1
|
Wei J, Li D, Long H, Han M. Immune microenvironment of tumor-draining lymph nodes: insights for immunotherapy. Front Immunol 2025; 16:1562797. [PMID: 40292299 PMCID: PMC12021902 DOI: 10.3389/fimmu.2025.1562797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 03/24/2025] [Indexed: 04/30/2025] Open
Abstract
Tumor-draining lymph nodes (TDLNs) play a crucial role in modulating tumor immune responses and influencing the efficacy of immunotherapy. However, our current understanding of the microenvironment within these lymph nodes remains limited. Tumors not only impair the anti-tumor activity of CD8+ T cells by creating an immunosuppressive microenvironment, but they also facilitate immune evasion and promote metastasis by altering the structure and function of TDLNs. Research has shown that tumor-specific memory CD8+ T cells (TTSM) within TDLNs are essential for the efficacy of immune checkpoint inhibitors, such as PD-1/PD-L1 blockers. Moreover, the abnormal structure of TDLNs, along with the presence of immunosuppressive cells-such as regulatory T cells (Tregs), regulatory B cells (Bregs), and immunosuppressive dendritic cells (DCs)-contributes to tumor-mediated immune evasion. Therefore, gaining a deeper understanding of the immune microenvironment within TDLNs is essential for improving the effectiveness of immunotherapies and developing novel therapeutic strategies. This review explores various TDLN-based therapeutic strategies, addressing the controversies surrounding lymph node dissection, the use of TDLNs as a source of tumor-infiltrating lymphocytes (TILs) for therapy, targeting immunosuppressive cells within TDLNs, and methods to reverse the structural abnormalities of TDLNs. These strategies offer valuable insights and potential directions for advancing tumor immunotherapy.
Collapse
Affiliation(s)
- Jiahuan Wei
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Daozhang Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Haixia Long
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
| | - Mei Han
- Department of Gynecologic Oncology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing, China
| |
Collapse
|
2
|
Naskar S, Sriraman N, Sarkar A, Mahajan N, Sarkar K. Tumor antigen presentation and the associated signal transduction during carcinogenesis. Pathol Res Pract 2024; 261:155485. [PMID: 39088877 DOI: 10.1016/j.prp.2024.155485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 08/03/2024]
Abstract
Numerous developments have been achieved in the study and treatment of cancer throughout the decades that it has been common. After decades of research, about 100 different kinds of cancer have been found, each with unique subgroups within certain organs. This has significantly expanded our understanding of the illness. A mix of genetic, environmental, and behavioral variables contribute to the complicated and diverse process of cancer formation. Mutations, or changes in the DNA sequence, are crucial to the development of cancer. These mutations have the ability to downregulate the expression and function of Major Histocompatibility Complex class I (MHC I) and MHCII receptors, as well as activate oncogenes and inactivate tumor suppressor genes. Cancer cells use this tactic to avoid being recognized by cytotoxic CD8+T lymphocytes, which causes issues with antigen presentation and processing. This review goes into great length into the PI3K pathway, changes to MHC I, and positive impacts of tsMHC-II on disease-free survival and overall survival and the involvement of dendritic cells (DCs) in different tumor microenvironments. The vital functions that the PI3K pathway and its link to the mTOR pathway are highlighted and difficulties in developing effective cancer targeted therapies and feedback systems has also been mentioned, where resistance mechanisms include RAS-mediated oncogenic changes and active PI3K signalling.
Collapse
Affiliation(s)
- Sohom Naskar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nawaneetan Sriraman
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Ankita Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Nitika Mahajan
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Koustav Sarkar
- Department of Biotechnology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India.
| |
Collapse
|
3
|
Lu Q, Kou D, Lou S, Ashrafizadeh M, Aref AR, Canadas I, Tian Y, Niu X, Wang Y, Torabian P, Wang L, Sethi G, Tergaonkar V, Tay F, Yuan Z, Han P. Nanoparticles in tumor microenvironment remodeling and cancer immunotherapy. J Hematol Oncol 2024; 17:16. [PMID: 38566199 PMCID: PMC10986145 DOI: 10.1186/s13045-024-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 03/15/2024] [Indexed: 04/04/2024] Open
Abstract
Cancer immunotherapy and vaccine development have significantly improved the fight against cancers. Despite these advancements, challenges remain, particularly in the clinical delivery of immunomodulatory compounds. The tumor microenvironment (TME), comprising macrophages, fibroblasts, and immune cells, plays a crucial role in immune response modulation. Nanoparticles, engineered to reshape the TME, have shown promising results in enhancing immunotherapy by facilitating targeted delivery and immune modulation. These nanoparticles can suppress fibroblast activation, promote M1 macrophage polarization, aid dendritic cell maturation, and encourage T cell infiltration. Biomimetic nanoparticles further enhance immunotherapy by increasing the internalization of immunomodulatory agents in immune cells such as dendritic cells. Moreover, exosomes, whether naturally secreted by cells in the body or bioengineered, have been explored to regulate the TME and immune-related cells to affect cancer immunotherapy. Stimuli-responsive nanocarriers, activated by pH, redox, and light conditions, exhibit the potential to accelerate immunotherapy. The co-application of nanoparticles with immune checkpoint inhibitors is an emerging strategy to boost anti-tumor immunity. With their ability to induce long-term immunity, nanoarchitectures are promising structures in vaccine development. This review underscores the critical role of nanoparticles in overcoming current challenges and driving the advancement of cancer immunotherapy and TME modification.
Collapse
Affiliation(s)
- Qiang Lu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, China
| | - Dongquan Kou
- Department of Rehabilitation Medicine, Chongqing Public Health Medical Center, Chongqing, China
| | - Shenghan Lou
- Department of Colorectal Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Milad Ashrafizadeh
- Department of General Surgery, Institute of Precision Diagnosis and Treatment of Digestive System Tumors, Carson International Cancer Center, Shenzhen University General Hospital, Shenzhen University, Shenzhen, 518055, Guangdong, China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250000, Shandong, China
| | - Amir Reza Aref
- Xsphera Biosciences, Translational Medicine Group, 6 Tide Street, Boston, MA, 02210, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA
| | - Israel Canadas
- Blood Cell Development and Function Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Yu Tian
- School of Public Health, Benedictine University, Lisle, USA
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC, V6H3Z6, Canada
| | - Pedram Torabian
- Cumming School of Medicine, Arnie Charbonneau Cancer Research Institute, University of Calgary, Calgary, AB, T2N 4Z6, Canada
- Department of Medical Sciences, University of Calgary, Calgary, AB, T2N 4Z6, Canada
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore
| | - Gautam Sethi
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore, 117600, Singapore.
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), 61 Biopolis Drive, Proteos, 138673, Singapore, Republic of Singapore
| | - Franklin Tay
- The Graduate School, Augusta University, 30912, Augusta, GA, USA
| | - Zhennan Yuan
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Peng Han
- Department of Oncology Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
- Key Laboratory of Tumor Immunology in Heilongjiang, Harbin, China.
| |
Collapse
|
4
|
Seaver K, Kourko O, Gee K, Greer PA, Basta S. IL-27 Improves Prophylactic Protection Provided by a Dead Tumor Cell Vaccine in a Mouse Melanoma Model. Front Immunol 2022; 13:884827. [PMID: 35529885 PMCID: PMC9069009 DOI: 10.3389/fimmu.2022.884827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
The protocol used to induce cell death for generating vaccines from whole tumor cells is a critical consideration that impacts vaccine efficacy. Here we compared how different protocols used to induce cell death impacted protection provided by a prophylactic whole tumor cell vaccine in a mouse melanoma model. We found that melanoma cells exposed to γ-irradiation or lysis combined with UV-irradiation (LyUV) provided better protection against tumor challenge than lysis only or cells exposed to UV-irradiation. Furthermore, we found that the immunoregulatory cytokine, IL-27 enhanced protection against tumor growth in a dose-dependent manner when combined with either LyUV or γ-irradiated whole tumor cell vaccine preparations. Taken together, this data supports the use of LyUV as a potential protocol for developing whole tumor cell prophylactic cancer vaccines. We also showed that IL-27 can be used at low doses as a potent adjuvant in combination with LyUV or γ-irradiation treated cancer cells to improve the protection provided by a prophylactic cancer vaccine in a mouse melanoma model.
Collapse
Affiliation(s)
- Kyle Seaver
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Olena Kourko
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Katrina Gee
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
| | - Peter A. Greer
- Department of Pathology and Molecular Medicine, Queen’s University, Kingston, ON, Canada
| | - Sameh Basta
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON, Canada
- *Correspondence: Sameh Basta,
| |
Collapse
|
5
|
The human anti-CD40 agonist antibody mitazalimab (ADC-1013; JNJ-64457107) activates antigen-presenting cells, improves expansion of antigen-specific T cells, and enhances anti-tumor efficacy of a model cancer vaccine in vivo. Cancer Immunol Immunother 2021; 70:3629-3642. [PMID: 33948686 PMCID: PMC8571159 DOI: 10.1007/s00262-021-02932-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/27/2021] [Indexed: 01/10/2023]
Abstract
Non-responders to checkpoint inhibitors generally have low tumor T cell infiltration and could benefit from immunotherapy that activates dendritic cells, with priming of tumor-reactive T cells as a result. Such therapies may be augmented by providing tumor antigen in the form of cancer vaccines. Our aim was to study the effects of mitazalimab (ADC-1013; JNJ-64457107), a human anti-CD40 agonist IgG1 antibody, on activation of antigen-presenting cells, and how this influences the priming and anti-tumor potential of antigen-specific T cells, in mice transgenic for human CD40. Mitazalimab activated splenic CD11c+ MHCII+ dendritic cells and CD19+ MHCII+ B cells within 6 h, with a return to baseline within 1 week. This was associated with a dose-dependent release of proinflammatory cytokines in the blood, including IP-10, MIP-1α and TNF-α. Mitazalimab administered at different dose regimens with ovalbumin protein showed that repeated dosing expanded ovalbumin peptide (SIINFEKL)-specific CD8+ T cells and increased the frequency of activated ICOS+ T cells and CD44hi CD62L- effector memory T cells in the spleen. Mitazalimab prolonged survival of mice bearing MB49 bladder carcinoma tumors and increased the frequency of activated granzyme B+ CD8+ T cells in the tumor. In the ovalbumin-transfected tumor E.G7-OVA lymphoma, mitazalimab administered with either ovalbumin protein or SIINFEKL peptide prolonged the survival of E.G7-OVA tumor-bearing mice, as prophylactic and therapeutic treatment. Thus, mitazalimab activates antigen-presenting cells, which improves expansion and activation of antigen-specific T cells and enhances the anti-tumor efficacy of a model cancer vaccine.
