1
|
Graham PT, Nowak AK, Cornwall SMJ, Larma I, Nelson DJ. The STING agonist, DMXAA, reduces tumor vessels and enhances mesothelioma tumor antigen presentation yet blunts cytotoxic T cell function in a murine model. Front Immunol 2022; 13:969678. [PMID: 36466911 PMCID: PMC9716460 DOI: 10.3389/fimmu.2022.969678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 10/20/2022] [Indexed: 08/22/2023] Open
Abstract
We assessed the murine Stimulator of Interferon Genes (STING) agonist, DMXAA, for anti-mesothelioma potential using the AE17-sOVA model that expresses ovalbumin (OVA) as a neo tumor antigen. Dose response experiments alongside testing different routes of administration identified a safe effective treatment regimen that induced 100% cures in mice with small or large tumors. Three doses of 25mg/kg DMXAA given intra-tumorally every 9 days induced tumor regression and long-term survival (>5 months). Re-challenge experiments showed that tumor-free mice developed protective memory. MTT and propidium-iodide assays showed that DMXAA exerted direct cytotoxic effects at doses >1mg/ml on the murine AE17 and AB1 mesothelioma cell lines. In-vivo studies using a CFSE-based in-vivo proliferation assay showed that DMXAA improved tumor-antigen presentation in tumor-draining lymph nodes, evidenced by OVA-specific OT-1 T cells undergoing more divisions. An in-vivo cytotoxic T lymphocyte (CTL) assay showed that DMXAA blunted the lytic quality of CTLs recognizing the dominant (SIINFEKL) and a subdominant (KVVRFDKL) OVA epitopes. DMXAA reduced tumor vessel size in-vivo and although the proportion of T cells infiltrating tumors reduced, the proportion of tumor-specific T cells increased. These data show careful dosing and treatment protocols reduce mesothelioma cell viability and modulate tumor vessels such that tumor-antigen specific CTLs access the tumor site. However, attempts to enhance DMXAA-induced anti-tumor responses by combination with an agonist anti-CD40 antibody or IL-2 reduced efficacy. These proof-of-concept data suggest that mesothelioma patients could benefit from treatment with a STING agonist, but combination with immunotherapy should be cautiously undertaken.
Collapse
Affiliation(s)
- Peter T. Graham
- School of Medicine, Curtin University, Bentley, WA, Australia
| | - Anna K. Nowak
- Medical School, University of Western Australia, Nedlands, WA, Australia
- National Centre for Asbestos Related Diseases, Nedlands, WA, Australia
- Institute of Respiratory Health, Nedlands, WA, Australia
| | | | - Irma Larma
- Becton Dickinson Pty Limited, Osborne Park, WA, Australia
| | - Delia J. Nelson
- School of Medicine, Curtin University, Bentley, WA, Australia
- Curtin Health Innovation Research Institute, Bentley, WA, Australia
| |
Collapse
|
2
|
Scholl JN, Dias CK, Muller L, Battastini AMO, Figueiró F. Extracellular vesicles in cancer progression: are they part of the problem or part of the solution? Nanomedicine (Lond) 2020; 15:2625-2641. [PMID: 33094653 DOI: 10.2217/nnm-2020-0256] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Extracellular vesicles (EVs) are released especially by cancer cells. They modulate the tumor microenvironment by interacting with immune cells while carrying immunosuppressive or immunostimulatory molecules. In this review, we will explore some conflicting reports regarding the immunological outcomes of EVs in cancer progression, in which they might initiate an antitumor immune response or an immunosuppressive response. Concerning immunosuppression, the role of tumor-derived EVs' in the adenosinergic system is underexplored. The enhancement of adenosine (ADO) levels in the tumor microenvironment impairs T-cell function and cytokine release. However, some tumor-derived EVs may deliver immunostimulatory factors, promoting immunogenic activity, even with ADO production. The modulatory role of ADO over the tumor progression represents a piece in an intricate microenvironment with anti and pro tumoral seesaw-like mechanisms.
Collapse
Affiliation(s)
- Juliete Nathali Scholl
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Camila Kehl Dias
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Laurent Muller
- Department of Otolaryngology, Head & Neck Surgery, University of Basel, Basel, 4031, Switzerland
| | - Ana Maria Oliveira Battastini
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| | - Fabrício Figueiró
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, 90035-003, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, 90035-003, Brazil
| |
Collapse
|
3
|
Rodriguez-Barbosa JI, Azuma M, Zelinskyy G, Perez-Simon JA, del Rio ML. Critical role of PD-L1 expression on non-tumor cells rather than on tumor cells for effective anti-PD-L1 immunotherapy in a transplantable mouse hematopoietic tumor model. Cancer Immunol Immunother 2020; 69:1001-1014. [DOI: 10.1007/s00262-020-02520-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 02/14/2020] [Indexed: 12/18/2022]
|
4
|
Neo-antigen specific T cell responses indicate the presence of metastases before imaging. Sci Rep 2019; 9:14640. [PMID: 31601975 PMCID: PMC6787183 DOI: 10.1038/s41598-019-51317-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Accepted: 09/24/2019] [Indexed: 12/03/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) causes 19% of all Australian cancer deaths, with a 5-year survival post-resection of around 60%. Post-operative recurrence is due to metastases that were undetectable pre-operatively, or growth of microscopic locoregional residual disease. However, post-operative imaging modalities typically only detect more advanced tumours; where PET-CT has a detection limit of 6–7 mm. Detection of small deposits of lung metastatic disease is of importance in order to facilitate early and potentially more effective treatment. In this study, in a murine model of lung metastatic disease, we explore whether neo-antigen specific T cells are a sensitive marker for the detection of lung cancer after primary tumour resection. We determine lung metastatic disease by histology, and then compare detection by PET-CT and neo-antigen specific T cell frequency. Detection of lung metastatic disease within the histology positive group by PET-CT and neo-antigen specific T cell frequency were 22.9% and 92.2%, respectively. Notably, neo-antigen specific T cells in the lung draining lymph node were indicative of metastatic disease (82.8 ± 12.9 spots/105 cells; mean ± SE), compared to healthy lung control (28.5 ± 8.6 spots/105 cells; mean ± SE). Potentially, monitoring tumour neo-antigen specific T cell profiles is a highly sensitive method for determining disease recurrence.
Collapse
|
5
|
Das K, Eisel D, Vormehr M, Müller-Decker K, Hommertgen A, Jäger D, Zörnig I, Feuerer M, Kopp-Schneider A, Osen W, Eichmüller SB. A transplantable tumor model allowing investigation of NY-BR-1-specific T cell responses in HLA-DRB1*0401 transgenic mice. BMC Cancer 2019; 19:914. [PMID: 31519152 PMCID: PMC6743128 DOI: 10.1186/s12885-019-6102-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 08/28/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND NY-BR-1 has been described as a breast cancer associated differentiation antigen with intrinsic immunogenicity giving rise to endogenous T and B cell responses. The current study presents the first murine tumor model allowing functional investigation of NY-BR-1-specific immune responses in vivo. METHODS A NY-BR-1 expressing tumor model was established in DR4tg mice based on heterotopic transplantation of stable transfectant clones derived from the murine H2 compatible breast cancer cell line EO771. Composition and phenotype of tumor infiltrating immune cells were analyzed by qPCR and FACS. MHC I binding affinity of candidate CTL epitopes predicted in silico was determined by FACS using the mutant cell line RMA-S. Frequencies of NY-BR-1 specific CTLs among splenocytes of immunized mice were quantified by FACS with an epitope loaded Db-dextramer. Functional CTL activity was determined by IFNγ catch or IFNγ ELISpot assays and statistical analysis was done applying the Mann Whitney test. Tumor protection experiments were performed by immunization of DR4tg mice with replication deficient recombinant adenovirus followed by s.c. challenge with NY-BR-1 expressing breast cancer cells. RESULTS Our results show spontaneous accumulation of CD8+ T cells and F4/80+ myeloid cells preferentially in NY-BR-1 expressing tumors. Upon NY-BR-1-specific immunization experiments combined with in silico prediction and in vitro binding assays, the first NY-BR-1-specific H2-Db-restricted T cell epitope could be identified. Consequently, flow cytometric analysis with fluorochrome conjugated multimers showed enhanced frequencies of CD8+ T cells specific for the newly identified epitope in spleens of immunized mice. Moreover, immunization with Ad.NY-BR-1 resulted in partial protection against outgrowth of NY-BR-1 expressing tumors and promoted intratumoral accumulation of macrophages. CONCLUSION This study introduces the first H2-Db-resctricted CD8+ T cell epitope-specific for the human breast cancer associated tumor antigen NY-BR-1. Our novel, partially humanized tumor model enables investigation of the interplay between HLA-DR4-restricted T cell responses and CTLs within their joint attack of NY-BR-1 expressing tumors.
Collapse
Affiliation(s)
- Krishna Das
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Virology, Innsbruck Medical University, Innsbruck, Austria.,Faculty of Biosciences, University Heidelberg, Heidelberg, Germany
| | - David Eisel
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Faculty of Biosciences, University Heidelberg, Heidelberg, Germany.,Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany
| | - Mathias Vormehr
- Biopharmaceutical New Technologies (BioNTech) Corporation, Mainz, Germany.,University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Karin Müller-Decker
- Core Facility Tumor Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Adriane Hommertgen
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Molecular & Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Jäger
- CCU Applied Tumor Immunity, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medical Oncology, National Center for Tumor Diseases (NCT) and University Hospital Heidelberg, Heidelberg, Germany
| | - Inka Zörnig
- Department of Medical Oncology, National Center for Tumor Diseases (NCT) and University Hospital Heidelberg, Heidelberg, Germany
| | - Markus Feuerer
- Institute of Immunology, Regensburg Center for Interventional Immunology (RCI), University Regensburg and University Hospital Regensburg, Regensburg, Germany
| | | | - Wolfram Osen
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stefan B Eichmüller
- Research Group GMP & T Cell Therapy, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
6
|
Opzoomer JW, Sosnowska D, Anstee JE, Spicer JF, Arnold JN. Cytotoxic Chemotherapy as an Immune Stimulus: A Molecular Perspective on Turning Up the Immunological Heat on Cancer. Front Immunol 2019; 10:1654. [PMID: 31379850 PMCID: PMC6652267 DOI: 10.3389/fimmu.2019.01654] [Citation(s) in RCA: 94] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 07/03/2019] [Indexed: 12/14/2022] Open
Abstract
Cytotoxic chemotherapeutics (CCTs) are widely used in the treatment of cancer. Although their mechanisms of action have been best understood in terms of targeting the apparatus of mitosis, an ability to stimulate anti-tumor immune responses is increasing the recognition of these agents as immunotherapies. Immune checkpoint blockade antibodies neutralize important, but specific, immune-regulatory interactions such as PD-1/PD-L1 and CTLA-4 to improve the anti-tumor immune response. However, CCTs can provide a broad-acting immune-stimulus against cancer, promoting both T-cell priming and recruitment to the tumor, which compliments the effects of immune checkpoint blockade. A key pathway in this process is "immunogenic cell death" (ICD) which occurs as a result of tumor cell endoplasmic reticulum stress and apoptosis elicited by CCTs. ICD involves a series of non-redundant signaling events which break tolerance and license anti-tumor antigen-specific T-cells, allowing CCTs to act as "in situ" tumor vaccination tools. Not all responses are tumor cell-intrinsic, as CCTs can also modulate the broader tumor microenvironment. This modulation occurs through preferential depletion of stromal cells which suppress and neutralize robust anti-tumor immune responses, such as myeloid cell populations and Tregs, while effector CD8+ and CD4+ T-cells and NK cells are relatively spared. The immune-stimulating effects of CCTs are dependent on chemotherapy class, dose and tumor cell sensitivity to the agent, highlighting the need to understand the underlying biology of these responses. This mini review considers the immune-stimulating effects of CCTs from a molecular perspective, specifically highlighting considerations for their utilization in the context of combinations with immunotherapy.
