37 results on '"Mithun Khattar"'
Search Results
2. Interleukin 21 (IL-21) regulates chronic allograft vasculopathy (CAV) in murine heart allograft rejection.
- Author
-
Mithun Khattar, Caitlin E Baum, Paul Schroder, Joshua D Breidenbach, Steven T Haller, Wenhao Chen, and Stanislaw Stepkowski
- Subjects
Medicine ,Science - Abstract
IL-21 is the most recently discovered common gamma-chain cytokine that promotes persistent T-cell responses in chronic infections, autoimmunity and cancer. However, the therapeutic potential of inhibiting the IL-21-BATF signaling axis, particularly in transplant rejection, remains unclear. We used heart transplant models to examine the effects of IL-21 blockade in prevention of chronic cardiac allograft vasculopathy (CAV) using genetic knock-out and therapeutic approaches. Both wild-type C57BL/6 and IL-21-/- strains acutely rejected Balb/c skin grafts and once immunized with this skin graft, rejected Balb/c heart allografts in an accelerated fashion. However, when transplanted with heart grafts from the class-II major histocompatibility complex mutant, B6bm12 mice; wild-type recipients developed CAV, while IL-21-/- recipients were protected, even at day 100 post-transplant. Similarly, BATF-/- recipients, lacking the transcription factor BATF responsible for IL-21 production, did not develop CAV in B6-bm12 heart allografts. Strikingly, in a transient treatment protocol, the development of CAV in wild-type recipients of B6-bm12 hearts allografts was blocked by the administration of IL-21 receptor fusion protein (R-Fc). Thus, we demonstrate that CAV is regulated at least in part by IL-21 signaling and its blockade by genetic approaches or therapy with IL-21R-Fc prevents CAV in mice.
- Published
- 2019
- Full Text
- View/download PDF
3. Interleukin-21 is a critical regulator of CD4 and CD8 T cell survival during priming under Interleukin-2 deprivation conditions.
- Author
-
Mithun Khattar, Yoshihiro Miyahara, Paul M Schroder, Aini Xie, Wenhao Chen, and Stanislaw M Stepkowski
- Subjects
Medicine ,Science - Abstract
Optimal T cell activation and expansion require binding of the common gamma-chain (γc) cytokine Interleukin-2 (IL-2) to its cognate receptor that in turn engages a γc/Janus tyrosine kinase (Jak)3 signaling pathway. Because of its restricted expression by antigen-activated T cells and its obligatory role in promoting their survival and proliferation, IL-2 has been considered as a selective therapeutic target for preventing T cell mediated diseases. However, in order to further explore IL-2 targeted therapy, it is critical to precisely understand its role during early events of T cell activation. In this study, we delineate the role of IL-2 and other γc cytokines in promoting the survival of CD4 and CD8 T cells during early phases of priming. Under IL-2 inhibitory conditions (by neutralizing anti-IL-2 mAbs), the survival of activated CD8⁺ T cells was reduced, whereas CD4⁺ T cells remained much more resistant. These results correlated with reduced Bcl-2 expression, and mitochondrial membrane potential in CD8⁺ T cells in comparison to CD4⁺ T cells. However, using transwell co-culture assays we have found that CD4⁺ T cells could rescue the survival of CD8⁺ T cells even under IL-2 deprived conditions via secretion of soluble factors. A cytokine screen performed on CD8⁺ T cells cultured alone revealed that IL-21, another γc cytokine, was capable of rescuing their survival under IL-2 deprivation. Indeed, blocking the IL-21 signaling pathway along with IL-2 neutralization resulted in significantly reduced survival of both CD4⁺ and CD8⁺ T cells. Taken together, we have shown that under IL-2 deprivation conditions, IL-21 may act as the major survival factor promoting T cell immune responses. Thus, investigation of IL-2 targeted therapies may need to be revisited to consider blockade of the IL-21 signaling pathways as an adjunct to provide more effective control of T cell immune responses.
- Published
- 2014
- Full Text
- View/download PDF
4. Transient combination therapy targeting the immune synapse abrogates T cell responses and prolongs allograft survival in mice.
