1
|
Kim S, Min H, Nah J, Jeong J, Park K, Kim W, Lee Y, Kim J, An J, Seong RH. Defective N-glycosylation in tumor-infiltrating CD8 + T cells impairs IFN-γ-mediated effector function. Immunol Cell Biol 2023; 101:610-624. [PMID: 37114567 DOI: 10.1111/imcb.12647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 01/23/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023]
Abstract
T cell-mediated antitumor immunity is modulated, in part, by N-glycosylation. However, the interplay between N-glycosylation and the loss of effector function in exhausted T cells has not yet been fully investigated. Here, we delineated the impact of N-glycosylation on the exhaustion of tumor-infiltrating lymphocytes in a murine colon adenocarcinoma model, focusing on the IFN-γ-mediated immune response. We found that exhausted CD8+ T cells downregulated the oligosaccharyltransferase complex, which is indispensable for N-glycan transfer. Concordant N-glycosylation deficiency in tumor-infiltrating lymphocytes leads to loss of antitumor immunity. Complementing the oligosaccharyltransferase complex restored IFN-γ production and alleviated CD8+ T cell exhaustion, resulting in reduced tumor growth. Thus, aberrant glycosylation induced in the tumor microenvironment incapacitates effector CD8+ T cells. Our findings provide insights into CD8+ T cell exhaustion by incorporating N-glycosylation to understand the characteristic loss of IFN-γ, opening new opportunities to amend the glycosylation status in cancer immunotherapies.
Collapse
Affiliation(s)
- Soyeon Kim
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Hyungyu Min
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Jinwoo Nah
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Jinguk Jeong
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Kyungsoo Park
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Wooseob Kim
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Youngjin Lee
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Jieun Kim
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Jungeun An
- Department of Life Sciences, Jeonbuk National University, Jeonju, Republic of Korea
| | - Rho Hyun Seong
- School of Biological Sciences and Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Turner CN, Mullins GN, Hoyer KK. CXCR5 +CD8 T cells: Potential immunotherapy targets or drivers of immune-mediated adverse events? Front Med (Lausanne) 2022; 9:1034764. [PMID: 36314014 PMCID: PMC9606409 DOI: 10.3389/fmed.2022.1034764] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/23/2022] [Indexed: 11/15/2022] Open
Abstract
CXCR5+CD8 T cells have attracted significant interest within multiple areas of immunology, cancer, and infection. This is in part due to their apparent dual functionality. These cells perform as cytotoxic cells in a variety of infection states including LCMV, HBV, HIV and SIV. However, CXCR5+CD8 T cells also associate with B cells in peripheral organs and function to stimulate B cell proliferation, antibody/B cell receptor class-switch, and antibody production. CXCR5+CD8 T cells are similar to CXCR5+CD4 T follicular helpers in their genetic make-up, B cell interactions, and functionality despite possessing elevated programmed cell death 1 and cytotoxic proteins. Within cancer CXCR5+CD8 T cells have risen as potential prognostic markers for overall survival and are functionally cytotoxic within tumor microenvironments. In inflammatory disease and autoimmunity, CXCR5+CD8 T cells are implicated in disease progression. During viral infection and cancer, CXCR5 expression on CD8 T cells generally is indicative of progenitor memory stem-like exhausted cells, which are more responsive to immune checkpoint blockade therapy. The use of immune checkpoint inhibitors to overcome immune exhaustion in cancer, and subsequent consequence of immune adverse events, highlights the dual nature of the cellular immune response. This review will detail the functionality of CXCR5+CD8 T cells in cancer and autoimmunity with potential repercussions during immune checkpoint blockade therapy discussed.
Collapse
Affiliation(s)
- Christi N. Turner
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Genevieve N. Mullins
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States
| | - Katrina K. Hoyer
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, United States,Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, CA, United States,Health Sciences Research Institute, University of California, Merced, Merced, CA, United States,*Correspondence: Katrina K. Hoyer
| |
Collapse
|
3
|
Leleux JA, Albershardt TC, Reeves R, James R, Krull J, Parsons AJ, ter Meulen J, Berglund P. Intratumoral expression of IL-12 from lentiviral or RNA vectors acts synergistically with TLR4 agonist (GLA) to generate anti-tumor immunological memory. PLoS One 2021; 16:e0259301. [PMID: 34855754 PMCID: PMC8638928 DOI: 10.1371/journal.pone.0259301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 10/15/2021] [Indexed: 11/20/2022] Open
Abstract
Systemic interleukin-12 (IL12) anti-tumor therapy is highly potent but has had limited utility in the clinic due to severe toxicity. Here, we present two IL12-expressing vector platforms, both of which can overcome the deficiencies of previous systemic IL12 therapies: 1) an integrating lentiviral vector, and 2) a self-replicating messenger RNA formulated with polyethyleneimine. Intratumoral administration of either IL12 vector platform resulted in recruitment of immune cells, including effector T cells and dendritic cells, and the complete remission of established tumors in multiple murine models. Furthermore, concurrent intratumoral administration of the synthetic TLR4 agonist glucopyranosyl lipid A formulated in a stable emulsion (GLA-SE) induced systemic memory T cell responses that mediated complete protection against tumor rechallenge in all survivor mice (8/8 rechallenged mice), whereas only 2/6 total rechallenged mice treated with intratrumoral IL12 monotherapy rejected the rechallenge. Taken together, expression of vectorized IL12 in combination with a TLR4 agonist represents a varied approach to broaden the applicability of intratumoral immune therapies of solid tumors.
Collapse
MESH Headings
- Animals
- CD8-Positive T-Lymphocytes/immunology
- Female
- Gene Expression Regulation
- Genetic Vectors/administration & dosage
- Genetic Vectors/pharmacology
- Glucosides/pharmacology
- Immunity, Innate/drug effects
- Immunity, Innate/genetics
- Immunologic Memory/drug effects
- Immunologic Memory/genetics
- Immunotherapy/methods
- Interferon-gamma/blood
- Interleukin-12/blood
- Interleukin-12/genetics
- Interleukin-12/immunology
- Lentivirus/genetics
- Lipid A/pharmacology
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/therapy
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Toll-Like Receptor 4/agonists
- Mice
Collapse
Affiliation(s)
- Jardin A. Leleux
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Tina C. Albershardt
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Rebecca Reeves
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Reice James
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Jordan Krull
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Andrea J. Parsons
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Jan ter Meulen
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| | - Peter Berglund
- Immune Design Corp., Seattle, WA, A wholly owned subsidiary of Merck & Co., Inc., Kenilworth, NJ, United States of America
| |
Collapse
|
4
|
Klarquist JS, Janssen EM. Melanoma-infiltrating dendritic cells: Limitations and opportunities of mouse models. Oncoimmunology 2021; 1:1584-1593. [PMID: 23264904 PMCID: PMC3525613 DOI: 10.4161/onci.22660] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The infiltration of melanoma lesions by dendritic cells (DCs) has been suggested to play a tumorigenic role due to the capacity of DCs to induce tumor tolerance and promote angiogenesis as well as metastasis. However, it has also been shown that tumor-infiltrating DCs (TIDCs) induce antitumor responses and hence may be targeted in cost-effective therapeutic approaches to obtain patient-specific DCs that present relevant tumor antigens, without the need for ex vivo DC expansion or tumor antigen identification. Unfortunately, little is known about the composition, nature and function of TIDCs found in human melanoma. The development of mouse melanoma models has greatly contributed to the molecular understanding of melanoma immunology in mice, but many questions on TIDCs remain unanswered. Here, we discuss current knowledge about melanoma TIDCs in various mouse models with regard to their translational potential and clinical relevance.
Collapse
Affiliation(s)
- Jared S Klarquist
- Division of Cellular and Molecular Immunology; Cincinnati Children's Hospital Research Foundation; University of Cincinnati College of Medicine; Cincinnati, OH USA
| | | |
Collapse
|
5
|
Verdon DJ, Mulazzani M, Jenkins MR. Cellular and Molecular Mechanisms of CD8 + T Cell Differentiation, Dysfunction and Exhaustion. Int J Mol Sci 2020; 21:ijms21197357. [PMID: 33027962 PMCID: PMC7582856 DOI: 10.3390/ijms21197357] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/01/2020] [Accepted: 10/02/2020] [Indexed: 02/07/2023] Open
Abstract
T cells follow a triphasic distinct pathway of activation, proliferation and differentiation before becoming functionally and phenotypically “exhausted” in settings of chronic infection, autoimmunity and in cancer. Exhausted T cells progressively lose canonical effector functions, exhibit altered transcriptional networks and epigenetic signatures and gain constitutive expression of a broad coinhibitory receptor suite. This review outlines recent advances in our understanding of exhausted T cell biology and examines cellular and molecular mechanisms by which a state of dysfunction or exhaustion is established, and mechanisms by which exhausted T cells may still contribute to pathogen or tumour control. Further, this review describes our understanding of exhausted T cell heterogeneity and outlines the mechanisms by which checkpoint blockade differentially engages exhausted T cell subsets to overcome exhaustion and recover T cell function.
Collapse
Affiliation(s)
- Daniel J. Verdon
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
| | - Matthias Mulazzani
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
| | - Misty R. Jenkins
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; (D.J.V.); (M.M.)
- Department of Medical Biology, The University of Melbourne, Parkville, VIC 3052, Australia
- Institute of Molecular Science, La Trobe University, Bundoora, VIC 3086, Australia
- Correspondence:
| |
Collapse
|
6
|
Walsh SR, Simovic B, Chen L, Bastin D, Nguyen A, Stephenson K, Mandur TS, Bramson JL, Lichty BD, Wan Y. Endogenous T cells prevent tumor immune escape following adoptive T cell therapy. J Clin Invest 2020; 129:5400-5410. [PMID: 31682239 DOI: 10.1172/jci126199] [Citation(s) in RCA: 76] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 09/03/2019] [Indexed: 12/20/2022] Open
Abstract
While the outcome of adoptive T cell therapy (ACT) is typically correlated with the functionality of the inoculated T cells, the role of the endogenous T cells is unknown. The success of checkpoint blockade therapy has demonstrated the potentially curative value of preexisting tumor-primed T cells in cancer treatment. Given the results from checkpoint blockade therapy, we hypothesized that endogenous T cells contribute to long-term survival following ACT. Here, we describe a therapeutic approach combining ACT with an oncolytic vaccine that allows simultaneous analysis of antitumor immunity mediated by transferred and endogenous T cells. We found that, in addition to promoting the expansion and tumor infiltration of the transferred T cells, oncolytic vaccines boosted tumor-primed host T cells. We determined that transferred T cells contributed to rapid destruction of large tumor masses while endogenous T cells concurrently prevented the emergence of antigen-loss variants. Moreover, while transferred T cells disappeared shortly after tumor regression, endogenous T cells secured long-term memory with a broad repertoire of antigen specificity. Our findings suggest that this combination strategy may exploit the full potential of ACT and tumor-primed host T cells to eliminate the primary tumor, prevent immune escape, and provide long-term protective memory.
