1
|
Gao J, Zhai Y, Lu W, Jiang X, Zhou J, Wu L, Du L, Ou C, Zhang X, He H, Zhu J, Zhang Z, Li M, Wu Y, Pan X. ROS-sensitive PD-L1 siRNA cationic selenide nanogels for self-inhibition of autophagy and prevention of immune escape. Bioact Mater 2024; 41:597-610. [PMID: 39280899 PMCID: PMC11393550 DOI: 10.1016/j.bioactmat.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 09/18/2024] Open
Abstract
In the field of cancer therapy, inhibiting autophagy has emerged as a promising strategy. However, pharmacological disruption of autophagy can lead to the upregulation of programmed death-ligand 1 (PD-L1), enabling tumor immune evasion. To address this issue, we developed innovative ROS-responsive cationic poly(ethylene imine) (PEI) nanogels using selenol chemistry-mediated multicomponent reaction (MCR) technology. This procedure involved simple mixing of low-molecular-weight PEI (LMW PEI), γ-selenobutylacetone (γ-SBL), and poly(ethylene glycol) methacrylate (PEGMA). Through high-throughput screening, we constructed a library of AxSeyOz nanogels and identified the optimized A1.8Se3O0.5/siPD-L1 nanogels, which exhibited a size of approximately 200 nm, excellent colloidal stability, and the most effective PD-L1 silencing efficacy. These nanogels demonstrated enhanced uptake by tumor cells, excellent oxidative degradation ability, and inhibited autophagy by alkalinizing lysosomes. The A1.8Se3O0.5/siPD-L1 nanogels significantly downregulated PD-L1 expression and increased the expression of major histocompatibility complex class I (MHC-I), resulting in robust proliferation of specific CD8+ T cells and a decrease in MC38 tumor growth. As a result, the A1.8Se3O0.5/siPD-L1 nanogels inhibited tumor growth through self-inhibition of autophagy, upregulation of MHC-I, and downregulation of PD-L1. Designed with dynamic diselenide bonds, the A1.8Se3O0.5/siPD-L1 nanogels showed synergistic antitumor efficacy through self-inhibition of autophagy and prevention of immune escape.
Collapse
Affiliation(s)
- Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Yonghua Zhai
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Weihong Lu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Xianghe Jiang
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Jingsheng Zhou
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Longhai Du
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Chunqing Ou
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Xinyi Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| | - Hanliang He
- The Department of Orthopedic Surgery, Dushu Lake Hospital Affiliated to Soochow University, Suzhou, 215028, China
| | - Jian Zhu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Zhengbiao Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
| | - Meiyun Li
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xiangqiang Pan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, Jiangsu Key Laboratory of Advanced Functional Polymer Design and Application, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| |
Collapse
|
2
|
Hou L, Zhang S, Yu W, Yang X, Shen M, Hao X, Ren X, Sun Q. Single-cell transcriptomics reveals tumor-infiltrating B cell function after neoadjuvant pembrolizumab and chemotherapy in non-small cell lung cancer. J Leukoc Biol 2024; 116:555-564. [PMID: 37931147 DOI: 10.1093/jleuko/qiad138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most pervasive lung cancer subtype. Recent studies have shown that immune checkpoint inhibitors achieved favorable clinical benefits in resectable NSCLC; however, the associated mechanism remains unclear. The role of T cells in antitumor immunity has received considerable attention, while the antitumor effects of tumor-infiltrating B cells (TIBs) in NSCLC remain poorly understood. Here, we conducted a single-cell RNA sequencing analysis of immune cells isolated from 12 patients with stage IIIA NSCLC to investigate B cell subtypes and their functions following neoadjuvant chemoimmunotherapy. We confirmed the simultaneous existence of the 4 B cell subtypes. Among them, memory B cells were found to be associated with a positive therapeutic effect to neoadjuvant chemoimmunotherapy. Furthermore, we found that G protein-coupled receptor 183 was most prevalent in memory B cells and associated with a positive therapeutic response. Multiplex immunofluorescence and flow cytometry experiments in an additional cohort of 22 treatment-naïve and 30 stage IIIA/IIIB NSCLC patients treated with neoadjuvant chemoimmunotherapy verified these findings. Overall, our analysis revealed the functions of TIBs and their potential effect on clinical treatment in NSCLC.
Collapse
Affiliation(s)
- Lingjie Hou
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
| | - Siyuan Zhang
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
| | - Wenwen Yu
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
| | - Xuena Yang
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
| | - Meng Shen
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
| | - Xishan Hao
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Haihe Laboratory of Cell Ecosystem, No. 10 Yuexin Road, Binhai District, Tianjin 300450, China
| | - Xiubao Ren
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Department of Biotherapy, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Haihe Laboratory of Cell Ecosystem, No. 10 Yuexin Road, Binhai District, Tianjin 300450, China
| | - Qian Sun
- National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Prevention and Therapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Tianjin's Clinical Research Center for Cancer, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Key Laboratory of Cancer Immunology and Biotherapy, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, No. 45 Binshui Road, Hexi District, Tianjin 300060, China
- Haihe Laboratory of Cell Ecosystem, No. 10 Yuexin Road, Binhai District, Tianjin 300450, China
| |
Collapse
|
3
|
Hannon G, Lesch ML, Gerber SA. Harnessing the Immunological Effects of Radiation to Improve Immunotherapies in Cancer. Int J Mol Sci 2023; 24:7359. [PMID: 37108522 PMCID: PMC10138513 DOI: 10.3390/ijms24087359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Ionizing radiation (IR) is used to treat 50% of cancers. While the cytotoxic effects related to DNA damage with IR have been known since the early 20th century, the role of the immune system in the treatment response is still yet to be fully determined. IR can induce immunogenic cell death (ICD), which activates innate and adaptive immunity against the cancer. It has also been widely reported that an intact immune system is essential to IR efficacy. However, this response is typically transient, and wound healing processes also become upregulated, dampening early immunological efforts to overcome the disease. This immune suppression involves many complex cellular and molecular mechanisms that ultimately result in the generation of radioresistance in many cases. Understanding the mechanisms behind these responses is challenging as the effects are extensive and often occur simultaneously within the tumor. Here, we describe the effects of IR on the immune landscape of tumors. ICD, along with myeloid and lymphoid responses to IR, are discussed, with the hope of shedding light on the complex immune stimulatory and immunosuppressive responses involved with this cornerstone cancer treatment. Leveraging these immunological effects can provide a platform for improving immunotherapy efficacy in the future.
Collapse
Affiliation(s)
- Gary Hannon
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Maggie L. Lesch
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Scott A. Gerber
- Department of Surgery, University of Rochester Medical Center, Rochester, NY 14642, USA; (G.H.); (M.L.L.)
- Center for Tumor Immunology Research, University of Rochester Medical Center, Rochester, NY 14642, USA
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
4
|
Saudi A, Banday V, Zirakzadeh AA, Selinger M, Forsberg J, Holmbom M, Henriksson J, Waldén M, Alamdari F, Aljabery F, Winqvist O, Sherif A. Immune-Activated B Cells Are Dominant in Prostate Cancer. Cancers (Basel) 2023; 15:cancers15030920. [PMID: 36765877 PMCID: PMC9913271 DOI: 10.3390/cancers15030920] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/22/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
B cells are multifaceted immune cells responding robustly during immune surveillance against tumor antigens by presentation to T cells and switched immunoglobulin production. However, B cells are unstudied in prostate cancer (PCa). We used flow cytometry to analyze B-cell subpopulations in peripheral blood and lymph nodes from intermediate-high risk PCa patients. B-cell subpopulations were related to clinicopathological factors. B-cell-receptor single-cell sequencing and VDJ analysis identified clonal B-cell expansion in blood and lymph nodes. Pathological staging was pT2 in 16%, pT3a in 48%, and pT3b in 36%. Lymph node metastases occurred in 5/25 patients (20%). Compared to healthy donors, the peripheral blood CD19+ B-cell compartment was significantly decreased in PCa patients and dominated by naïve B cells. The nodal B-cell compartment had significantly increased fractions of CD19+ B cells and switched memory B cells. Plasmablasts were observed in tumor-draining sentinel lymph nodes (SNs). VDJ analysis revealed clonal expansion in lymph nodes. Thus, activated B cells are increased in SNs from PCa patients. The increased fraction of switched memory cells and plasmablasts together with the presence of clonally expanded B cells indicate tumor-specific T-cell-dependent responses from B cells, supporting an important role for B cells in the protection against tumors.