Collapse
Affiliation(s)
- James W Opzoomer
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Dominika Sosnowska
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - Joanne E Anstee
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - James F Spicer
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| | - James N Arnold
- Faculty of Life Sciences and Medicine, School of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
7
|
Pearce H, Hutton P, Chaudhri S, Porfiri E, Patel P, Viney R, Moss P. Spontaneous CD4 + and CD8 + T-cell responses directed against cancer testis antigens are present in the peripheral blood of testicular cancer patients. Eur J Immunol 2017; 47:1232-1242. [PMID: 28555838 PMCID: PMC5519936 DOI: 10.1002/eji.201646898] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/17/2017] [Accepted: 05/22/2017] [Indexed: 12/11/2022]
Abstract
Cancer/testis antigen (CTAg) expression is restricted to spermatogenic cells in an immune‐privileged site within the testis. However, these proteins are expressed aberrantly by malignant cells and T‐cell responses against CTAgs develop in many cancer patients. We investigated the prevalence, magnitude and phenotype of CTAg‐specific T cells in the blood of patients with testicular germ cell tumors (TGCTs). CD8+ and CD4+ T‐cell responses against MAGE‐A family antigens were present in 44% (20/45) of patients’ samples assayed by ex vivo IFN‐γ ELISPOT. The presence of MAGE‐specific CD8+ T cells was further determined following short‐term in vitro expansion through the use of pMHC‐I multimers containing known immunogenic peptides. Longitudinal analysis revealed that the frequency of MAGE‐specific T cells decreased by 89% following orchidectomy suggesting that persistence of tumor antigen is required to sustain CTAg‐specific T‐cell immunity. Notably, this decrease correlated with a decline in the global effector/memory T‐cell pool following treatment. Spontaneous T‐cell immunity against CTAg proteins therefore develops in many patients with testicular cancer and may play an important role in the excellent clinical outcome of patients with this tumor subtype.
Collapse
Affiliation(s)
- Hayden Pearce
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Paul Hutton
- University Hospitals NHS Foundation Trust, Birmingham, UK
| | | | - Emilio Porfiri
- University Hospitals NHS Foundation Trust, Birmingham, UK.,Institute of Cancer and Genomics, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Prashant Patel
- University Hospitals NHS Foundation Trust, Birmingham, UK.,Institute of Cancer and Genomics, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Richard Viney
- University Hospitals NHS Foundation Trust, Birmingham, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.,University Hospitals NHS Foundation Trust, Birmingham, UK
| |
Collapse
|
8
|
Nelson DJ, Clark B, Munyard K, Williams V, Groth D, Gill J, Preston H, Chan A. A review of the importance of immune responses in luminal B breast cancer. Oncoimmunology 2017; 6:e1282590. [PMID: 28405507 DOI: 10.1080/2162402x.2017.1282590] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/10/2017] [Accepted: 01/10/2017] [Indexed: 12/14/2022] Open
Abstract
Historically, the immune environment was not considered an important target for breast cancer treatment. However, the association of lymphocytic infiltrates in triple negative and HER-2 over-amplified breast cancer subtypes with better outcomes, has provoked interest in evaluating the role of the immune system in the luminal B subtype that accounts for 39% of breast cancers and has a poor patient prognosis. It is unknown which immunosuppressive cell types or molecules (e.g., checkpoint molecules) are relevant, or where measurement is most informative. We hypothesize that a profound immunosuppressive tumor and/or lymph node milieu is prognostic and impacts on responses to therapies.
Collapse
Affiliation(s)
- Delia J Nelson
- School of Biomedical Sciences, Curtin University, Bentley, Perth, WA, Australia; CHIRI Biosciences, Curtin University, Perth, WA, Australia
| | - Briony Clark
- School of Biomedical Sciences, Curtin University, Bentley, Perth, WA, Australia; CHIRI Biosciences, Curtin University, Perth, WA, Australia
| | - Kylie Munyard
- School of Biomedical Sciences, Curtin University, Bentley, Perth, WA, Australia; CHIRI Biosciences, Curtin University, Perth, WA, Australia
| | - Vincent Williams
- School of Biomedical Sciences, Curtin University , Bentley, Perth, WA, Australia
| | - David Groth
- School of Biomedical Sciences, Curtin University, Bentley, Perth, WA, Australia; CHIRI Biosciences, Curtin University, Perth, WA, Australia
| | - Jespal Gill
- Western Diagnostics Pathology , Myaree, Perth, WA, Australia
| | - Henry Preston
- Western Diagnostics Pathology , Myaree, Perth, WA, Australia
| | - Arlene Chan
- Breast Cancer Research Centre-WA, Hollywood Private Hospital, Nedlands, WA, Australia; Curtin Medical School, Curtin University, Perth, WA, Australia
| |
Collapse
|
9
|
Oda SK, Strauch P, Fujiwara Y, Al-Shami A, Oravecz T, Tigyi G, Pelanda R, Torres RM. Lysophosphatidic acid inhibits CD8 T cell activation and control of tumor progression. Cancer Immunol Res 2015; 1:245-55. [PMID: 24455753 DOI: 10.1158/2326-6066.cir-13-0043-t] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
CD8 T lymphocytes are able to eliminate nascent tumor cells through a process referred to as immune surveillance. However, multiple inhibitory mechanisms within the tumor microenvironment have been described that impede tumor rejection by CD8 T cells, including increased signaling by inhibitory receptors. Lysophosphatidic acid (LPA) is a bioactive lysophospholipid that has been shown repeatedly to promote diverse cellular processes benefiting tumorigenesis. Accordingly, the increased expression of LPA and LPA receptors is a common feature of diverse tumor cell lineages and can result in elevated systemic LPA levels. LPA is recognized by at least 6 distinct G-protein-coupled receptors and several of which are expressed by T cells, although the precise role of LPA signaling in CD8 T cell activation and function has not been defined. Here, we demonstrate that LPA signaling via the LPA5 receptor expressed by CD8 T cells suppresses antigen receptor signaling, cell activation and proliferation in vitro and in vivo. Importantly, in a mouse melanoma model tumor-specific CD8 T cells that are LPA5-deficient are able to control tumor growth significantly better than wild-type tumor-specific CD8 T cells. Together, these data suggest that the production of LPA by tumors serves not only in an autocrine manner to promote tumorigenesis but also as a mechanism to suppress adaptive immunity and highlights a potential novel target for cancer treatment.
Collapse
Affiliation(s)
- Shannon K Oda
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado
| | - Pamela Strauch
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado
| | - Yuko Fujiwara
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, Tennessee
| | | | | | - Gabor Tigyi
- Department of Physiology, University of Tennessee Health Science Center Memphis, Memphis, Tennessee
| | - Roberta Pelanda
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado
| | - Raul M Torres
- Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado
| |
Collapse
|
10
|
Creaney J, Ma S, Sneddon SA, Tourigny MR, Dick IM, Leon JS, Khong A, Fisher SA, Lake RA, Lesterhuis WJ, Nowak AK, Leary S, Watson MW, Robinson BW. Strong spontaneous tumor neoantigen responses induced by a natural human carcinogen. Oncoimmunology 2015; 4:e1011492. [PMID: 26140232 PMCID: PMC4485777 DOI: 10.1080/2162402x.2015.1011492] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 01/16/2015] [Accepted: 01/21/2015] [Indexed: 12/26/2022] Open
Abstract
A key to improving cancer immunotherapy will be the identification of tumor-specific "neoantigens" that arise from mutations and augment the resultant host immune response. In this study we identified single nucleotide variants (SNVs) by RNA sequencing of asbestos-induced murine mesothelioma cell lines AB1 and AB1-HA. Using the NetMHCpan 2.8 algorithm, the theoretical binding affinity of predicted peptides arising from high-confidence, exonic, non-synonymous SNVs was determined for the BALB/c strain. The immunoreactivity to 20 candidate mutation-carrying peptides of increased affinity and the corresponding wild-type peptides was determined using interferon-γ ELISPOT assays and lymphoid organs of non-manipulated tumor-bearing mice. A strong endogenous immune response was demonstrated to one of the candidate neoantigens, Uqcrc2; this response was detected in the draining lymph node and spleen. Antigen reactive cells were not detected in non-tumor bearing mice. The magnitude of the response to the Uqcrc2 neoantigen was similar to that of the strong influenza hemagglutinin antigen, a model tumor neoantigen. This work confirms that the approach of RNAseq plus peptide prediction and ELISPOT testing is sufficient to identify natural tumor neoantigens.
Collapse
Affiliation(s)
- Jenette Creaney
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia ; Australian Mesothelioma Tumour Bank; Sir Charles Gairdner Hospital ; Nedlands, Australia
| | - Shaokang Ma
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia
| | - Sophie A Sneddon
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia
| | - Michelle R Tourigny
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia
| | - Ian M Dick
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia ; Australian Mesothelioma Tumour Bank; Sir Charles Gairdner Hospital ; Nedlands, Australia
| | - Justine S Leon
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia ; Australian Mesothelioma Tumour Bank; Sir Charles Gairdner Hospital ; Nedlands, Australia
| | - Andrea Khong
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia
| | - Scott A Fisher
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia
| | - Richard A Lake
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia
| | - W Joost Lesterhuis
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia
| | - Anna K Nowak
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia ; Department of Oncology; Sir Charles Gairdner Hospital ; Nedlands, Australia
| | - Shay Leary
- Institute for Immunology and Infectious Diseases; Murdoch University ; Murdoch, Australia
| | - Mark W Watson
- Institute for Immunology and Infectious Diseases; Murdoch University ; Murdoch, Australia
| | - Bruce W Robinson
- National Centre for Asbestos Related Diseases; School of Medicine and Pharmacology; University of Western Australia ; Nedlands, Australia ; Australian Mesothelioma Tumour Bank; Sir Charles Gairdner Hospital ; Nedlands, Australia ; Department of Respiratory Medicine; Sir Charles Gairdner Hospital ; Nedlands, Australia
| |
Collapse
|
11
|
Abstract
We aimed to determine if the tumor microenvironment could be turned into a “self”-vaccine site. We show that provoking a local inflammatory response modulates endothelia to permit the infiltration of innate and adaptive effector cells which collaborate to eradicate the inflamed tumor and other tumor deposits, and provide long-term protection.