Collapse
|
6
|
McDonnell AM, Currie AJ, Brown M, Kania K, Wylie B, Cleaver A, Lake R, Robinson BWS. Tumor cells, rather than dendritic cells, deliver antigen to the lymph node for cross-presentation. Oncoimmunology 2021; 1:840-846. [PMID: 23162751 PMCID: PMC3489739 DOI: 10.4161/onci.20493] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
It is widely accepted that generation of tumor specific CD8+ T-cell responses occur via cross-priming; however the source of tumor antigen for this event is unknown. We examined the source and form of tumor antigen required for cross-presentation in the local lymph node (LN) using a syngeneic mouse tumor model expressing a marker antigen. We found that cross-presentation of this model tumor antigen in the LN is dependent on continuous traffic of antigen from the tumor site, but without any detectable migration of tumor resident dendritic cells (DCs). Instead, small numbers of tumor cells metastasize to local LNs where they are exposed to a localized CTL attack, resulting in delivery of tumor antigen into the cross-presentation pathway.
Collapse
Affiliation(s)
- Alison M McDonnell
- School of Medicine and Pharmacology; The University of Western Australia; Crawley, Australia ; National Centre for Asbestos Related Diseases; The University of Western Australia; Crawley, Australia
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Mpakali A, Stratikos E. The Role of Antigen Processing and Presentation in Cancer and the Efficacy of Immune Checkpoint Inhibitor Immunotherapy. Cancers (Basel) 2021; 13:E134. [PMID: 33406696 PMCID: PMC7796214 DOI: 10.3390/cancers13010134] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 02/07/2023] Open
Abstract
Recent clinical successes of cancer immunotherapy using immune checkpoint inhibitors (ICIs) are rapidly changing the landscape of cancer treatment. Regardless of initial impressive clinical results though, the therapeutic benefit of ICIs appears to be limited to a subset of patients and tumor types. Recent analyses have revealed that the potency of ICI therapies depends on the efficient presentation of tumor-specific antigens by cancer cells and professional antigen presenting cells. Here, we review current knowledge on the role of antigen presentation in cancer. We focus on intracellular antigen processing and presentation by Major Histocompatibility class I (MHCI) molecules and how it can affect cancer immune evasion. Finally, we discuss the pharmacological tractability of manipulating intracellular antigen processing as a complementary approach to enhance tumor immunogenicity and the effectiveness of ICI immunotherapy.
Collapse
Affiliation(s)
- Anastasia Mpakali
- National Centre for Scientific Research Demokritos, Agia Paraskevi, 15341 Athens, Greece
| | - Efstratios Stratikos
- National Centre for Scientific Research Demokritos, Agia Paraskevi, 15341 Athens, Greece
- Laboratory of Biochemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis Zographou, 15784 Athens, Greece
| |
Collapse
|
8
|
Burn OK, Prasit KK, Hermans IF. Modulating the Tumour Microenvironment by Intratumoural Injection of Pattern Recognition Receptor Agonists. Cancers (Basel) 2020; 12:E3824. [PMID: 33352882 PMCID: PMC7765936 DOI: 10.3390/cancers12123824] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Signalling through pattern recognition receptors (PRRs) leads to strong proinflammatory responses, enhancing the activity of antigen presenting cells and shaping adaptive immune responses against tumour associated antigens. Unfortunately, toxicities associated with systemic administration of these agonists have limited their clinical use to date. Direct injection of PRR agonists into the tumour can enhance immune responses by directly modulating the cells present in the tumour microenvironment. This can improve local antitumour activity, but importantly, also facilitates systemic responses that limit tumour growth at distant sites. As such, this form of therapy could be used clinically where metastatic tumour lesions are accessible, or as neoadjuvant therapy. In this review, we summarise current preclinical data on intratumoural administration of PRR agonists, including new strategies to optimise delivery and impact, and combination studies with current and promising new cancer therapies.
Collapse
Affiliation(s)
- Olivia K. Burn
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6042, New Zealand; (O.K.B.); (K.K.P.)
- Maurice Wilkins Centre, Private Bag 92019, Auckland 1042, New Zealand
| | - Kef K. Prasit
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6042, New Zealand; (O.K.B.); (K.K.P.)
- Maurice Wilkins Centre, Private Bag 92019, Auckland 1042, New Zealand
| | - Ian F. Hermans
- Malaghan Institute of Medical Research, P.O. Box 7060, Wellington 6042, New Zealand; (O.K.B.); (K.K.P.)
- Maurice Wilkins Centre, Private Bag 92019, Auckland 1042, New Zealand
| |
Collapse
|
9
|
Wang L, Zeng W, Wang L, Wang Z, Yin X, Qin Y, Zhang F, Zhang C, Liang W. Naringenin Enhances the Antitumor Effect of Therapeutic Vaccines by Promoting Antigen Cross-Presentation. THE JOURNAL OF IMMUNOLOGY 2020; 204:622-631. [PMID: 31871020 DOI: 10.4049/jimmunol.1900278] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 11/21/2019] [Indexed: 11/19/2022]
Abstract
Dendritic cells (DCs) can internalize and cross-present exogenous Ags to CD8+ T cells for pathogen or tumor cell elimination. Recently, growing evidences suggest the possible immunoregulatory role of flavonoids through modulating the Ag presentation of DCs. In this study, we report that naringenin, a grapefruit-derived flavonoid, possesses the ability to increase the Ag cross-presentation in both murine DC line DC2.4 as well as bone marrow-derived DCs, and naringenin-induced moderate intracellular oxidative stress that contributed to the disruption of lysosomal membrane enhanced Ag leakage to cytosol and cross-presentation. Moreover, in a murine colon adenocarcinoma model, naringenin induced more CD103+ DCs infiltration into tumor and facilitated the activation of CD8+ T cells and strengthened the performance of therapeutic E7 vaccine against TC-1 murine lung cancer. Our investigations may inspire novel thoughts for vaccine design and open a new field of potential applications of flavonoids as immunomodulators to improve host protection against infection and tumor.
Collapse
Affiliation(s)
- Luoyang Wang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China; and.,Department of Chemical Engineering, Tsinghua University, Beijing 100101, China
| | - Wenfeng Zeng
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; .,University of Chinese Academy of Sciences, Beijing 100101, China; and
| | - Luyao Wang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China; and
| | - Zihao Wang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China; and
| | - Xiaozhe Yin
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China; and
| | - Yan Qin
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China; and
| | - Fayun Zhang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China; and
| | - Chunling Zhang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100101, China; and
| | - Wei Liang
- Protein and Peptide Pharmaceutical Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; .,University of Chinese Academy of Sciences, Beijing 100101, China; and
| |
Collapse
|
10
|
Chang H, Zou Z, Wang Q, Li J, Jin H, Yin Q, Xing D. Targeting and Specific Activation of Antigen-Presenting Cells by Endogenous Antigen-Loaded Nanoparticles Elicits Tumor-Specific Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1900069. [PMID: 31921548 PMCID: PMC6947714 DOI: 10.1002/advs.201900069] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 08/03/2019] [Indexed: 05/19/2023]
Abstract
Immunotherapy has shown tremendous promise for improving cancer treatment. Unfortunately, antigen-presenting cells (APCs) in cancer patients cannot effectively recognize and process tumor antigens to activate host immune responses. In this study, an approach is developed to improve cancer immunotherapy that utilizes endogenous antigen-carrying nanoparticles (EAC-NPs), which encompasses a set of antigens isolated from solid tumors and adjuvants. The EAC-NPs specifically target APCs and subsequently result in enhanced T cell responses and improved antitumor efficacy. Mechanistic studies reveal that the EAC-NPs enhance and prolong the presence of immune compounds in APCs, which ensure persistent antigen loading and stimulation, induce a rapid proliferation of CD4+ and CD8+ T cells, and significantly increase the ratios of intratumoral CD4+ T/Treg and CD8+ T/Treg. The work using nanotechnology provides a promising strategy in improving antitumor immunity by enhancing the immunogenicity and presentation of tumor self-antigens for cancer immunotherapy.
Collapse
Affiliation(s)
- Hao‐Cai Chang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Zheng‐Zhi Zou
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Qiu‐Hong Wang
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Jie Li
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Huan Jin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Qian‐Xia Yin
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| | - Da Xing
- MOE Key Laboratory of Laser Life Science & Institute of Laser Life ScienceSouth China Normal UniversityGuangzhou510631China
- College of BiophotonicsSouth China Normal UniversityGuangzhou510631China
| |
Collapse
|
11
|
Neeve SC, Robinson BWS, Fear VS. The role and therapeutic implications of T cells in cancer of the lung. Clin Transl Immunology 2019; 8:e1076. [PMID: 31485330 PMCID: PMC6712517 DOI: 10.1002/cti2.1076] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/05/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022] Open
Abstract
Lung cancer remains the leading cause of cancer-related death worldwide. The disease is classified into two major subtypes, small-cell lung cancer (SCLC) and the more prevalent non-small-cell lung cancer (NSCLC). First-line conventional therapies, such as chemotherapy, radiotherapy and surgery, have offered limited benefit, and patient prognosis remains poor with post-treatment recurrences representing a major cause of morbidity. Consequently, there is an urgent need for improved therapeutic options. Historically, NSCLC has been considered a non-immunogenic disease. However, increased understanding of tumor-immune interactions has challenged this paradigm in both lung and other malignancies, with cancer elimination by tumor-specific T cells increasingly well described in a myriad of solid tumors. Recent evidence has demonstrated that absent or weak anticancer responses are likely a product of tumor-derived immunosuppression. This knowledge, along with a greater appreciation for the role of T cells in lung cancer elimination, has driven development of novel immunotherapeutic approaches which are demonstrating remarkable clinical efficacy. This review examines the role of T cells in lung cancer, discussing the direction and clinical significance of current and future immunotherapeutic strategies.