Collapse
|
7
|
Frey AB. The Inhibitory Signaling Receptor Protocadherin-18 Regulates Tumor-Infiltrating CD8 + T-cell Function. Cancer Immunol Res 2017; 5:920-928. [PMID: 28874354 DOI: 10.1158/2326-6066.cir-17-0187] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/30/2017] [Accepted: 08/28/2017] [Indexed: 11/16/2022]
Abstract
Cancers are infiltrated with antitumor CD8+ T cells that arise during tumor growth, but are defective in effector phase functions because of the suppressive microenvironment. The reactivation of TILs can result in tumor destruction, showing that lytic dysfunction in CD8+ tumor-infiltrating lymphocytes (TIL) permits tumor growth. Like all memory T cells, TILs express inhibitory signaling receptors (aka checkpoint inhibitor molecules) that downregulate TCR-mediated signal transduction upon TIL interaction with cells expressing cognate ligands, thereby restricting cell activation and preventing the effector phase. Previously, we identified a novel murine CD8+ TIL inhibitory signaling receptor, protocadherin-18, and showed that it interacts with p56lck kinase to abrogate proximal TCR signaling. Here, we show that TILs from mice deleted in protocadherin-18 had enhanced antitumor activity and that coblockade of PD-1 and protocadherin-18 in wild-type mice significantly enhanced TIL effector phase function. These results define an important role for protocadherin-18 in antitumor T-cell activity. Cancer Immunol Res; 5(10); 920-8. ©2017 AACR.
Collapse
Affiliation(s)
- Alan B Frey
- Department of Cell Biology and Perlmutter Cancer Center, New York University Langone School of Medicine, New York, New York.
| |
Collapse
|
8
|
Waugh KA, Leach SM, Moore BL, Bruno TC, Buhrman JD, Slansky JE. Molecular Profile of Tumor-Specific CD8+ T Cell Hypofunction in a Transplantable Murine Cancer Model. THE JOURNAL OF IMMUNOLOGY 2016; 197:1477-88. [PMID: 27371726 DOI: 10.4049/jimmunol.1600589] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 06/09/2016] [Indexed: 12/21/2022]
Abstract
Mechanisms of self-tolerance often result in CD8(+) tumor-infiltrating lymphocytes (TIL) with a hypofunctional phenotype incapable of tumor clearance. Using a transplantable colon carcinoma model, we found that CD8(+) T cells became tolerized in <24 h in an established tumor environment. To define the collective impact of pathways suppressing TIL function, we compared genome-wide mRNA expression of tumor-specific CD8(+) T cells from the tumor and periphery. Notably, gene expression induced during TIL hypofunction more closely resembled self-tolerance than viral exhaustion. Differential gene expression was refined to identify a core set of genes that defined hypofunctional TIL; these data comprise the first molecular profile of tumor-specific TIL that are naturally responding and represent a polyclonal repertoire. The molecular profile of TIL was further dissected to determine the extent of overlap and distinction between pathways that collectively restrict T cell functions. As suggested by the molecular profile of TIL, protein expression of inhibitory receptor LAG-3 was differentially regulated throughout prolonged late-G1/early-S phase of the cell cycle. Our data may accelerate efficient identification of combination therapies to boost anti-tumor function of TIL specifically against tumor cells.
Collapse
Affiliation(s)
| | - Sonia M Leach
- Center for Genes, Environment and Health, National Jewish Health, Denver, CO 80206
| | - Brandon L Moore
- University of Colorado School of Medicine, Aurora, CO 80045; and
| | - Tullia C Bruno
- University of Colorado School of Medicine, Aurora, CO 80045; and
| | | | - Jill E Slansky
- University of Colorado School of Medicine, Aurora, CO 80045; and
| |
Collapse
|
9
|
Frey AB. Suppression of T cell responses in the tumor microenvironment. Vaccine 2015; 33:7393-7400. [PMID: 26403368 DOI: 10.1016/j.vaccine.2015.08.096] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 07/06/2015] [Accepted: 08/05/2015] [Indexed: 12/29/2022]
Abstract
The immune system recognizes protein antigens expressed in transformed cells evidenced by accumulation of antigen-specific T cells in tumor and tumor draining lymph nodes. However, despite demonstrable immune response, cancers grow progressively suggesting that priming of antitumor immunity is insufficiently vigorous or that antitumor immunity is suppressed, or both. Compared to virus infection, antitumor T cells are low abundance that likely contributes to tumor escape and enhancement of priming is a long-sought goal of experimental vaccination therapy. Furthermore, patient treatment with antigen-specific T cells can in some cases overcome deficient priming and cause tumor regression supporting the notion that low numbers of T cells permits tumor outgrowth. However, tumor-induced suppression of antitumor immune response is now recognized as a significant factor contributing to cancer growth and reversal of the inhibitory influences within the tumor microenvironment is a major research objective. Multiple cell types and factors can inhibit T cell functions in tumors and may be grouped in two general classes: T cell intrinsic and T cell extrinsic. T cell intrinsic factors are exemplified by T cell expression of cell surface inhibitory signaling receptors that, after contact with cells expressing a cognate ligand, inactivate proximal T Cell Receptor-mediated signal transduction therein rendering T cells dysfunctional. T cell extrinsic factors are more diverse in nature and are produced by tumors and various non-tumor cells in the tumor microenvironment. These include proteins secreted by tumor or stromal cells, highly reactive soluble oxygen and nitrogen species, cytokines, chemokines, gangliosides, and toxic metabolites. These factors may restrict T cell entrance into the tumor parenchyma, cause inactivation of effector phase T cell functions, or induce T cell apoptosis ultimately causing diminished cancer elimination. Here, we review the contributions of inhibitory factors to tumor T cell dysfunction leading to tumor escape.
Collapse
Affiliation(s)
- Alan B Frey
- Department of Cell Biology, Perlmutter Cancer Center, New York University Langone School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
10
|
Lima L, Ferreira JA, Tavares A, Oliveira D, Morais A, Videira PA, Medeiros R, Santos L. FASL polymorphism is associated with response to bacillus Calmette-Guérin immunotherapy in bladder cancer. Urol Oncol 2013; 32:44.e1-7. [PMID: 23948181 DOI: 10.1016/j.urolonc.2013.05.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Revised: 05/21/2013] [Accepted: 05/22/2013] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Deregulation of FAS/FASL system may lead to immune escape and influence bacillus Calmette-Guérin (BCG) immunotherapy outcome, which is currently the gold standard adjuvant treatment for high-risk non-muscle invasive bladder tumors. Among other events, functional promoter polymorphisms of FAS and FASL genes may alter their transcriptional activity. Therefore, we aim to evaluate the role of FAS and FASL polymorphisms in the context of BCG therapy, envisaging the validation of these biomarkers to predict response. PATIENTS AND METHODS DNA extracted from peripheral blood from 125 patients with bladder cancer treated with BCG therapy was analyzed by Polymerase Chain Reaction-Restriction Fragment Length Polymorphism for FAS-670 A/G and FASL-844 T/C polymorphisms. FASL mRNA expression was analyzed by real-time Polymerase Chain Reaction. RESULTS Carriers of FASL-844 CC genotype present a decreased recurrence-free survival after BCG treatment when compared with FASL-844 T allele carriers (mean 71.5 vs. 97.8 months, P = 0.030) and have an increased risk of BCG treatment failure (Hazard Ratio = 1.922; 95% Confidence Interval: [1.064-3.471]; P = 0.030). Multivariate analysis shows that FASL-844 T/C and therapeutics scheme are independent predictive markers of recurrence after treatment. The evaluation of FASL gene mRNA levels demonstrated that patients carrying FASL-844 CC genotype had higher FASL expression in bladder tumors (P = 0.0027). Higher FASL levels were also associated with an increased risk of recurrence after BCG treatment (Hazard Ratio = 2.833; 95% Confidence Interval: [1.012-7.929]; P = 0.047). FAS-670 A/G polymorphism analysis did not reveal any association with BCG therapy outcome. CONCLUSIONS Our results suggest that analysis of FASL-844 T/C, but not FAS-670 A/G polymorphisms, may be used as a predictive marker of response to BCG immunotherapy.
Collapse
Affiliation(s)
- Luís Lima
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida, Porto, Portugal; ICBAS, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal; Nucleo de Investigação em Farmácia-Centro de Investigação em Saúde e Ambiente (CISA), School of Allied Health Sciences-Polytechnic Institute of Oporto, Porto, Portugal; LPCC, Research Department-Portuguese League Against Cancer (NRNorte), Porto, Portugal.
| | - José A Ferreira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida, Porto, Portugal; QOPNA, Mass Spectrometry Center of the University of Aveiro, Campus de Santiago, Aveiro, Portugal
| | - Ana Tavares
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida, Porto, Portugal; Department of Pathology, Portuguese Institute of Oncology, Porto, Portugal
| | - Daniela Oliveira
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida, Porto, Portugal
| | - António Morais
- Department of Urology, Portuguese Institute of Oncology, Porto, Portugal
| | - Paula A Videira
- CEDOC, Department of Immunology, Faculdade de Ciências Médicas, FCM, Universidade Nova de Lisboa, Lisboa, Portugal
| | - Rui Medeiros
- LPCC, Research Department-Portuguese League Against Cancer (NRNorte), Porto, Portugal; Molecular Oncology Group, Portuguese Institute of Oncology, Porto, Portugal; Department of Pathology and Molecular Immunology, ICBAS, Abel Salazar Biomedical Sciences Institute, University of Porto, Porto, Portugal; Health Faculty of University Fernando Pessoa, Porto, Portugal
| | - Lúcio Santos
- Experimental Pathology and Therapeutics Group, Portuguese Institute of Oncology, Rua Dr. António Bernardino de Almeida, Porto, Portugal; Health Faculty of University Fernando Pessoa, Porto, Portugal; Department of Surgical Oncology, Portuguese Institute of Oncology, Porto, Portugal
| |
Collapse
|
11
|
Urdinguio RG, Fernandez AF, Moncada-Pazos A, Huidobro C, Rodriguez RM, Ferrero C, Martinez-Camblor P, Obaya AJ, Bernal T, Parra-Blanco A, Rodrigo L, Santacana M, Matias-Guiu X, Soldevilla B, Dominguez G, Bonilla F, Cal S, Lopez-Otin C, Fraga MF. Immune-dependent and independent antitumor activity of GM-CSF aberrantly expressed by mouse and human colorectal tumors. Cancer Res 2012; 73:395-405. [PMID: 23108143 DOI: 10.1158/0008-5472.can-12-0806] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Granulocyte-macrophage colony-stimulating factor (GM-CSF/CSF2) is a cytokine produced in the hematologic compartment that may enhance antitumor immune responses, mainly by activation of dendritic cells. Here, we show that more than one-third of human colorectal tumors exhibit aberrant DNA demethylation of the GM-CSF promoter and overexpress the cytokine. Mouse engraftment experiments with autologous and homologous colon tumors engineered to repress the ectopic secretion of GM-CSF revealed the tumor-secreted GM-CSF to have an immune-associated antitumor effect. Unexpectedly, an immune-independent antitumor effect was observed that depended on the ectopic expression of GM-CSF receptor subunits by tumors. Cancer cells expressing GM-CSF and its receptor did not develop into tumors when autografted into immunocompetent mice. Similarly, 100% of the patients with human colon tumors that overexpressed GM-CSF and its receptor subunits survived at least 5 years after diagnosis. These data suggest that expression of GM-CSF and its receptor subunits by colon tumors may be a useful marker for prognosis as well as for patient stratification in cancer immunotherapy.