Collapse
Affiliation(s)
- Delia J Nelson
- Immunology and Cancer Group; School of Biomedical Sciences; Curtin University; Kent St.; Bentley; Perth, WA Australia ; Western Australian Biomedical Research Institute; Bentley; Perth, WA Australia ; Curtin Health Innovation Research Institute; Bentley; Perth, WA Australia
| |
Collapse
|
12
|
Murphy KA, Erickson JR, Johnson CS, Seiler CE, Bedi J, Hu P, Pluhar GE, Epstein AL, Ohlfest JR. CD8+ T cell-independent tumor regression induced by Fc-OX40L and therapeutic vaccination in a mouse model of glioma. THE JOURNAL OF IMMUNOLOGY 2013; 192:224-33. [PMID: 24293627 DOI: 10.4049/jimmunol.1301633] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Despite the growing number of preclinical and clinical trials focused on immunotherapy for the treatment of malignant gliomas, the prognosis for this disease remains grim. Although some promising advances have been made, the immune response stimulated as a result of immunotherapeutic protocols has been inefficient at complete tumor elimination, primarily due to our lack of understanding of the necessary effector functions of the immune system. We previously demonstrated that a tumor lysate vaccine/Fc-OX40L therapy is capable of inducing enhanced survival and tumor elimination in the GL261 mouse glioma model. The following experiments were performed to determine the mechanism(s) of action of this therapy that elicits a potent antitumor immune response. The evidence subsequently outlined indicates a CD8(+) T cell-independent and CD4(+) T cell-, NK cell-, and B cell-dependent means of prolonged survival. CD8(+) T cell-independent tumor clearance is surprising considering the current focus of many cancer immunotherapy protocols. These results provide evidence for CD8(+) T cell-independent means of antitumor response and should lead to additional examination of the potential manipulation of this mechanism for future treatment strategies.
Collapse
Affiliation(s)
- Katherine A Murphy
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Jackaman C, Majewski D, Fox SA, Nowak AK, Nelson DJ. Chemotherapy broadens the range of tumor antigens seen by cytotoxic CD8(+) T cells in vivo. Cancer Immunol Immunother 2012; 61:2343-56. [PMID: 22714286 PMCID: PMC11029427 DOI: 10.1007/s00262-012-1307-4] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Accepted: 06/04/2012] [Indexed: 12/22/2022]
Abstract
Cytotoxic chemotherapies may expose the immune system to high levels of tumor antigens and expand the CD8(+) T-cell response to include weak or subdominant antigens. Here, we evaluated the in vivo CTL response to tumor antigens using a murine mesothelioma tumor cell line transfected with a neotumor antigen, ovalbumin, that contains a known hierarchy of epitopes for MHC class I molecules. We show that as tumors progress, effector CTLs are generated in vivo that focus on the dominant epitope SIINFEKL, although a weak response was seen to one (KVVRFDKL) subdominant epitope. These CTLs did not prevent tumor growth. Cisplatin treatment slowed tumor growth, slightly improved in vivo SIINFEKL presentation to T cells and reduced SIINFEKL-CTL activity. However, the CTL response to KVVRFDKL was amplified, and a response to another subdominant epitope, NAIVFKGL, was revealed. Similarly, gemcitabine cured most mice, slightly enhanced SIINFEKL presentation, reduced SIINFEKL-CTL activity yet drove a significant CTL response to NAIVFKGL, but not KVVRFDKL. These NAIVFKGL-specific CTLs secreted IFNγ and proliferated in response to in vitro NAIVFKGL stimulation. IL-2 treatment during chemotherapy refocused the response to SIINFEKL and simultaneously degraded the cisplatin-driven subdominant CTL response. These data show that chemotherapy reveals weaker tumor antigens to the immune system, a response that could be rationally targeted. Furthermore, while integrating IL-2 into the chemotherapy regimen interfered with the hierarchy of the response, IL-2 or other strategies that support CTL activity could be considered upon completion of chemotherapy.
Collapse
Affiliation(s)
- Connie Jackaman
- Immunology and Cancer Group, School of Biomedical Sciences, Curtin University, Kent St., Bentley, Perth, WA 6102 Australia
- Western Australia Biomedical Research Institute, Bentley, Perth, WA 6102 Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA 6102 Australia
| | - David Majewski
- Immunology and Cancer Group, School of Biomedical Sciences, Curtin University, Kent St., Bentley, Perth, WA 6102 Australia
- Western Australia Biomedical Research Institute, Bentley, Perth, WA 6102 Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA 6102 Australia
| | - Simon A. Fox
- Western Australia Biomedical Research Institute, Bentley, Perth, WA 6102 Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA 6102 Australia
- School of Pharmacy, Curtin University, Kent St., Perth, WA 6102 Australia
| | - Anna K. Nowak
- School of Medicine and Pharmacology, University of Western Australia, Nedlands Perth, WA 6009 Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Perth, WA 6009 Australia
| | - Delia J. Nelson
- Immunology and Cancer Group, School of Biomedical Sciences, Curtin University, Kent St., Bentley, Perth, WA 6102 Australia
- Western Australia Biomedical Research Institute, Bentley, Perth, WA 6102 Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA 6102 Australia
| |
Collapse
|
14
|
Fransen MF, Arens R, Melief CJ. Local targets for immune therapy to cancer: Tumor draining lymph nodes and tumor microenvironment. Int J Cancer 2012; 132:1971-6. [DOI: 10.1002/ijc.27755] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Accepted: 07/18/2012] [Indexed: 12/22/2022]
|
15
|
Jackaman C, Nelson DJ. Intratumoral interleukin-2/agonist CD40 antibody drives CD4+ -independent resolution of treated-tumors and CD4+ -dependent systemic and memory responses. Cancer Immunol Immunother 2012; 61:549-60. [PMID: 22002241 PMCID: PMC11029634 DOI: 10.1007/s00262-011-1120-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 09/23/2011] [Indexed: 12/21/2022]
Abstract
Targeting interleukin-2 (IL-2) and/or agonist anti-CD40 antibody (Ab) into tumors represents an effective vaccination strategy that avoids systemic toxicity and resolves treated-site tumors. Here, we examined IL-2 and/or anti-CD40 Ab-driven local versus systemic T cell function and the installation of T cell memory. Single tumor studies showed that IL-2 induced a potent CD4+ and CD8+ T cell response that was limited to the draining lymph node and treated-site tumor, and lymph node tumor-specific CD8+ T cells did not upregulate CD44. A two-tumor model showed that while IL-2-treated-site tumors resolved, distal tumors continued to grow, implying limited systemic immunity. In contrast, anti-CD40 Ab treatment with or without IL-2 expanded the systemic T cell response to non-draining lymph nodes, and distal tumors resolved. Tumor-specific T cells in lymph nodes of anti-CD40 Ab ± IL-2-treated mice upregulated CD44, demonstrating activation and transition to effector/memory migratory cells. While CD40-activated CD4+ T cells were not required for eradicating treated-site tumors, they, plus CD8+ T cells, were crucial for removing distal tumors. Rechallenge/depletion experiments showed that the effector/memory phase required the presence of previously CD40/IL-2-activated CD4+ and CD8+ T cells to prevent recurrence. These novel findings show that different T cell effector mechanisms can operate for the eradication of local treated-site tumors versus untreated distal tumors and that signaling through CD40 generates a whole of body, effector/memory CD4+ and CD8+ T cell response that is amplified and prolonged via IL-2. Thus, successful immunotherapy needs to generate collaborating CD4+ and CD8+ T cells for a complete long-term protective cure.
Collapse
Affiliation(s)
- Connie Jackaman
- School of Biomedical Sciences, Immunology and Cancer Group, Curtin University, Kent St., Bentley, Perth, WA 6102 Australia
- Western Australia Biomedical Research Institute, Bentley, Perth, WA 6102 Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA 6102 Australia
| | - Delia J. Nelson
- School of Biomedical Sciences, Immunology and Cancer Group, Curtin University, Kent St., Bentley, Perth, WA 6102 Australia
- Western Australia Biomedical Research Institute, Bentley, Perth, WA 6102 Australia
- Curtin Health Innovation Research Institute, Bentley, Perth, WA 6102 Australia
| |
Collapse
|
16
|
McDonnell AM, Nowak AK, Lake RA. Contribution of the immune system to the chemotherapeutic response. Semin Immunopathol 2011; 33:353-67. [PMID: 21274535 DOI: 10.1007/s00281-011-0246-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2010] [Accepted: 01/11/2011] [Indexed: 01/28/2023]
Abstract
The immune system plays an important role in the surveillance of neoplastic cells by eliminating them before they manifest as full-blown cancer. Despite this, tumors do develop in the presence of a functioning immune system. Conventional chemotherapy and its ability to directly kill tumor cells is one of the most effective weapons in the fight against cancer, however, increasing evidence suggests that the therapeutic efficacy of some cytotoxic drugs relies on their capacity to interact with the immune system. Killing of tumor cells in a manner that favors their capture by immune cells or selective targeting of immunosuppressive pathways by specific chemotherapies promotes the generation of an effective anti-cancer response; however, this alone is rarely sufficient to cause elimination of advanced disease. An understanding of the immunological events occurring in both animal models and patients undergoing chemotherapy will guide decisions for the development of appropriate combinations and scheduling for the integration of chemotherapy with immunotherapy.
Collapse
Affiliation(s)
- Alison M McDonnell
- National Centre for Asbestos-Related Diseases and School of Medicine and Pharmacology, The University of Western Australia, Perth, 6009 Western Australia, Australia
| | | | | |
Collapse
|
17
|
Kraman M, Bambrough PJ, Arnold JN, Roberts EW, Magiera L, Jones JO, Gopinathan A, Tuveson DA, Fearon DT. Suppression of Antitumor Immunity by Stromal Cells Expressing Fibroblast Activation Protein-. Science 2010; 330:827-30. [PMID: 21051638 DOI: 10.1126/science.1195300] [Citation(s) in RCA: 884] [Impact Index Per Article: 63.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Matthew Kraman
- Wellcome Trust Immunology Unit, Department of Medicine, University of Cambridge, Medical Research Council Centre, Hills Road, Cambridge CB2 2QH, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Steer HJ, Lake RA, Nowak AK, Robinson BWS. Harnessing the immune response to treat cancer. Oncogene 2010; 29:6301-13. [PMID: 20856204 DOI: 10.1038/onc.2010.437] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
It is well established that the immune system has the capacity to attack malignant cells. During malignant transformation cells acquire numerous molecular and biochemical changes that render them potentially vulnerable to immune cells. Yet it is self-evident that a growing tumour has managed to evade these host defence mechanisms. The exact ways in which the immune system interacts with tumour cells and how cancers are able to escape immunological eradication have only recently started to be fully elucidated. Understanding the relationship between the tumour and the anti-tumour immune response and how this can be altered with conventional treatments and immune-targeted therapies is crucial to developing new treatments for patients with cancer. In this review, focusing on the anti-tumour T-cell response, we summarize our understanding of how tumours, cancer treatments and the immune system interact, how tumours evade the immune response and how this process could be manipulated for the benefit of patients with cancer.