Collapse
Affiliation(s)
- Samuel C Neeve
- National Centre for Asbestos Related Diseases (NCARD)Lv5 QQ Block (M503)QEII Medical CentreThe University of Western AustraliaPerthWAAustralia
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
| | - Bruce WS Robinson
- National Centre for Asbestos Related Diseases (NCARD)Lv5 QQ Block (M503)QEII Medical CentreThe University of Western AustraliaPerthWAAustralia
- Centre for Respiratory HealthThe University of Western AustraliaPerthWAAustralia
| | - Vanessa S Fear
- National Centre for Asbestos Related Diseases (NCARD)Lv5 QQ Block (M503)QEII Medical CentreThe University of Western AustraliaPerthWAAustralia
- School of Biomedical SciencesThe University of Western AustraliaPerthWAAustralia
- Telethon Kids InstitutePerthWAAustralia
| |
Collapse
|
12
|
Strategies for Targeting Cancer Immunotherapy Through Modulation of the Tumor Microenvironment. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00113-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
13
|
Cellular therapy approaches harnessing the power of the immune system for personalized cancer treatment. Semin Immunol 2019; 42:101306. [DOI: 10.1016/j.smim.2019.101306] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 09/17/2019] [Indexed: 12/30/2022]
|
14
|
Garcia-Bates TM, Palma ML, Shen C, Gambotto A, Macatangay BJC, Ferris RL, Rinaldo CR, Mailliard RB. Contrasting Roles of the PD-1 Signaling Pathway in Dendritic Cell-Mediated Induction and Regulation of HIV-1-Specific Effector T Cell Functions. J Virol 2019; 93:e02035-18. [PMID: 30541848 PMCID: PMC6384070 DOI: 10.1128/jvi.02035-18] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 11/30/2018] [Indexed: 12/25/2022] Open
Abstract
Eliciting highly functional CD8+ cytotoxic T lymphocyte (CTL) responses against a broad range of epitopes will likely be required for immunotherapeutic control of HIV-1 infection. However, the combination of CTL exhaustion and the ability of HIV-1 to rapidly establish CTL escape variants presents major hurdles toward this goal. Our previous work highlighted the use of monocyte-derived, mature, high-interleukin-12 (IL-12)-producing type 1 polarized dendritic cells (MDC1) to selectively induce more potent effector CTLs derived from naive, rather than memory, CD8+ T cell precursors isolated from HIV-1-positive participants in the Multicenter AIDS Cohort Study. In this study, we report that these highly stimulatory antigen-presenting cells also express enhanced levels of the coinhibitory molecule programmed cell death ligand 1 (PD-L1), the ligand for PD-1, which is further upregulated upon subsequent stimulation with the CD4+ T helper cell-derived factor CD40L. Interestingly, blocking the PD-1 signaling pathway during MDC1 induction of HIV-1-specific CTL responses inhibited the priming, activation, and differentiation of naive CD8+ T cells into effector T cells expressing high levels of T-box transcription factor (T-bethi) and eomesodermin (Eomes+). In contrast, PD-1 blockade enhanced the overall magnitude of memory HIV-specific CTL responses and reversed the exhausted memory phenotype from a T-betlow/Eomes+ to a T-bethi/Eomes+ phenotype. These results indicate that the PD-L1/PD-1 signaling pathway has a previously unappreciated dual role in the induction and regulation of HIV-1-specific CTL immunity, which is greatly determined by the context and differentiation stage of the responsive CD8+ T cells.IMPORTANCE Targeting the PD-1/PD-L1 immune checkpoint axis with signaling inhibitors has proven to be a powerful immunotherapeutic strategy to enhance the functional quality and survival of existing antigen-specific effector T cells. However, our study demonstrates that the context and timing of PD-1 signaling in T cells greatly impact the outcome of the effector response. In particular, we show that PD-1 activation plays a positive role during the DC-mediated initiation stage of the primary T cell response, while it serves as an inhibitory mechanism during the effector phase of the response. Therefore, caution should be taken in the design of therapies that include targeting of the PD-1/PD-L1 signaling pathway in order to avoid potential negative impacts on the induction of de novo T cell responses.
Collapse
Affiliation(s)
- Tatiana M Garcia-Bates
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Mariana L Palma
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| | - Chengli Shen
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Andrea Gambotto
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Bernard J C Macatangay
- Department of Medicine, Division of Infectious Diseases, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Charles R Rinaldo
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Robbie B Mailliard
- Department of Infectious Diseases and Microbiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
15
|
Diao J, Gu H, Tang M, Zhao J, Cattral MS. Tumor Dendritic Cells (DCs) Derived from Precursors of Conventional DCs Are Dispensable for Intratumor CTL Responses. THE JOURNAL OF IMMUNOLOGY 2018; 201:1306-1314. [PMID: 29997124 DOI: 10.4049/jimmunol.1701514] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Accepted: 06/05/2018] [Indexed: 12/13/2022]
Abstract
The success of adoptive CTL therapy for cancer depends on interactions between tumor-infiltrating CTLs and cancer cells as well as other cells and molecules in the tumor microenvironment. Tumor dendritic cells (DCs) comprise several subsets: CD103+CD11b- DC1 and CD11b+CD64- DC2, which originate from circulating precursors of conventional DCs, and CD11b+CD64+ DC3, which arise from monocytes. It remains controversial which of these subset(s) promotes intratumor CTL proliferation, expansion, and function. To address this issue, we used the Zbtb46-DTR-transgenic mouse model to selectively deplete DC1 and DC2 from tumors and lymphoid tissues. Wild-type and Zbtb46-DTR bone marrow chimeras were inoculated with B16 melanoma cells that express OVA and were treated with OT-1 CTLs. We found that depletion of DCs derived from precursors of conventional DCs in Zbtb46-DTR bone marrow chimeras abolished CTL proliferation and expansion in tumor-draining lymph nodes. By contrast, intratumor CTL accumulation, proliferation, and IFN-γ expression were unaffected by their absence. We found that adoptive cell therapy increases the frequency of monocyte-derived tumor DC3, which possess the capacity to cross-present tumor Ags and induce CTL proliferation. Our findings support the specialized roles of different DC subsets in the regulation of antitumor CTL responses.
Collapse
Affiliation(s)
- Jun Diao
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2N2, Canada; and
| | - Hongtao Gu
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2N2, Canada; and
| | - Michael Tang
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2N2, Canada; and
| | - Jun Zhao
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2N2, Canada; and
| | - Mark S Cattral
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario M5G 2N2, Canada; and .,Department of Surgery, University of Toronto, Toronto, Ontario M5G 2N2, Canada
| |
Collapse
|
16
|
Vaccine therapy in hematologic malignancies. Blood 2018; 131:2640-2650. [DOI: 10.1182/blood-2017-11-785873] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/04/2018] [Indexed: 02/06/2023] Open
Abstract
Abstract
Immune-based therapy has emerged as a paradigm shift in cancer therapy with dramatic responses observed in previously incurable disease. Cancer vaccines are being developed to disrupt tumor-associated tolerance and activate and selectively expand tumor-specific lymphocytes within the native effector cell repertoire while maintaining immune-regulatory protection against autoimmunity. Although individual antigen approaches result in immune response with a suggestion of clinical effect in some settings, broader efficacy may be dependent on presentation of multiple antigens that capture clonal diversity presented in the context of functionally potent antigen-presenting cells. The use of whole cell–based strategies such as dendritic cell/tumor fusions have yielded provocative results in single-arm studies and are currently being explored in multicenter randomized trials. The posttransplant setting is a potentially promising platform for vaccination due to cytoreduction and relative depletion of inhibitory accessory cells fostering greater immune responsiveness. Integration of these efforts with other immunotherapeutic strategies and agents that target the tumor microenvironment is being studied in an effort to generate durable immunologic responses with clinically meaningful impact on disease.
Collapse
|
17
|
Abstract
Although immunotherapy has been at the forefront of cancer therapy for the last several years, better clinical responses are still desired. Interleukin-33 is perhaps one of the most overlooked antitumor cytokines. Its ability to promote type 1 immune responses, which control tumor growth in preclinical animal models is overshadowed by its association with type 2 immunity and poor prognosis in some human cancers. Accumulating evidence shows that IL-33 is a powerful new tool for restoring and enhancing the body's natural antitumor immunity cycle. Furthermore, the antitumor mechanisms of IL-33 are two-fold, as it can directly boost CD8+ T cell function and restore dendritic cell dysfunction in vivo. Mechanistic studies have identified a novel pathway induced by IL-33 and its receptor ST2 in which dendritic cells avoid dysfunction and retain cross-priming abilities in tumor-bearing conditions. Here, we also comment on IL-33 data in human cancers and explore the idea that endogenous IL-33 may not deserve its reputation for promoting tumor growth. In fact, tumors may hijack the IL-33/ST2 axis to avoid immune surveillance and escape antitumor immunity.
