Todryk SM, Bangma CH, Kraaij R. A prostate cancer vaccine comprising whole cells secreting IL-7, effective against subcutaneous challenge, requires local GM-CSF for intra-prostatic efficacy. Cancer Immunol Immunother 2009; 58:373-81. [PMID: 18641982 PMCID: PMC11031066 DOI: 10.1007/s00262-008-0560-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2008] [Accepted: 07/01/2008] [Indexed: 10/21/2022]
Abstract
A panel of cytokine-secreting RM-9 prostate cancer cells were tested as whole cell vaccines to determine their capacity to evoke an anti-prostate cancer immune response. In our model, vaccines secreting mGM-CSF or mIL-7 resulted in the highest increase in circulating T lymphocytes after vaccination, prolonged survival and, in a proportion of animals, tumor-free survival. Anti-tumor effects were more evident after a subcutaneous RM-9 challenge than after an intraprostatic challenge. However, when the RM-9/mGM-CSF cell line was used as intraprostatic tumor challenge, protection after RM-9/mIL-7 vaccination was restored.
Collapse
Affiliation(s)
- Caroline Schroten-Loef
- Erasmus MC, Department of Urology, Josephine Nefkens Institute, Room Be355A, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Corrina M. A. de Ridder
- Erasmus MC, Department of Urology, Josephine Nefkens Institute, Room Be355A, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Suzanne Reneman
- Erasmus MC, Department of Urology, Josephine Nefkens Institute, Room Be355A, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Marije Crezee
- Erasmus MC, Department of Urology, Josephine Nefkens Institute, Room Be355A, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Angus Dalgleish
- Department of Oncology, St. George’s University of London, London, UK
| | - Stephen M. Todryk
- School of Applied Sciences, University of Northumbria, Newcastle-upon-Tyne, UK
| | - Chris H. Bangma
- Erasmus MC, Department of Urology, Josephine Nefkens Institute, Room Be355A, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Robert Kraaij
- Erasmus MC, Department of Urology, Josephine Nefkens Institute, Room Be355A, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| |
Collapse
|
27
|
Li B, VanRoey M, Wang C, Chen THT, Korman A, Jooss K. Anti-programmed death-1 synergizes with granulocyte macrophage colony-stimulating factor--secreting tumor cell immunotherapy providing therapeutic benefit to mice with established tumors. Clin Cancer Res 2009; 15:1623-34. [PMID: 19208793 DOI: 10.1158/1078-0432.ccr-08-1825] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose of the present study was to evaluate granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapy, which is known to stimulate potent and long-lasting antigen-specific immune responses, in combination with PD-1 blockade, which has been shown to augment cellular immune responses. EXPERIMENTAL DESIGN Survival studies were done in the B16 melanoma and CT26 colon carcinoma tumor models. Immune monitoring studies were done in the B16 model. GM-CSF-secreting tumor cell immunotherapy was administered s.c. and the anti-PD-1 antibody was administered i.p. RESULTS The studies reported here show that combining PD-1 blockade with GM-CSF-secreting tumor cell immunotherapy prolonged the survival of tumor-bearing animals compared with animals treated with either therapy alone. Prolonged survival correlated with strong antigen-specific T-cell responses detected by tetramer staining and an in vivo CTL assay, higher secretion levels of proinflammatory cytokines by splenocytes, and the persistence of functional CD8+ T cells in the tumor microenvironment. Furthermore, in the biweekly multiple treatment setting, repeated antigen-specific T-cell expansion was only observed following administration of the cellular immunotherapy with the PD-1 blockade and not when the cellular immunotherapy or PD-1 blockade was used as monotherapy. CONCLUSION The combination of PD-1 blockade with GM-CSF-secreting tumor cell immunotherapy leads to significantly improved antitumor responses by augmenting the tumor-reactive T-cell responses induced by the cellular immunotherapy. Readministration of the cellular immunotherapy with the anti-PD-1 antibody in subsequent immunotherapy cycles was required to reactivate these T-cell responses.
Collapse
Affiliation(s)
- Betty Li
- Cell Genesys, Inc., South San Francisco, California 94080, USA
| | | | | | | | | | | |
Collapse
|
28
|
Li B, VanRoey M, Triebel F, Jooss K. Lymphocyte activation gene-3 fusion protein increases the potency of a granulocyte macrophage colony-stimulating factor-secreting tumor cell immunotherapy. Clin Cancer Res 2008; 14:3545-54. [PMID: 18519788 DOI: 10.1158/1078-0432.ccr-07-5200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The purpose of the present study was to evaluate granulocyte macrophage colony-stimulating factor (GM-CSF)-secreting tumor cell immunotherapy, which is known to stimulate a potent and long-lasting antigen-specific immune response in combination with lymphocyte activation gene-3 fusion protein (LAG-3Ig), which has been shown to act as an adjuvant for priming T helper type 1 and cytotoxic T-cell responses. EXPERIMENTAL DESIGN Survival and immune monitoring studies were done in the B16 melanoma model. GM-CSF-secreting tumor cell immunotherapy was administered as a single s.c. injection and LAG-3Ig was administered s.c. at the immunotherapy site. RESULTS The studies reported here show that combining LAG-3Ig with GM-CSF-secreting tumor cell immunotherapy prolonged the survival of tumor-bearing animals compared with animals treated with either therapy alone. Prolonged survival correlated with increased numbers of systemic IFN gamma-secreting CD8+ T cells and a significantly increased infiltration of activated effector CD8+ T cells into the tumor. Moreover, an increase in antigen-specific IgG1 humoral responses was detected in serum of animals injected with the combination therapy compared with animals injected with either therapy alone. CONCLUSION LAG-3Ig combined with a GM-CSF-secreting tumor cell immunotherapy stimulated both cellular and humoral antitumor immune responses that correlated with prolonged survival in tumor-bearing animals.