Collapse
Affiliation(s)
- H J Steer
- School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia.
| | | | | | | |
Collapse
|
19
|
Thompson ED, Enriquez HL, Fu YX, Engelhard VH. Tumor masses support naive T cell infiltration, activation, and differentiation into effectors. J Exp Med 2010; 207:1791-804. [PMID: 20660615 PMCID: PMC2916130 DOI: 10.1084/jem.20092454] [Citation(s) in RCA: 192] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Accepted: 06/16/2010] [Indexed: 12/31/2022] Open
Abstract
Studies of T cell responses to tumors have focused on the draining lymph node (LN) as the site of activation. We examined the tumor mass as a potential site of activation after adoptive transfer of naive tumor-specific CD8 T cells. Activated CD8 T cells were present in tumors within 24 h of adoptive transfer and proliferation of these cells was also evident 4-5 d later in mice treated with FTY720 to prevent infiltration of cells activated in LNs. To confirm that activation of these T cells occurred in the tumor and not the tumor-draining LNs, we used mice lacking LNs. Activated and proliferating tumor-infiltrating lymphocytes were evident in these mice 24 h and 4 d after naive cell transfer. T cells activated within tumors acquired effector function that was evident both ex vivo and in vivo. Both cross-presenting antigen presenting cells within the tumor and tumor cells directly presenting antigen activated these functional CD8 effectors. We conclude that tumors support the infiltration, activation, and effector differentiation of naive CD8 T cells, despite the presence of immunosuppressive mechanisms. Thus, targeting of T cell activation to tumors may present a tool in the development of cancer immunotherapy.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- Antigen-Presenting Cells/immunology
- CD11a Antigen/metabolism
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/pathology
- Cell Differentiation/immunology
- Cell Movement/drug effects
- Cell Movement/immunology
- Cell Proliferation
- DNA-Binding Proteins/genetics
- Fingolimod Hydrochloride
- Granzymes/metabolism
- Hyaluronan Receptors/metabolism
- Immunosuppressive Agents/pharmacology
- Integrin alpha4/metabolism
- Interferon-gamma/metabolism
- Lymph Nodes/cytology
- Lymph Nodes/drug effects
- Lymph Nodes/immunology
- Lymph Nodes/pathology
- Lymphocyte Activation/immunology
- Lysosomal Membrane Proteins/metabolism
- Melanoma, Experimental/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Monophenol Monooxygenase/immunology
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Ovalbumin/immunology
- Peptide Fragments/immunology
- Propylene Glycols/pharmacology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Sphingosine/analogs & derivatives
- Sphingosine/pharmacology
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- beta 2-Microglobulin/genetics
Collapse
Affiliation(s)
- Elizabeth D Thompson
- Department of Microbiology and Carter Immunology Center, University of Virginia, Charlottesville, VA 22908, USA
| | | | | | | |
Collapse
|
20
|
Abstract
Cross-priming is an important mechanism to activate cytotoxic T lymphocytes (CTLs) for immune defence against viruses and tumours. Although it was discovered more than 25 years ago, we have only recently gained insight into the underlying cellular and molecular mechanisms, and we are just beginning to understand its physiological importance in health and disease. Here we summarize current concepts on the cross-talk between the immune cells involved in CTL cross-priming and on its role in antimicrobial and antitumour defence, as well as in immune-mediated diseases.
Collapse
|
21
|
Contassot E, Preynat-Seauve O, French L, Huard B. Lymph node tumor metastases: more susceptible than primary tumors to CD8+ T-cell immune destruction. Trends Immunol 2009; 30:569-73. [DOI: 10.1016/j.it.2009.08.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Revised: 08/25/2009] [Accepted: 08/28/2009] [Indexed: 01/21/2023]
|
22
|
Murillo O, Dubrot J, Palazón A, Arina A, Azpilikueta A, Alfaro C, Solano S, Ochoa MC, Berasain C, Gabari I, Pérez-Gracia JL, Berraondo P, Hervás-Stubbs S, Melero I. In vivo depletion of DC impairs the anti-tumor effect of agonistic anti-CD137 mAb. Eur J Immunol 2009; 39:2424-36. [PMID: 19662633 DOI: 10.1002/eji.200838958] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Anti-CD137 mAb are capable of inducing tumor rejection in several syngeneic murine tumor models and are undergoing clinical trials for cancer. The anti-tumor effect involves co-stimulation of tumor-specific CD8(+) T cells. Whether antigen cross-presenting DC are required for the efficacy of anti-CD137 mAb treatment has never been examined. Here we show that the administration of anti-CD137 mAb eradicates EG7-OVA tumors by a strictly CD8beta(+) T-cell-dependent mechanism that correlates with increased CTL activity. Ex vivo analyses to determine the identity of the draining lymph node cell type responsible for tumor antigen cross-presentation revealed that CD11c(+) cells, most likely DC, are the main players in this tumor model. A minute number of tumor cells, revealed by the presence of OVA cDNA, reach tumor-draining lymph nodes. Direct antigen presentation by tumor cells themselves also participates in anti-OVA CTL induction. Using CD11c diphtheria toxin receptor-green fluorescent protein-->C57BL/6 BM chimeric mice, which allow for sustained ablation of DC with diphtheria toxin, we confirmed the involvement of DC in tumor antigen cross-presentation in CTL induction against OVA(257-264) epitope and in the antitumor efficacy induced by anti-CD137 mAb.
Collapse
Affiliation(s)
- Oihana Murillo
- Gene Therapy Unit, Centro de Investigación Médica Aplicada, Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Le TT, Gardner J, Hoang-Le D, Schmidt CW, MacDonald KP, Lambley E, Schroder WA, Ogbourne SM, Suhrbier A. Immunostimulatory cancer chemotherapy using local ingenol-3-angelate and synergy with immunotherapies. Vaccine 2009; 27:3053-62. [DOI: 10.1016/j.vaccine.2009.03.025] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 03/11/2009] [Accepted: 03/15/2009] [Indexed: 01/17/2023]
|
24
|
Broomfield SA, van der Most RG, Prosser AC, Mahendran S, Tovey MG, Smyth MJ, Robinson BWS, Currie AJ. Locally Administered TLR7 Agonists Drive Systemic Antitumor Immune Responses That Are Enhanced by Anti-CD40 Immunotherapy. THE JOURNAL OF IMMUNOLOGY 2009; 182:5217-24. [DOI: 10.4049/jimmunol.0803826] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
25
|
Jackaman C, Lew AM, Zhan Y, Allan JE, Koloska B, Graham PT, Robinson BWS, Nelson DJ. Deliberately provoking local inflammation drives tumors to become their own protective vaccine site. Int Immunol 2008; 20:1467-79. [DOI: 10.1093/intimm/dxn104] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
26
|
Sheu BC, Kuo WH, Chen RJ, Huang SC, Chang KJ, Chow SN. Clinical significance of tumor-infiltrating lymphocytes in neoplastic progression and lymph node metastasis of human breast cancer. Breast 2008; 17:604-10. [PMID: 18656354 DOI: 10.1016/j.breast.2008.06.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2007] [Revised: 01/30/2008] [Accepted: 06/09/2008] [Indexed: 12/15/2022] Open
Abstract
To investigate the clinical significance of tumor-infiltrating lymphocytes (TILs) within the tumor milieu, we quantitatively measured and compared the subpopulations of TILs in 24 patients with stage I-III breast carcinoma. Peripheral blood mononuclear cells (PBMCs), normal breast parenchyma-infiltrating lymphocytes (NILs), and TILs were isolated from tissue specimens and quantified by flow cytometry. The results showed that increased proportion of CD8(+) T cells, with decreased proportion of CD4(+) T cells, was significant in gated CD3(+) TILs as compared to autologous NILs or PBMCs (P<0.001). The tumor-infiltrating CD8(+) T cells significantly increased with stage progression, reflected in a more strongly decreased CD4/CD8 percentage (P=0.003). The CD4/CD8 percentage of TILs was strongly correlated with lymphovascular permeation and subsequent lymph node metastasis (P<0.001). Increased percentages of tumor-infiltrating CD8(+) T cells with decreased CD4/CD8 percentages are of prognostic importance for cancer progression in human breast cancer.
Collapse
Affiliation(s)
- Bor-Ching Sheu
- Department of Obstetrics & Gynecology, College of Medicine and The Hospital, National Taiwan University, Taipei 100, Taiwan.
| | | | | | | | | | | |
Collapse
|
27
|
Müller-Hermelink N, Braumüller H, Pichler B, Wieder T, Mailhammer R, Schaak K, Ghoreschi K, Yazdi A, Haubner R, Sander CA, Mocikat R, Schwaiger M, Förster I, Huss R, Weber WA, Kneilling M, Röcken M. TNFR1 signaling and IFN-gamma signaling determine whether T cells induce tumor dormancy or promote multistage carcinogenesis. Cancer Cell 2008; 13:507-18. [PMID: 18538734 DOI: 10.1016/j.ccr.2008.04.001] [Citation(s) in RCA: 233] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2004] [Revised: 05/23/2007] [Accepted: 04/08/2008] [Indexed: 12/12/2022]
Abstract
Immune responses may arrest tumor growth by inducing tumor dormancy. The mechanisms leading to either tumor dormancy or promotion of multistage carcinogenesis by adaptive immunity are poorly characterized. Analyzing T antigen (Tag)-induced multistage carcinogenesis in pancreatic islets, we show that Tag-specific CD4+ T cells home selectively into the tumor microenvironment around the islets, where they either arrest or promote transition of dysplastic islets into islet carcinomas. Through combined TNFR1 signaling and IFN-gamma signaling, Tag-specific CD4+ T cells induce antiangiogenic chemokines and prevent alpha(v)beta(3) integrin expression, tumor angiogenesis, tumor cell proliferation, and multistage carcinogenesis, without destroying Tag-expressing islet cells. In the absence of either TNFR1 signaling or IFN-gamma signaling, the same T cells paradoxically promote angiogenesis and multistage carcinogenesis. Thus, tumor-specific T cells can directly survey multistage carcinogenesis through cytokine signaling.
Collapse
MESH Headings
- Animals
- Antigens, Viral, Tumor/genetics
- Antigens, Viral, Tumor/metabolism
- Blood Glucose/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD4-Positive T-Lymphocytes/transplantation
- Cell Movement
- Cell Proliferation
- Cell Survival
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/immunology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/pathology
- Cells, Cultured
- GTPase-Activating Proteins/genetics
- GTPase-Activating Proteins/metabolism
- Immunotherapy/methods
- Insulinoma/blood supply
- Insulinoma/genetics
- Insulinoma/immunology
- Insulinoma/metabolism
- Insulinoma/pathology
- Insulinoma/therapy
- Integrin alphaVbeta3/metabolism
- Interferon-gamma/metabolism
- Mice
- Mice, Inbred C3H
- Mice, Knockout
- Mice, Transgenic
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Pancreatic Neoplasms/blood supply
- Pancreatic Neoplasms/genetics
- Pancreatic Neoplasms/immunology
- Pancreatic Neoplasms/metabolism
- Pancreatic Neoplasms/pathology
- Pancreatic Neoplasms/therapy
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction
- Th1 Cells/immunology
- Th1 Cells/pathology
- Time Factors
- Whole-Body Irradiation
Collapse
Affiliation(s)
- Nele Müller-Hermelink
- Department of Dermatology, Eberhard Karls University, Liebermeisterstrasse 25, 72076 Tübingen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Currie AJ, van der Most RG, Broomfield SA, Prosser AC, Tovey MG, Robinson BWS. Targeting the effector site with IFN-alphabeta-inducing TLR ligands reactivates tumor-resident CD8 T cell responses to eradicate established solid tumors. THE JOURNAL OF IMMUNOLOGY 2008; 180:1535-44. [PMID: 18209049 DOI: 10.4049/jimmunol.180.3.1535] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Effective antitumor CD8 T cell responses may be activated by directly targeting the innate immune system within tumors. We investigated this response by injecting a range of TLR agonists into established tumors using a mouse model of malignant mesothelioma stably transduced with the hemagglutinin (HA) gene as a marker Ag (AB1-HA). Persistent delivery of the dsRNA mimetic poly(I:C) into established AB1-HA tumors resulted in complete tumor resolution in 40% of mice, with the remaining mice also showing a significant delay in tumor progression. Experiments in athymic nude mice along with CD8 depletion and IFN-alphabeta blocking studies revealed that tumor resolution required both CD8 T cells and type I IFN induction, and was associated with local changes in MHC class I expression. Surprisingly, however, tumor resolution was not associated with systemic dissemination or tumor infiltration of effector CD8 T cells. Instead, the antitumor response was critically dependent on the reactivation of tumor-resident CD8 T cell responses. These studies suggest that, once reactivated, pre-existing local CD8 T cell responses are sufficient to resolve established tumors and that in situ type I IFN is a determining factor.