Collapse
Affiliation(s)
- Donye Dominguez
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine–Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Zhang
- Robert H. Lurie Comprehensive Cancer Center, Department of Medicine–Division of Hematology/Oncology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
18
|
|
19
|
Lipid bodies containing oxidatively truncated lipids block antigen cross-presentation by dendritic cells in cancer. Nat Commun 2017; 8:2122. [PMID: 29242535 PMCID: PMC5730553 DOI: 10.1038/s41467-017-02186-9] [Citation(s) in RCA: 206] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 11/08/2017] [Indexed: 01/15/2023] Open
Abstract
Cross-presentation is a critical function of dendritic cells (DCs) required for induction of antitumor immune responses and success of cancer immunotherapy. It is established that tumor-associated DCs are defective in their ability to cross-present antigens. However, the mechanisms driving these defects are still unknown. We find that impaired cross-presentation in DCs is largely associated with defect in trafficking of peptide-MHC class I (pMHC) complexes to the cell surface. DCs in tumor-bearing hosts accumulate lipid bodies (LB) containing electrophilic oxidatively truncated (ox-tr) lipids. These ox-tr-LB, but not LB present in control DCs, covalently bind to chaperone heat shock protein 70. This interaction prevents the translocation of pMHC to cell surface by causing the accumulation of pMHC inside late endosomes/lysosomes. As a result, tumor-associated DCs are no longer able to stimulate adequate CD8 T cells responses. In conclusion, this study demonstrates a mechanism regulating cross-presentation in cancer and suggests potential therapeutic avenues.
Collapse
|
20
|
Grant M, Bollard CM. Developing T-cell therapies for lymphoma without receptor engineering. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2017; 2017:622-631. [PMID: 29222313 PMCID: PMC6142576 DOI: 10.1182/asheducation-2017.1.622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
T-cell therapy has emerged from the bench for the treatment of patients with lymphoma. Responses to T-cell therapeutics are regulated by multiple factors, including the patient's immune system status and disease stage. Outside of engineering of chimeric antigen receptors and artificial T-cell receptors, T-cell therapy can be mediated by ex vivo expansion of antigen-specific T cells targeting viral and/or nonviral tumor-associated antigens. These approaches are contributing to enhanced clinical responses and overall survival. In this review, we summarize the available T-cell therapeutics beyond receptor engineering for the treatment of patients with lymphoma.
Collapse
Affiliation(s)
- Melanie Grant
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, DC; and
| | - Catherine M. Bollard
- Center for Cancer and Immunology Research, Children’s National Health System, Washington, DC; and
- Departments of Pediatrics and Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC
| |
Collapse
|
21
|
Developing T-cell therapies for lymphoma without receptor engineering. Blood Adv 2017; 1:2579-2590. [PMID: 29296911 DOI: 10.1182/bloodadvances.2017009886] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 09/14/2017] [Indexed: 12/19/2022] Open
Abstract
T-cell therapy has emerged from the bench for the treatment of patients with lymphoma. Responses to T-cell therapeutics are regulated by multiple factors, including the patient's immune system status and disease stage. Outside of engineering of chimeric antigen receptors and artificial T-cell receptors, T-cell therapy can be mediated by ex vivo expansion of antigen-specific T cells targeting viral and/or nonviral tumor-associated antigens. These approaches are contributing to enhanced clinical responses and overall survival. In this review, we summarize the available T-cell therapeutics beyond receptor engineering for the treatment of patients with lymphoma.
Collapse
|
22
|
Grant ML, Bollard CM. Cell therapies for hematological malignancies: don't forget non-gene-modified t cells! Blood Rev 2017; 32:203-224. [PMID: 29198753 DOI: 10.1016/j.blre.2017.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 11/13/2017] [Accepted: 11/24/2017] [Indexed: 11/26/2022]
Abstract
Cell therapy currently performs an important role in the treatment of patients with various hematological malignancies. The response to the cell therapy is regulated by multiple factors including the patient's immune system status, genetic profile, stage at diagnosis, age, and underlying disease. Cell therapy that does not require genetic manipulation can be mediated by donor lymphocyte infusion strategies, selective depletion in the post-transplant setting and the ex vivo expansion of antigen-specific T cells. For hematologic malignancies, cell therapy is contributing to enhanced clinical responses and overall survival and the immune response to cell therapy is predictive of response in multiple cancer types. In this review we summarize the available T cell therapeutics that do not rely on gene engineering for the treatment of patients with blood cancers.
Collapse
Affiliation(s)
- Melanie L Grant
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Health System, Washington, DC, USA
| | - Catherine M Bollard
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Health System, Washington, DC, USA; Department of Microbiology, Immunology, and Tropical Medicine, The George Washington University, Washington, DC, USA.
| |
Collapse
|
23
|
Doorduijn EM, Sluijter M, Marijt KA, Querido BJ, van der Burg SH, van Hall T. T cells specific for a TAP-independent self-peptide remain naïve in tumor-bearing mice and are fully exploitable for therapy. Oncoimmunology 2017; 7:e1382793. [PMID: 29399388 PMCID: PMC5790395 DOI: 10.1080/2162402x.2017.1382793] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 09/15/2017] [Accepted: 09/16/2017] [Indexed: 12/20/2022] Open
Abstract
Cancers frequently evade immune-recognition by lowering peptide:MHC-I complexes on their cell surface. Limited peptide supply due to TAP-deficiency results in such MHC-Ilow immune-escape variants. Previously, we reported on a category of TAP-independent self-peptides, called TEIPP, with selective presentation by these tumors. Here we demonstrate that in contrast to T cells specific for conventional tumor antigens, TEIPP-directed T cells remain naïve in mice bearing immune-escaped tumors. This unaffected state was caused by low levels of MHC-I on the tumors and the failure to cross-present low levels of antigenic protein by host APCs. Importantly, increased levels of MHC-I, antigen or co-stimulation resulted in potent activation of TEIPP-specific T cells via direct presentation. Genetic knockdown by CRISPR/Cas9 technology of the relevant MHC-I allele in tumor cells indeed abrogated T cell activation. Vaccine-mediated priming of TEIPP-specific T cells induced efficient homing to MHC-Ilow tumors and subsequently protected mice against outgrowth of their MHC-Ilow tumor. Thus, our data open up the search of TEIPP-specific T cells in cancer patients to explore their application against MHC-Ilow tumor cells.
Collapse
Affiliation(s)
- Elien M Doorduijn
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Marjolein Sluijter
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Koen A Marijt
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Bianca J Querido
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Thorbald van Hall
- Department of Medical Oncology, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| |
Collapse
|
24
|
Cabo M, Offringa R, Zitvogel L, Kroemer G, Muntasell A, Galluzzi L. Trial Watch: Immunostimulatory monoclonal antibodies for oncological indications. Oncoimmunology 2017; 6:e1371896. [PMID: 29209572 PMCID: PMC5706611 DOI: 10.1080/2162402x.2017.1371896] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022] Open
Abstract
The goal of cancer immunotherapy is to establish new or boost pre-existing anticancer immune responses that eradicate malignant cells while generating immunological memory to prevent disease relapse. Over the past few years, immunomodulatory monoclonal antibodies (mAbs) that block co-inhibitory receptors on immune effectors cells - such as cytotoxic T lymphocyte-associated protein 4 (CTLA4), programmed cell death 1 (PDCD1, best known as PD-1) - or their ligands - such as CD274 (best known as PD-L1) - have proven very successful in this sense. As a consequence, many of such immune checkpoint blockers (ICBs) have already entered the clinical practice for various oncological indications. Considerable attention is currently being attracted by a second group of immunomodulatory mAbs, which are conceived to activate co-stimulatory receptors on immune effector cells. Here, we discuss the mechanisms of action of these immunostimulatory mAbs and summarize recent progress in their preclinical and clinical development.
Collapse
Affiliation(s)
- Mariona Cabo
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Rienk Offringa
- Department of General Surgery, Heidelberg University Hospital, Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
- DKFZ-Bayer Joint Immunotherapeutics Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - Laurence Zitvogel
- Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- INSERM, U1015, Villejuif, France
- Center of Clinical Investigations in Biotherapies of Cancer (CICBT) 1428, Villejuif, France
- Université Paris Sud/Paris XI, Le Kremlin-Bicêtre, France
| | - Guido Kroemer
- Université Paris Descartes/Paris V, France
- Université Pierre et Marie Curie/Paris VI, Paris
- Equipe 11 labellisée Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- INSERM, U1138, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
- Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP; Paris, France
| | - Aura Muntasell
- Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Lorenzo Galluzzi
- Université Paris Descartes/Paris V, France
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| |
Collapse
|
25
|
Delivering safer immunotherapies for cancer. Adv Drug Deliv Rev 2017; 114:79-101. [PMID: 28545888 DOI: 10.1016/j.addr.2017.05.011] [Citation(s) in RCA: 221] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2017] [Revised: 05/05/2017] [Accepted: 05/17/2017] [Indexed: 12/14/2022]
Abstract
Cancer immunotherapy is now a powerful clinical reality, with a steady progression of new drug approvals and a massive pipeline of additional treatments in clinical and preclinical development. However, modulation of the immune system can be a double-edged sword: Drugs that activate immune effectors are prone to serious non-specific systemic inflammation and autoimmune side effects. Drug delivery technologies have an important role to play in harnessing the power of immune therapeutics while avoiding on-target/off-tumor toxicities. Here we review mechanisms of toxicity for clinically-relevant immunotherapeutics, and discuss approaches based in drug delivery technology to enhance the safety and potency of these treatments. These include strategies to merge drug delivery with adoptive cellular therapies, targeting immunotherapies to tumors or select immune cells, and localizing therapeutics intratumorally. Rational design employing lessons learned from the drug delivery and nanomedicine fields has the potential to facilitate immunotherapy reaching its full potential.