Collapse
Affiliation(s)
- Aws Saudi
- Department of Urology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
- Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
| | - Viqar Banday
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umea University, 901 85 Umea, Sweden
- Department of Clinical Microbiology, Immunology, Umea University, 901 85 Umeå, Sweden
| | | | - Martin Selinger
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), 901 87 Umeå, Sweden
- Department of Molecular Biology, Umeå Centre for Microbial Research, 6K and 6L, Umeå University, 901 87 Umeå, Sweden
| | - Jon Forsberg
- Department of Urology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
| | - Martin Holmbom
- Department of Urology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
| | - Johan Henriksson
- The Laboratory for Molecular Infection Medicine Sweden (MIMS), 901 87 Umeå, Sweden
- Department of Molecular Biology, Umeå Centre for Microbial Research, 6K and 6L, Umeå University, 901 87 Umeå, Sweden
| | - Mauritz Waldén
- Department of Urology, Central Hospital of Karlstad, 652 30 Karlstad, Sweden
| | - Farhood Alamdari
- Department of Urology, Västmanland Hospital, 721 89 Västerås, Sweden
| | - Firas Aljabery
- Department of Urology, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
- Department of Clinical and Experimental Medicine, Medical Faculty, Linköping University, 581 85 Linköping, Sweden
| | - Ola Winqvist
- ABClabs, BioClinicum, Campus Solna, 171 76 Stockholm, Sweden
| | - Amir Sherif
- Department of Surgical and Perioperative Sciences, Urology and Andrology, Umea University, 901 85 Umea, Sweden
- Correspondence:
| |
Collapse
|
5
|
Qin Y, Lu F, Lyu K, Chang AE, Li Q. Emerging concepts regarding pro- and anti tumor properties of B cells in tumor immunity. Front Immunol 2022; 13:881427. [PMID: 35967441 PMCID: PMC9366002 DOI: 10.3389/fimmu.2022.881427] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 07/07/2022] [Indexed: 12/26/2022] Open
Abstract
Controversial views regarding the roles of B cells in tumor immunity have existed for several decades. However, more recent studies have focused on its positive properties in antitumor immunity. Many studies have demonstrated a close association of the higher density of intratumoral B cells with favorable outcomes in cancer patients. B cells can interact with T cells as well as follicular dendritic cells within tertiary lymphoid structures, where they undergo a series of biological events, including clonal expansion, somatic hypermutation, class switching, and tumor-specific antibody production, which may trigger antitumor humoral responses. After activation, B cells can function as effector cells via direct tumor-killing, antigen-presenting activity, and production of tumor-specific antibodies. At the other extreme, B cells can obtain inhibitory functions by relevant stimuli, converting to regulatory B cells, which serve as an immunosuppressive arm to tumor immunity. Here we summarize our current understanding of the bipolar properties of B cells within the tumor immune microenvironment and propose potential B cell-based immunotherapeutic strategies, which may help promote cancer immunotherapy.
Collapse
Affiliation(s)
- You Qin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
| | - Furong Lu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Kexing Lyu
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Alfred E. Chang
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Qiao Li, ; Alfred E. Chang,
| | - Qiao Li
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Qiao Li, ; Alfred E. Chang,
| |
Collapse
|
6
|
Newnes HV, Armitage JD, Audsley KM, Bosco A, Waithman J. Directing the Future Breakthroughs in Immunotherapy: The Importance of a Holistic Approach to the Tumour Microenvironment. Cancers (Basel) 2021; 13:cancers13235911. [PMID: 34885021 PMCID: PMC8656826 DOI: 10.3390/cancers13235911] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Immunotherapies have changed the way we treat cancer and, while some patients have benefitted greatly, there are still those that do not respond to therapy. Understanding why some patients respond to therapy and others do not is critical in developing new immunotherapeutic strategies. The increasing awareness of the importance of investigating the tumour in its entirety, including the surrounding tissue and role of various immune cells is helping to differentiate responders and non-responders. In addition, the resolution gained by the development of sophisticated bioinformatic technologies allows for a deeper understanding of the complex roles of individual cells in the tumour. This advancement will be critical for the development of novel therapies to treat cancer. Abstract Immunotherapy has revolutionised the treatment of cancers by exploiting the immune system to eliminate tumour cells. Despite the impressive response in a proportion of patients, clinical benefit has been limited thus far. A significant focus to date has been the identification of specific markers associated with response to immunotherapy. Unfortunately, the heterogeneity between patients and cancer types means identifying markers of response to therapy is inherently complex. There is a growing appreciation for the role of the tumour microenvironment (TME) in directing response to immunotherapy. The TME is highly heterogeneous and contains immune, stromal, vascular and tumour cells that all communicate and interact with one another to form solid tumours. This review analyses major cell populations present within the TME with a focus on their diverse and often contradictory roles in cancer and how this informs our understanding of immunotherapy. Furthermore, we discuss the role of integrated omics in providing a comprehensive view of the TME and demonstrate the potential of leveraging multi-omics to decipher the underlying mechanisms of anti-tumour immunity for the development of novel immunotherapeutic strategies.
Collapse
|
7
|
Shi Y. PLAN B for immunotherapy: Promoting and leveraging anti-tumor B cell immunity. J Control Release 2021; 339:156-163. [PMID: 34563591 DOI: 10.1016/j.jconrel.2021.09.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 09/19/2021] [Accepted: 09/21/2021] [Indexed: 12/19/2022]
Abstract
Current immuno-oncology primarily focuses on adaptive cellular immunity mediated by T lymphocytes. The other important lymphocytes, B cells, are largely ignored in cancer immunotherapy. B cells are generally considered to be responsible for humoral immune response to viral and bacterial infections. The role of B cells in cancer immunity has long been under debate. Recently, increasing evidence from both preclinical and clinical research has shown that B cells can also induce potent anti-cancer immunity, via humoral and cellular immune responses. Yet it is unclear how to efficiently integrate B cell immunity in cancer immunotherapy. In the current perspective, anti-tumor immunity of B cells is discussed regarding antibody production, antigen presentation, cytokine release and contribution to intratumoral tertiary lymphoid structures. Afterwards, immunosuppressive regulatory phenotypes of B cells are summarized. Furthermore, strategies to activate and modulate B cells using nanomedicines and biomaterials are discussed. This article provides a unique perspective on "PLAN B" (promoting and leveraging anti-tumor B cell immunity) using nanomedicines and biomaterials for cancer immunotherapy. This is envisaged to form a new research direction with the potential to reach the next breakthrough in immunotherapy.
Collapse
Affiliation(s)
- Yang Shi
- Institute for Experimental Molecular Imaging, Uniklinik RWTH Aachen and Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, Aachen 52074, Germany.
| |
Collapse
|
8
|
Activated B Cells and Plasma Cells Are Resistant to Radiation Therapy. Int J Radiat Oncol Biol Phys 2021; 112:514-528. [PMID: 34474108 DOI: 10.1016/j.ijrobp.2021.08.037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE B cells play a key role in outcomes of cancer patients and responses to checkpoint blockade immunotherapies. However, the effect of radiation therapy on B cell populations is poorly understood. Here we characterize the effects of radiation on the development, survival, and phenotype of physiological B-cell subsets. METHODS AND MATERIALS Naïve and immunized tumor bearing and nontumor bearing mice were treated with large-field or focal stereotactic radiation and distinct B-cell subsets of varying developmental stages were analyzed by flow cytometry and real-time reverse transcription polymerase chain reaction. RESULTS We first report that focal stereotactic radiation is highly superior to large-field radiation at inducing tumor infiltration of B cells, CD8+ T cells, and macrophages. We observed that radiation affects B cell development in the bone marrow, increasing frequencies of early pro-B cells and late pro-B cells while inducing upregulation of programmed cell death protein 1. We then demonstrate that class switched B cells and plasma cells are highly resistant to radiation therapy compared with naïve B cells and upregulate activation markers programmed cell death 1 ligand 2 and major histocompatibility complex class II) after radiation. Mechanistically, radiation upregulates Xbp1 and Bcl6 in plasma cells, conferring radioresistance. Furthermore, using an immunization approach, we demonstrate that radiation enhances activation-induced cytidine deaminase mediated class switching and somatic hypermutation in primed B cells. CONCLUSIONS These data demonstrate that stereotactic radiation is superior to large-field radiation at inducing infiltration of immune cells into tumors and that plasma cells and class switched B cells are highly resistant to radiation therapy. These results represent the most comprehensive analysis of the effects of radiation on B cells to date and identify novel mechanisms by which radiation modulates immune cells within the tumor microenvironment.