Collapse
Affiliation(s)
- Andrew J Currie
- National Research Centre for Asbestos Related Diseases, QEII Medical Centre, Verdun Street, Nedlands, Perth 6009, Western Australia.
| | | | | | | | | | | |
Collapse
|
29
|
An allogeneic hybrid-cell fusion vaccine against canine mammary cancer. Vet Immunol Immunopathol 2008; 123:289-304. [PMID: 18423623 DOI: 10.1016/j.vetimm.2008.02.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2007] [Revised: 11/29/2007] [Accepted: 02/13/2008] [Indexed: 11/22/2022]
Abstract
Mammary cancer is among the most prevalent of canine tumors frequently resulting in death due to metastatic disease. Most tumors fail to raise an effective immune reaction making improving immune recognition a priority. Hybrid-cell fusion strategies have been employed to load dendritic cell populations with tumor cell antigens to stimulate immune recognition; however, recovery, heterogeneity and quality of primary cells from patients present enormous challenges. We employed allogeneic cell lines to develop an improved hybrid-cell fusion strategy and evaluated immune reactions in normal laboratory beagles. Such a strategy relies on enhanced immune recognition of allogeneic tumor cell antigens by antigen presenting cells. Optimized PEG-promoted fusions between uniquely stained canine mammary tumor CMT12 or CMT28 cells and a dendritic cell-like DH82 cell fusion partner resulted in greater than 40% hybrid-cell fusion populations by flow cytometry and fluorescence microscopy. Hybrid-cell fusions were delivered by direct ultrasound guided injection into popliteal lymph nodes of laboratory beagles. Only hybrid-cell fusions provided statistically significant enhancement of cell-mediated immunity ((51)Cr-release assay) compared to innate reactions in naïve vehicle injected dogs while dogs vaccinated with either single cell component alone did not. Vaccination with hybrid-cell fusions enhanced IFN-gamma expression in sorted CD8+ and CD4+ cells but not in CD4-/CD8- cells consistent with a CTL response. Cell-mediated immune assays revealed strong reactions against matched (vaccine component) CMT cells and unmatched CMT cells indicative of an immune response to mammary cancer antigens common to both cell lines. These results provide proof of principle for development of an allogeneic vaccination strategy against canine mammary cancer.
Collapse
|
30
|
Perez-Diez A, Joncker NT, Choi K, Chan WFN, Anderson CC, Lantz O, Matzinger P. CD4 cells can be more efficient at tumor rejection than CD8 cells. Blood 2007; 109:5346-54. [PMID: 17327412 PMCID: PMC1890845 DOI: 10.1182/blood-2006-10-051318] [Citation(s) in RCA: 333] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Researchers designing antitumor treatments have long focused on eliciting tumor-specific CD8 cytotoxic T lymphocytes (CTL) because of their potent killing activity and their ability to reject transplanted organs. The resulting treatments, however, have generally been surprisingly poor at inducing complete tumor rejection, both in experimental models and in the clinic. Although a few scattered studies suggested that CD4 T "helper" cells might also serve as antitumor effectors, they have generally been studied mostly for their ability to enhance the activity of CTL. In this mouse study, we compared monoclonal populations of tumor-specific CD4 and CD8 T cells as effectors against several different tumors, and found that CD4 T cells eliminated tumors that were resistant to CD8-mediated rejection, even in cases where the tumors expressed major histocompatibility complex (MHC) class I molecules but not MHC class II. MHC class II expression on host tissues was critical, suggesting that the CD4 T cells act indirectly. Indeed, the CD4 T cells partnered with NK cells to obtain the maximal antitumor effect. These findings suggest that CD4 T cells can be powerful antitumor effector cells that can, in some cases, outperform CD8 T cells, which are the current "gold standard" effector cell in tumor immunotherapy.
Collapse
Affiliation(s)
- Ainhoa Perez-Diez
- Ghost Lab, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
31
|
Yang MY, Zetler PM, Prins RM, Khan-Farooqi H, Liau LM. Immunotherapy for patients with malignant glioma: from theoretical principles to clinical applications. Expert Rev Neurother 2007; 6:1481-94. [PMID: 17078788 DOI: 10.1586/14737175.6.10.1481] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Malignant gliomas are the most common type of primary brain tumor and are in great need of novel therapeutic approaches. Advances in treatment have been very modest, significant improvement in survival has been lacking for many decades and prognosis remains dismal. Despite 'gross total' surgical resections and currently available radio-chemotherapy, malignant gliomas inevitably recur due to reservoirs of notoriously invasive tumor cells that infiltrate adjacent and nonadjacent areas of normal brain parenchyma. In principle, the immune system is uniquely qualified to recognize and target these infiltrative pockets of tumor cells, which have generally eluded conventional treatment approaches. In the span of the last 10 years, our understanding of the cancer-immune system relationship has increased exponentially, and yet, we are only beginning to tease apart the intricacies of the CNS and immune cell interactions. This article reviews the complex associations of the immune system with brain tumors. We provide an overview of currently available treatment options for malignant gliomas, existing gaps in our knowledge of brain tumor immunology, and molecular techniques and targets that might be exploited for improved patient stratification and design of 'custom immunotherapeutics'. We will also examine major new immunotherapy approaches that are being actively investigated to treat patients with malignant glioma, and identify some current and future research priorities in this area.
Collapse
Affiliation(s)
- Meng-Yin Yang
- Division of Neurosurgery, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095-6901, USA.
| | | | | | | | | |
Collapse
|
32
|
Abstract
Lymph nodes that lie immediately downstream of tumors [tumor-draining lymph nodes (TDLNs)] undergo profound alterations due to the presence of the upstream tumor. The antigen-presenting cell population in TDLNs becomes modified such that tumor-derived antigens are cross-presented by host cells in a tolerizing fashion. In addition, the number and suppressor activity of regulatory T cells (Tregs) are increased in the TDLN. Emerging evidence suggests that some of these Tregs may be generated de novo against specific tumor-derived antigens, and thus they arise as a direct consequence of antigen presentation in the TDLN. Others may represent Tregs against self-antigens, which undergo preferential activation in the tolerogenic milieu of the TDLN. The TDLN thus becomes an anatomic context in which presentation of new antigens not only fails to elicit a protective immune response but also actively creates systemic tolerance. In this regard, the TDLN displays features analogous to classical immune privilege. Accumulating evidence thus suggests that the TDLNs, although small in size, may exert a profound tolerizing influence on the rest of the immune system. These mechanisms will need to be interrupted in order for clinical anti-tumor immunotherapy to be successful.
Collapse
Affiliation(s)
- David H Munn
- Immunotherapy Center, Department of Pediatrics, Medical College of Georgia, Augusta, GA 30912, USA.
| | | |
Collapse
|
33
|
Hargadon KM, Brinkman CC, Sheasley-O'neill SL, Nichols LA, Bullock TNJ, Engelhard VH. Incomplete differentiation of antigen-specific CD8 T cells in tumor-draining lymph nodes. THE JOURNAL OF IMMUNOLOGY 2006; 177:6081-90. [PMID: 17056534 DOI: 10.4049/jimmunol.177.9.6081] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CD8 T cells lacking effector activity have been recovered from lymphoid organs of mice and patients with progressing tumors. We explored the basis for lack of effector activity in tumor-bearing mice by evaluating Ag presentation and CD8 T cell function in lymphoid organs over the course of tumor outgrowth. Early after tumor injection, cross-presentation by bone marrow-derived APC was necessary for T cell activation, inducing proliferation and differentiation into IFN-gamma-producing, cytolytic effectors. At later stages of outgrowth, tumor metastasized to draining lymph nodes. Both cross- and direct presentation occurred, but T cell differentiation induced by either modality was incomplete (proliferation without cytokine production). T cells within tumor-infiltrated nodes differentiated appropriately if Ag was presented by activated, exogenous dendritic cells. Thus, activated T cells lacking effector function develop through incomplete differentiation in the lymph nodes of late-stage tumor-bearing mice, rather than through suppression of previously differentiated cells.
Collapse
Affiliation(s)
- Kristian M Hargadon
- Department of Microbiology and Carter Immunology Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | | | | | |
Collapse
|
34
|
Bundell CS, Jackaman C, Suhrbier A, Robinson BWS, Nelson DJ. Functional endogenous cytotoxic T lymphocytes are generated to multiple antigens co-expressed by progressing tumors; after intra-tumoral IL-2 therapy these effector cells eradicate established tumors. Cancer Immunol Immunother 2006; 55:933-47. [PMID: 16283304 PMCID: PMC11030810 DOI: 10.1007/s00262-005-0086-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2005] [Accepted: 09/14/2005] [Indexed: 11/24/2022]
Abstract
Tumors contain many antigens that may be recognized by the immune system. It is not known whether these antigens, and the epitopes within these antigens, can all be recognized by the anti-tumor immune response or if such responses are restricted to a few dominant epitopes. Effector function of endogenous cytotoxic T lymphocytes (CTL) generated during tumor progression has previously been assessed by indirect, ex vivo assays, which often focused on a single antigen. Therefore, we evaluated the endogenous in vivo CTL response to multiple neo tumor antigens using murine Lewis lung carcinoma tumor cells transfected with ovalbumin or a polyepitope construct. Both express multiple MHC class I-restricted epitopes. Ovalbumin contains a known hierarchy of epitopes for given MHC molecules, whilst the polyepitope expresses a number of dominant epitopes. We show that as tumors progress, potent effector CTL are generated in vivo that are restricted to dominant epitopes; we did not see the responses to subdominant or cryptic epitopes. Our data show that the CTL recognizing tumor antigens vary in their lytic capacity, as the CTL responding to two of the four epitopes were particularly potent killers. The presence of these effector CTLs did not prevent tumor growth. However, intra-tumoral IL-2 treatment altered the potency, but not the hierarchy, of these CTL such that they mediated tumor regression. These results have implications for immunotherapy protocols.