Collapse
|
26
|
Achkar T, Tarhini AA. The use of immunotherapy in the treatment of melanoma. J Hematol Oncol 2017; 10:88. [PMID: 28434398 PMCID: PMC5402170 DOI: 10.1186/s13045-017-0458-3] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 03/29/2017] [Indexed: 11/13/2022] Open
Abstract
Patients with advanced melanoma have a compromised anti-tumor immune response leading to tumor immune tolerance and a tumor microenvironment conducive to disease progression. Immunotherapy that successfully overcomes this tumor-mediated immune suppression has made the greatest impact in the management of this disease over the past few years. This progress through immunotherapy builds upon earlier successes that interferon-α had in the treatment of melanoma in the adjuvant setting, as well as that of high-dose interleukin-2 in advanced melanoma. The development of immune checkpoint inhibitors has led to dramatic clinical activity in advanced melanoma. In particular, anti-CTLA4 and anti-PD1 monoclonal antibodies have taken us forward into the realm of longer survival and durable responses with the possibility of cure in a continuously increasing proportion of patients. Combination immunotherapeutic strategies and novel immunotherapeutic agents are being tested at an accelerated pace where the outlook for long-term survival benefits for the majority of patients appears brighter than ever.
Collapse
Affiliation(s)
- Tala Achkar
- University of Pittsburgh, Pittsburgh, PA, USA.,University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Room 555, Pittsburgh, PA, 15232, USA
| | - Ahmad A Tarhini
- University of Pittsburgh, Pittsburgh, PA, USA. .,University of Pittsburgh Cancer Institute, 5150 Centre Avenue, Room 555, Pittsburgh, PA, 15232, USA.
| |
Collapse
|
27
|
Tang M, Diao J, Cattral MS. Molecular mechanisms involved in dendritic cell dysfunction in cancer. Cell Mol Life Sci 2017; 74:761-776. [PMID: 27491428 PMCID: PMC11107728 DOI: 10.1007/s00018-016-2317-8] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 07/15/2016] [Accepted: 07/21/2016] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DC) play a pivotal role in the tumor microenvironment (TME). As the primary antigen-presenting cells in the tumor, DCs modulate anti-tumor responses by regulating the magnitude and duration of infiltrating cytotoxic T lymphocyte responses. Unfortunately, due to the immunosuppressive nature of the TME, as well as the inherent plasticity of DCs, tumor DCs are often dysfunctional, a phenomenon that contributes to immune evasion. Recent progresses in our understanding of tumor DC biology have revealed potential molecular targets that allow us to improve tumor DC immunogenicity and cancer immunotherapy. Here, we review the molecular mechanisms that drive tumor DC dysfunction. We discuss recent advances in our understanding of tumor DC ontogeny, tumor DC subset heterogeneity, and factors in the tumor microenvironment that affect DC recruitment, differentiation, and function. Finally, we describe potential strategies to optimize tumor DC function in the context of cancer therapy.
Collapse
Affiliation(s)
- Michael Tang
- Toronto General Hospital Research Institute, University Health Network, Peter Munk Building, 11-173, 585 University Ave., Toronto, ON, M5G 2N2, Canada
| | - Jun Diao
- Toronto General Hospital Research Institute, University Health Network, Peter Munk Building, 11-173, 585 University Ave., Toronto, ON, M5G 2N2, Canada
| | - Mark S Cattral
- Toronto General Hospital Research Institute, University Health Network, Peter Munk Building, 11-173, 585 University Ave., Toronto, ON, M5G 2N2, Canada.
- Department of Surgery, University of Toronto, Toronto, ON, M5G 2N2, Canada.
| |
Collapse
|
28
|
Dominguez D, Ye C, Geng Z, Chen S, Fan J, Qin L, Long A, Wang L, Zhang Z, Zhang Y, Fang D, Kuzel TM, Zhang B. Exogenous IL-33 Restores Dendritic Cell Activation and Maturation in Established Cancer. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2017; 198:1365-1375. [PMID: 28011934 PMCID: PMC5263113 DOI: 10.4049/jimmunol.1501399] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 11/22/2016] [Indexed: 12/16/2022]
Abstract
The role of IL-33, particularly in tumor growth and tumor immunity, remains ill-defined. We show that exogenous IL-33 can induce robust antitumor effect through a CD8+ T cell-dependent mechanism. Systemic administration of rIL-33 alone was sufficient to inhibit growth of established tumors in transplant and de novo melanoma tumorigenesis models. Notably, in addition to a direct action on CD8+ T cell expansion and IFN-γ production, rIL-33 therapy activated myeloid dendritic cells (mDCs) in tumor-bearing mice, restored antitumor T cell activity, and increased Ag cross-presentation within the tumor microenvironment. Furthermore, combination therapy consisting of rIL-33 and agonistic anti-CD40 Abs demonstrated synergistic antitumor activity. Specifically, MyD88, an essential component of the IL-33 signaling pathway, was required for the IL-33-mediated increase in mDC number and upregulation in expression of costimulatory molecules. Importantly, we identified that the IL-33 receptor ST2, MyD88, and STAT1 cooperate to induce costimulatory molecule expression on mDCs in response to rIL-33. Thus, our study revealed a novel IL-33-ST2-MyD88-STAT1 axis that restores mDC activation and maturation in established cancer and, thereby, the magnitude of antitumor immune responses, suggesting a potential use of rIL-33 as a new immunotherapy option to treat established cancer.
Collapse
Affiliation(s)
- Donye Dominguez
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Cong Ye
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Zhe Geng
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Siqi Chen
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Jie Fan
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Lei Qin
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Alan Long
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Long Wang
- Cancer Therapy and Research Center, Department of Medicine, University of Texas Health Science Center, San Antonio, TX 78229
| | - Zhuoli Zhang
- Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Yi Zhang
- Biotherapy Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China; and
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Timothy M Kuzel
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Bin Zhang
- Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611;
| |
Collapse
|
29
|
The immunotherapy era of myeloma: monoclonal antibodies, vaccines, and adoptive T-cell therapies. Blood 2016; 128:1679-87. [DOI: 10.1182/blood-2016-05-636357] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/17/2016] [Indexed: 01/06/2023] Open
Abstract
Abstract
The treatment of multiple myeloma has evolved significantly over the last decades from primarily alkylator-based chemotherapeutic agents with minimal efficacy to the introduction of more effective agents including immune modulators and proteasome inhibitors, which have changed the landscape of therapy for this disease. We are now entering a new era that will increasingly integrate immunotherapy into standard treatment. This review discusses the current immune-based strategies currently approved, as well as various immune approaches being actively investigated including monoclonal antibodies, checkpoint inhibitors, vaccines, and adoptive T-cell therapies.
Collapse
|
30
|
Aldarouish M, Wang C. Trends and advances in tumor immunology and lung cancer immunotherapy. J Exp Clin Cancer Res 2016; 35:157. [PMID: 27686848 PMCID: PMC5043622 DOI: 10.1186/s13046-016-0439-3] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/24/2016] [Indexed: 02/06/2023] Open
Abstract
Among several types of tumor, lung cancer is considered one of the most fatal and still the main cause of cancer-related deaths. Although chemotherapeutic agents can improve survival and quality of life compared with symptomatic treatment, cancers usually still progress after chemotherapy and are often aggravated by serious side effects. In the last few years there has been a growing interest in immunotherapy for lung cancer based on promising preliminary results in achieving meaningful and durable treatments responses with minimal manageable toxicity. This article is divided into two parts, the first part discusses the role of human immune system in controlling and eradicating cancer and the mechanisms of immune response evasion by tumor. The second part reviews the recent progress made in immunotherapy for lung cancer with results from trials evaluating therapeutic vaccines in addition to immune checkpoint blockade, specifically cytotoxic T lymphocyte associated protein 4, programmed death receptor 1 pathway, using monoclonal antibodies.
Collapse
Affiliation(s)
- Mohanad Aldarouish
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu Province, People's Republic of China.
| | - Cailian Wang
- Department of Oncology, Zhongda Hospital, School of Medicine, Southeast University, 87 Dingjiaqiao Rd, Nanjing, Jiangsu Province, People's Republic of China.
| |
Collapse
|
31
|
Abstract
The central nervous system (CNS) possesses powerful local and global immunosuppressive capabilities that modulate unwanted inflammatory reactions in nervous tissue. These same immune-modulatory mechanisms are also co-opted by malignant brain tumors and pose a formidable challenge to brain tumor immunotherapy. Routes by which malignant gliomas coordinate immunosuppression include the mechanical and functional barriers of the CNS; immunosuppressive cytokines and catabolites; immune checkpoint molecules; tumor-infiltrating immune cells; and suppressor immune cells. The challenges to overcoming tumor-induced immunosuppression, however, are not unique to the brain, and several analogous immunosuppressive mechanisms also exist for primary tumors outside of the CNS. Ultimately, the immune responses in the CNS are linked and complementary to immune processes in the periphery, and advances in tumor immunotherapy in peripheral sites may therefore illuminate novel approaches to brain tumor immunotherapy, and vice versa.
Collapse
Affiliation(s)
- Powell Perng
- Department of Neurosurgery, School of Medicine, Johns Hopkins University , Baltimore, MD , USA
| | - Michael Lim
- Department of Neurosurgery, School of Medicine, Johns Hopkins University , Baltimore, MD , USA
| |
Collapse
|
32
|
Chen P, Luo S, Wen YJ, Li YH, Li J, Wang YS, Du LC, Zhang P, Tang J, Yang DB, Hu HZ, Zhao X, Wei YQ. Low-dose paclitaxel improves the therapeutic efficacy of recombinant adenovirus encoding CCL21 chemokine against murine cancer. Cancer Sci 2015; 105:1393-401. [PMID: 25230206 PMCID: PMC4462366 DOI: 10.1111/cas.12537] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 02/05/2023] Open
Abstract
Secondary lymphoid tissue chemokine (SLC/CCL21), one of the CC chemokines, exerts potent antitumor immunity by co-localizing T cells and dendritic cells at the tumor site and is currently tested against human solid tumors. Here, we investigated whether the combination of recombinant adenovirus encoding murine CCL21 (Ad-mCCL21) with low-dose paclitaxel would improve therapeutic efficacy against murine cancer. Immunocompetent mice bearing B16-F10 melanoma or 4T1 breast carcinoma were treated with either Ad-mCCL21, paclitaxel, or both agents together. Our results showed that Ad-mCCL21 + low-dose paclitaxel more effectively reduced the growth of tumors as compared with either treatment alone and significantly prolonged survival time of the tumor-bearing animals. These antitumor effects of the combined therapy were linked to altered cytokine network at the tumor site, enhanced apoptosis of tumor cells, and decreased formation of new vessels in tumors. Importantly, the combined therapy elicited a strong therapeutic antitumor immunity, which could be partly abrogated by the depletion of CD4+ or CD8+ T lymphocytes. Collectively, these preclinical evaluations may provide a combined strategy for antitumor immunity and should be considered for testing in clinical trials.