Collapse
|
9
|
Qin Y, Peng F, Ai L, Mu S, Li Y, Yang C, Hu Y. Tumor-infiltrating B cells as a favorable prognostic biomarker in breast cancer: a systematic review and meta-analysis. Cancer Cell Int 2021; 21:310. [PMID: 34118931 PMCID: PMC8199375 DOI: 10.1186/s12935-021-02004-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 06/01/2021] [Indexed: 01/01/2023] Open
Abstract
Background Tumor-infiltrating B lymphocytes (TIL-Bs) is a heterogeneous population of lymphocytes. The prognostic value of TIL-Bs in patients with breast cancer remains controversial. Here we conducted this meta-analysis to clarify the association of TIL-Bs with outcomes of patients with breast cancer. Methods We searched PubMed, Embase, and Web of Science to identify relevant studies assessing the prognostic significance of TIL-Bs in patients with breast cancer. Fixed- or random-effects models were used to evaluate the pooled hazard ratios (HRs) for overall survival (OS), breast cancer-specific survival (BCSS), disease-free survival (DFS), and relapse-free survival (RFS) in breast cancer. Results
A total of 8 studies including 2628 patients were included in our study. Pooled analyses revealed that high level of TIL-Bs was associated with longer OS (pooled HR = 0.42, 95% CI 0.24–0.60), BCSS (pooled HR = 0.66, 95% CI 0.47–0.85), and DFS/RFS (pooled HR = 0.41, 95% CI 0.27–0.55). Conclusions This meta-analysis suggests that TIL-Bs could be a promising prognostic marker for breast cancer. Novel therapeutic strategies for breast cancer treatment could be developed by enhancement of B cell-mediated antitumor immunity. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-021-02004-9.
Collapse
Affiliation(s)
- You Qin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fei Peng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Hubei, 430022, Wuhan, China
| | - Lisha Ai
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Hubei, 430022, Wuhan, China.
| | - Shidai Mu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Hubei, 430022, Wuhan, China
| | - Yuting Li
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chensu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Hubei, 430022, Wuhan, China.
| |
Collapse
|
10
|
Tumor-Infiltrating CD20 + B Lymphocytes: Significance and Prognostic Implications in Oral Cancer Microenvironment. Cancers (Basel) 2021; 13:cancers13030395. [PMID: 33494389 PMCID: PMC7865920 DOI: 10.3390/cancers13030395] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/18/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The complex interplay between the different cellular components in the tumor microenvironment (TME) dynamically modulates the antitumor immune response. This study investigates the prognostic relevance of CD20+ tumor-infiltrating B lymphocytes in oral squamous cell carcinoma (OSCC), and also possible relationships with other immune subtypes and key players within the oral TME. Abstract Immunohistochemical analysis of stromal/tumoral CD20+ B lymphocytes was performed in 125 OSCC patients. Correlations with immune profiles CD4+, CD8+, and FOXP3+ tumor-infiltrating lymphocytes (TILs), tumoral PD-L1, and stem-related factors NANOG and SOX2 were assessed, and also associations with clinical data and patient survival. There was a strong positive correlation between the infiltration of CD20+ B lymphocytes and other immune profiles (i.e., CD4+, CD8+, and FOXP3+ TILs, and CD68+ and CD163+ macrophages) both in stroma and tumor nests. Strikingly, CD20+ TILs were inversely correlated with NANOG/SOX2 expression. Stromal CD20+ TILs were significantly associated with T classification and second primary tumors. A stratified survival analysis showed that tumoral CD20+ TILs were significantly associated with prognosis in male and younger patients, with tobacco or alcohol consumption, high tumoral CD8+ TILs, low tumoral infiltration by CD68+ macrophages, positive PD-L1 expression, and negative NANOG/SOX2. Multivariate Cox analysis further revealed clinical stage and tumoral CD20+ TILs independently associated with disease-specific survival (HR = 2.42, p = 0.003; and HR = 0.57, p = 0.04, respectively). In conclusion, high CD20+ TIL density emerges as an independent good prognostic factor in OSCC, suggesting a role in antitumor immunity. This study also uncovered an inverse correlation between CD20+ TILs and CSC marker expression.
Collapse
|
11
|
Norouzian M, Mehdipour F, Balouchi Anaraki S, Ashraf MJ, Khademi B, Ghaderi A. Atypical Memory and Regulatory B Cell Subsets in Tumor Draining Lymph Nodes of Head and Neck Squamous Cell Carcinoma Correlate with Good Prognostic Factors. Head Neck Pathol 2019; 14:645-656. [PMID: 31691165 PMCID: PMC7413970 DOI: 10.1007/s12105-019-01095-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 10/30/2019] [Indexed: 12/29/2022]
Abstract
Research on the role of B cells in the development and modulation of antitumor immunity has increased in recent years; however, knowledge about B cell phenotype and function in tumor draining lymph nodes (TDLNs) is still incomplete. This study aimed to investigate changes in the phenotypic profile of B cells in TDLNs of head and neck squamous cell carcinoma (HNSCC) during disease progression. Mononuclear cells were isolated from TDLNs and stained with antibodies for CD19 and other B cell-related markers and analyzed by flow cytometry. CD19+ B cells comprised 38.6 ± 8.9% of lymphocytes in TDLNs of HNSCC. Comparison of metastatic and non-metastatic LNs disclosed no significant differences in the frequencies of B cell subsets including antigen-experienced, naïve, switched, unswitched, atypical memory, marginal zone-like B cells, and B cells with regulatory phenotypes. The percentage of atypical memory (CD27-IgM-IgD-) B cells was significantly higher in patients with tongue SCC with no involved LNs (p = 0.033) and correlated inversely with the number of involved LNs. The frequency of CD24hiCD38hi B cells was significantly higher in non-metastatic LNs of patients with grade I compared to grade II (p = 0.016), and the percentage of CD5+ B cells decreased as tumors progressed from stage III to IV (p = 0.008). Our data show that in TDLNs of HNSCC, the frequency of B cells with atypical memory and regulatory phenotypes was significantly associated with good prognostic factors; however, their function remains to be investigated.
Collapse
Affiliation(s)
- Marzieh Norouzian
- grid.412571.40000 0000 8819 4698Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fereshteh Mehdipour
- grid.412571.40000 0000 8819 4698Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, P.O. Box: 71345-3119, Shiraz, Iran
| | - Sima Balouchi Anaraki
- grid.412571.40000 0000 8819 4698Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Javad Ashraf
- grid.412571.40000 0000 8819 4698Department of Oral Pathology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Bijan Khademi
- grid.412571.40000 0000 8819 4698Department of Otolaryngology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Abbas Ghaderi
- grid.412571.40000 0000 8819 4698Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran ,grid.412571.40000 0000 8819 4698Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, P.O. Box: 71345-3119, Shiraz, Iran
| |
Collapse
|
12
|
Zhao KL, Yang XJ, Jin HZ, Zhao L, Hu JL, Qin WJ. Double-edge Role of B Cells in Tumor Immunity: Potential Molecular Mechanism. Curr Med Sci 2019; 39:685-689. [PMID: 31612383 DOI: 10.1007/s11596-019-2092-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/03/2019] [Indexed: 01/01/2023]
Abstract
B cells are a heterogeneous population, which have distinct functions of antigen presentation, activating T cells, and secreting antibodies, cytokines as well as protease. It is supposed that the balance among these B cells subpopulation (resting B cells, activated B cells, Bregs, and other differentiated B cells) will determine the ultimate role of B cells in tumor immunity. There has been increasing evidence supporting opposite roles of B cells in tumor immunity, though there are no general acceptable phenotypes for them. Recent years, a new designated subset of B cells identified as Bregs has emerged from immunosuppressive and/or regulatory functions in tumor immune responses. Therefore, transferring activated B cells would be possible to become a promising strategy against tumor via conquering the immunosuppressive status of B cells in future. Understanding the potential mechanism of double-edge role of B cells will help researchers utilize activated B cells to improve their anti-tumor response. Moreover, the molecular pathways related to B cell differentiation are involved in its tumor-promoting effect, such as NF-κB, STAT3, BTK. So, we review the molecular and signaling pathway mechanisms of B cells involved in both tumor-promoting and tumor-suppressive immunity, in order to help researchers optimize B cells to fight cancer better.