Collapse
Affiliation(s)
- Rocio G Urdinguio
- Cancer Epigenetics Laboratory, HUCA, Institute of Oncology of Asturias (IUOPA), Universidad de Oviedo, Oviedo, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Bezouška K, Kubínková Z, Stříbný J, Volfová B, Pompach P, Kuzma M, Šírová M, Říhová B. Dimerization of an immunoactivating peptide derived from mycobacterial hsp65 using N-hydroxysuccinimide based bifunctional reagents is critical for its antitumor properties. Bioconjug Chem 2012; 23:2032-41. [PMID: 22988810 DOI: 10.1021/bc300056x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We have shown previously that a short pentapeptide derived from the mycobacterial heat shock protein hsp65 can be highly activating for the immune system based on its strong reactivity with the early activation antigen of lymphocytes CD69. Here, we investigated an optimal form of presentation of this antigen to the cells of the immune system. Four different forms of the dimerized heptapeptide LELTEGY, and of the control inactive dimerized heptapeptide LELLEGY that both contained an extra UV active glycine-tyrosine sequence, were prepared using dihydroxysuccinimidyl oxalate (DSO), dihydroxysuccinimidyl tartarate (DST), dihydroxysuccinimidyl glutarate (DSG), and dihydroxysuccinimidyl suberate (DSS), respectively. Heptapeptides dimerized through DST and DSG linkers had optimal activity in CD69 precipitation assay. Moreover, dimerization of active heptapeptide resulted in a remarkable increase in its proliferation activity and production of cytokines in vitro. Furthermore, while DST and DSG dimerized heptapeptides both significantly enhanced the cytotoxicity of natural killer cells in vitro, only the DSG dimerized compound was active in suppressing growth of melanoma tumors in mice and in enhancing the cytotoxic activity of tumor infiltrating lymphocytes ex vivo. Thus, while the dimerization of the immunoactive peptide caused a dramatic increase in its immunoactivating properties, its in vivo anticancer properties were influenced by the chemical nature of linker used for its dimerization.
Collapse
Affiliation(s)
- Karel Bezouška
- Department of Biochemistry, Faculty of Science, Charles University Prague, Hlavova 8, CZ-12840 Praha 2, Czech Republic.
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Tumor-induced CD8+ T-cell dysfunction in lung cancer patients. Clin Dev Immunol 2012; 2012:741741. [PMID: 23118782 PMCID: PMC3483679 DOI: 10.1155/2012/741741] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 08/28/2012] [Accepted: 09/04/2012] [Indexed: 12/21/2022]
Abstract
Lung cancer is the leading cause of cancer deaths worldwide and one of the most common types of cancers. The limited success of chemotherapy and radiotherapy regimes have highlighted the need to develop new therapies like antitumor immunotherapy. CD8+ T-cells represent a major arm of the cell-mediated anti-tumor response and a promising target for developing T-cell-based immunotherapies against lung cancer. Lung tumors, however, have been considered to possess poor immunogenicity; even so, lung tumor-specific CD8+ T-cell clones can be established that possess cytotoxicity against autologous tumor cells. This paper will focus on the alterations induced in CD8+ T-cells by lung cancer. Although memory CD8+ T-cells infiltrate lung tumors, in both tumor-infiltrating lymphocytes (TILs) and malignant pleural effusions, these cells are dysfunctional and the effector subset is reduced. We propose that chronic presence of lung tumors induces dysfunctions in CD8+ T-cells and sensitizes them to activation-induced cell death, which may be associated with the poor clinical responses observed in immunotherapeutic trials. Getting a deeper knowledge of the evasion mechanisms lung cancer induce in CD8+ T-cells should lead to further understanding of lung cancer biology, overcome tumor evasion mechanisms, and design improved immunotherapeutic treatments for lung cancer.
Collapse
|
14
|
Weiss JM, Bilate AM, Gobert M, Ding Y, Curotto de Lafaille MA, Parkhurst CN, Xiong H, Dolpady J, Frey AB, Ruocco MG, Yang Y, Floess S, Huehn J, Oh S, Li MO, Niec RE, Rudensky AY, Dustin ML, Littman DR, Lafaille JJ. Neuropilin 1 is expressed on thymus-derived natural regulatory T cells, but not mucosa-generated induced Foxp3+ T reg cells. ACTA ACUST UNITED AC 2012; 209:1723-42, S1. [PMID: 22966001 PMCID: PMC3457733 DOI: 10.1084/jem.20120914] [Citation(s) in RCA: 489] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropilin-1 surface expression discriminates between nT reg cells with stable expression and Nrp1 low iT reg cells showing inducible expression under inflammatory conditions. Foxp3 activity is essential for the normal function of the immune system. Two types of regulatory T (T reg) cells express Foxp3, thymus-generated natural T reg (nT reg) cells, and peripherally generated adaptive T reg (iT reg) cells. These cell types have complementary functions. Until now, it has not been possible to distinguish iT reg from nT reg cells in vivo based solely on surface markers. We report here that Neuropilin 1 (Nrp1) is expressed at high levels by most nT reg cells; in contrast, mucosa-generated iT reg and other noninflammatory iT reg cells express low levels of Nrp1. We found that Nrp1 expression is under the control of TGF-β. By tracing nT reg and iT reg cells, we could establish that some tumors have a very large proportion of infiltrating iT reg cells. iT reg cells obtained from highly inflammatory environments, such as the spinal cords of mice with spontaneous autoimmune encephalomyelitis (EAE) and the lungs of mice with chronic asthma, express Nrp1. In the same animals, iT reg cells in secondary lymphoid organs remain Nrp1low. We also determined that, in spontaneous EAE, iT reg cells help to establish a chronic phase of the disease.
Collapse
Affiliation(s)
- Jonathan M Weiss
- Kimmel Center for Biology and Medicine at the Skirball Institute, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Prado-Garcia H, Romero-Garcia S, Morales-Fuentes J, Aguilar-Cazares D, Lopez-Gonzalez JS. Activation-induced cell death of memory CD8+ T cells from pleural effusion of lung cancer patients is mediated by the type II Fas-induced apoptotic pathway. Cancer Immunol Immunother 2012; 61:1065-80. [PMID: 22159518 PMCID: PMC11028981 DOI: 10.1007/s00262-011-1165-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 11/15/2011] [Indexed: 12/21/2022]
Abstract
Lung cancer is the second most common form of cancer and the leading cause of cancer death worldwide. Pleural effusions, containing high numbers of mononuclear and tumor cells, are frequent in patients with advanced stages of lung cancer. We reported that in pleural effusions from primary lung cancer, the CD8+ T cell subpopulation, and particularly the terminally differentiated subset, is reduced compared to that of non-malignant effusions. We analyzed the participation of activation-induced cell death (AICD) and extrinsic pathways (type I or II) as mechanisms for the decrease in pleural effusion CD8+ T cell subpopulation. Pleural effusion or peripheral blood CD4+ and CD8+ T cells, from lung cancer patients, were stimulated with anti-CD3 antibody and analyzed for (a) apoptosis by annexin-V-binding and TUNEL assay, (b) transcript levels of Fas ligand (FasL) and TRAIL by real-time RT-PCR, (c) expression of FasL and TRAIL, measured as integrated mean fluorescence intensities (iMFI) by flow cytometry, (d) expression of Bcl-2 and BIM molecules, measured as MFI, and (e) apoptosis inhibition using caspase-8 and -9 inhibitors. Pleural effusion CD8+ T cells, but not CD4+ T cells, from cancer patients underwent AICD. Blocking FasL/Fas pathway protected from AICD. Upregulation of FasL and TRAIL expressions was found in pleural effusion CD8+ T cells, which also showed a subset of Bcl-2 low cells. In memory CD8+ T cells, AICD depended on both extrinsic and intrinsic apoptotic pathways. Hence, in the pleural space of lung cancer patients, AICD might compromise the antitumor function of CD8+ T cells.
Collapse
Affiliation(s)
- Heriberto Prado-Garcia
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias "Ismael Cosío Villegas", Mexico City, Mexico.
| | | | | | | | | |
Collapse
|
16
|
Vazquez-Cintron EJ, Monu NR, Burns JC, Blum R, Chen G, Lopez P, Ma J, Radoja S, Frey AB. Protocadherin-18 is a novel differentiation marker and an inhibitory signaling receptor for CD8+ effector memory T cells. PLoS One 2012; 7:e36101. [PMID: 22567129 PMCID: PMC3342238 DOI: 10.1371/journal.pone.0036101] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Accepted: 03/26/2012] [Indexed: 01/22/2023] Open
Abstract
CD8+ tumor infiltrating T cells (TIL) lack effector-phase functions due to defective proximal TCR-mediated signaling previously shown to result from inactivation of p56lck kinase. We identify a novel interacting partner for p56lck in nonlytic TIL, Protocadherin-18 (‘pcdh18’), and show that pcdh18 is transcribed upon in vitro or in vivo activation of all CD8+ central memory T cells (CD44+CD62LhiCD127+) coincident with conversion into effector memory cells (CD44+CD62LloCD127+). Expression of pcdh18 in primary CD8+ effector cells induces the phenotype of nonlytic TIL: defective proximal TCR signaling, cytokine secretion, and cytolysis, and enhanced AICD. pcdh18 contains a motif (centered at Y842) shared with src kinases (QGQYQP) that is required for the inhibitory phenotype. Thus, pcdh18 is a novel activation marker of CD8+ memory T cells that can function as an inhibitory signaling receptor and restrict the effector phase.
Collapse
Affiliation(s)
- Edwin J. Vazquez-Cintron
- Department of Cell Biology, New York University Langone School of Medicine New York, New York, United States of America
| | - Ngozi R. Monu
- New York University Cancer Institute, New York University Langone School of Medicine New York, New York, United States of America
| | - Jeremy C. Burns
- Department of Cell Biology, New York University Langone School of Medicine New York, New York, United States of America
| | - Roy Blum
- New York University Cancer Institute, New York University Langone School of Medicine New York, New York, United States of America
| | - Gregory Chen
- Department of Cell Biology, New York University Langone School of Medicine New York, New York, United States of America
| | - Peter Lopez
- Department of Pathology, New York University Langone School of Medicine New York, New York, United States of America
| | - Jennifer Ma
- Center for Cancer and Immunology Research, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia, United States of America
| | - Sasa Radoja
- Center for Cancer and Immunology Research, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's Research Institute, Children's National Medical Center, Washington, District of Columbia, United States of America
| | - Alan B. Frey
- Department of Cell Biology, New York University Langone School of Medicine New York, New York, United States of America
- New York University Cancer Institute, New York University Langone School of Medicine New York, New York, United States of America
- * E-mail:
| |
Collapse
|
17
|
Vazquez-Cintron EJ, Monu NR, Frey AB. Tumor-induced disruption of proximal TCR-mediated signal transduction in tumor-infiltrating CD8+ lymphocytes inactivates antitumor effector phase. THE JOURNAL OF IMMUNOLOGY 2011; 185:7133-40. [PMID: 21127315 DOI: 10.4049/jimmunol.1001157] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The presence in cancer tissue of Ag-specific, activated tumor infiltrating CD8(+) T cells proves that tumors express Ags capable of eliciting immune response. Therefore, in general, tumor escape from immune-mediated clearance is not attributable to immunological ignorance. However, tumor-infiltrating lymphocytes are defective in effector phase function, demonstrating tumor-induced immune suppression that likely underlies tumor escape. Since exocytosis of lytic granules is dependent upon TCR-mediated signal transduction, it is a reasonable contention that tumors may induce defective signal transduction in tumor infiltrating T cells. In this review, we consider the biochemical basis for antitumor T cell dysfunction, focusing on the role of inhibitory signaling receptors in restricting TCR-mediated signaling in tumor-infiltrating lymphocytes.