Collapse
Affiliation(s)
- Ping Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, China; National Institutes for Food and Drug Control, Beijing, China; Chengdu Institute of Biological Products Co., Ltd, Chengdu, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The quality of the host immune response in patients with advanced melanoma is compromised with a bias towards Th2-type polarization and a tumor microenvironment that facilitates disease progression. Overcoming tumor-induced immune suppression through strategies that build upon the immunomodulatory qualities and clinical activity of interferon-α as demonstrated in the melanoma adjuvant setting is a major clinical need. The recent advances in the field of immune checkpoint modulation and the unprecedented clinical activity in advanced melanoma opens the door on novel combinations that may overcome tumor tolerogenic mechanisms that are known to suppress the potent anti-tumor impact of interferon (IFN)-α. Promising preliminary data suggest that such combinations may move the clinical management of advanced melanoma into the next level, beyond what is currently seen with immune checkpoint blockers alone.
Collapse
Affiliation(s)
- Imran Rafique
- University of Pittsburgh Medical Center, Pittsburgh, PA
| | - John M Kirkwood
- University of Pittsburgh Medical Center, Pittsburgh, PA; University of Pittsburgh Cancer Institute, Pittsburgh, PA
| | - Ahmad A Tarhini
- University of Pittsburgh Medical Center, Pittsburgh, PA; University of Pittsburgh Cancer Institute, Pittsburgh, PA.
| |
Collapse
|
34
|
Shao K, Singha S, Clemente-Casares X, Tsai S, Yang Y, Santamaria P. Nanoparticle-based immunotherapy for cancer. ACS NANO 2015; 9:16-30. [PMID: 25469470 DOI: 10.1021/nn5062029] [Citation(s) in RCA: 333] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The design of nanovaccines capable of triggering effective antitumor immunity requires an understanding of how the immune system senses and responds to threats, including pathogens and tumors. Equally important is an understanding of the mechanisms employed by tumor cells to evade immunity and an appreciation of the deleterious effects that antitumor immune responses can have on tumor growth, such as by skewing tumor cell composition toward immunologically silent tumor cell variants. The immune system and tumors engage in a tug-of-war driven by competition where promoting antitumor immunity or tumor cell death alone may be therapeutically insufficient. Nanotechnology affords a unique opportunity to develop therapeutic compounds than can simultaneously tackle both aspects, favoring tumor eradication. Here, we review the current status of nanoparticle-based immunotherapeutic strategies for the treatment of cancer, ranging from antigen/adjuvant delivery vehicles (to professional antigen-presenting cell types of the immune system) to direct tumor antigen-specific T-lymphocyte-targeting compounds and their combinations thereof.
Collapse
Affiliation(s)
- Kun Shao
- Julia McFarlane Diabetes Research Centre (JMDRC) and Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases and Hotchkiss Brain Institute, Cummings School of Medicine, University of Calgary , Calgary, Alberta T2N 4N1 Canada
| | | | | | | | | | | |
Collapse
|
35
|
McDonnell AM, Lesterhuis WJ, Khong A, Nowak AK, Lake RA, Currie AJ, Robinson BWS. Tumor-infiltrating dendritic cells exhibit defective cross-presentation of tumor antigens, but is reversed by chemotherapy. Eur J Immunol 2014; 45:49-59. [PMID: 25316312 DOI: 10.1002/eji.201444722] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Revised: 09/19/2014] [Accepted: 10/10/2014] [Indexed: 11/11/2022]
Abstract
Cross-presentation defines the unique capacity of an APC to present exogenous Ag via MHC class I molecules to CD8(+) T cells. DCs are specialized cross-presenting cells and as such have a critical role in antitumor immunity. DCs are routinely found within the tumor microenvironment, but their capacity for endogenous or therapeutically enhanced cross-presentation is not well characterized. In this study, we examined the tumor and lymph node DC cross-presentation of a nominal marker tumor Ag, HA, expressed by the murine mesothelioma tumor AB1-HA. We found that tumors were infiltrated by predominantly CD11b(+) DCs with a semimature phenotype that could not cross-present tumor Ag, and therefore, were unable to induce tumor-specific T-cell activation or proliferation. Although tumor-infiltrating DCs were able to take up, process, and cross-present exogenous cell-bound and soluble Ags, this was significantly impaired relative to lymph node DCs. Importantly, however, systemic chemotherapy using gemcitabine reversed the defect in Ag cross-presentation of tumor DCs. These data demonstrate that DC cross-presentation within the tumor microenvironment is defective, but can be reversed by chemotherapy. These results have important implications for anticancer therapy, particularly regarding the use of immunotherapy in conjunction with cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Alison M McDonnell
- National Centre for Asbestos Related Diseases, School of Medicine and Pharmacology, The University of Western Australia, Nedlands, WA, Australia
| | | | | | | | | | | | | |
Collapse
|
36
|
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: 4.4] [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
|
37
|
Abstract
Signal peptide (SP) domains have a common motif but also sequence specific features. This knowledge was mainly ignored by immunologists who considered SP as generic, short-lived, targeting sequences. Consequently, while SP-derived MHC class I, class II and HLA-E epitopes have been isolated, their use as antigen-specific vaccine candidates (VCs) was mostly neglected. Recently we demonstrated the rational of selecting entire SP domains as multi-epitope long peptide VCs based on their high T and B-cell epitope densities. This review summarizes preclinical and clinical results demonstrating the various advantages of human SP domain VCs derived from both bacterial and tumor antigens. Such vaccine design provides for a straightforward, yet unique immunotherapeutic means of generating robust, non-toxic, diversified, combined antigen-specific CD4+/CD8+ T/B-cell immunity, irrespective of patient HLA repertoire also in disease associated transporter-associated with antigen processing (TAP) deficiencies. Subsequent clinical trials will further assess the full potential of this approach.
Collapse
Key Words
- ADCC, antibody-dependent cell-mediated cytotoxicity
- AE, adverse events
- APC, antigen presenting cells
- DC, dendritic cells
- ER, endoplasmic reticulum
- ImMucin
- LP, long peptide
- MHC
- MHC, major histocompatibility complex
- MM, multiple myeloma
- MUC1
- PBMC, peripheral blood mononuclear cells
- SP, signal peptide
- SPP, signal peptide peptidase
- SPase, signal peptidase
- T-cell
- TAA, tumor associated antigen
- TAP, transporter-associated with antigen processing
- VC, vaccine candidate
- antibodies
- cancer
- hGM-CSF, human granulocyte-macrophage colony-stimulating factor
- long peptide
- signal peptide
- tuberculosis
- vaccine
Collapse
|
38
|
Kovjazin R, Carmon L. The use of signal peptide domains as vaccine candidates. Hum Vaccin Immunother 2014. [DOI: 10.4161/hv.29549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|
39
|
Bossi G, Buisson S, Oates J, Jakobsen BK, Hassan NJ. ImmTAC-redirected tumour cell killing induces and potentiates antigen cross-presentation by dendritic cells. Cancer Immunol Immunother 2014; 63:437-48. [PMID: 24531387 PMCID: PMC11029007 DOI: 10.1007/s00262-014-1525-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 01/30/2014] [Indexed: 10/25/2022]
Abstract
Antigen cross-presentation by dendritic cells (DCs) is thought to play a critical role in driving a polyclonal and durable T cell response against cancer. It follows, therefore, that the capacity of emerging immunotherapeutic agents to orchestrate tumour eradication may depend on their ability to induce antigen cross-presentation. ImmTACs [immune-mobilising monoclonal TCRs (T cell receptors) against cancer] are a new class of soluble bi-specific anti-cancer agents that combine pico-molar affinity TCR-based antigen recognition with T cell activation via a CD3-specific antibody fragment. ImmTACs specifically recognise human leucocyte antigen (HLA)-restricted tumour-associated antigens, presented by cancer cells, leading to T cell redirection and a potent anti-tumour response. Using an ImmTAC specific for a HLA-A*02-restricted peptide derived from the melanoma antigen gp100 (termed IMCgp100), we here observe that ImmTAC-driven melanoma-cell death leads to cross-presentation of melanoma antigens by DCs. These, in turn, can activate both melanoma-specific T cells and polyclonal T cells redirected by IMCgp100. Moreover, activation of melanoma-specific T cells by cross-presenting DCs is enhanced in the presence of IMCgp100; a feature that serves to increase the prospect of breaking tolerance in the tumour microenvironment. The mechanism of DC cross-presentation occurs via 'cross-dressing' which involves the rapid and direct capture by DCs of membrane fragments from dying tumour cells. DC cross-presentation of gp100-peptide-HLA complexes was visualised and quantified using a fluorescently labelled soluble TCR. These data demonstrate how ImmTACs engage with the innate and adaptive components of the immune system enhancing the prospect of mediating an effective and durable anti-tumour response in patients.