Collapse
Affiliation(s)
- Christine S. Bundell
- School of Medicine and Pharmacology, University of Western Australia, 4th Floor, G Block, Queen Elizabeth II Medical Centre, Perth, WA 6009 Australia
| | - Connie Jackaman
- School of Medicine and Pharmacology, University of Western Australia, 4th Floor, G Block, Queen Elizabeth II Medical Centre, Perth, WA 6009 Australia
| | - Andreas Suhrbier
- Queensland Institute of Medical Research, Post Office Royal Brisbane Hospital, Brisbane, QLD 4029 Australia
| | - Bruce W. S. Robinson
- School of Medicine and Pharmacology, University of Western Australia, 4th Floor, G Block, Queen Elizabeth II Medical Centre, Perth, WA 6009 Australia
- West Australian Institute for Medical Research, Queen Elizabeth II Medical Centre, Perth, WA 6009 Australia
| | - Delia J. Nelson
- School of Medicine and Pharmacology, University of Western Australia, 4th Floor, G Block, Queen Elizabeth II Medical Centre, Perth, WA 6009 Australia
- Western Australian Biomedical Research Institute, Kent St., Curtin University, Bentley, WA 6102 Australia
- School of Biomedical Sciences, Kent St., Curtin University, Bentley, WA 6102 Australia
| |
Collapse
|
35
|
Joncker NT, Helft J, Jacquet A, Premel V, Lantz O. Intratumor CD4 T-Cell Accumulation Requires Stronger Priming than for Expansion and Lymphokine Secretion. Cancer Res 2006; 66:5443-51. [PMID: 16707473 DOI: 10.1158/0008-5472.can-05-3526] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
T cells need to migrate to and accumulate inside tumors before mediating rejection of the tumor. The number of specific T cells inside tumors may depend on the efficiency of priming in the draining lymph node (DLN), intratumor deletion, suppressive phenomena, or both. We used monoclonal anti-male antigen CD4 (Marilyn) T cells and tumor cell lines expressing or not the corresponding antigen (Dby) to analyze CD4 T-cell accumulation in tumors. Priming by MHC II(+) or MHC II(-) male splenocytes or Dby(+) tumor cells induced similar Marilyn T-cell expansion in the DLN and recirculation in other lymph nodes and capacity to produce IFN-gamma. However, intratumor accumulation was different for each priming condition. In mice with Dby(-) tumors, MHC II(+) male splenocyte priming induced greater, although not statistically significant, Marilyn T-cell accumulation in the tumors than MHC II(-) male splenocyte priming. In mice with Dby(+) tumors, priming in the tumor DLN induced less Marilyn T-cell intratumor accumulation than priming by MHC II(+) male splenocytes. We saw comparable differences for Marilyn T-cell accumulation in gut lamina propria, suggesting that priming affects effector T-cell accumulation in inflamed tissues. Mature dendritic cells were loaded with graded doses of Dby peptide to control for antigen-presenting cell characteristics during priming. We observed similar proliferation, with higher concentrations inducing higher intratumor accumulation. Thus, intratumor accumulation requires stronger stimulation than for proliferation or the capacity to secrete lymphokines. In this system, priming intensity alone can explain the number of intratumor T cells without having to call for intratumor deletion or suppression phenomena.
Collapse
Affiliation(s)
- Nathalie T Joncker
- Laboratoire d'Immunologie pré-clinique, Institut National de la Sante et de la Recherche Medicale U653, Institut Curie, Paris, France
| | | | | | | | | |
Collapse
|
36
|
Castano AP, Liu Q, Hamblin MR. A green fluorescent protein-expressing murine tumour but not its wild-type counterpart is cured by photodynamic therapy. Br J Cancer 2006; 94:391-7. [PMID: 16421588 PMCID: PMC2361144 DOI: 10.1038/sj.bjc.6602953] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The ideal cancer treatment should both destroy the primary tumour and at the same time educate the immune system to recognise the tumour as foreign so that distant metastases will also be eradicated. Photodynamic therapy (PDT) involves the i.v. administration of photosensitisers followed by illumination of the tumour with red light producing reactive oxygen species that eventually cause vascular shutdown and tumour cell death by apoptosis and necrosis. Anti-tumour immunity is stimulated after PDT due to the acute inflammatory response, generation of tumour-specific antigens, and induction of heat-shock proteins. Green fluorescent protein (GFP) is used as an optical reporter to noninvasively image the progression of mouse tumours, and in addition, may act as a foreign (jellyfish) antigen. We asked whether GFP-expressing tumours could be used to monitor the response of tumour-bearing mice to PDT, and whether the tumour response differed when a nonimmunogenic tumour cell line was transduced with GFP. We injected RIF-1 or RIF-1 EGFP (stably transduced with a retroviral vector) cells in the leg of C3H/HeN mice and both the cells and tumour grew equally well. We used PDT with benzoporphyrin derivative and a short drug-light interval. There were complete cures and 100% mouse survival of RIF-1 EGFP while RIF-1 wild-type tumours all recurred. Cured mice were resistant to rechallenge with RIF-1 EGFP cells and a rechallenge with wild-type RIF-1 cells grew significantly slower. There was also slower RIF-1 EGFP rechallenge growth but no rejection when RIF-1 EGFP tumours were surgically removed. There was a low rate of PDT cure of tumours when RIF-1 cells were transduced with an empty retroviral vector. The presence of antibodies against EGFP in mouse serum suggests EGFP can act as a foreign antigen and PDT can then stimulate a long-term memory immune response.
Collapse
Affiliation(s)
- A P Castano
- BAR414, Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - Q Liu
- BAR414, Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
| | - M R Hamblin
- BAR414, Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA
- Department of Dermatology, Harvard Medical School, Boston, MA, USA
- Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
- BAR414, Wellman Center for Photomedicine, Massachusetts General Hospital, 40 Blossom Street, Boston, MA 02114, USA. E-mail:
| |
Collapse
|
37
|
Needham DJ, Lee JX, Beilharz MW. Intra-tumoural regulatory T cells: a potential new target in cancer immunotherapy. Biochem Biophys Res Commun 2006; 343:684-91. [PMID: 16563349 DOI: 10.1016/j.bbrc.2006.03.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2006] [Accepted: 03/03/2006] [Indexed: 12/18/2022]
Abstract
We hypothesised that T(reg) cells preferentially expand/infiltrate inside murine mesotheliomas. Immunotherapy based on the manipulation of T(reg) cell populations should therefore be targeted to the tumour site. The AE17 murine mesothelioma model was used for this study. Both intra-tumoural T(reg) cells and those in the periphery of tumour-bearing mice were identified by flow cytometry. The effect on tumour growth of intra-tumoural depletion of T(reg) cells using the PC61 anti-CD25 mAb was then examined. We identified CD4+ T(reg) cells co-expressing both the CD25 cell surface marker and the transcription factor Foxp3 within murine mesotheliomas. These intra-tumoural T(reg) cells increase significantly as a percentage of total CD4+ T cells within the tumour as it grows. We showed that the depletion of intra-tumoural T(reg) cells with anti-CD25 mAb injected directly into the tumours can cause significantly reduced tumour growth. Localised, intra-tumoural depletion of T(reg) cells is a new, clinically relevant treatment option for established tumours.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/therapeutic use
- Cell Proliferation
- Female
- Forkhead Transcription Factors/metabolism
- Lymphocyte Depletion
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Mesothelioma/drug therapy
- Mesothelioma/immunology
- Mesothelioma/pathology
- Mice
- Mice, Inbred C57BL
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Receptors, Interleukin-2/analysis
- Receptors, Interleukin-2/antagonists & inhibitors
- Receptors, Interleukin-2/immunology
- T-Lymphocyte Subsets/drug effects
- T-Lymphocyte Subsets/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Demelza J Needham
- Discipline of Microbiology and Immunology, School of Biomedical, Biomolecular and Chemical Sciences, The University of Western Australia, Perth, WA, Australia
| | | | | |
Collapse
|
38
|
Chiou SH, Sheu BC, Chang WC, Huang SC, Hong-Nerng H. Current concepts of tumor-infiltrating lymphocytes in human malignancies. J Reprod Immunol 2005; 67:35-50. [PMID: 16111767 DOI: 10.1016/j.jri.2005.06.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumor-infiltrating lymphocytes (TILs) develop as manifestations of the recognition and defense against malignant cells by the host immune system. TILs were literally defined as "tumor-infiltrating lymphocytes", which a posteriori locate within the tumor tissues. Although such cells can be found, they fail to control the growth of tumor. Many have proposed diverse mechanisms for dysfunction of TILs with regard to the roles of immunosurveillance against cancer. However, only a few cancer types, e.g. melanoma, have seen the benefits brought by activating these cells for immunotherapy. Functional defects of TILs have been linked to abnormalities of signaling molecules; however, there is conflicting data. The death of TILs was attributed to expression of cancer-derived FasL, PD-1 and RCAS1, and cancer-induced activation-induced cell death (AICD). Confirmed by studies using TILs and animal models, the compromise of tumor-specific immune responses was thought to result from not only mechanisms of clonal anergy but also exhaustion and/or deletion. Furthermore, functional cytotoxic CD8(+) TILs might be rendered incompetent by cancer-induced up-regulation of inhibitory NK receptors or proximal signaling abnormalities. Additionally, immune privilege was partly attributed to recruitment of regulatory T cells to the tumor sites. The failure of IL-2 signaling, which stands at the center of T cell functionalities, had been linked to the enzymatic activity of cancer-derived matrix metalloproteinases (MMPs). Finally, the exploitation of IDO expression, an important enzyme in pregnancy-related immunosuppression, by cancer cells might play a role in tumor immunity. The disparity of cancer types, origin, developmental stages and individual genetic backgrounds likely account for differences, or even contradictions, which might be the reason why immunotherapy works only on a few cancer types. Delineating the mechanisms behind functional defects of TILs can help not only boost chances of the development of a successful cure but understand the not fully identified roles played by immune system in the face of malignancies.
Collapse
Affiliation(s)
- Shin-Heng Chiou
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, 7 Chung-Shan South Road, Taipei 100, Taiwan
| | | | | | | | | |
Collapse
|
39
|
Liu K, Caldwell SA, Abrams SI. Immune Selection and Emergence of Aggressive Tumor Variants as Negative Consequences of Fas-Mediated Cytotoxicity and Altered IFN-γ-Regulated Gene Expression. Cancer Res 2005; 65:4376-88. [PMID: 15899830 DOI: 10.1158/0008-5472.can-04-4269] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Antitumor responses can be induced in patients via active or adoptive immunotherapy, yet complete tumor eradication occurs infrequently. This paradox in tumor immunology led us to address two questions: (a) Does an antitumor response, which is intended to destroy the aberrant target population, also at the same time select for aggressive tumor variants (ATV) in vivo? (b) If this process does occur, what is the contribution of the perforin- or Fas-mediated effector mechanism in the immune selection of such ATV? Here, in an experimental mouse lung metastasis model, we showed that ATV generated either naturally in vivo or in vitro by anti-Fas selection resembled each other biologically and genetically as judged by enhanced tumor growth and genome-scale gene expression profiling, respectively. Furthermore, ATV that survived CTL adoptive immunotherapy displayed an even more profound loss of Fas expression and function as well as enhanced malignant proficiency in vivo. ATV, however, retained sensitivity to perforin-mediated lysis in vitro. Lastly, such ATV displayed a diminished responsiveness in their expression of IFN-gamma-regulated genes, including those mechanistically linked to Fas-mediated death (i.e., Fas and caspase-1). Overall, we showed that (a) immune selection did occur in vivo and played an important role in the emergence of ATV, (b) ATV bearing a Fas-resistant phenotype was a chief consequence of immune selection, and (c) an overall diminished responsiveness of IFN-gamma-regulated gene expression was characteristic of ATV. Thus, in this model, Fas-mediated cytotoxicity, in concert with IFN-gamma-regulated gene expression, mechanistically constituted significant determinants of immune selection of ATV in vivo.