Collapse
Affiliation(s)
- Kai-Liang Zhao
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiao-Jia Yang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Hong-Zhong Jin
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liang Zhao
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jian-Li Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Wen-Juan Qin
- Department of Radiation Oncology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, 361001, China.
| |
Collapse
|
13
|
Chakraborty D, Pati S, Bose S, Dhar S, Dutta S, Sa G. Cancer immunotherapy: present scenarios and the future of immunotherapy. THE NUCLEUS 2019. [DOI: 10.1007/s13237-019-00273-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
|
14
|
Affiliation(s)
- Grace J Yuen
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge MA 02139
| | - Ezana Demissie
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge MA 02139
| | - Shiv Pillai
- Ragon Institute of MGH, MIT and Harvard, Harvard Medical School, Cambridge MA 02139
| |
Collapse
|
15
|
Prince MEP, Zhou L, Moyer JS, Tao H, Lu L, Owen J, Etigen M, Zheng F, Chang AE, Xia J, Wolf G, Wicha MS, Huang S, Ren X, Li Q. Evaluation of the immunogenicity of ALDH(high) human head and neck squamous cell carcinoma cancer stem cells in vitro. Oral Oncol 2018; 59:30-42. [PMID: 27424180 DOI: 10.1016/j.oraloncology.2016.05.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 05/04/2016] [Accepted: 05/20/2016] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To establish the concept that the antigenicity/immunogenicity of ALDH(high) human head and neck squamous cell carcinoma (HNSCC) cancer stem cells (CSC) is distinct from that of ALDH(low) non-CSCs. METHODS We generated CSC-loaded dendritic cells (DCs) to sensitize autologous peripheral blood T, B lymphocytes to react with CSCs using human HNSCC samples in vitro. RESULTS From peripheral blood collected from patients with HNSCC, we obtained PBMCs. DCs generated from the PBMC and pulsed with the lysate of ALDH(high) cells isolated from cultured HNSCC cells (CSC-DC) could sensitize autologous T, B lymphocytes in vitro, which was evident by cytokine production, CTL activity, and antibody secretion of these primed T, B cells in response to ALDH(high) CSCs. In contrast, DCs pulsed with lysate of ALDH(low) cells (ALDH(low)-DC) resulted in limited sensitization/priming of autologous T, B lymphocytes to produce IFNγ, GM-CSF; lyse CSCs, and secrete IgM and IgG in response to ALDH(high) CSCs. These results demonstrated significant differences in the antigenicity/immunogenicity between ALDH(high) CSCs vs. ALDH(low) cells isolated from the tumor specimen of patients with HNSCC, which indicates the existence of unique CSC antigens in the ALDH(high) population. CONCLUSION It is feasible to generate DCs from the PBMCs and isolate ALDH(high) CSCs from cultured tumor cells of the patients with HNSCC to prepare CSC-DC vaccines that can induce anti-HNSCC CSC cellular and humoral immunity, indicating its potential clinical application to treat patients with HNSCC.
Collapse
Affiliation(s)
- Mark E P Prince
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Li Zhou
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,Department of Immunology, Biotherapy Center ,Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Jeffrey S Moyer
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Huimin Tao
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lu
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - John Owen
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Martin Etigen
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Fang Zheng
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA.,Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Alfred E Chang
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Jianchuan Xia
- State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Gregory Wolf
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Max S Wicha
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| | - Shiang Huang
- Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiubao Ren
- Department of Immunology, Biotherapy Center ,Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Qiao Li
- University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan, USA
| |
Collapse
|
16
|
Xia Y, Tao H, Hu Y, Chen Q, Chen X, Xia L, Zhou L, Wang Y, Bao Y, Huang S, Ren X, Lundy SK, Dai F, Li Q, Chang AE. IL-2 augments the therapeutic efficacy of adoptively transferred B cells which directly kill tumor cells via the CXCR4/CXCL12 and perforin pathways. Oncotarget 2018; 7:60461-60474. [PMID: 27528023 PMCID: PMC5312396 DOI: 10.18632/oncotarget.11124] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 07/26/2016] [Indexed: 01/07/2023] Open
Abstract
We previously reported that antitumor B cells directly kill tumor cells via the Fas/FasL pathway and are regulated by IL-10. In this study, we defined additional mechanisms involved in B cell antitumor immunity. Administration of IL-2 significantly augmented the therapeutic efficacy of adoptively transferred tumor-draining lymph node (TDLN) B cells which express IL- 2R. Culture supernatant of purified B splenocytes harvested from the mice that received adoptive transfer of 4T1 TDLN B cells plus IL-2 administration produced larger amounts of IgG which bound to 4T1, resulting in 4T1 lysis. Furthermore, we detected CXCR4 expression on 4T1 TDLN B cells, and 4T1 tumor cells produced its ligand CXCL12. Transwell experiments demonstrated the chemoattraction of CXCR4-expressing 4T1 TDLN B cells towards CXCL12- producing 4T1 cells. Blockade of CXCR4 using a CXCR4-specific inhibitor, AMD3100, significantly reduced the killing of 4T1 tumor cells by 4T1 TDLN B cells. Blockade of FasL and CXCR4 concurrently inhibited B cell-mediated direct killing of tumor cells in an additive manner, indicating that both Fas/FasL and CXCL12/CXCR4 pathways are involved in the direct killing of 4T1 cells by 4T1 TDLN B cells. TDLN B cells produced perforin. Additional transwell experiments showed that effector B cells could directly kill tumor cells in cell-cell contact via the Fas/FasL and CXCR4/CXCL12 pathways as well as perforin, while without cell contact, perforin secreted by B cells led to tumor cell cytotoxicity. These findings underscore the diversity of function by which B cells can play an important role in the host immune response to tumor.
Collapse
Affiliation(s)
- Yang Xia
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA.,The No.1 People's Hospital of Hefei, Hefei, China
| | - Huimin Tao
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA.,Hubei Province Stem Cell Research & Appling Center, Wuhan Union Hospital, Wuhan, China.,Current address: Fuda Cancer Hospital, Jinan University School of Medicine and Fuda Cancer Institute, Guangzhou, China
| | - Yangyang Hu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA.,Hubei Province Stem Cell Research & Appling Center, Wuhan Union Hospital, Wuhan, China
| | - Quanning Chen
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of General Surgery, Tongji Hospital of Tongji University, Shanghai, China
| | - Xin Chen
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Oncology, Wuhan University, Renmin Hospital, Wuhan, China
| | - Leiming Xia
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA.,The No.1 People's Hospital of Hefei, Hefei, China
| | - Li Zhou
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA.,Department of Biotherapy, Tianjin University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Yi Wang
- The No.1 People's Hospital of Hefei, Hefei, China
| | - Yangyi Bao
- The No.1 People's Hospital of Hefei, Hefei, China
| | - Shiang Huang
- Hubei Province Stem Cell Research & Appling Center, Wuhan Union Hospital, Wuhan, China
| | - Xiubao Ren
- Department of Biotherapy, Tianjin University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key laboratory of Cancer Immunology and Biotherapy, Tianjin, China
| | - Steven K Lundy
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Fu Dai
- The No.1 People's Hospital of Hefei, Hefei, China
| | - Qiao Li
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Alfred E Chang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
17
|
Huang Y, Li D, Qin DY, Gou HF, Wei W, Wang YS, Wei YQ, Wang W. Interleukin-armed chimeric antigen receptor-modified T cells for cancer immunotherapy. Gene Ther 2017; 25:192-197. [DOI: 10.1038/gt.2017.81] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 04/10/2017] [Accepted: 07/28/2017] [Indexed: 01/01/2023]
|
18
|
Padera TP, Meijer EFJ, Munn LL. The Lymphatic System in Disease Processes and Cancer Progression. Annu Rev Biomed Eng 2016; 18:125-58. [PMID: 26863922 DOI: 10.1146/annurev-bioeng-112315-031200] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Advances in our understanding of the structure and function of the lymphatic system have made it possible to identify its role in a variety of disease processes. Because it is involved not only in fluid homeostasis but also in immune cell trafficking, the lymphatic system can mediate and ultimately alter immune responses. Our rapidly increasing knowledge of the molecular control of the lymphatic system will inevitably lead to new and effective therapies for patients with lymphatic dysfunction. In this review, we discuss the molecular and physiological control of lymphatic vessel function and explore how the lymphatic system contributes to many disease processes, including cancer and lymphedema.