Collapse
Affiliation(s)
- Edwin J Vazquez-Cintron
- Department of Cell Biology, New York University Langone Medical Center, 550 First Avenue, New York, NY 10016, USA
| | | | | |
Collapse
|
18
|
Liu YY, Sun LC, Wei JJ, Li D, Yuan Y, Yan B, Liang ZH, Zhu HF, Xu Y, Li B, Song CW, Liao SJ, Lei Z, Zhang GM, Feng ZH. Tumor Cell-Released TLR4 Ligands Stimulate Gr-1+CD11b+F4/80+ Cells to Induce Apoptosis of Activated T Cells. THE JOURNAL OF IMMUNOLOGY 2010; 185:2773-82. [DOI: 10.4049/jimmunol.1000772] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
19
|
In vivo targeting and growth inhibition of the A20 murine B-cell lymphoma by an idiotype-specific peptide binder. Blood 2010; 116:226-38. [DOI: 10.1182/blood-2009-11-253617] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Abstract
B-cell lymphoma is a clonal expansion of neoplastic cells that may result in fatal outcomes. Here, we report the in vivo targeting and growth inhibition of aggressive A20 murine B-cell lymphoma by idiotype-specific peptide pA20-36. pA20-36 was selected from random peptide libraries and bound specifically to the B-cell receptor (BCR) of A20 cells in mice engrafted with A20 lymphoma, as shown by histology and positron emission tomographic analysis. BCR cross-linking of A20 cells with pA20-36 resulted in massive apoptosis of targeted tumor cells and in an increased survival of the diseased animals without any detectable evidence of toxicity. The pA20-36 treatment reverted the immune suppression of the tumor microenvironment as shown by reduced expression of vascular endothelial growth factor, interleukin-10, and transforming growth factor-β cytokines together with a lower number of CD11b+Gr-1+ inhibitor myeloid-derived suppressor cells and Foxp3+CD4+ Treg cells. Furthermore, pA20-36 treatment was associated with an increased number of tumor-infiltrating, activated CD8+ T cells that exerted a tumor-specific cytolytic activity. These findings show that a short peptide that binds specifically to the complementarity-determining regions of the A20 BCR allows in vivo detection of neoplastic cells together with significant inhibition of tumor growth in vivo.
Collapse
|
20
|
Wang S, Chen L. Immunobiology of cancer therapies targeting CD137 and B7-H1/PD-1 cosignal pathways. Curr Top Microbiol Immunol 2010; 344:245-67. [PMID: 20582531 DOI: 10.1007/82_2010_81] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cancer immunotherapy is finally entering a new era with manipulation of cosignaling pathways as a therapeutic approach, for which the principle was proved nearly two decades ago. In addition to CTLA-4, CD137 and B7-H1/PD-1 pathways are two new targets in the stage. CD137 pathway is costimulatory and its agonistic antibody delivers potent signal to drive T cell growth and activation. On the other hand, blockade of B7-H1/PD-1 pathway with antagonistic antibody has shown to protect ongoing T cell responses from impairment by immune evasion mechanism in cancer microenvironment. With these tools in hand, a mechanism-based design of combined immunotherapy with high efficacy is becoming a reality.
Collapse
Affiliation(s)
- Shengdian Wang
- Center for Infection and Immunity, Institute for Biophysics, Chinese Academy of Sciences, Beijing, China
| | | |
Collapse
|
21
|
Simpson-Abelson MR, Purohit VS, Pang WM, Iyer V, Odunsi K, Demmy TL, Yokota SJ, Loyall JL, Kelleher RJ, Balu-Iyer S, Bankert RB. IL-12 delivered intratumorally by multilamellar liposomes reactivates memory T cells in human tumor microenvironments. Clin Immunol 2009; 132:71-82. [PMID: 19395317 PMCID: PMC2693480 DOI: 10.1016/j.clim.2009.03.516] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2008] [Revised: 02/16/2009] [Accepted: 03/17/2009] [Indexed: 01/07/2023]
Abstract
Using a novel loading technique, IL-12 is reported here to be efficiently encapsulated within large multilamellar liposomes. The preclinical efficacy of the cytokine loaded liposomes to deliver IL-12 into human tumors and to reactive tumor-associated T cells in situ is tested using a human tumor xenograft model. IL-12 is released in vivo from these liposomes in a biologically active form when injected into tumor xenografts that are established by the subcutaneous implantation of non-disrupted pieces of human lung, breast or ovarian tumors into immunodeficient mice. The histological architecture of the original tumor tissue, including tumor-associated leukocytes, tumor cells and stromal cells is preserved anatomically and the cells remain functionally responsive to cytokines in these xenografts. The local and sustained release of IL-12 into the tumor microenvironment reactivates tumor-associated quiescent effector memory T cells to proliferate, produce and release IFN-gamma resulting in the killing of tumor cells in situ. Very little IL-12 is detected in the serum of mice for up to 5 days after an intratumoral injection of the IL-12 liposomes. We conclude that IL-12 loaded large multilamellar liposomes provide a safe method for the local and sustained delivery of IL-12 to tumors and a therapeutically effective way of reactivating existing tumor-associated T cells in human solid tumor microenvironments. The potential of this local in situ T cell re-stimulation to induce a systemic anti-tumor immunity is discussed.
Collapse
Affiliation(s)
- Michelle R. Simpson-Abelson
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| | - Vivek S. Purohit
- Department of Pharmaceutics, State University at Buffalo, New York
| | - Wing Man Pang
- Department of Pharmaceutics, State University at Buffalo, New York
| | - Vandana Iyer
- Department of Pharmaceutics, State University at Buffalo, New York
| | - Kunle Odunsi
- Department of Gynecologic Oncology, Roswell Park Cancer Institute, Buffalo, New York
| | - Todd L Demmy
- Department of Thoracic Surgery, Roswell Park Cancer Institute, Buffalo, New York
| | - Sandra J. Yokota
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| | - Jenni L. Loyall
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| | - Raymond J. Kelleher
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| | - Sathy Balu-Iyer
- Department of Pharmaceutics, State University at Buffalo, New York
| | - Richard B. Bankert
- Department of Microbiology and Immunology and The Witebsky Center for Microbial Pathogenesis and Immunology, State University of New York at Buffalo
| |
Collapse
|
22
|
Mortarini R, Vegetti C, Molla A, Arienti F, Ravagnani F, Maurichi A, Patuzzo R, Santinami M, Anichini A. Impaired STAT phosphorylation in T cells from melanoma patients in response to IL-2: association with clinical stage. Clin Cancer Res 2009; 15:4085-94. [PMID: 19509154 DOI: 10.1158/1078-0432.ccr-08-3323] [Citation(s) in RCA: 24] [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
PURPOSE To assess the extent of signal transducer and activator of transcription (STAT) activation in response to interleukin 2 (IL-2) in melanoma patients' T cells, along with clinical stage of tumor progression. EXPERIMENTAL DESIGN T lymphocytes from peripheral blood of healthy donors and of American Joint Committee on Cancer stage I to IV melanoma patients, as well as from metastatic lymph nodes of patients, were evaluated for responsiveness to IL-2. CFSE assays and single-cell phospho-STAT-specific flow cytometry screening were used. Results. T cells from advanced melanoma patients, in comparison with healthy donors, showed reduced proliferation to IL-2 and IL-15, but not to anti-CD3 monoclonal antibody. Impaired response occurred in CCR7(+) and CCR7(-) T-cell subsets, but not in CD3(-) CD8(+) natural killer (NK) cells, and was not explained by induction of apoptosis, increased cytokine consumption, or altered IL-2R subunit expression in patients' T lymphocytes. By phospho-specific flow cytometry, defective STAT1 and STAT5 activation in response to IL-2 was found mainly in T lymphocytes from peripheral blood and/or tumor site of American Joint Committee on Cancer stage III and IV patients, compared with stage I and II patients and to donors, and in melanoma antigen-specific T cells isolated from metastatic lymph nodes. At tumor site, impaired STAT activation in T cells did not correlate with frequency of CD4(+) CD25(+) Foxp3(+) T cells. Serum from advanced melanoma patients inhibited IL-2-dependent STAT activation in donors' T cells and a neutralizing monoclonal antibody to transforming growth factor beta1 counteracted such inhibition. CONCLUSIONS These results provide evidence for development of impaired STAT signaling in response to IL-2, along with clinical evolution of the disease, in melanoma patients' T cells.
Collapse
Affiliation(s)
- Roberta Mortarini
- Department of Experimental Oncology, Human Tumors Immunobiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lin YC, Chang LY, Huang CT, Peng HM, Dutta A, Chen TC, Yeh CT, Lin CY. Effector/memory but not naive regulatory T cells are responsible for the loss of concomitant tumor immunity. THE JOURNAL OF IMMUNOLOGY 2009; 182:6095-104. [PMID: 19414761 DOI: 10.4049/jimmunol.0803829] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The phenomenon of concomitant tumor immunity involves a tumor-bearing host rejecting another similar tumor at a distant site and suggests the existence of tumor-specific immunity. Loss of this immunity may contribute to tumor metastasis. However, mechanisms underlying the loss of concomitant immunity are largely unknown. We set up a concomitant tumor immunity model in which this immunity is gradually lost as the primary tumor progresses. We found that CD8(+) T cells, especially tumor-infiltrating CD8(+) T cells, from mice that lost concomitant tumor immunity, possessed potent antitumor properties and strongly expressed effector molecules. Furthermore, effector/memory regulatory T cells (Treg cells, CD103(+)CD4(+)Foxp3(+) T cells) increased as the primary tumor progressed. They initially accumulated around the tumor and in the spleen at later points. Not only did these cells more greatly express killing molecules, they also suppressed the functions of tumor-bearing CD8(+) T cells in vitro and in vivo. Finally, we show that these effector/memory Treg cells inhibit concomitant tumor immunity in vivo. Taken together, data suggest that effector/memory Treg cells are responsible for the loss of concomitant tumor immunity associated with tumor progression.