Collapse
Affiliation(s)
- Kebin Liu
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892-1402, USA
| | | | | |
Collapse
|
40
|
Jenkinson SR, Williams NA, Morgan DJ. The Role of Intercellular Adhesion Molecule-1/LFA-1 Interactions in the Generation of Tumor-Specific CD8+ T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2005; 174:3401-7. [PMID: 15749873 DOI: 10.4049/jimmunol.174.6.3401] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The activation of naive CD4+ T cells requires both TCR engagement and a second costimulatory signal mediated by the interaction of CD28 with CD80/CD86 expressed on professional APC. However, the situation for naive CD8+ T cells is less clear. Although evidence indicates that induction of CD8+ T cell responses is also dependent on professional APC, the ability of some tumors, which do not express CD80/CD86, to induce CTL suggests that other pathways of costimulation exist for the activation of CD8+ T cells. We examined the ability of tumor cells expressing different levels of a tumor-specific Ag to directly prime CD8+ T cells. We demonstrate that CD8+ T cells are directly activated by tumor cells in a CD80/CD86-CD28 independent manner. In this system, costimulation requires ICAM-1/LFA-1 interaction. This results in the generation of CTL capable of inhibiting tumor growth in vivo, and maintaining long-term survival.
Collapse
Affiliation(s)
- S Rhiannon Jenkinson
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, Bristol, United Kingdom
| | | | | |
Collapse
|
41
|
Hirano F, Kaneko K, Tamura H, Dong H, Wang S, Ichikawa M, Rietz C, Flies DB, Lau JS, Zhu G, Tamada K, Chen L. Blockade of B7-H1 and PD-1 by Monoclonal Antibodies Potentiates Cancer Therapeutic Immunity. Cancer Res 2005. [DOI: 10.1158/0008-5472.1089.65.3] [Citation(s) in RCA: 239] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Abstract
Contemporary approaches for vaccination and immunotherapy are often capable of eliciting strong T-cell responses against tumor antigens. However, such responses are not parallel to clinical tumor regression. The development of evasion mechanisms within tumor microenvironment may be responsible for poor therapeutic responses. We report here that constitutive or inducible expression of B7-H1, a B7 family molecule widely expressed by cancers, confers resistance to therapeutic anti-CD137 antibody in mice with established tumors. The resistance is accompanied with failure of antigen-specific CD8+ CTLs to destroy tumor cells without impairment of CTL function. Blockade of B7-H1 or PD-1 by specific monoclonal antibodies could reverse this resistance and profoundly enhance therapeutic efficacy. Our findings support that B7-H1/PD-1 forms a molecular shield to prevent destruction by CTLs and implicate new approaches for immunotherapy of human cancers.
Collapse
Affiliation(s)
- Fumiya Hirano
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
| | - Katsumi Kaneko
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
| | - Hideto Tamura
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
| | - Haidong Dong
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
| | - Shengdian Wang
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
- 2The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Masao Ichikawa
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
- 2The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Cecilia Rietz
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
- 2The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Dallas B. Flies
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
- 2The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Julie S. Lau
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
| | - Gefeng Zhu
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
- 2The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Koji Tamada
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
- 2The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - Lieping Chen
- 1Department of Immunology, Mayo Clinic, Rochester, Minnesota and
- 2The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| |
Collapse
|
42
|
Jackson DC, Lau YF, Le T, Suhrbier A, Deliyannis G, Cheers C, Smith C, Zeng W, Brown LE. A totally synthetic vaccine of generic structure that targets Toll-like receptor 2 on dendritic cells and promotes antibody or cytotoxic T cell responses. Proc Natl Acad Sci U S A 2004; 101:15440-5. [PMID: 15489266 PMCID: PMC523460 DOI: 10.1073/pnas.0406740101] [Citation(s) in RCA: 198] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2004] [Indexed: 12/29/2022] Open
Abstract
A simple generic peptide-based vaccine structure that targets Toll-like receptor 2-expressing dendritic cells and causes their activation is described. The vaccines are totally synthetic, serve as their own adjuvant, and are composed of (i) a single helper T cell epitope, (ii) a target epitope that is either recognized by CD8+ T cells or B cells, and (iii) a Toll-like receptor 2-targeting lipid moiety, S-[2,3-bis(palmitoyloxy)propyl]cysteine, that is situated between the peptide epitopes to form a branched configuration. The different CD8+ T cell epitopes examined were from (i) influenza virus, (ii) the intracellular bacterium Listeria monocytogenes, and (iii) ovalbumin as a model tumor antigen. Vaccines containing a B cell epitope from gastrin or luteinizing hormone-releasing hormone as a B cell epitope were also examined for their ability to elicit antibody against the parent hormones. Each of the vaccines was capable of inducing either CD8+ T cell or antibody-mediated immune responses. The lipidated vaccines, but not the nonlipidated vaccines, were able to mediate protection against viral or bacterial infection and mediate prophylactic and therapeutic anticancer activity. The two hormone-based vaccines induced high antibody titers, which in the case of luteinizing hormone-releasing hormone resulted in abrogation of reproductive function. These results highlight the utility of simple, totally synthetic, epitope-based vaccines.
Collapse
Affiliation(s)
- David C Jackson
- Cooperative Research Centre for Vaccine Technology and Department of Microbiology and Immunology, University of Melbourne, Parkville, Victoria 3010, Australia.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Lenarczyk A, Le TTT, Drane D, Malliaros J, Pearse M, Hamilton R, Cox J, Luft T, Gardner J, Suhrbier A. ISCOM based vaccines for cancer immunotherapy. Vaccine 2004; 22:963-74. [PMID: 15161073 DOI: 10.1016/j.vaccine.2003.09.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Immunostimulating complex (ISCOM) vaccines are particulate antigen delivery vehicles composed of saponin, cholesterol, phospholipid and immunogen. Here we illustrate that ISCOM-based vaccines represent an attractive modality for the development of anti-cancer vaccines. Using murine models and a model cancer antigen, ISCOM vaccines were shown to induce potent CD8 T cell responses, to mediate protection in three different tumor models, to promote Th1-biased immunity, and to induce CD8 T cell responses in the absence of CD4+ T cell help. The former three activities were also found to be substantially improved when the vaccine antigen was associated with the ISCOM structure. Furthermore, the presence in vivo of pre-existing antibodies against the vaccine antigen did not inhibit CD8 T cell induction by the ISCOM vaccine. Although vaccination was effective against challenge with vaccine-antigen expressing tumors, no activity against neighboring vaccine-antigen negative tumor cells was observed, indicating that determinant spreading or bystander activity does not lead to significant anti-cancer activity.
Collapse
MESH Headings
- Animals
- Antibodies, Neoplasm/blood
- Antigens, Neoplasm/chemistry
- Antigens, Neoplasm/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/chemistry
- Cancer Vaccines/immunology
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/prevention & control
- Epitopes
- Female
- ISCOMs/chemistry
- ISCOMs/immunology
- Immunoglobulin G/immunology
- Injections, Subcutaneous
- Melanoma, Experimental/immunology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Ovalbumin/chemistry
- Ovalbumin/immunology
- Time Factors
- Vaccination
Collapse
Affiliation(s)
- Aleksandra Lenarczyk
- Cooperative Research Center for Vaccine Technology, Queensland Institute of Medical Research and the University of Queensland, Queensland 4029, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, Messina JL, Chandler P, Koni PA, Mellor AL. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest 2004. [PMID: 15254595 DOI: 10.1172/jci200421583] [Citation(s) in RCA: 321] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
One mechanism contributing to immunologic unresponsiveness toward tumors may be presentation of tumor antigens by tolerogenic host APCs. We show that mouse tumor-draining LNs (TDLNs) contained a subset of plasmacytoid DCs (pDCs) that constitutively expressed immunosuppressive levels of the enzyme indoleamine 2,3-dioxygenase (IDO). Despite comprising only 0.5% of LN cells, these pDCs in vitro potently suppressed T cell responses to antigens presented by the pDCs themselves and also, in a dominant fashion, suppressed T cell responses to third-party antigens presented by nonsuppressive APCs. Adoptive transfer of DCs from TDLNs into naive hosts created profound local T cell anergy, specifically toward antigens expressed by the transferred DCs. Anergy was prevented by targeted disruption of the IDO gene in the DCs or by administration of the IDO inhibitor drug 1-methyl-D-tryptophan to recipient mice. Within the population of pDCs, the majority of the functional IDO-mediated suppressor activity segregated with a novel subset of pDCs coexpressing the B-lineage marker CD19. We hypothesize that IDO-mediated suppression by pDCs in TDLNs creates a local microenvironment that is potently suppressive of host antitumor T cell responses.
Collapse
Affiliation(s)
- David H Munn
- Department of Pediatrics, Institute for Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia 30912, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Munn DH, Sharma MD, Hou D, Baban B, Lee JR, Antonia SJ, Messina JL, Chandler P, Koni PA, Mellor AL. Expression of indoleamine 2,3-dioxygenase by plasmacytoid dendritic cells in tumor-draining lymph nodes. J Clin Invest 2004; 114:280-90. [PMID: 15254595 PMCID: PMC449750 DOI: 10.1172/jci21583] [Citation(s) in RCA: 505] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2004] [Accepted: 05/25/2004] [Indexed: 12/13/2022] Open
Abstract
One mechanism contributing to immunologic unresponsiveness toward tumors may be presentation of tumor antigens by tolerogenic host APCs. We show that mouse tumor-draining LNs (TDLNs) contained a subset of plasmacytoid DCs (pDCs) that constitutively expressed immunosuppressive levels of the enzyme indoleamine 2,3-dioxygenase (IDO). Despite comprising only 0.5% of LN cells, these pDCs in vitro potently suppressed T cell responses to antigens presented by the pDCs themselves and also, in a dominant fashion, suppressed T cell responses to third-party antigens presented by nonsuppressive APCs. Adoptive transfer of DCs from TDLNs into naive hosts created profound local T cell anergy, specifically toward antigens expressed by the transferred DCs. Anergy was prevented by targeted disruption of the IDO gene in the DCs or by administration of the IDO inhibitor drug 1-methyl-D-tryptophan to recipient mice. Within the population of pDCs, the majority of the functional IDO-mediated suppressor activity segregated with a novel subset of pDCs coexpressing the B-lineage marker CD19. We hypothesize that IDO-mediated suppression by pDCs in TDLNs creates a local microenvironment that is potently suppressive of host antitumor T cell responses.