Collapse
Affiliation(s)
- Timothy P Padera
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | - Eelco F J Meijer
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, and Cancer Center, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114;
| |
Collapse
|
19
|
Tao H, Lu L, Xia Y, Dai F, Wang Y, Bao Y, Lundy SK, Ito F, Pan Q, Zhang X, Zheng F, Shu G, Fang B, Jiang J, Xia J, Huang S, Li Q, Chang AE. Antitumor effector B cells directly kill tumor cells via the Fas/FasL pathway and are regulated by IL-10. Eur J Immunol 2015; 45:999-1009. [PMID: 25545618 PMCID: PMC4414939 DOI: 10.1002/eji.201444625] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 12/01/2014] [Accepted: 12/19/2014] [Indexed: 11/05/2022]
Abstract
We have previously reported that adoptive transfer of tumor-draining lymph node (TDLN) B cells confers tumor regression in a spontaneous pulmonary metastasis mouse model of breast cancer. In this study, we identified IL-10-producing cells within these B cells, and found that IL-10 removal, either by using IL-10(-/-) TDLN B cells or by systemic neutralization of IL-10, significantly augmented the therapeutic efficacy of adoptively transferred TDLN B cells. Depletion of IL-10 in B-cell adoptive transfers significantly increased CTLs and B-cell activity of PBMCs and splenic cells in the recipient. Activated TDLN B cells express Fas ligand, which was further enhanced by coculture of these TDLN B cells with 4T1 tumor cells. Effector B cells killed tumor cells directly in vitro in an antigen specific and Fas ligand-dependent manner. Trafficking of TDLN B cells in vivo suggested that they were recruited to the tumor and lung as well as secondary lymphoid organs. These findings further define the biological function of antitumor effector B cells, which may offer alternative cellular therapies to cancer.
Collapse
Affiliation(s)
- Huimin Tao
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Lu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yang Xia
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Fu Dai
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yi Wang
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yangyi Bao
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Steven K. Lundy
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Fumito Ito
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| | - Qin Pan
- Wuhan University School of Medicine, Wuhan, China
| | | | - Fang Zheng
- Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guoshun Shu
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- Second Xiangya Hospital, Changsha, China
| | - Bingmu Fang
- Lishui People’s Hospital of Zhejiang Province, China
| | - Jinhong Jiang
- Lishui People’s Hospital of Zhejiang Province, China
| | - Jianchuang Xia
- State Key Laboratory of Oncology in Southern China and Department of Experimental Research, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shiang Huang
- Hubei Province Stem Cell Research & Appling Center, Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qiao Li
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
- The Third Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Alfred E. Chang
- Department of Surgery, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
20
|
Mu CY, Qin PX, Qu QX, Chen C, Huang JA. Soluble CD40 in plasma and malignant pleural effusion with non-small cell lung cancer: A potential marker of prognosis. Chronic Dis Transl Med 2015; 1:36-41. [PMID: 29062985 PMCID: PMC5643771 DOI: 10.1016/j.cdtm.2015.02.010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Soluble CD40 (sCD40) is a potential modulator for both antitumor responses and CD40-based immunotherapy; however the levels and significance of sCD40 in non-small cell lung cancer (NSCLC) patients with malignant pleural effusion are unknown. METHODS Forty-eight patients with lung cancer were treated in our institutions from January 2008 to January 2010. Peripheral blood and pleural effusion samples were collected from each subject. sCD40 levels in plasma and malignant pleural effusions supernatant were measured. The CD40L expression on CD3t T-cells was confirmed by flow cytometric direct immunofluorescence analysis. All patients were followed up after the study ended on January 1, 2010. RESULTS Patients with malignant pleural effusion of NSCLC had elevated circulating and pleural effusion levels of sCD40, and these elevated sCD40 levels were associated with advanced diseases and a poor prognosis. CONCLUSIONS These findings indicate that elevated sCD40 may have a role in modulating antitumor responses and may also be a useful prognostic marker.
Collapse
Affiliation(s)
- Chuan-Yong Mu
- Respiratory Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Pang-Xue Qin
- Gastrointestinal Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Qiu-Xia Qu
- Biotechnology Research Institute, Soochow University, 48 Renmin Road, Suzhou, Jiangsu 215007, China
| | - Cheng Chen
- Respiratory Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jian-An Huang
- Respiratory Department, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
- Corresponding author.
| |
Collapse
|
21
|
Pereira ER, Jones D, Jung K, Padera TP. The lymph node microenvironment and its role in the progression of metastatic cancer. Semin Cell Dev Biol 2015; 38:98-105. [PMID: 25620792 DOI: 10.1016/j.semcdb.2015.01.008] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/16/2022]
Abstract
Lymph nodes are initial sites for cancer metastasis in many solid tumors. However, their role in cancer progression is still not completely understood. Emerging evidence suggests that the lymph node microenvironment provides hospitable soil for the seeding and proliferation of cancer cells. Resident immune and stromal cells in the lymph node express and secrete molecules that may facilitate the survival of cancer cells in this organ. More comprehensive studies are warranted to fully understand the importance of the lymph node in tumor progression. Here, we will review the current knowledge of the role of the lymph node microenvironment in metastatic progression.
Collapse
Affiliation(s)
- Ethel R Pereira
- E.L. Steele Laboratory, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - Dennis Jones
- E.L. Steele Laboratory, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - Keehoon Jung
- E.L. Steele Laboratory, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA
| | - Timothy P Padera
- E.L. Steele Laboratory, Department of Radiation Oncology, Harvard Medical School and Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
22
|
Microenvironment of tumor-draining lymph nodes: opportunities for liposome-based targeted therapy. Int J Mol Sci 2014; 15:20209-39. [PMID: 25380524 PMCID: PMC4264163 DOI: 10.3390/ijms151120209] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 02/07/2023] Open
Abstract
The World Health Organization (WHO) recently reported that the total number of global cancer cases in 2013 reached 14 million, a 10% rise since 2008, while the total number of cancer deaths reached 8.2 million, a 5.2% increase since 2008. Metastasis is the major cause of death from cancer, accounting for 90% of all cancer related deaths. Tumor-draining lymph nodes (TDLN), the sentinel nodes, are the first organs of metastasis in several types of cancers. The extent of metastasis in the TDLN is often used in disease staging and prognosis evaluation in cancer patients. Here, we describe the microenvironment of the TDLN and review the recent literature on liposome-based therapies directed to immune cells within the TDLN with the intent to target cancer cells.
Collapse
|
23
|
Zirakzadeh AA, Marits P, Sherif A, Winqvist O. Multiplex B cell characterization in blood, lymph nodes, and tumors from patients with malignancies. THE JOURNAL OF IMMUNOLOGY 2013; 190:5847-55. [PMID: 23630345 DOI: 10.4049/jimmunol.1203279] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
B lymphocytes contribute to immune surveillance, by tumor-specific Abs and Ag presentation to T lymphocytes, but are insufficiently studied in humans. In this article, we report a flow cytometric investigation of B lymphocyte subpopulations in blood, lymph nodes (LNs), and malignant tissues from 20 patients operated on because of advanced solid tumors. The CD19(+) compartment in peripheral blood was essentially unaltered in patients, as compared with healthy control subjects. In metastatic LNs, signs of B lymphocyte activation were observed, as evidenced by increased proportions of plasmablasts and CD86-expressing cells. In tumor-infiltrating B lymphocytes (TIL-B), both switched memory cells and plasmablasts were expanded, as compared with nonmalignant epithelium. Moreover, pronounced skewing of Igλ/Igκ ratio was evident among TIL-Bs. By spectratype analysis on IgH, we confirmed a monoclonal expansion of the Vh7 family in TIL-B, also present in a tumor-associated LN. Sequencing the clonally expanded Vh7 revealed signs of somatic hypermutation. In conclusion, B lymphocytes in cancer patients exhibit signs of activation in tumor-associated tissues, likely induced by recognition of tumor Ags. Increased numbers of switched memory cells and plasmablasts in combination with clonal expansion and signs of somatic hypermutation suggest a CD4(+) T lymphocyte-dependent antitumoral response, which may be exploited for immunotherapy.