Collapse
Affiliation(s)
- Yung-Chang Lin
- School of Medicine, College of Medicine, Chang Gung University, Kweishan, Taoyuan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
SUMMARY Despite the expression of antigens by tumor cells, spontaneous immune-mediated rejection of cancer seems to be a rare event. T-cell receptor engagement by peptide/major histocompatibility complexes constitutes the main signal for the activation of naive T cells but is not sufficient to initiate a productive generation and maintenance of effector cells. Full activation of T cells requires additional signals driven by costimulatory molecules present on activated antigen-presenting cells but rarely on tumors. Following the discovery of B7-1 (CD80), several other costimulatory molecules have been shown to contribute to T-cell activation and have relevance for improving anti-tumor immunity. Moreover, increasing the understanding of coinhibitory receptors has highlighted key additional pathways that can dominantly inhibit anti-tumor T-cell function. Improving positive costimulation, and interfering with negative regulation, continues to represent an attractive immunotherapeutic approach for the treatment of cancer. This review focuses upon those pathways with the highest potential for clinical application in human cancer patients.
Collapse
Affiliation(s)
- Gregory Driessens
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - Justin Kline
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| | - Thomas F. Gajewski
- Department of Pathology, The University of Chicago, Chicago, IL, USA
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
25
|
Abstract
The virological synapse (VS) is a specialized molecular structure that facilitates the transfer of certain lymphotropic viruses into uninfected T cells. However, the role of the VS in the transfer of nonlymphotropic viruses into T cells is unknown. Herpes simplex virus (HSV) has been shown in vitro to infect T cells and modulate T-cell receptor function, thereby suppressing T-cell antiviral function. However, whether such infection of T cells occurs in vivo is unknown. Here, we examined whether T-cell infection could be observed in human HSV disease and investigated the mechanism of HSV entry into T cells. We found that HSV-infected T cells were readily detectable during human disease, suggesting that infection and modulation of T-cell function plays a role in human immunopathology. HSV infection of both CD4(+) and CD8(+) T cells occurred much more efficiently via direct cell-to-cell spread from infected fibroblasts than by cell-free virus. Activation of T cells increased their permissivity to HSV infection. Cell-to-cell spread to T cells did not require HSV glycoproteins E and I (gE and gI), which are critical for cell-to-cell spread between epithelial cells. Transfer of HSV to T cells required gD, and the four known entry receptors appear to be contributing to viral entry, with a dominant role for the herpesvirus entry mediator and nectin-1. VS-like structures enriched in activated lymphocyte function-associated antigen 1 (LFA-1) were observed at the point of contact between HSV-infected fibroblasts and T cells. Consistent with spread occurring via the VS, transfer of HSV was increased by activation of LFA-1, and cell-to-cell spread could be inhibited by antibodies to LFA-1 or gD. Taken together, these results constitute the first demonstration of VS-dependent cell-to-cell spread for a predominantly nonlymphotropic virus. Furthermore, they support an important role for infection and immunomodulation of T cells in clinical human disease. Targeting of the VS might allow selective immunopotentiation during infections with HSV or other nonlymphotropic viruses.
Collapse
|
26
|
Abstract
The immune response to cancer has been long recognized, including both innate and adaptive responses, showing that the immune system can recognize protein products of genetic and epigenetic changes in transformed cells. The accumulation of antigen-specific T cells within the tumor, the draining lymph node, and the circulation, either in newly diagnosed patients or resultant from experimental immunotherapy, proves that tumors produce antigens and that priming occurs. Unfortunately, just as obviously, tumors grow, implying that anti-tumor immune responses are either not sufficiently vigorous to eliminate the cancer or that anti-tumor immunity is suppressed. Both possibilities are supported by current data. In experimental animal models of cancer and also in patients, systemic immunity is usually not dramatically suppressed, because tumor-bearing animals and patients develop T-cell-dependent immune responses to microbes and to either model antigens or experimental cancer vaccines. However, inhibition of specific anti-tumor immunity is common, and several possible explanations of tolerance to tumor antigens or tumor-induced immunesuppression have been proposed. Inhibition of effective anti-tumor immunity results from the tumor or the host response to tumor growth, inhibiting the activation, differentiation, or function of anti-tumor immune cells. As a consequence, anti-tumor T cells cannot respond productively to developmental, targeting, or activation cues. While able to enhance the number and phenotype of anti-tumor T cells, the modest success of immunotherapy has shown the necessity to attempt to reverse tolerance in anti-tumor T cells, and the vanguard of experimental therapy now focuses on vaccination in combination with blockade of immunosuppressive mechanisms. This review discusses several potential mechanisms by which anti-tumor T cells may be inhibited in function.
Collapse
Affiliation(s)
- Alan B Frey
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA.
| | | |
Collapse
|
27
|
Tseng-Rogenski SS, Arredouani MS, Neeley YC, Lu B, Chinnaiyan AM, Sanda MG. Fas-mediated T cell deletion potentiates tumor antigen-specific tolerance in a mouse model of prostate cancer. Cancer Immunol Immunother 2008; 57:1357-65. [PMID: 18265979 PMCID: PMC11030269 DOI: 10.1007/s00262-008-0471-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2007] [Accepted: 01/29/2008] [Indexed: 01/08/2023]
Abstract
A pivotal obstacle to cancer immunotherapy is peripheral T cell tolerance to tumor-associated antigens (TAAs). Tolerance induction among mature T cells in the periphery operates through a variety of mechanisms, including anergy and apoptosis. Although Fas-FasL-mediated apoptosis is a well-defined tolerance inducing mechanism, direct evidence of its interference with TAA-specific immunity in vivo is still lacking. In this report, we used the TRAMP mouse, which expresses SV40 large T antigen (Tag) preferentially in the prostate and develops prostate tumors, as a model system to address the role of Fas-mediated apoptosis in regulating peripheral T cell tolerance. Using RT-PCR and tetramer staining to quantify TAA-specific TCR-expressing cytolytic T lymphocytes (CTLs), we have shown the presence of TAA-specific CTLs at higher levels in TRAMP mice than in syngeneic C57Bl/6 mice. Tag-specific immunization led to the expansion of Tag-specific CTLs in C57Bl/6 mice, and to their elimination in TRAMP mice. Interestingly, in TRAMP mice with deficient Fas (Hybrid TRAMP-lpr/lpr), Tag-specific CTL elimination in response to Tag immunization did not take place. The results of cytolytic-function assays were consistent with induction and elimination patterns of TAA-specific CTLs and those of RT-PCR and tetramer staining. In conclusion, our data show that Fas-mediated TAA-specific CTL apoptosis contributes to T cell tolerance and suggest that such tolerance could be potentiated following TAA-specific immunization.
Collapse
Affiliation(s)
- Stephanie S. Tseng-Rogenski
- Departments of Urology and Pathology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109 USA
| | - Mohamed S. Arredouani
- Urology Division, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| | - Yilin C. Neeley
- Departments of Urology and Pathology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109 USA
| | - Bin Lu
- Urology Division, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| | - Arul M. Chinnaiyan
- Departments of Urology and Pathology, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI 48109 USA
| | - Martin G. Sanda
- Urology Division, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115 USA
| |
Collapse
|
28
|
Wallace A, Kapoor V, Sun J, Mrass P, Weninger W, Heitjan DF, June C, Kaiser LR, Ling LE, Albelda SM. Transforming growth factor-beta receptor blockade augments the effectiveness of adoptive T-cell therapy of established solid cancers. Clin Cancer Res 2008; 14:3966-74. [PMID: 18559619 PMCID: PMC2491721 DOI: 10.1158/1078-0432.ccr-08-0356] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE Adoptive cellular immunotherapy is a promising approach to eradicate established tumors. However, a significant hurdle in the success of cellular immunotherapy involves recently identified mechanisms of immune suppression on cytotoxic T cells at the effector phase. Transforming growth factor-beta (TGF-beta) is one of the most important of these immunosuppressive factors because it affects both T-cell and macrophage functions. We thus hypothesized that systemic blockade of TGF-beta signaling combined with adoptive T-cell transfer would enhance the effectiveness of the therapy. EXPERIMENTAL DESIGN Flank tumors were generated in mice using the chicken ovalbumin-expressing thymoma cell line, EG7. Splenocytes from transgenic OT-1 mice (whose CD8 T cells recognize an immunodominant peptide in chicken ovalbumin) were activated in vitro and adoptively transferred into mice bearing large tumors in the presence or absence of an orally available TGF-beta receptor-I kinase blocker (SM16). RESULTS We observed markedly smaller tumors in the group receiving the combination of SM16 chow and adoptive transfer. Additional investigation revealed that TGF-beta receptor blockade increased the persistence of adoptively transferred T cells in the spleen and lymph nodes, increased numbers of adoptively transferred T cells within tumors, increased activation of these infiltrating T cells, and altered the tumor microenvironment with a significant increase in tumor necrosis factor-alpha and decrease in arginase mRNA expression. CONCLUSIONS We found that systemic blockade of TGF-beta receptor activity augmented the antitumor activity of adoptively transferred T cells and may thus be a useful adjunct in future clinical trials.
Collapse
Affiliation(s)
- Africa Wallace
- Thoracic Oncology Research Laboratory, University of Pennsylvania, Pennsylvania, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Prado-Garcia H, Aguilar-Cazares D, Meneses-Flores M, Morales-Fuentes J, Lopez-Gonzalez JS. Lung carcinomas do not induce T-cell apoptosis via the Fas/Fas ligand pathway but down-regulate CD3 epsilon expression. Cancer Immunol Immunother 2008; 57:325-36. [PMID: 17668204 PMCID: PMC11030893 DOI: 10.1007/s00262-007-0372-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2007] [Accepted: 07/11/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND Non-small cell lung carcinoma (NSCLC) patients have impaired cellular immune responses. It has been hypothesized that tumor cells expressing Fas Ligand (FasL) induce in T lymphocytes: (a) apoptosis (tumor counterattack) and (b) down-regulation of CD3zeta expression. However, the hypothesis of tumor counterattack is still controversial. METHODS We analyzed FasL expression on NSCLC cell lines and on tumor cells from lung adenocarcinoma patients by flow cytometry and immunocytochemistry. FasL mRNA expression was detected in NSCLC cell lines using RT-PCR, and functional FasL was evaluated on Fas-expressing Jurkat T-cells by annexin-V-FITC staining and by SubG(1) peak detection. Also, the proapoptotic effect of microvesicles released from NSCLC cell lines in Jurkat T-cells was studied. Alterations in the expression levels of CD3zeta, CD3epsilon, and CD28 [measured as mean fluorescence intensity (MFI)] were determined in Jurkat T-cells after co-culture with NSCLC cell lines or tumor-derived microvesicles. Furthermore, the expression levels of CD3zeta and CD3epsilon in CD4+T and CD8+T lymphocytes from lung adenocarcinoma patients was studied. RESULTS Our results indicate that NSCLC cells neither FasL expressed nor induced apoptosis in Jurkat T-cells. Tumor-derived microvesicles did not induce apoptosis in Jurkat T-cells. In contrast, NSCLC cell lines down-regulated CD3epsilon but not CD3zeta chain expression in Jurkat T-cells; this effect was induced by soluble factors but not by microvesicles. In lung adenocarcinoma patients, significant decreases of MFI values for CD3epsilon, but not CD3zeta, were found in CD4+T and CD8+T cells from pleural effusion compared to peripheral blood and in peripheral blood of patients compared to healthy donors. CONCLUSIONS Our data do not support the tumor counterattack hypothesis for NSCLC. Nonetheless, down-regulation of CD3epsilon in T-cells induced by NSCLC cells might lead to T-cell dysfunction.