Collapse
Affiliation(s)
- David H Munn
- Department of Pediatrics, Institute for Molecular Medicine and Genetics, Medical College of Georgia, Augusta, Georgia 30912, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Jackaman C, Bundell CS, Kinnear BF, Smith AM, Filion P, van Hagen D, Robinson BWS, Nelson DJ. IL-2 intratumoral immunotherapy enhances CD8+ T cells that mediate destruction of tumor cells and tumor-associated vasculature: a novel mechanism for IL-2. THE JOURNAL OF IMMUNOLOGY 2004; 171:5051-63. [PMID: 14607902 DOI: 10.4049/jimmunol.171.10.5051] [Citation(s) in RCA: 163] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Therapeutic use of IL-2 can generate antitumor immunity; however, a variety of different mechanisms have been reported. We injected IL-2 intratumorally (i.t.) at different stages of growth, using our unique murine model of mesothelioma (AE17; and AE17 transfected with secretory OVA (AE17-sOVA)), and systematically analyzed real-time events as they occurred in vivo. The majority of mice with small tumors when treatment commenced displayed complete tumor regression, remained tumor free for >2 mo, and survived rechallenge with AE17 tumor cells. However, mice with large tumors at the start of treatment failed to respond. Timing experiments showed that IL-2-mediated responses were dependent upon tumor size, not on the duration of disease. Although i.t. IL-2 did not alter tumor Ag presentation in draining lymph nodes, it did enhance a previously primed, endogenous, tumor-specific in vivo CTL response that coincided with regressing tumors. Both CD4(+) and CD8(+) cells were required for IL-2-mediated tumor eradication, because IL-2 therapy failed in CD4(+)-depleted, CD8(+)-depleted, and both CD4(+)- and CD8(+)-depleted C57BL/6J animals. Tumor-infiltrating CD8(+) T cells, but not CD4(+) T cells, increased in association with a marked reduction in tumor-associated vascularity. Destruction of blood vessels required CD8(+) T cells, because this did not occur in nude mice or in CD8(+)-depleted C57BL/6J mice. These results show that repeated doses of i.t. (but not systemic) IL-2 mediates tumor regression via an enhanced endogenous tumor-specific CTL response concomitant with reduced vasculature, thereby demonstrating a novel mechanism for IL-2 activity.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/therapeutic use
- Animals
- Antigen Presentation
- Antigens, CD/biosynthesis
- Antigens, Neoplasm/administration & dosage
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Antineoplastic Agents/administration & dosage
- Antineoplastic Agents/therapeutic use
- B7-2 Antigen
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- Cell Death/immunology
- Cell Division/genetics
- Cell Division/immunology
- Cell Line, Tumor
- Cytotoxicity, Immunologic
- Egg Proteins/administration & dosage
- Egg Proteins/genetics
- Egg Proteins/pharmacokinetics
- Female
- Graft Rejection/immunology
- Graft Rejection/pathology
- Graft Rejection/physiopathology
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/therapeutic use
- Histocompatibility Antigens Class I/metabolism
- Immunodominant Epitopes/biosynthesis
- Immunodominant Epitopes/genetics
- Immunodominant Epitopes/metabolism
- Immunotherapy, Active/methods
- Injections, Intralesional
- Injections, Intraperitoneal
- Interleukin-2/administration & dosage
- Interleukin-2/therapeutic use
- Lymphocytes, Tumor-Infiltrating/pathology
- Membrane Glycoproteins/biosynthesis
- Mesothelioma/blood supply
- Mesothelioma/mortality
- Mesothelioma/therapy
- Mesothelioma/ultrastructure
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Mice, Transgenic
- Necrosis
- Neoplasm Transplantation
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/prevention & control
- Ovalbumin/administration & dosage
- Ovalbumin/genetics
- Ovalbumin/pharmacokinetics
- Peptide Fragments
- Survival Rate
- T-Lymphocyte Subsets/immunology
Collapse
Affiliation(s)
- Connie Jackaman
- School of Medicine and Pharmacology, University of Western Australia
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Mukherjee S, Upham JW, Ramshaw I, Bundell C, van Bruggen I, Robinson BWS, Nelson DJ. Dendritic cells infected with a vaccinia virus interleukin-2 vector secrete high levels of IL-2 and can become efficient antigen presenting cells that secrete high levels of the immunostimulatory cytokine IL-12. Cancer Gene Ther 2003; 10:591-602. [PMID: 12872141 DOI: 10.1038/sj.cgt.7700604] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Dendritic cell (DC) therapies using DC presenting tumor antigen/s can induce CD8(+) CTL that mediate tumor eradication, nonetheless many patients remain unresponsive. Thus, cytokine gene vectors applied to DC may amplify these responses. Herein, we examined the responses that monocyte-derived DC (at different maturational stages) make when infected with a vaccinia virus-interleukin-2 (VV-IL-2) vector in vitro. VV-IL-2-infected DC secreted significant levels of bioactive IL-2 and maintained their antigen presentation function. However, we show that DC are exquisitely sensitive to their local antigenic microenvironment, and that responses generated by one antigen can be altered by another. VV-IL-2 infection of immature DC led to DC activation (upregulation of CD80, CD86 and class II surface molecules) when the virus was propagated through xenogeneic, but not syngeneic, mammalian cells; these DC secreted IL-10 and tumor necrosis factor-alpha (TNF-alpha), but not IL-12. In contrast, after VV-IL-2 infection (regardless of their mammalian cellular context), IFNgamma/LPS-matured DC inevitably downregulated their antigen presenting machinery. In conclusion, immunostimulatory DC can be generated by VV-IL-2, but this depends upon (i) infecting immature DC only, (ii) the mammalian cells through which the virus is prepared and (iii) individual donors; hence donors must be screened to assess their specific responses.
Collapse
Affiliation(s)
- Sutapa Mukherjee
- Department of Medicine, University of Western Australia, Perth, Australia
| | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
There is extensive plasticity in the T-cell response to antigen. Helper CD4(+) T cells, cytotoxic CD8(+) T cells, the progression from naïve to effector and memory T cells, and differentiation into Th1, Tc1, Th2 and Tc2 subsets have long been recognized. More recently it has become apparent that T-cell populations display additional diversity in terms of phenotype, anatomical distribution and effector function.
Collapse
Affiliation(s)
- David L Woodland
- Trudeau Institute, 100 Algonquin Avenue, Saranac Lake, New York, NY 12983, USA.
| | | |
Collapse
|
49
|
Nowak AK, Lake RA, Marzo AL, Scott B, Heath WR, Collins EJ, Frelinger JA, Robinson BWS. Induction of tumor cell apoptosis in vivo increases tumor antigen cross-presentation, cross-priming rather than cross-tolerizing host tumor-specific CD8 T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4905-13. [PMID: 12734333 DOI: 10.4049/jimmunol.170.10.4905] [Citation(s) in RCA: 349] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cross-presentation of cell-bound Ags from established, solid tumors to CD8 cells is efficient and likely to have a role in determining host response to tumor. A number of investigators have predicted that when tumor Ags are derived from apoptotic cells either no response, due to Ag "sequestration," or CD8 cross-tolerance would ensue. Because the crucial issue of whether this happens in vivo has never been addressed, we induced apoptosis of established hemagglutinin (HA)-transfected AB1 tumors in BALB/c mice using the apoptosis-inducing reagent gemcitabine. This shrank the tumor by approximately 80%. This induction of apoptosis increased cross-presentation of HA to CD8 cells yet neither gross deletion nor functional tolerance of HA-specific CD8 cells were observed, based on tetramer analysis, proliferation of specific CD8 T cells, and in vivo CTL activity. Interestingly, apoptosis primed the host for a strong antitumor response to a second, virus-generated HA-specific signal in that administration of an HA-expressing virus after gemcitabine administration markedly decreased tumor growth compared with viral administration without gemcitabine. Thus tumor cell apoptosis in vivo neither sequesters tumor Ags nor cross-tolerizes tumor-specific CD8 cells. This observation has fundamental consequences for the development of tumor immunotherapy protocols and for understanding T cell reactivity to tumors and the in vivo immune responses to apoptotic cells.
Collapse
MESH Headings
- Animals
- Antigen Presentation/drug effects
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Apoptosis/drug effects
- Apoptosis/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Clonal Anergy/drug effects
- Clonal Anergy/immunology
- Clonal Deletion/drug effects
- Clonal Deletion/immunology
- Cytotoxicity, Immunologic/drug effects
- Deoxycytidine/administration & dosage
- Deoxycytidine/analogs & derivatives
- Dose-Response Relationship, Immunologic
- Epitopes, T-Lymphocyte/immunology
- Epitopes, T-Lymphocyte/metabolism
- Growth Inhibitors/administration & dosage
- Hemagglutinin Glycoproteins, Influenza Virus/immunology
- Hemagglutinin Glycoproteins, Influenza Virus/metabolism
- Immunization
- Injections, Intraperitoneal
- Mesothelioma/drug therapy
- Mesothelioma/immunology
- Mesothelioma/pathology
- Mesothelioma/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Transgenic
- Tumor Cells, Cultured
- Up-Regulation/drug effects
- Up-Regulation/immunology
- Gemcitabine
Collapse
Affiliation(s)
- Anna K Nowak
- Tumor Immunology Group, Department of Medicine, University of Western Australia and Western Australian Institute of Medical Research, Perth, Australia
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Schüler T, Blankenstein T. Cutting edge: CD8+ effector T cells reject tumors by direct antigen recognition but indirect action on host cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:4427-31. [PMID: 12707316 DOI: 10.4049/jimmunol.170.9.4427] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD8(+) effector T cells recognize malignant cells by monitoring their surface for the presence of tumor-derived peptides bound to MHC class I molecules. In addition, tumor-derived Ags can be cross-presented to CD8(+) effector T cells by APCs. IFN-gamma production by CD8(+) T cells is often critical for tumor rejection. However, it remained unclear whether 1) CD8(+) T cells secrete IFN-gamma in response to Ag recognition on tumor cells or APCs and 2) whether IFN-gamma mediates its antitumor effect by acting on host or tumor cells. We show in this study that CD8(+) effector T cells can reject tumors in bone marrow-chimeric mice incapable of cross-presenting Ag by bone marrow-derived APCs and that tumor rejection required host cells to express IFN-gammaR. Together, CD8(+) effector T cells recognize Ag directly on tumor cells, and this recognition is sufficient to reject tumors by IFN-gamma acting on host cells.
Collapse
MESH Headings
- Animals
- Antigen Presentation/immunology
- Antigen-Presenting Cells/immunology
- Antigen-Presenting Cells/metabolism
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/metabolism
- Bone Marrow Cells/immunology
- Bone Marrow Cells/metabolism
- CD8-Positive T-Lymphocytes/immunology
- Cells, Cultured
- Graft Rejection/immunology
- Interferon-gamma/metabolism
- Interferon-gamma/physiology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasm Transplantation
- Ovalbumin/immunology
- Ovalbumin/metabolism
- Receptors, Interferon/biosynthesis
- Receptors, Interferon/physiology
- Tumor Cells, Cultured
- Interferon gamma Receptor
Collapse
Affiliation(s)
- Thomas Schüler
- Max Delbrück Center for Molecular Medicine, Berlin, Germany.
| | | |
Collapse
|