Collapse
Affiliation(s)
- A Ali Zirakzadeh
- Department of Medicine, Unit of Translational Immunology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | | | | | | |
Collapse
|
24
|
B cells are critical to T-cell-mediated antitumor immunity induced by a combined immune-stimulatory/conditionally cytotoxic therapy for glioblastoma. Neoplasia 2012; 13:947-60. [PMID: 22028620 DOI: 10.1593/neo.11024] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Revised: 08/19/2011] [Accepted: 08/23/2011] [Indexed: 12/25/2022] Open
Abstract
We have demonstrated that modifying the tumor microenvironment through intratumoral administration of adenoviral vectors (Ad) encoding the conditional cytotoxic molecule, i.e., HSV1-TK and the immune-stimulatory cytokine, i.e., fms-like tyrosine kinase 3 ligand (Flt3L) leads to T-cell-dependent tumor regression in rodent models of glioblastoma. We investigated the role of B cells during immune-mediated glioblastoma multiforme regression. Although treatment with Ad-TK+Ad-Flt3L induced tumor regression in 60% of wild-type (WT) mice, it completely failed in B-cell-deficient Igh6(-/-) mice. Tumor-specific T-cell precursors were detected in Ad-TK+Ad-Flt3L-treated WT mice but not in Igh6(-/-) mice. The treatment also failed in WT mice depleted of total B cells or marginal zone B cells. Because we could not detect circulating antibodies against tumor cells and the treatment was equally efficient in WT mice and in mice with B-cell-specific deletion of Prdm 1 (encoding Blimp-1), in which B cells are present but unable to fully differentiate into antibody-secreting plasma cells, tumor regression in this model is not dependent on B cells' production of tumor antigen-specific immunoglobulins. Instead, B cells seem to play a role as antigen-presenting cells (APCs). Treatment with Ad-TK+Ad-Flt3L led to an increase in the number of B cells in the cervical lymph nodes, which stimulated the proliferation of syngeneic T cells and induced clonal expansion of antitumor T cells. Our data show that B cells act as APCs, playing a critical role in clonal expansion of tumor antigen-specific T cells and brain tumor regression.
Collapse
|
25
|
Ladányi A, Kiss J, Mohos A, Somlai B, Liszkay G, Gilde K, Fejös Z, Gaudi I, Dobos J, Tímár J. Prognostic impact of B-cell density in cutaneous melanoma. Cancer Immunol Immunother 2011; 60:1729-38. [PMID: 21779876 PMCID: PMC11028465 DOI: 10.1007/s00262-011-1071-x] [Citation(s) in RCA: 144] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2011] [Accepted: 07/06/2011] [Indexed: 12/25/2022]
Abstract
Studies on the prognostic importance of tumor-infiltrating lymphocytes have mainly focused on T cells, while little is known about the role of tumor-infiltrating B lymphocytes. We investigated the prevalence of CD20(+) B cells by immunohistochemistry in primary melanoma samples of 106 patients and analyzed in relation to clinicopathological parameters and patients' survival. The majority of samples contained a significant amount of B lymphocytes, predominantly dispersed in the stroma surrounding tumor deposits (mean peritumoral and intratumoral densities: 178.7 ± 156.1 vs. 4.9 ± 6.9 cells/mm², respectively). B cells organized in follicle-like aggregates were also observed in 26% of the samples. B-cell density correlated with that of activated (CD25(+) or OX40(+)) T lymphocytes. Infiltration by CD20(+) lymphocytes did not correlate with tumor thickness, while the presence of B-cell aggregates was observed more frequently in thick melanomas. On the other hand, B-cell infiltration was more pronounced in nonmetastatic or lymph node metastatic tumors, compared to visceral metastatic ones. Accordingly, high number of these cells provided significant survival advantage (P = 0.0391 and P = 0.0136 for intra- and peritumoral infiltration, respectively). Furthermore, combination of peritumoral B-cell density with the number of activated T lymphocytes identified patient subgroups with different disease outcome, which was most favorable in the case of high density, while very poor in the case of low density of both cell types. Multivariate survival analysis identified tumor thickness and CD20(+)/OX40(+) cell density combination as significant independent prognostic factors. Taken together, our results show correlation between low number of CD20(+) B lymphocytes and melanoma progression, indicating a possible role of tumor-infiltrating B cells in antitumoral immune response. It was also reflected in better outcome of the disease since the density of B lymphocytes alone as well as in combination with that of activated T cells proved of prognostic importance in patients with malignant melanoma.
Collapse
Affiliation(s)
- Andrea Ladányi
- Center of Surgical and Molecular Tumor Pathology, National Institute of Oncology, 7-9. Ráth György u., Budapest 1122, Hungary.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Li Q, Lao X, Pan Q, Ning N, Yet J, Xu Y, Li S, Chang AE. Adoptive transfer of tumor reactive B cells confers host T-cell immunity and tumor regression. Clin Cancer Res 2011; 17:4987-95. [PMID: 21690573 DOI: 10.1158/1078-0432.ccr-11-0207] [Citation(s) in RCA: 117] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE We investigated the antitumor reactivity of adoptively transferred effector B cells and the mechanisms by which they may mediate tumor regression in a spontaneous metastases model. EXPERIMENTAL DESIGN 4T1 breast cancer cells were inoculated into the flanks of syngeneic Balb/C mice to prime draining lymph nodes. Tumor-draining lymph nodes (TDLN) were harvested and B cells activated ex vivo with lipopolysaccharide and anti-CD40 monoclonal antibody. These activated B cells were adoptively transferred into mice inoculated with 4T1 tumor in the mammary fat pad. The induction of host T-cell immunity was evaluated. RESULTS Activated 4T1 TDLN B cells secreted immunoglobulin G (IgG) in response to tumor cells which was immunologically specific. These activated B cells were capable of mediating specific lysis of tumor cells in vitro. Transfer of these activated B cells alone mediated the inhibition of spontaneous metastases to the lung. Examination of the host revealed that the transfer of these B cells resulted in the induction of tumor-specific T-cell immunity as measured by cytotoxicity and cytokine (IFNγ and granulocyte-macrophage colony-stimulating factor) production. The combined transfer of activated T and B cells from TDLN resulted in tumor regression, which was greater than either cell population alone, with host B cells capable of producing IgG that mediated lysis of tumor in the presence of complement. CONCLUSIONS We have found that appropriately primed B cells can mediate tumor regression by itself and confers host T-cell antitumor immunity. Furthermore, effector B cells can serve as a useful adjunct in adoptive T-cell therapy.
Collapse
Affiliation(s)
- Qiao Li
- Division of Surgical Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Li Q, Teitz-Tennenbaum S, Donald EJ, Li M, Chang AE. In vivo sensitized and in vitro activated B cells mediate tumor regression in cancer adoptive immunotherapy. THE JOURNAL OF IMMUNOLOGY 2009; 183:3195-203. [PMID: 19667089 DOI: 10.4049/jimmunol.0803773] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Adoptive cellular immunotherapy utilizing tumor-reactive T cells has proven to be a promising strategy for cancer treatment. However, we hypothesize that successful treatment strategies will have to appropriately stimulate not only cellular immunity, but also humoral immunity. We previously reported that B cells in tumor-draining lymph nodes (TDLNs) may function as APCs. In this study, we identified TDLN B cells as effector cells in an adoptive immunotherapy model. In vivo primed and in vitro activated TDLN B cells alone mediated effective (p < 0.05) tumor regression after adoptive transfer into two histologically distinct murine pulmonary metastatic tumor models. Prior lymphodepletion of the host with either chemotherapy or whole-body irradiation augmented the therapeutic efficacy of the adoptively transferred TDLN B cells in the treatment of s.c. tumors as well as metastatic pulmonary tumors. Furthermore, B cell plus T cell transfers resulted in substantially more efficient antitumor responses than B cells or T cells alone (p < 0.05). Activated TDLN B cells conferred strong humoral responses to tumor. This was evident by the production of IgM, IgG, and IgG2b, which bound specifically to tumor cells and led to specific tumor cell lysis in the presence of complement. Collectively, these data indicate that in vivo primed and in vitro activated B cells can be employed as effector cells for cancer therapy. The synergistic antitumor efficacy of cotransferred activated B effector cells and T effector cells represents a novel approach for cancer adoptive immunotherapy.
Collapse
Affiliation(s)
- Qiao Li
- Division of Surgical Oncology, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | | | | | |
Collapse
|
28
|
Yu J, Tian R, Xiu B, Yan J, Jia R, Zhang L, Chang AE, Song H, Li Q. Antitumor activity of T cells generated from lymph nodes draining the SEA-expressing murine B16 melanoma and secondarily activated with dendritic cells. Int J Biol Sci 2009; 5:135-46. [PMID: 19173035 PMCID: PMC2631223 DOI: 10.7150/ijbs.5.135] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Accepted: 01/19/2009] [Indexed: 01/21/2023] Open
Abstract
The successful use of tumor-draining lymph nodes (TDLN) as a source of effector cells for cancer immunotherapy depends largely on the immunogenicity of the tumor drained by the lymph nodes as well as the methods for secondary in vitro T cell activation and expansion. We transferred the bacterial superantigen staphylococcal enterotoxin A (SEA) gene into B16 murine melanoma tumor cells, and used them to induce TDLN (SEA TDLN) in syngeneic hosts. Wild-type (wt) TDLN induced by parental B16 tumor was used as a control. In vitro, SEA TDLN cells proliferated more vigorously, produced more IFNγ and demonstrated higher CTL activity than wt TDLN cells when activated with anti-CD3/anti-CD28/IL-2. In vivo, SEA TDLN cells mediated tumor eradication more effectively than similarly activated wt TDLN cells (p<0.01). Furthermore, use of dendritic cells (DC) plus tumor antigen in vitro in addition to anti-CD3/anti-CD28/IL-2 stimulation further amplified the immune function and therapeutic efficacy of SEA TDLN cells. DC-stimulated SEA TDLN cells eliminated nearly 90% of the pulmonary metastasis in mice bearing established B16 melanoma micrometastases. These results indicate that enforced expression of superantigen SEA in poorly immunogenic tumor cells can enhance their immunogenicity as a vaccine in vivo. The combined use of genetically modified tumor cells as vaccine to induce TDLN followed by secondary stimulation using antigen-presenting cells and tumor antigen in a sequential immunization/activation procedure may represent a unique method to generate more potent effector T cells for adoptive immunotherapy of cancer.