Collapse
Affiliation(s)
- Giovanna Bossi
- Immunocore Ltd, 57 Jubilee Avenue Milton Park, Abingdon, Oxon OX14 4RX UK
| | - Sandrine Buisson
- Immunocore Ltd, 57 Jubilee Avenue Milton Park, Abingdon, Oxon OX14 4RX UK
| | - Joanne Oates
- Immunocore Ltd, 57 Jubilee Avenue Milton Park, Abingdon, Oxon OX14 4RX UK
| | - Bent K. Jakobsen
- Immunocore Ltd, 57 Jubilee Avenue Milton Park, Abingdon, Oxon OX14 4RX UK
| | - Namir J. Hassan
- Immunocore Ltd, 57 Jubilee Avenue Milton Park, Abingdon, Oxon OX14 4RX UK
| |
Collapse
|
40
|
James BR, Brincks EL, Kucaba TA, Boon L, Griffith TS. Effective TRAIL-based immunotherapy requires both plasmacytoid and CD8α dendritic cells. Cancer Immunol Immunother 2014; 63:685-97. [PMID: 24711083 DOI: 10.1007/s00262-014-1548-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2013] [Accepted: 03/25/2014] [Indexed: 11/26/2022]
Abstract
It is now appreciated that there are distinct subsets of dendritic cells (DC) with specialized functions. Plasmacytoid DC (pDC) and CD8α DC can contribute to the priming, activation and function of antitumor CD8 T cells; however, their specific roles and necessity in stimulating antitumor immunity are not clearly understood. We examined the importance of pDC and CD8α DC during immunotherapy of an orthotopic model of metastatic renal cell carcinoma. Immunotherapy that utilizes a recombinant adenovirus encoding tumor necrosis factor-related apoptosis-inducing ligand (Ad5-TRAIL) in combination with an immunostimulatory CpG-containing oligodeoxynucleotide (CpG) resulted in the clearance of primary and metastatic tumors in wild-type (WT) replete BALB/c mice and prolonged survival. In comparison, mice deficient in either pDC (accomplished using a depleting mAb specific for PDCA1) or CD8α DC (through utilization of CD8α DC-deficient Batf3(-/-) BALB/c mice) had uncontrolled tumor growth and high mortality after Ad5-TRAIL/CpG administration. The ineffectiveness of Ad5-TRAIL/CpG therapy in the anti-PDCA1-treated and Batf3(-/-) BALB/c mice was marked by an altered activation phenotype of the DC, as well as significantly reduced expression of type I IFN-stimulated genes and IL-15/IL-15R complex production. In addition, pDC-depleted and Batf3(-/-) BALB/c mice had significantly decreased effector CD8 T cell infiltration in the primary tumor site compared with WT mice after therapy. These data collectively suggest that pDC and CD8α DC carry out independent, but complementary, roles that are necessary to initiate an efficacious antitumor immune response after Ad5-TRAIL/CpG therapy.
Collapse
Affiliation(s)
- Britnie R James
- Department of Urology, University of Minnesota, 3-125 CCRB, 2231 6th St. SE, Minneapolis, MN, 55455, USA
| | | | | | | | | |
Collapse
|
41
|
Ramakrishnan R, Tyurin VA, Tuyrin VA, Veglia F, Condamine T, Amoscato A, Mohammadyani D, Johnson JJ, Zhang LM, Klein-Seetharaman J, Celis E, Kagan VE, Gabrilovich DI. Oxidized lipids block antigen cross-presentation by dendritic cells in cancer. THE JOURNAL OF IMMUNOLOGY 2014; 192:2920-31. [PMID: 24554775 DOI: 10.4049/jimmunol.1302801] [Citation(s) in RCA: 213] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Cross-presentation is one of the main features of dendritic cells (DCs), which is critically important for the development of spontaneous and therapy-inducible antitumor immune responses. Patients, at early stages of cancer, have normal presence of DCs. However, the difficulties in the development of antitumor responses in patients with low tumor burden raised the question of the mechanisms of DC dysfunction. In this study, we found that, in differentiated DCs, tumor-derived factors blocked the cross-presentation of exogenous Ags without inhibiting the Ag presentation of endogenous protein or peptides. This effect was caused by intracellular accumulation of different types of oxidized neutral lipids: triglycerides, cholesterol esters, and fatty acids. In contrast, the accumulation of nonoxidized lipids did not affect cross-presentation. Oxidized lipids blocked cross-presentation by reducing the expression of peptide-MHC class I complexes on the cell surface. Thus, this study suggests the novel role of oxidized lipids in the regulation of cross-presentation.
Collapse
Affiliation(s)
- Rupal Ramakrishnan
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Fraser CK, Brown MP, Diener KR, Hayball JD. Unravelling the complexity of cancer–immune system interplay. Expert Rev Anticancer Ther 2014; 10:917-34. [DOI: 10.1586/era.10.66] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
43
|
Tesone AJ, Svoronos N, Allegrezza MJ, Conejo-Garcia JR. Pathological mobilization and activities of dendritic cells in tumor-bearing hosts: challenges and opportunities for immunotherapy of cancer. Front Immunol 2013; 4:435. [PMID: 24339824 PMCID: PMC3857526 DOI: 10.3389/fimmu.2013.00435] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Accepted: 11/22/2013] [Indexed: 12/22/2022] Open
Abstract
A common characteristic of solid tumors is the pathological recruitment of immunosuppressive myeloid cells, which in certain tumors includes dendritic cells (DCs). DCs are of particular interest in the field of cancer immunotherapy because they induce potent and highly specific anti-tumor immune responses, particularly in the early phase of tumorigenesis. However, as tumors progress, these cells can be transformed into regulatory cells that contribute to an immunosuppressive microenvironment favoring tumor growth. Therefore, controlling DC phenotype has the potential to elicit effective anti-tumor responses while simultaneously weakening the tumor’s ability to protect itself from immune attack. This review focuses on the dual nature of DCs in the tumor microenvironment, the regulation of DC phenotype, and the prospect of modifying DCs in situ as a novel immunotherapeutic approach.
Collapse
Affiliation(s)
- Amelia J Tesone
- Tumor Microenvironment and Metastasis Program, Wistar Institute , Philadelphia, PA , USA
| | | | | | | |
Collapse
|
44
|
Heinrich B, Goepfert K, Delic M, Galle PR, Moehler M. Influence of the oncolytic parvovirus H-1, CTLA-4 antibody tremelimumab and cytostatic drugs on the human immune system in a human in vitro model of colorectal cancer cells. Onco Targets Ther 2013; 6:1119-27. [PMID: 23986643 PMCID: PMC3754820 DOI: 10.2147/ott.s49371] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Introduction Tumor-directed and immune-system-stimulating therapies are of special interest in cancer treatment. Here, we demonstrate the potential of parvovirus H-1 (H-1PV) to efficiently kill colorectal cancer cells and induce immunogenicity of colorectal tumors by inducing maturation of dendritic cells (DCs) alone and also in combination with cytostatic drugs in vitro. Using our cell culture model, we have additionally investigated the effects of anti-CTLA-4 (cytotoxic T-lymphocyte-associated antigen 4) receptor antibody tremelimumab on this process. Materials and methods Colon carcinoma cell lines were treated with different concentrations of cytostatic drugs or tremelimumab or were infected with H-1PV in different multiplicities of infection (MOIs), and viability was determined using MTT assays. Expression of CTLA-4 in colon carcinoma cell lines was measured by FACScan™. For the coculture model, we isolated monocytes using adherence, and differentiation into immature DCs (iDCs) was stimulated using interleukin-4 and granulocyte-macrophage colony-stimulating factor. Maturation of iDCs into mature DCs (mDCs) was induced by a cytokine cocktail. SW480 colon carcinoma cells were infected with H-1PV or treated with cytostatic drugs. Drug treated and H-1PV-infected SW480 colon carcinoma cells were cocultured with iDCs and expression of maturation markers was measured using FACScan™. Cytokine measurements were performed using enzyme-linked immunosorbent assay. Results Colon carcinoma cells SW480 were potently infected and killed by H-1PV. CTLA-4 expression in SW480 cells increased after infection with H-1PV and also after treatment with cytostatic drugs. Tremelimumab had no influence on viability of the colon carcinoma cell line. There was no maturation of iDCs after coculture with SW480; instead, H-1PV-infected or drug pretreated SW480 induced maturation. Cytokine production was higher for H-1PV-infected cells but was not significantly enhanced by tremelimumab treatment alone or in combination. Addition of tremelimumab did not interfere with the maturation process as measured by markers of maturation as well as by determination of cytokine levels. Conclusion By enhancing both cell death and immunogenicity of tumors, H-1PV is of special interest for tumor-directed therapy. These features make it a promising candidate for clinical application in human colorectal cancer. As tremelimumab does not significantly interfere with this process, an interesting therapeutic combination of active enhancement of tumor immunogenicity and independent masking of the CTLA-4 silencing process on tumor cells is highlighted.
Collapse
Affiliation(s)
- Bernd Heinrich
- University Medical Center of the Johannes Gutenberg University Mainz, 1st Department of Internal Medicine, Langenbeckstrasse, Mainz, Germany
| | | | | | | | | |
Collapse
|
45
|
Abstract
Better understanding of the underlying principles of tumor biology and immunology, enhanced by recent insights into the mechanisms of immune recognition, regulation, and tumor escape has provided new approaches for cancer immunotherapy. This article reviews the current status and future directions of cancer immunotherapy, with a focus on the recent encouraging results from immune-modulating antibodies and adoptive cell therapy.