Collapse
Affiliation(s)
- Heriberto Prado-Garcia
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Dolores Aguilar-Cazares
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Manuel Meneses-Flores
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Jorge Morales-Fuentes
- Servicio Clinico 3, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| | - Jose Sullivan Lopez-Gonzalez
- Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Tlalpan 4502, Col. Seccion XVI, CP 14080 Mexico City, Mexico
| |
Collapse
|
30
|
Monu N, Frey AB. Suppression of proximal T cell receptor signaling and lytic function in CD8+ tumor-infiltrating T cells. Cancer Res 2008; 67:11447-54. [PMID: 18056473 DOI: 10.1158/0008-5472.can-07-1441] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CD8(+) tumor-infiltrating lymphocytes (TIL) lack in vivo and in vitro lytic function due to a signaling deficit characterized by failure to flux calcium or activate tyrosine kinase activity upon contact with cognate tumor cells. Although CD3 zeta is phosphorylated by conjugation in vitro with cognate tumor cells, showing that TIL are triggered, PLC gamma-1, LAT, and ZAP70 are not activated and LFA-1 is not affinity-matured, and because p56(lck) is required for LFA-1 activation, this implies that the signaling blockade is very proximal. Here, we show that TIL signaling defects are transient, being reversed upon purification and brief culture in vitro, implying a fast-acting "switch". Biochemical analysis of purified nonlytic TIL shows that contact with tumor cells causes transient activation of p56(lck) ( approximately 10 s) which is rapidly inactivated. In contrast, tumor-induced activation of p56(lck) in lytic TIL is sustained coincident with downstream TCR signaling and lytic function. Shp-1 is robustly active in nonlytic TIL compared with lytic TIL, colocalizes with p56(lck) in nonlytic TIL, and inhibition of Shp-1 activity in lytic TIL in vitro blocks tumor-induced defective TIL cytolysis. Collectively, our data support the notion that contact of nonlytic TIL with tumor cells, and not with tumor-infiltrating myeloid-derived suppressor cells, causes activation of Shp-1 that rapidly dephosphorylates the p56(lck) activation motif (Y394), thus inhibiting effector phase functions.
Collapse
Affiliation(s)
- Ngozi Monu
- Department of Cell Biology, New York University Cancer Institute, New York University School of Medicine, New York, New York 10016, USA
| | | |
Collapse
|
31
|
Norian LA, Allen PM. Rapid maturation of effector T cells in tumors, but not lymphoid organs, during tumor regression. PLoS One 2007; 2:e821. [PMID: 17786193 PMCID: PMC1950566 DOI: 10.1371/journal.pone.0000821] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2007] [Accepted: 08/09/2007] [Indexed: 12/18/2022] Open
Abstract
Increasing the efficacy of adoptively transferred, tumor antigen specific T cells is a major goal of immunotherapy. Clearly, a more thorough understanding of the effector phase of T cell responses, within the tumor site itself, would be beneficial. To examine this issue, we adoptively transferred tumor antigen-specific effector T cells into tumor-bearing mice, then performed kinetic evaluations of their phenotype, function, and survival in tumors, draining lymph nodes (dLNs), and spleens during regression of murine fibrosarcomas. Effector function in tumors was quantitated through the use of a novel intratumoral cytolytic assay. This approach revealed dynamic changes in the phenotype, cytolytic capacity, and viability of tumor infiltrating effector T cells during the course of tumor regression. Over a period of days, T cells within tumors rapidly transitioned from a CD25(hi)/CD27(hi) to a CD25(low)/CD27(low) phenotype and displayed an increase in cytolytic capacity, indicative of effector maturation. Simultaneously, however, the viability of maturing T cells within tumors diminished. In contrast, transferred T cells trafficking through lymphoid organs were much more static, as they maintained a stable phenotype, robust cytolytic activity, and high viability. Therefore, there exists a marked phenotypic and functional divergence between tumor-infiltrating effector T cells and their counterparts in lymphoid organs. Our results indicate that the population of tumor-infiltrating T cells is unique in experiencing rapid effector maturation post-transfer, and suggest that strategies aimed at prolonging the survival of CD25(low)/CD27(low) full effectors, which displayed the highest levels of intratumoral cytolytic activity, should enhance the efficacy of T cell based tumor immunotherapies.
Collapse
Affiliation(s)
- Lyse A Norian
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri, United States of America.
| | | |
Collapse
|
32
|
Kilinc MO, Aulakh KS, Nair RE, Jones SA, Alard P, Kosiewicz MM, Egilmez NK. Reversing tumor immune suppression with intratumoral IL-12: activation of tumor-associated T effector/memory cells, induction of T suppressor apoptosis, and infiltration of CD8+ T effectors. THE JOURNAL OF IMMUNOLOGY 2007; 177:6962-73. [PMID: 17082611 DOI: 10.4049/jimmunol.177.10.6962] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A single intratumoral injection of IL-12 and GM-CSF-loaded slow-release microspheres induces T cell-dependent eradication of established primary and metastatic tumors in a murine lung tumor model. To determine how the delivery of cytokines directly to the microenvironment of a tumor nodule induces local and systemic antitumor T cell activity, we characterized therapy-induced phenotypic and functional changes in tumor-infiltrating T cell populations. Analysis of pretherapy tumors demonstrated that advanced primary tumors were infiltrated by CD4+ and CD8+ T cells with an effector/memory phenotype and CD4+CD25+Foxp3+ T suppressor cells. Tumor-associated effector memory CD8+ T cells displayed impaired cytotoxic function, whereas CD4+CD25+Foxp3+ cells effectively inhibited T cell proliferation demonstrating functional integrity. IL-12/GM-CSF treatment promoted a rapid up-regulation of CD43 and CD69 on CD8+ effector/memory T cells, augmented their ability to produce IFN-gamma, and restored granzyme B expression. Importantly, treatment also induced a concomitant and progressive loss of T suppressors from the tumor. Further analysis established that activation of pre-existing effector memory T cells was short-lived and that both the effector/memory and the suppressor T cells became apoptotic within 4 days of treatment. Apoptotic death of pre-existing effector/memory and suppressor T cells was followed by infiltration of the tumor with activated, nonapoptotic CD8+ effector T lymphocytes on day 7 posttherapy. Both CD8+ T cell activation and T suppressor cell purge were mediated primarily by IL-12 and required IFN-gamma. This study provides important insight into how local IL-12 therapy alters the immunosuppressive tumor milieu to one that is immunologically active, ultimately resulting in tumor regression.
Collapse
MESH Headings
- Adenocarcinoma, Bronchiolo-Alveolar/immunology
- Adenocarcinoma, Bronchiolo-Alveolar/pathology
- Adenocarcinoma, Bronchiolo-Alveolar/therapy
- Animals
- Apoptosis/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/therapeutic use
- Cell Death/immunology
- Cell Line, Tumor
- Cell Movement/immunology
- Cells, Cultured
- Female
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use
- Immunologic Memory
- Injections, Intralesional
- Interleukin-12/administration & dosage
- Interleukin-12/therapeutic use
- Interleukin-2 Receptor alpha Subunit/biosynthesis
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Lymphocyte Activation/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Microspheres
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/pathology
Collapse
Affiliation(s)
- Mehmet O Kilinc
- J.G. Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY 40202, 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: 2.5] [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
|
Broderick L, Bankert RB. Memory T cells in human tumor and chronic inflammatory microenvironments: sleeping beauties re-awakened by a cytokine kiss. Immunol Invest 2006; 35:419-36. [PMID: 16916760 DOI: 10.1080/08820130600755066] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Human tumors often progress and spread in spite of the presence of large numbers of CD4+ and CD8+ T cells with activated or memory cell phenotypes. The T cells in the microenvironment of human lung tumors fail to be activated in response to stimulation via the T cell receptor and CD28 under conditions that fully activate T cells derived from the peripheral blood of the cancer patients. A combination of regulatory mechanisms which are also observed in a variety of different chronic inflammatory conditions may contribute to the T cell unresponsiveness, and to their inability to respond to and kill tumor cells. The non-responsiveness of memory T cells isolated from human lung tumors and non-malignant chronic inflammatory tissues can be reversed in vitro by a brief pulse with IL-12, and the local and sustained release of exogenous IL-12 into the microenvironment of human tumor xenografts in SCID mice re-activates the tumor-associated T cells in situ. In the later case, the T cells proliferate, secrete interferon-gamma and initiate a cascade of events that culminate in the eradication of tumor cells from the xenograft. In transplantable and spontaneously developing tumors of mice the injection of a single tumor nodule with IL-12 loaded biodegradable microspheres activates tumor-associated T cells to kill tumor cells in situ, and provokes a systemic anti-tumor response that results in the eradication of distant metastatic tumor nodules that are not treated with the cytokine. These mice exhibit a systemic tumor specific immunity as they resist a second challenge with the same (but not a different) tumor. These findings suggest that it will be possible to provoke a systemic anti-tumor immunity in cancer patients by the direct injection of IL-12 loaded biodegradable microspheres or liposomes to locally deliver very low but sustained doses of IL-12 into a single tumor site. This strategy which is based upon the ability of IL-12 to re-activate tumor-associated T cells is termed in situ tumor vaccination.
Collapse
Affiliation(s)
- Lori Broderick
- Department of Microbiology and Immunology, State University of New York at Buffalo, Buffalo, New York 14214, USA
| | | |
Collapse
|
35
|
Shen DT, Ma JSY, Mather J, Vukmanovic S, Radoja S. Activation of primary T lymphocytes results in lysosome development and polarized granule exocytosis in CD4+and CD8+subsets, whereas expression of lytic molecules confers cytotoxicity to CD8+T cells. J Leukoc Biol 2006; 80:827-37. [PMID: 16891618 DOI: 10.1189/jlb.0603298] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Lytic granule exocytosis is the major cytotoxic mechanism used by CD8(+) cytotoxic lymphocytes. CD8(+) T cells acquire this effector function in the process characterized by lysosomal biogenesis, induction of expression of cytolytic molecules, and their selective sorting into the lysosomal vesicles. However, temporal relation of these differentiation stages during T cell activation has not been defined precisely. Also, although CD4(+) T cells typically do not express lytic molecules as a consequence of activation, and therefore, do not acquire granule exocytosis-mediated lytic function, it is not clear whether CD4(+) T cells are able to degranulate. By using in vitro TCR stimulation of primary mouse lymphocytes, we found that polyclonally activated CD4(+) T cells degranulate upon TCR ligation and polarize enlarged lysosomal granules in response to target cell recognition, despite the lack of granule exocytosis-mediated cytotoxicity. Upon TCR stimulation, resting CD8(+) T cells rapidly express lytic molecules and acquire potent lytic function early in activation. Maximal cytolytic potential, however, depends on enlargement of lysosomal granules during the subsequent activation stages. Thus, polyclonal TCR stimulation of resting T cells results in development of lysosomal granules and their release upon TCR engagement in CD4(+) and CD8(+) T cells, but only CD8(+) T cells acquire lytic function as a result of induction of expression of lytic molecules.