Collapse
Affiliation(s)
- Jiyun Yu
- Institute of Basic Medical Sciences, Academy of Military Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Iuchi T, Teitz-Tennenbaum S, Huang J, Redman BG, Hughes SD, Li M, Jiang G, Chang AE, Li Q. Interleukin-21 augments the efficacy of T-cell therapy by eliciting concurrent cellular and humoral responses. Cancer Res 2008; 68:4431-41. [PMID: 18519706 DOI: 10.1158/0008-5472.can-07-5530] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Interleukin (IL)-21 modulates T-cell-associated, B-cell-associated, and natural killer cell-associated immunity. However, the potential of IL-21 to simultaneously stimulate cellular and humoral antitumor responses and the mechanisms involved have not yet been adequately explored. In this report, we examined the immune-modulating effect of IL-21 when used in vitro and its adjuvant effects when administrated concomitantly with T-cell transfer for cancer therapy. Use of IL-21 in concert with IL-2 in culture up-regulated both type 1 and type 2 cytokine production of activated tumor-draining lymph node cells and enhanced their therapeutic efficacy. Administration of IL-21 and IL-2 as an adjuvant to T-cell transfer resulted in simultaneously elicited cellular and humoral responses. This concurrent response has led to effective regression of established pulmonary metastatic tumors and s.c. tumors. T-cell transfer plus IL-21/IL-2 administration conferred systemic immunity to the treated hosts. This was evident by the induction of protective immunity against tumor rechallenge, expansion of memory T cells, and significantly elevated serum levels of IFN gamma and IL-10. Furthermore, we observed significantly enhanced tumor-associated antibody response after T-cell + IL-2 + IL-21 therapy. Cytotoxic antibody subclass IgG2b increased strikingly in the sera of treated animals; they bound specifically to MCA205 tumor cells, and such immune sera mediated tumor cell lysis in the presence of complement. Use of B-cell-deficient mice provided direct evidence that humoral responses contribute to T-cell + IL-2 + IL-21-elicited antitumor immunity. Collectively, these findings provide a rationale to evaluate the use of IL-21 in T-cell therapy of human cancers.
Collapse
Affiliation(s)
- Takekazu Iuchi
- Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0666, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Genetically engineered T cells expressing a HER2-specific chimeric receptor mediate antigen-specific tumor regression. Cancer Gene Ther 2008; 15:382-92. [PMID: 18292797 DOI: 10.1038/cgt.2008.5] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In this report, we developed a chimeric receptor (N29gamma chR) involving the single chain Fv (scFv) derived from N29 monoclonal antibody (mAb) specific for p185HER2 and characterized the therapeutic efficacy of primary T cells engineered to express N29gamma chR in two histologically distinct murine tumor models. Murine breast (MT901) and fibrosarcoma (MCA207) cancer cell lines were engineered to express human HER2 as targets. Administration of N29gamma chR-expressing T cells eliminated 3-day pulmonary micrometastases of MT901/HER2 and MCA207/HER2 but not parental tumor cells. A 5 to 8-fold increased dose of N29gamma T cells was required to mediate regression of advanced 8-day macrometastases. Exogenous administration of interleukin-2 (IL-2) after N29gamma T-cell transfer was dispensable for treatment of 3-day micrometastases, but was required for mediating regression of well-established 8-day macrometastases. Moreover, fractionated CD8 T cells expressing N29gamma chR suppressed HER2-positive tumor cell growth after adoptive transfer independent of CD4(+) cells. These data indicate that genetically modified T cells expressing a HER2-targeting chimeric receptor can mediate antigen-specific regression of preestablished metastatic cancers in a cell dose-dependent fashion. Systemic administration of IL-2 augments the therapeutic efficacy of these genetically engineered T cells in advanced diseases. These results are relevant to the implication of genetically redirected T cells in clinical cancer immunotherapy.
Collapse
|
31
|
Kroon HM, Li Q, Teitz-Tennenbaum S, Whitfield JR, Noone AM, Chang AE. 4-1BB Costimulation of Effector T Cells for Adoptive Immunotherapy of Cancer: Involvement of Bcl Gene Family Members. J Immunother 2007; 30:406-16. [PMID: 17457215 DOI: 10.1097/cji.0b013e31802eecc6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
We previously reported that in vitro costimulation of murine MCA 205 tumor-draining lymph node (TDLN) cells through a third signal, 4-1BB (CD137), in addition to CD3 and CD28 engagement significantly increases T-cell yield and amplifies antitumor responses in adoptive therapy. The increased T-cell yield seemed to be related to inhibition of activation-induced cell death. In this study, using real time-polymerase chain reaction and intracellular staining, we tested our hypothesis that antiapoptotic Bcl gene members are modulated in 4-1BB ligated TDLN cells. TDLN cells activated through 4-1BB in conjunction with CD3/CD28 demonstrated elevated Bcl-2 and Bcl-xL gene and protein expression compared with CD3/CD28 activation. Furthermore, Bcl-2 and/or Bcl-xL inhibition abrogated 4-1BB-conferred rescue of activation-induced cell death in TDLN cells, and as a result, 4-1BB-enhanced TDLN cell yield was abolished. Congenic mice were used as donors for TDLN cells labeled with CFSE to evaluate proliferation and persistence of activated cells after intravenous adoptive transfer. The effector function of transferred cells was assessed by determining the incidence of interferon-gamma-producing cells in response to tumor stimulation in serial blood samples drawn from treated mice using intracellular cytokine staining. CD28 and CD28/4-1BB costimulation significantly enhanced in vivo proliferation and survival of the infused cells compared with CD3 activation. 4-1BB coligation augmented the proliferation and effector function of the infused cells compared with both CD3 and CD3/CD28-activated cells. Characterizing the function of signaling molecules involved in T-cell activation pathways may allow optimization of conditions in the generation of effector T cells for cancer immunotherapy.
Collapse
Affiliation(s)
- Hidde M Kroon
- Division of Surgical Oncology, Surgery Department, University of Michigan, Ann Arbor, MI 48109-0932, USA
| | | | | | | | | | | |
Collapse
|
32
|
Huang J, Wang Y, Guo J, Lu H, Lin X, Ma L, Teitz-Tennenbaum S, Chang AE, Li Q. Radiation-induced apoptosis along with local and systemic cytokine elaboration is associated with DC plus radiotherapy-mediated renal cell tumor regression. Clin Immunol 2007; 123:298-310. [PMID: 17449328 DOI: 10.1016/j.clim.2007.02.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2006] [Revised: 02/14/2007] [Accepted: 02/16/2007] [Indexed: 02/07/2023]
Abstract
Utilizing melanoma and sarcoma tumor models syngeneic to C57BL/6 mice, we previously reported the antitumor effects of intratumoral (i.t.) administration of dendritic cells (DC) combined with localized radiotherapy (RT). However, the mechanisms underlying the augmented therapeutic effects have yet to be fully defined. Using the BALB/c host, we explored in this study the capacity of RT to augment the therapeutic efficacy of DC in the syngeneic renal cell cancer, Renca. I.t. DC administration combined with RT inhibited tumor growth in a synergistic manner. This extends our previous findings using a different host strain and two histologically distinct tumor models. More importantly, we provide evidence in this report that RT induced significant apoptosis and necrosis in Renca tumor cells, which involved down-regulated expression of Bcl-2 and a concurrent up-regulated expression of Bax. We also found significantly elevated expression of TNFalpha in RT plus DC-treated Renca tumors. Furthermore, splenocytes isolated from DC plus RT-treated mice elaborated higher levels of IL-2, IL-4, IFNgamma and IgG, IgM in response to tumor cells compared with splenocytes from monotherapy-treated hosts. These data support the conclusion that radiotherapy enhanced DC vaccination by inducing tumor cell apoptosis in BABL/c host, and the significantly augmented therapeutic efficacy by RT+DC treatment was associated with an increased local production of TNFalpha as well as an amplified systemic antitumor responses conferred by the combined therapy. I.t. DC administration in concert with localized RT may represent a promising novel regimen for human cancer therapy.