Collapse
Affiliation(s)
- Betty Li
- Cell Genesys, Inc., 500 Forbes Boulevard, South San Francisco, CA 94080, USA.
| | | | | | | |
Collapse
|
29
|
Sportès C, Hakim FT, Memon SA, Zhang H, Chua KS, Brown MR, Fleisher TA, Krumlauf MC, Babb RR, Chow CK, Fry TJ, Engels J, Buffet R, Morre M, Amato RJ, Venzon DJ, Korngold R, Pecora A, Gress RE, Mackall CL. Administration of rhIL-7 in humans increases in vivo TCR repertoire diversity by preferential expansion of naive T cell subsets. ACTA ACUST UNITED AC 2008; 205:1701-14. [PMID: 18573906 PMCID: PMC2442646 DOI: 10.1084/jem.20071681] [Citation(s) in RCA: 364] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Interleukin-7 (IL-7) is a homeostatic cytokine for resting T cells with increasing serum and tissue levels during T cell depletion. In preclinical studies, IL-7 therapy exerts marked stimulating effects on T cell immune reconstitution in mice and primates. First-in-human clinical studies of recombinant human IL-7 (rhIL-7) provided the opportunity to investigate the effects of IL-7 therapy on lymphocytes in vivo. rhIL-7 induced in vivo T cell cycling, bcl-2 up-regulation, and a sustained increase in peripheral blood CD4+ and CD8+ T cells. This T cell expansion caused a significant broadening of circulating T cell receptor (TCR) repertoire diversity independent of the subjects' age as naive T cells, including recent thymic emigrants (RTEs), expanded preferentially, whereas the proportions of regulatory T (T reg) cells and senescent CD8+ effectors diminished. The resulting composition of the circulating T cell pool more closely resembled that seen earlier in life. This profile, distinctive among cytokines under clinical development, suggests that rhIL-7 therapy could enhance and broaden immune responses, particularly in individuals with limited naive T cells and diminished TCR repertoire diversity, as occurs after physiological (age), pathological (human immunodeficiency virus), or iatrogenic (chemotherapy) lymphocyte depletion.
Collapse
Affiliation(s)
- Claude Sportès
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Ryan AA, Wozniak TM, Shklovskaya E, O’Donnell MA, Fazekas de St. Groth B, Britton WJ, Triccas JA. Improved Protection against Disseminated Tuberculosis byMycobacterium bovisBacillus Calmette-Guérin Secreting Murine GM-CSF Is Associated with Expansion and Activation of APCs. THE JOURNAL OF IMMUNOLOGY 2007; 179:8418-24. [DOI: 10.4049/jimmunol.179.12.8418] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
31
|
Dereuddre-Bosquet N, Vaslin B, Delache B, Brochard P, Clayette P, Aubenque C, Morre M, Assouline B, Le Grand R. Rapid modifications of peripheral T-cell subsets that express CD127 in macaques treated with recombinant IL-7. J Med Primatol 2007; 36:228-37. [PMID: 17669211 DOI: 10.1111/j.1600-0684.2007.00240.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND Interleukin-7 (IL-7) is a key regulator of thymopoiesis and T-cell homeostasis, which increases blood T-cell number by enhancing thymic output of naive cells and peripheral proliferation. METHODS We explored the effects of unglycosylated recombinant simian IL-7 (rsIL-7) administration on peripheral T-cell subpopulations in healthy macaques. RESULTS RsIL-7 was well tolerated. Mean half-life ranged between 9.3 and 13.9 hours. Blood CD3(+)CD4(+) and CD3(+)CD8(+) lymphocyte counts decreased rapidly after each rsIL-7 administration, the duration of these effects being dependent on the frequency of administration. At treatment completion, the increased of CD3(+) lymphocytes was marked at 100 microg/kg every 2 days. CD3(+) lymphocytes that harbour the alpha chain of IL-7 receptor (CD127) and CD3(+)CD8(+) lymphocytes that expressed the proliferation marker Ki-67 exhibited a similar initial profile. The expression of the anti-apoptotic marker Bcl-2 increased in CD3(+) lymphocytes during the treatment and post-treatment period in a dose/frequency dependent manner. CONCLUSION RsIL-7 was well tolerated in macaques and induces rapid modifications of T-cells that express CD127.
Collapse
|