Collapse
Affiliation(s)
- Fumito Ito
- Department of Surgery, University of Michigan Health System, 3410 Cancer Center/5932, 1500 East Medical Center Drive, Ann Arbor, MI 48109-5932, USA
| | | |
Collapse
|
46
|
Hargadon KM. Tumor-altered dendritic cell function: implications for anti-tumor immunity. Front Immunol 2013; 4:192. [PMID: 23874338 PMCID: PMC3708450 DOI: 10.3389/fimmu.2013.00192] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/27/2013] [Indexed: 01/20/2023] Open
Abstract
Dendritic cells (DC) are key regulators of both innate and adaptive immunity, and the array of immunoregulatory functions exhibited by these cells is dictated by their differentiation, maturation, and activation status. Although a major role for these cells in the induction of immunity to pathogens has long been appreciated, data accumulated over the last several years has demonstrated that DC are also critical regulators of anti-tumor immune responses. However, despite the potential for stimulation of robust anti-tumor immunity by DC, tumor-altered DC function has been observed in many cancer patients and tumor-bearing animals and is often associated with tumor immune escape. Such dysfunction has significant implications for both the induction of natural anti-tumor immune responses as well as the efficacy of immunotherapeutic strategies that target endogenous DC in situ or that employ exogenous DC as part of anti-cancer immunization maneuvers. In this review, the major types of tumor-altered DC function will be described, with emphasis on recent insights into the mechanistic bases for the inhibition of DC differentiation from hematopoietic precursors, the altered programing of DC precursors to differentiate into myeloid-derived suppressor cells or tumor-associated macrophages, the suppression of DC maturation and activation, and the induction of immunoregulatory DC by tumors, tumor-derived factors, and tumor-associated cells within the milieu of the tumor microenvironment. The impact of these tumor-altered cells on the quality of the overall anti-tumor immune response will also be discussed. Finally, this review will also highlight questions concerning tumor-altered DC function that remain unanswered, and it will address factors that have limited advances in the study of this phenomenon in order to focus future research efforts in the field on identifying strategies for interfering with tumor-associated DC dysfunction and improving DC-mediated anti-tumor immunity.
Collapse
Affiliation(s)
- Kristian M Hargadon
- Hargadon Laboratory, Department of Biology, Hampden-Sydney College , Hampden-Sydney, VA , USA
| |
Collapse
|
47
|
Yorty JL. Short-term corticosterone treatment decreases the early CD8+ T cell response to simian virus 40 tumor antigen but has no impact on the late CD8+ T cell response. Brain Behav Immun 2013; 28:139-48. [PMID: 23164951 DOI: 10.1016/j.bbi.2012.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2012] [Revised: 11/09/2012] [Accepted: 11/09/2012] [Indexed: 10/27/2022] Open
Abstract
CD8+ T cells (T(CD8)) help control tumor growth in vivo through recognition of distinct tumor antigens and cytolysis of tumor cells. The T(CD8) immune response in C57BL/6 mice to the Simian Virus 40 oncoprotein, large tumor antigen (Tag), targets multiple epitopes and is well-characterized. Epitope IV, an H-2K(b)-restricted epitope, is immunodominant while epitope I, an H-2D(b)-restricted epitope is subdominant. GCs alter many aspects of T cell function. Indeed, the current studies demonstrate that exposure of mice to the immunosuppressive GC, corticosterone (CORT), over the entire course of the primary immune response limits activation of endogenous Tag-specific T(CD8). Even short-term CORT treatment from day -1 to day +2 post-immunization significantly reduced splenic size and the absolute number of Tag-specific T(CD8) on day 6 post-immunization. In vivo killing activity was also reduced. However, by day 10 post-immunization, the peak of the immune response, the absolute number of Tag-specific T(CD8) and their in vivo killing of epitope I or epitope IV-expressing target cells had recovered in CORT treated mice. Adoptive transfer of transgenic T cells post-CORT removal demonstrated that CORT decreased the ability of the endogenous antigen-presenting cells to induce proliferation of the exogenous transgenic T cells. Combined, these studies have implications about the timing of clinical steroid treatment relative to immunization or adoptive transfer for cancer immunotherapy.
Collapse
Affiliation(s)
- Jodi L Yorty
- Department of Biology, Elizabethtown College, One Alpha Drive, Elizabethtown, PA 17022, United States.
| |
Collapse
|
48
|
Sieben M, Schäfer P, Dinsart C, Galle PR, Moehler M. Activation of the human immune system via toll-like receptors by the oncolytic parvovirus H-1. Int J Cancer 2012; 132:2548-56. [PMID: 23151948 DOI: 10.1002/ijc.27938] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 10/05/2012] [Indexed: 01/09/2023]
Abstract
This study aimed to investigate the function of toll-like receptors (TLRs) during oncolytic parvovirus H-1 (H-1PV)-induced human immune responses. First, the role of TLRs in the activation of the NFκB transcription factor was characterized; second, the immunologic effects of H-1PV-induced tumor cell lysates (TCL) on human antitumor immune responses were evaluated. A human ex vivo model was used to study immune responses with dendritic cells (DCs). Human embryonic kidney cells (HEK293) transfected to stably express TLRs were used as potential human DC equivalents to further investigate the role of specific TLRs during immune activation. TLR3 and TLR9 were activated by H-1PV infection, which correlated with NFκB translocation to the nucleus and a reduced cytoplasmic IκB expression. Using a TLR-signaling reporter plasmid (pNiFty-Luc), NFκB activity was increased following H-1PV infection. In addition, human DCs coincubated with H-1PV-induced TCL demonstrated increased TLR3 and TLR9 expression. These data suggest that H-1PV-induced TCL stimulate human DCs at least in part through TLR-dependent signaling pathways. Thus, DC maturation occurred through exposure to H-1PV-induced TCL through TLR-signaling leading to NFκB-dependent activation of the adaptive immune system as indicated by the increased expression of CD86, TLR3 and TLR9. Furthermore, the transcription of various cytokines indicates the activation of immune response, therefore the production of the proinflammatory cytokine TNF-α was determined. Here, H-1PV-induced TCL significantly enhanced the TNF-α level by DCs after coculture. H-1PV oncolytic virotherapy enhances immune priming by different effects on DCs and generates antitumor immunity. These findings potentially offer a new approach to tumor therapy.
Collapse
Affiliation(s)
- Maike Sieben
- First Department of Internal Medicine, University Medical Center of the Johannes Gutenberg University of Mainz, Mainz, Germany
| | | | | | | | | |
Collapse
|
49
|
Khong A, Nelson DJ, Nowak AK, Lake RA, Robinson BWS. The use of agonistic anti-CD40 therapy in treatments for cancer. Int Rev Immunol 2012; 31:246-66. [PMID: 22804570 DOI: 10.3109/08830185.2012.698338] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Agonistic anti-CD40 antibody is a potent stimulator of anti-tumor immune responses due to its action on both immune and tumor cells. It has the ability to "precondition" dendritic cells, allowing them to prime effective cytotoxic T-cell responses. Thus, anti-CD40 antibody provides an ideal therapy for combination with traditional cancer treatments (i.e., chemotherapy, surgery) in order to elicit immune-mediated anti-tumor effects. This review summarizes the mechanisms of action of agonistic anti-CD40, the use of mouse models to investigate its effects and combinations with other therapies in vivo, and current clinical trials combining humanized anti-CD40 antibody with chemotherapy and/or other immunotherapies.
Collapse
Affiliation(s)
- Andrea Khong
- School of Medicine and Pharmacology, University of Western Australia, Perth, Australia
| | | | | | | | | |
Collapse
|
50
|
Yan X, Doffek K, Yin C, Krein M, Phillips M, Sugg SL, Johnson B, Shilyansky J. Annexin-V promotes anti-tumor immunity and inhibits neuroblastoma growth in vivo. Cancer Immunol Immunother 2012; 61:1917-27. [PMID: 22476407 PMCID: PMC11029734 DOI: 10.1007/s00262-012-1250-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2011] [Accepted: 03/21/2012] [Indexed: 10/28/2022]
Abstract
The goal of the current study is to determine the effects of blocking phosphatidylserine (PS) on the growth of neuroblastoma in mice. PS, an anionic phospholipid restricted to the cytoplasmic surface of plasma membranes in most cells, is externalized to the surface of apoptotic cells. PS has been shown to induce immune tolerance to self-antigens. PS can also be found on the surface of live cells and in particular tumor cells. Annexin-V (AnV) is a protein that specifically binds and blocks PS. To determine the effects of blocking PS with AnV on tumor growth and immunogenicity, mice were inoculated with AGN2a, a poorly immunogenic murine neuroblastoma that expresses high level of PS on the cell surface. Survival and anti-tumor T cell response were determined. AGN2a were engineered to secrete AnV. Secreted protein effectively blocked tumor PS. 40 % of mice inoculated with AnV-expressing AGN2a cells survived free of tumor, whereas none of the mice inoculated with control cells survived (p = 0.0062). The benefits of AnV were lost when mice were depleted of T cells. The findings suggest that AnV could protect mice from tumor challenge through an immune mediated mechanism. Mice were then immunized with irradiated AnV-secreting or control cells, and challenged with wild-type AGN2a cells. AnV-secreting cell vaccine protected 80 % of mice from AGN2a challenge, while control cell vaccine prevented tumor growth in only 30 % of animals (p = 0.012). ELISPOT analysis demonstrated that AnV-secreting cell vaccine induced a greater frequency of interferon-gamma producing splenic T cells. T cells isolated from mice immunized with AnV-secreting but not control vaccine lysed AGN2a. In summary, AnV blocked PS, enhanced T cell mediated tumor immunity, and inhibited tumor growth.
Collapse
Affiliation(s)
- Xiaocai Yan
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Kara Doffek
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Chaobo Yin
- Department of Surgery, Carver School of Medicine, University of Iowa, 200 Hawkins Ave., Iowa City, IA 52242 USA
| | - Michael Krein
- Department of Surgery, Carver School of Medicine, University of Iowa, 200 Hawkins Ave., Iowa City, IA 52242 USA
| | - Michael Phillips
- Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Sonia L. Sugg
- Department of Surgery, Carver School of Medicine, University of Iowa, 200 Hawkins Ave., Iowa City, IA 52242 USA
| | - Bryon Johnson
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI 53226 USA
| | - Joel Shilyansky
- Department of Surgery, Carver School of Medicine, University of Iowa, 200 Hawkins Ave., Iowa City, IA 52242 USA
| |
Collapse
|