Collapse
Affiliation(s)
- David T Shen
- Children's National Medical Center, Center for Cancer and Immunology, Children's Research Institute, Washington, D.C. 20010, USA
| | | | | | | | | |
Collapse
|
36
|
Koneru M, Monu N, Schaer D, Barletta J, Frey AB. Defective adhesion in tumor infiltrating CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2006; 176:6103-11. [PMID: 16670319 DOI: 10.4049/jimmunol.176.10.6103] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD8(+) tumor-infiltrating lymphocytes (TIL) are defective in cytolysis due to tumor-induced inhibition of proximal TCR-mediated signaling, a defect that is relieved upon purification and brief culture. We show in this study that frequency of conjugation in vitro of nonlytic TIL with tumor cells is low in comparison with their lytic counterparts, and the strength of interaction and duration of conjugation are also reduced. Previous reports show that p56(lck) activation is required for TCR-initiated LFA-1 avidity up-regulation, raising the question: is low LFA-1 avidity the basis of reduced TIL conjugation frequency? When stimulated with phorbol ester, nonlytic TIL bind purified ICAM-1 equivalently as lytic TIL, suggesting that LFA-1 can be activated if proximal TCR signaling is bypassed. However, when treated with phorbol ester, the conjugation frequency of nonlytic TIL does not increase. CD2 and CD8 also mediate T cell adhesion to cognate target cells and are both expressed at lower levels in nonlytic TIL in addition to being excluded from the immune synapse formed upon conjugation. Collectively, these results imply that adhesion defects in nonlytic TIL result from a combination of decreased cell surface levels of adhesion molecules, deficient LFA-1 activation, and the failure to recruit essential adhesion receptors to the membrane contact site formed with cognate target cells.
Collapse
Affiliation(s)
- Mythili Koneru
- Department of Cell Biology, and Kaplan Cancer Center, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA
| | | | | | | | | |
Collapse
|
37
|
Sun T, Zhou Y, Li H, Han X, Shi Y, Wang L, Miao X, Tan W, Zhao D, Zhang X, Guo Y, Lin D. FASL -844C polymorphism is associated with increased activation-induced T cell death and risk of cervical cancer. ACTA ACUST UNITED AC 2005; 202:967-74. [PMID: 16186185 PMCID: PMC2213165 DOI: 10.1084/jem.20050707] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The FAS receptor-ligand system plays a key role in regulating apoptotic cell death, and corruption of this signaling pathway has been shown to participate in tumor-immune escape and carcinogenesis. We have recently demonstrated (Sun, T., X. Miao, X. Zhang, W. Tan, P. Xiong, and D. Lin. 2004. J. Natl. Cancer Inst. 96:1030-1036; Zhang, X., X. Miao, T. Sun, W. Tan, S. Qu, P. Xiong, Y. Zhou, and D. Lin. 2005. J. Med. Genet. 42:479-484) that functional polymorphisms in FAS and FAS ligand (FASL) are associated with susceptibility to lung cancer and esophageal cancer; however, the mechanisms underlying this association have not been elucidated. We show that the FAS -1377G, FAS -670A, and FASL -844T variants are expressed more highly on ex vivo-stimulated T cells than the FAS -1377A, FAS -670G, and FASL -844C variants. Moreover, activation-induced cell death (AICD) of T cells carrying the FASL -844C allele was increased. We also found a threefold increased risk of cervical cancer among subjects with the FASL -844CC genotype compared with those with the -844TT genotype in a case-control study in Chinese women. Together, these observations suggest that genetic polymorphisms in the FAS-FASL pathway confer host susceptibility to cervical cancers, which might be caused by immune escape of tumor cells because of enhanced AICD of tumor-specific T cells.
Collapse
Affiliation(s)
- Tong Sun
- Department of Etiology and Carcinogenesis, Cancer Institute and Hospital, China
| | | | | | | | | | | | | | | | | | | | | | | |
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: 3.3] [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
|
Görgün G, Holderried TAW, Zahrieh D, Neuberg D, Gribben JG. Chronic lymphocytic leukemia cells induce changes in gene expression of CD4 and CD8 T cells. J Clin Invest 2005; 115:1797-805. [PMID: 15965501 PMCID: PMC1150284 DOI: 10.1172/jci24176] [Citation(s) in RCA: 240] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2004] [Accepted: 05/10/2005] [Indexed: 01/26/2023] Open
Abstract
To examine the impact of tumors on the immune system, we compared global gene expression profiles of peripheral blood T cells from previously untreated patients with B cell chronic lymphocytic leukemia (CLL) with those from age-matched healthy donors. Although the cells analyzed were not part of the malignant clone, analysis revealed differentially expressed genes, mainly involved in cell differentiation in CD4 cells and defects in cytoskeleton formation, vesicle trafficking, and cytotoxicity in CD8 cells of the CLL patients. In coculture experiments using CLL cells and T cells from healthy allogeneic donors, similar defects developed in both CD4 and CD8 cells. These changes were induced only with direct contact and were not cytokine mediated. Identification of the specific pathways perturbed in the T cells of cancer-bearing patients will allow us to assess steps to repair these defects, which will likely be required to enhance antitumor immunity.
Collapse
MESH Headings
- Adult
- Aged
- Aged, 80 and over
- Base Sequence
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Case-Control Studies
- Chemokines/genetics
- Cytoskeletal Proteins/genetics
- DNA, Neoplasm/genetics
- Female
- Gene Expression Profiling
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Male
- Middle Aged
- Models, Biological
- Oligonucleotide Array Sequence Analysis
- Receptors, Chemokine/genetics
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Güllü Görgün
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | | | | | | |
Collapse
|
40
|
Kusmartsev S, Gabrilovich DI. STAT1 signaling regulates tumor-associated macrophage-mediated T cell deletion. THE JOURNAL OF IMMUNOLOGY 2005; 174:4880-91. [PMID: 15814715 DOI: 10.4049/jimmunol.174.8.4880] [Citation(s) in RCA: 309] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
It is well established that tumor progression is associated with the accumulation of myeloid suppressive cells, which in mice include Gr-1+ immature myeloid cells and F4/80+ macrophages. The paradox is that with the exception of terminal stages of the disease or chemotherapy treatment, tumor-bearing mice or cancer patients do not display a profound systemic immune suppression. We therefore raised the question as to whether myeloid cell-mediated T cell suppression is controlled at a local level at the site of the tumor. We have demonstrated that after adoptive transfer to tumor-bearing recipients, Gr-1+ (immature myeloid cells) freshly isolated from spleens of tumor-bearing mice become F4/80+ tumor-associated macrophages (TAM). These TAM, but not F4/80+ macrophages or Gr-1+ cells freshly isolated from spleens of tumor-bearing or naive mice were able to inhibit T cell-mediated immune response in vitro via induction of T cell apoptosis. Arginase and NO were both responsible for the apoptotic mechanism, and were seen only in TAM, but not in freshly isolated Gr1+ cells. Using the analysis of STAT activity in combination with STAT knockout mice, we have determined that STAT1, but not STAT3 or STAT6, was responsible for TAM-suppressive activity.
Collapse
Affiliation(s)
- Sergei Kusmartsev
- H. Lee Moffitt Cancer Center, University of South Florida, Tampa, FL 33612, USA.
| | | |
Collapse
|
41
|
Bronte V, Kasic T, Gri G, Gallana K, Borsellino G, Marigo I, Battistini L, Iafrate M, Prayer-Galetti T, Pagano F, Viola A. Boosting antitumor responses of T lymphocytes infiltrating human prostate cancers. ACTA ACUST UNITED AC 2005; 201:1257-68. [PMID: 15824085 PMCID: PMC2213151 DOI: 10.1084/jem.20042028] [Citation(s) in RCA: 292] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Immunotherapy may provide valid alternative therapy for patients with hormone-refractory metastatic prostate cancer. However, if the tumor environment exerts a suppressive action on antigen-specific tumor-infiltrating lymphocytes (TIL), immunotherapy will achieve little, if any, success. In this study, we analyzed the modulation of TIL responses by the tumor environment using collagen gel matrix–supported organ cultures of human prostate carcinomas. Our results indicate that human prostatic adenocarcinomas are infiltrated by terminally differentiated cytotoxic T lymphocytes that are, however, in an unresponsive status. We demonstrate the presence of high levels of nitrotyrosines in prostatic TIL, suggesting a local production of peroxynitrites. By inhibiting the activity of arginase and nitric oxide synthase, key enzymes of L-arginine metabolism that are highly expressed in malignant but not in normal prostates, reduced tyrosine nitration and restoration of TIL responsiveness to tumor were achieved. The metabolic control exerted by the tumor on TIL function was confirmed in a transgenic mouse prostate model, which exhibits similarities with human prostate cancer. These results identify a novel and dominant mechanism by which cancers induce immunosuppression in situ and suggest novel strategies for tumor immunotherapy.
Collapse
Affiliation(s)
- Vincenzo Bronte
- Department of Oncology and Surgical Sciences, University of Padova, 35100 Padova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Koneru M, Schaer D, Monu N, Ayala A, Frey AB. Defective Proximal TCR Signaling Inhibits CD8+ Tumor-Infiltrating Lymphocyte Lytic Function. THE JOURNAL OF IMMUNOLOGY 2005; 174:1830-40. [PMID: 15699109 DOI: 10.4049/jimmunol.174.4.1830] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
CD8+ tumor-infiltrating lymphocytes (TIL) are severely deficient in cytolysis, a defect that may permit tumor escape from immune-mediated destruction. Because lytic function is dependent upon TCR signaling, we have tested the hypothesis that primary TIL have defective signaling by analysis of the localization and activation status of TIL proteins important in TCR-mediated signaling. Upon conjugate formation with cognate target cells in vitro, TIL do not recruit granzyme B+ granules, the microtubule-organizing center, F-actin, Wiskott-Aldrich syndrome protein, nor proline rich tyrosine kinase-2 to the target cell contact site. In addition, TIL do not flux calcium nor demonstrate proximal tyrosine kinase activity, deficiencies likely to underlie failure to fully activate the lytic machinery. Confocal microscopy and fluorescence resonance energy transfer analyses demonstrate that TIL are triggered by conjugate formation in that the TCR, p56lck, CD3zeta, LFA-1, lipid rafts, ZAP70, and linker for activation of T cells localize at the TIL:tumor cell contact site, and CD43 and CD45 are excluded. However, proximal TCR signaling is blocked upon conjugate formation because the inhibitory motif of p56lck is rapidly phosphorylated (Y505) and COOH-terminal Src kinase is recruited to the contact site, while Src homology 2 domain-containing protein phosphatase 2 is cytoplasmic. Our data support a novel mechanism explaining how tumor-induced inactivation of proximal TCR signaling regulates lytic function of antitumor T cells.