Collapse
Affiliation(s)
- Jianhua Huang
- Department of Immunology, The PLA General Hospital, Beijing, China, and Division of Surgical Oncology, University of Michigan, Ann Arbor 48109-0666, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Peng Z, Chen L, Li R, Li L, Dong YF, Zhang L. Autologous dendritic cells enhance anti-tumor activity of tumor-draining lymph node cells from colonic adenocarcinoma patients in vitro. Shijie Huaren Xiaohua Zazhi 2006; 14:1843-1846. [DOI: 10.11569/wcjd.v14.i18.1843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the enhancement of the anti-tumor activity of tumor-draining lymph node (TDLN) cells induced by autologous dendritic cells (DCs) in vitro.
METHODS: Peripheral blood mononuclear cells (PBMCs) were separated from the patients with colonic adenocarcinoma and then induced with granulocyte-macrophage colony-stimulating factor (GM-CSF), interleukin-4 (IL-4) and TNF-α for maturation. Thereafter, the autologous tumor lysate was used to sensitize DCs. TDLN cells were isolated from regional lymph nodes of the patients, purified, and incubated with interleukin-2 (IL-2). Finally, the TDLN cells were activated by the sensitized DCs or autologous tumor lysate, respectively, and the TDLN cells without activation served as controls. The cytotoxic activity of the activated TDLN cells to colonic adenocarcinoma cell line LS174T and human melanoma cell line A375 were comparatively analyzed.
RESULTS: The cytotoxicities of the DCs-activated TDLN cells to LS174T cells were significantly stronger than those of TDLN cells activated by autologous tumor lysate or without activation (56.13% ± 7.33% vs 42.46% ± 7.68%, 33.50% ± 7.00%, P < 0.001, as the ratio of TDLN cells to tumor cells was 20∶1; 44.85% ± 6.50% vs 30.50% ± 9.17%, 26.75% ± 8.88%, P < 0.001, as the ratio of TDLN cells to tumor cells was 10∶1). However, for A375 cells, the cytotoxic activities had no significant difference among the TDLN cells activated by DCs and autologous tumor lysate, and without activation.
CONCLUSION: The specific anti-tumor activity of TDLN cells can be enhanced by autologous DCs in vitro.
Collapse
|
34
|
Aricò E, Wang E, Tornesello ML, Tagliamonte M, Lewis GK, Marincola FM, Buonaguro FM, Buonaguro L. Immature monocyte derived dendritic cells gene expression profile in response to Virus-Like Particles stimulation. J Transl Med 2005; 3:45. [PMID: 16384534 PMCID: PMC1360684 DOI: 10.1186/1479-5876-3-45] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Accepted: 12/29/2005] [Indexed: 11/10/2022] Open
Abstract
We have recently developed a candidate HIV-1 vaccine model based on HIV-1 Pr55gag Virus-Like Particles (HIV-VLPs), produced in a baculovirus expression system and presenting a gp120 molecule from an Ugandan HIV-1 isolate of the clade A (HIV-VLPAs). The HIV-VLPAs induce in Balb/c mice systemic and mucosal neutralizing Antibodies as well as cytotoxic T lymphocytes, by intra-peritoneal as well as intra-nasal administration. Moreover, we have recently shown that the baculovirus-expressed HIV-VLPs induce maturation and activation of monocyte-derived dendritic cells (MDDCs) which, in turn, produce Th1- and Th2-specific cytokines and stimulate in vitro a primary and secondary response in autologous CD4+ T cells. In the present manuscript, the effects of the baculovirus-expressed HIV-VLPAs on the genomic transcriptional profile of MDDCs obtained from normal healthy donors have been evaluated. The HIV-VLPA stimulation, compared to both PBS and LPS treatment, modulate the expression of genes involved in the morphological and functional changes characterizing the MDDCs activation and maturation. The results of gene profiling analysis here presented are highly informative on the global pattern of gene expression alteration underlying the activation of MDDCs by HIV-VLPAs at the early stages of the immune response and may be extremely helpful for the identification of exclusive activation markers.
Collapse
Affiliation(s)
- Eleonora Aricò
- Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892-1502, USA
- Department of Cell Biolology and Neurosciences, Istituto Superiore di Sanita, Rome, Italy
| | - Ena Wang
- Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892-1502, USA
| | - Maria Lina Tornesello
- Lab. Viral Oncogenesis and Immunotherapies & AIDS Reference Center, Department of Experimental Oncology, Istituto Nazionale Tumori "Fond. G. Pascale", 80131 Napoli, Italy
| | - Maria Tagliamonte
- Lab. Viral Oncogenesis and Immunotherapies & AIDS Reference Center, Department of Experimental Oncology, Istituto Nazionale Tumori "Fond. G. Pascale", 80131 Napoli, Italy
| | - George K Lewis
- Institute of Human Virology, University of Maryland Biotechnology Institute
- Department of Microbiology and Immunology, University of Maryland School of Medicine, University of Maryland Baltimore, Baltimore, MD 21201, USA
| | - Francesco M Marincola
- Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892-1502, USA
| | - Franco M Buonaguro
- Lab. Viral Oncogenesis and Immunotherapies & AIDS Reference Center, Department of Experimental Oncology, Istituto Nazionale Tumori "Fond. G. Pascale", 80131 Napoli, Italy
| | - Luigi Buonaguro
- Lab. Viral Oncogenesis and Immunotherapies & AIDS Reference Center, Department of Experimental Oncology, Istituto Nazionale Tumori "Fond. G. Pascale", 80131 Napoli, Italy
- Institute of Human Virology, University of Maryland Biotechnology Institute
| |
Collapse
|
35
|
Xu Z, Shen SN, Qian XP, Yu LX, Liu BR. Construction and expression of a eukaryotic expression plasmid containing human CD40 ligand in human hepatocellular carcinoma cell line HepG2 and its effect on apoptosis. Shijie Huaren Xiaohua Zazhi 2005; 13:2413-2419. [DOI: 10.11569/wcjd.v13.i20.2413] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To construct and express a eukaryotic expression plasmid containing human CD40 ligand (CD40L) in human hepatocellular carcinoma cell line HepG2 for the biological function study of CD40L on HepG2 cells.
METHODS: Human CD40L cDNA was synthesized by reverse transcription-polymerase chain reaction (RT-PCR) with the specific primers from the RNA of human peripheral blood monocyte (PBMC) and directly ligated into the eukaryotic expression vector pcDNATM3.1/myc-His(-)A through digestion with specific restriction endonuclease. The recombined plasmid was transformed into the E. coli DH5α to amplify CD40L gene. Then HepG2 cells were divided into 4 groups. The cells in group A were transfected with the recombined plasmid, and group B with the blank plasmid (not containing CD40L cDNA). The cells in group C were just normally cultured, and the ones in group D weren't transfected but added G418 as the control for transfection. The expression of CD40L and CD40 were detected by RT-PCR and fluorescent activated cell sorter (FACS) in HepG2 cells, and the apoptosis, cell cycle, as well as Fas expression, were measured by FACS.
RESULTS: The full-length human CD40L cDNA was successfully cloned into the eukaryotic vector pcDNATM3.1/myc-His(-)A. The recombinant plasmid was stably transfected into group A and the rate of CD40L expression was 39.7%. There was no CD40L expression in group B and C. The rates of CD40 expression in group A, B and C were 15.4%, 31.7% and 28.5%, respectively. The apoptotic rate of group A was 45.0±0.3%, but neither group B or C showed obvious apoptosis (P <0.01). Compared with group C, a larger proportion of cells in group A was restrained at G0/G1 phase (90.4±1.3% vs 60.6±1.5%, P <0.01), while the proportions of the cells in S phase (6.32±1.0% vs 12.0±0.7%) and G2/M phase (3.3±0.7% vs 27.3±1.2%) were reduced (both P <0.01). The expression of Fas in group A showed marked increase as compared with that in group B and C (27.8±1.5% vs 3.2±0.8%, 4.2±1.0%, respectively, both P <0.01).
CONCLUSION: The eukaryotic expression plasmid pcDNATM3.1/myc-His(-)A can be stably expressed in HepG2 cell line. CD40L-induced apoptosis of HepG2 is correlated with the increased expression of Fas and cell cycle arrest.
Collapse
|