Collapse
MESH Headings
- Actins/deficiency
- Actins/metabolism
- Animals
- CD2 Antigens/metabolism
- CD3 Complex/metabolism
- CD8 Antigens/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Calcium/metabolism
- Cell Line, Tumor
- Cell Separation
- Cytoplasmic Granules/immunology
- Cytoplasmic Granules/metabolism
- Cytotoxicity, Immunologic
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/antagonists & inhibitors
- Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism
- Lymphocytes, Tumor-Infiltrating/enzymology
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Mice
- Mice, Inbred C57BL
- Phosphorylation
- Phosphotyrosine/metabolism
- Protein Transport/immunology
- Protein-Tyrosine Kinases/deficiency
- Protein-Tyrosine Kinases/metabolism
- Receptor-CD3 Complex, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/deficiency
- Receptors, Antigen, T-Cell/physiology
- Signal Transduction/immunology
- ZAP-70 Protein-Tyrosine Kinase
Collapse
Affiliation(s)
- Mythili Koneru
- Department of Cell Biology and Kaplan Cancer Center, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | |
Collapse
|
43
|
Chang CJ, Tai KF, Roffler S, Hwang LH. The Immunization Site of Cytokine-Secreting Tumor Cell Vaccines Influences the Trafficking of Tumor-Specific T Lymphocytes and Antitumor Efficacy against Regional Tumors. THE JOURNAL OF IMMUNOLOGY 2004; 173:6025-32. [PMID: 15528337 DOI: 10.4049/jimmunol.173.10.6025] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Tumor cells engineered to secrete cytokines, referred to as tumor cell vaccines, can often generate systemic antitumor immunity and, in many cases, cause tumor regression. We compared the efficacy of s.c. immunization or intrahepatic immunization of GM-CSF-expressing tumor cell vaccines on the growth of s.c. or orthotopic liver tumors. A chemically transformed hepatic epithelial cell line, GP7TB, derived from Fischer 344 rats, was used to generate tumor models and tumor cell vaccines. Our results demonstrated that two s.c. injections of an irradiated tumor cell vaccine significantly controlled the growth of s.c. tumors, but was completely ineffective against orthotopic liver tumors. Effector cell infiltration in liver tumors was markedly reduced compared with s.c. tumors. Enhanced apoptosis of some effector cells was observed in the liver tumors compared with the s.c. tumors. Furthermore, the T cells induced by s.c. immunization preferentially migrated to s.c. tumor sites, as demonstrated by adoptive transfer experiments. In contrast, intrahepatic immunization, using parental tumor cells admixed with adenoviruses carrying the GM-CSF gene, yielded significantly better therapeutic effects on the liver tumors than on the s.c. tumors. Adoptive transfer experiments further confirmed that the T cells induced by liver immunization preferentially migrated to the liver tumor sites. Our results demonstrate that distinct T cell populations are induced by different immunization routes. Thus, the homing behavior of T cells depends on the route of immunization and is an important factor determining the efficacy of immunotherapy for regional tumors.
Collapse
MESH Headings
- Adenoviridae/genetics
- Animals
- Apoptosis/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cancer Vaccines/radiation effects
- Cancer Vaccines/therapeutic use
- Cell Count
- Cell Line, Tumor
- Cell Proliferation/radiation effects
- Cytokines/metabolism
- Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage
- Granulocyte-Macrophage Colony-Stimulating Factor/immunology
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/immunology
- Growth Inhibitors/radiation effects
- Growth Inhibitors/therapeutic use
- Immunotherapy, Adoptive/methods
- Injections, Subcutaneous
- Liver/immunology
- Liver/pathology
- Liver Neoplasms, Experimental/immunology
- Liver Neoplasms, Experimental/metabolism
- Liver Neoplasms, Experimental/pathology
- Liver Neoplasms, Experimental/prevention & control
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Male
- Neoplasm Transplantation
- Rats
- Rats, Inbred F344
- Skin/immunology
- Skin/pathology
- Splenic Neoplasms/immunology
- Splenic Neoplasms/pathology
- Splenic Neoplasms/prevention & control
Collapse
Affiliation(s)
- Chun-Jung Chang
- Graduate Institute of Microbiology, National Taiwan University College of Medicine, 7 Chung-Shan South Road, Taipei 10016
| | | | | | | |
Collapse
|
44
|
Derweesh IH, Tannenbaum CS, Rayman PA, Finke JH. Mechanisms of immune dysfunction in renal cell carcinoma. Cancer Treat Res 2003; 116:29-51. [PMID: 14650824 DOI: 10.1007/978-1-4615-0451-1_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/27/2023]
Affiliation(s)
- Ithaar H Derweesh
- Department of Immunology, Lerner Research Institute, Glickman Urological Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | | | | | | |
Collapse
|
45
|
Ando H, Saio M, Tamakawa N, Ohe N, Nakayama T, Yu H, Kaku Y, Iwama T, Shinoda J, Sakai N, Takami T. Failure of B7.1-modified tumor to evoke full activation of CD8+ tumor-infiltrating lymphocytes in the central nervous system: prevention of parental tumor growth in the subcutaneous environment. J Neurosurg 2002; 97:432-40. [PMID: 12186473 DOI: 10.3171/jns.2002.97.2.0432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT It is well known that the central nervous system (CNS) is an immunologically privileged site. To characterize CD8+ tumor-infiltrating lymphocytes (TILs) recovered from the CNS, the authors compared these cells with TILs recovered from subcutaneous tissue by using a B7.1 gene-modified tumor implantation model. METHODS The authors established a B7.1 gene-modified EL4 murine lymphoma cell line (EL4-B7.1) and implanted the cells into the CNS to observe the duration of tumor-free survival. Although EL4-B7.1 cells were completely rejected in a subcutaneous implantation model, 40% of animals died after the CNS implantation (all animals in which the parent tumor was implanted died within 16 days). Therefore, the authors isolated TILs from each implantation site and analyzed the expressions of activation antigens CD25 and CD69 by performing the anti-CD8 magnetic beads separation method and flow cytometric analysis. After implantation of the parent tumor, there was no difference in the number of TILs from each site (CD25 1.7-3.2%, CD69 21.9-34.3%). After implantation of the B7.1-modified tumor, the CD25-expressing TIL population from the subcutaneous site was 4.68 times higher than that from the CNS site (17.8% compared with 3.8%). Based on these findings, the authors used a mitomycin C-treated EL4-B7.1 subcutaneous vaccination with various protocols. Vaccination before tumor challenge was sufficient to prevent the development of the tumor. For animals with established tumor, the vaccination protocol was able to prolong host survival (p = 0.0053). CONCLUSIONS The data clearly demonstrate that the CNS environment fails to activate CD8+ TILs fully. These are the first data indicating in detail a difference between CD8+ TILs from the CNS and those from other sites based on a B7.1-modified tumor model.
Collapse
Affiliation(s)
- Hiromichi Ando
- Department of Neurosurgery, Gifu University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Keilholz U, Weber J, Finke JH, Gabrilovich DI, Kast WM, Disis ML, Kirkwood JM, Scheibenbogen C, Schlom J, Maino VC, Lyerly HK, Lee PP, Storkus W, Marincola F, Worobec A, Atkins MB. Immunologic monitoring of cancer vaccine therapy: results of a workshop sponsored by the Society for Biological Therapy. J Immunother 2002; 25:97-138. [PMID: 12074049 DOI: 10.1097/00002371-200203000-00001] [Citation(s) in RCA: 222] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The Society for Biological Therapy held a Workshop last fall devoted to immune monitoring for cancer immunotherapy trials. Participants included members of the academic and pharmaceutical communities as well as the National Cancer Institute and the Food and Drug Administration. Discussion focused on the relative merits and appropriate use of various immune monitoring tools. Six breakout groups dealt with assays of T-cell function, serologic and proliferation assays to assess B cell and T helper cell activity, and enzyme-linked immunospot assay, tetramer, cytokine flow cytometry, and reverse transcription polymerase chain reaction assays of T-cell immunity. General conclusions included: (1) future vaccine studies should be designed to determine whether T-cell dysfunction (tumor-specific and nonspecific) correlated with clinical outcome; (2) tetramer-based assays yield quantitative but not functional data (3) enzyme-linked immunospot assays have the lowest limit of detection (4) cytokine flow cytometry have a higher limit of detection than enzyme-linked immunospot assay, but offer the advantages of speed and the ability to identify subsets of reactive cells; (5) antibody tests are simple and accurate and should be incorporated to a greater extent in monitoring plans; (6) proliferation assays are imprecise and should not be emphasized in future studies; (7) the reverse transcription polymerase chain reaction assay is a promising research approach that is not ready for widespread application; and (8)there is a critical need to validate these assays as surrogates for vaccine potency and clinical effect. Current data and opinion support the use of a functional assay like the enzyme-linked immunospot assay or cytokine flow cytometry in combination with a quantitative assay like tetramers for immune monitoring. At present, assays appear to be most useful as measures of vaccine potency. Careful immune monitoring in association with larger scale clinical trials ultimately may enable the correlation of monitoring results with clinical benefit.
Collapse
|
47
|
Radoja S, Saio M, Schaer D, Koneru M, Vukmanovic S, Frey AB. CD8(+) tumor-infiltrating T cells are deficient in perforin-mediated cytolytic activity due to defective microtubule-organizing center mobilization and lytic granule exocytosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:5042-51. [PMID: 11673513 DOI: 10.4049/jimmunol.167.9.5042] [Citation(s) in RCA: 121] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tumor-infiltrating lymphocytes (TIL) are well known to be functionally impaired typified by the inability to lyse cognate tumor cells in vitro. We have investigated the basis for defective TIL lytic function in transplantable murine tumor models. CD8(+) TIL are nonlytic immediately on isolation even though they express surface activation markers, contain effector phase cytokine mRNAs, and contain perforin and granzyme B proteins which are packaged into lytic granules. Ag-specific lytic capability is rapidly recovered if purified TIL are briefly cultured in vitro and tumor lysis is perforin-, but not Fas ligand mediated. In response to TCR ligation of nonlytic TIL in vitro, proximal and distal signaling events are normal; calcium flux is rapid; mitogen-activated protein/extracellular signal-related kinase kinase, extracellular regulatory kinase 2, phosphoinositide-3 kinase, and protein kinase C are activated; and IL-2 and IFN-gamma is secreted. However, on conjugate formation between nonlytic TIL and cognate tumor cells in vitro, the microtubule-organizing center (MTOC) does not localize to the immunological synapse, thereby precluding exocytosis of preformed lytic granules and accounting for defective TIL lytic function. Recovery of TCR-mediated, activation-dependent MTOC mobilization and lytic activity requires proteasome function, implying the existence of an inhibitor of MTOC mobilization. Our findings show that the regulated release of TIL cytolytic granules is defective despite functional TCR-mediated signal transduction.
Collapse
Affiliation(s)
- S Radoja
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | |
Collapse
|