- Author
-
Paul M Schroder, Mithun Khattar, Ronghai Deng, Aini Xie, Wenhao Chen, and Stanislaw M Stepkowski
- Subjects
Medicine ,Science - Abstract
T cells play a major role in allograft rejection, which occurs after T cell activation by the engagement of several functional molecules to form an immune synapse with alloantigen presenting cells. In this study, the immune synapse was targeted using mAbs directed to the TCR beta-chain (TCRβ) and lymphocyte function-associated antigen-1 (LFA1) to induce long-term allograft survival. Evaluation of antigen-specific T cell responses was performed by adoptively transferring CFSE labeled transgenic OT-II cells into wild-type mice and providing OVA peptide by intravenous injection. Graft survival studies were performed in mice by transplanting BALB/c ear skins onto the flanks of C57BL/6 recipients. The anti-TCRβ plus anti-LFA1 mAb combination (but not either mAb alone) abrogated antigen-specific T cell responses invitro and invivo. Transient combination therapy with these agents resulted in significantly prolonged skin allograft survival in mice (51±10 days; p
- Published
- 2013
- Full Text
- View/download PDF
5. Introduction
- Author
-
Mithun Khattar and Karuppiah Kannan
- Published
- 2022
- Full Text
- View/download PDF
6. 390 Digital spatial profiling and antigen-dependent phenotypic analysis of IL15-engineered tumor-infiltrating lymphocytes (cytoTIL15® therapy) in an allogeneic melanoma PDX model
- Author
-
Rachel Burga, Zheng Ao, Arman Aksoy, Scott Lajoie, Kyle Pedro, Jack Tremblay, Gauri Kulkarni, Alonso Villasmil Ocando, Benjamin Primack, Meghan Langley, Theresa Ross, Jeremy Tchaicha, Michelle Ols, Jan ter Meulen, and Mithun Khattar
- Published
- 2022
- Full Text
- View/download PDF
7. 369 Enhancers of innate and adaptive immunity combine with membrane bound IL15 to increase the efficacy of tumor infiltrating lymphocyte (TIL) therapy for tumors with immunosuppressive microenvironments
- Author
-
Carmela Passaro, Balazs Koscso, Sean Smith, Violet Young, Theresa Ross, Benjamin Primack, Natasha Ly, Patricia Timpug, Shabnam Davoodi, Rachel Burga, Gauri Kulkarni, Scott Lajoie, Meghan Langley, Nirzari Shah, Dexue Sun, Dan Jun Li, Raina Duan, Arman Aksoy, Mithun Khattar, Jeremy Tchaicha, Dhruv Sethi, Jan ter Meulen, and Michelle Ols
- Published
- 2022
- Full Text
- View/download PDF
8. 166 Genetically engineered tumor-infiltrating lymphocytes (cytoTIL15) exhibit IL-2-independent persistence and anti-tumor efficacy against melanoma in vivo
- Author
-
Sunandan Saha, Rachel A. Burga, Meghan Langley, Michelle Ols, Jack Tremblay, Stanley Tam, Jeremy Hatem Tchaicha, Benjamin Primack, Colleen Foley, Gabriel Helmlinger, Theresa Ross, Sethi Dhruv Kam, Emily Brideau, Shyam Subramanian, Alonso Villasmil Ocando, Kyle Pedro, Mithun Khattar, Scott Lajoie, Gary Vanasse, Dan Thornton, Jan ter Meulen, and Gwen Wilmes
- Subjects
Pharmacology ,Antitumor activity ,Cancer Research ,Tumor-infiltrating lymphocytes ,Genetically engineered ,Melanoma ,Immunology ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Biology ,medicine.disease ,Persistence (computer science) ,Oncology ,In vivo ,medicine ,Cancer research ,Molecular Medicine ,Immunology and Allergy ,RC254-282 - Abstract
BackgroundAdoptive cell therapy with tumor-infiltrating lymphocytes (TILs) has demonstrated tremendous promise in clinical trials for patients with solid or metastatic tumors.1 However, current TIL therapy requires systemic administration of IL-2 to promote TIL survival, and IL-2-associated toxicities greatly limit patient eligibility and reduce the long-term clinical benefit of TIL therapy.2 3 Unlike IL-2, which promotes T cell exhaustion, IL-15 maintains antigen-independent TIL persistence through homeostatic proliferation and supports CD8+ T cell anti-tumor activity without stimulating regulatory T cells. We designed genetically engineered TILs to express a regulated form of membrane-bound IL-15 (mbIL15) for tunable long-term persistence, leading to enhanced efficacy and safety for the treatment of patients with solid tumors.MethodsObsidian’s cytoDRiVE™ platform includes small human protein sequences called drug responsive domains (DRD)s that enable regulated expression of a fused target protein under control of FDA-approved, bioavailable small molecule ligands. cytoTIL15 contains TILs engineered with mbIL15 under the control of a carbonic-anhydrase-2 DRD, controlled by the ligand acetazolamide (ACZ). After isolation from tumors, TILs were transduced and expanded in vitro through a proprietary TIL expansion process. cytoTIL15 were immunophenotyped and assessed for in vitro antigen-independent survival and co-cultured with tumor cells to assess polyfunctionality and cytotoxicity. In vivo TIL persistence and anti-tumor efficacy was evaluated through adoptive transfer of TILs into immunodeficient NSG mice, either naïve or implanted with subcutaneous patient-derived-xenograft (PDX) tumors.Results cytoTIL15 and conventional IL2-dependent TILs isolated from melanoma tumor samples expanded to clinically relevant numbers over 14 days. Throughout expansion, cytoTIL15 were enriched for CD8+ T cells and acquired enhanced memory-like characteristics, while maintaining diverse TCRVβ sub-family representation. cytoTIL15 demonstrated enhanced potency over conventional TILs, as measured by increased polyfunctionality and cytotoxicity against tumor and PDX lines in vitro (figure 1A). In a 10-day antigen-independent in vitro assay, cytoTIL15 persisted at greater frequencies than conventional TILs in the absence of IL-2 (figure 1B; *p30 days following adoptive cell transfer (figure 1C). Importantly, cytoTIL15 achieved significant tumor control in a human PDX model (figure 1D), which correlated with increased TIL accumulation in secondary lymphoid organs.Abstract 166 Figure 1cytoTIL15 demonstrate superior persistence. cytoTIL15 is an engineered TIL product expressing regulatable mbIL15. (A) cytoTIL15 demonstrate enhanced in vitro cytotoxicity after co-culture with melanoma tumor lines (representative data from 3 TIL donors). (B) cytoTIL15 have improved persistence in antigen- and IL2- independent culture conditions in vitro compared to conventional TILs cultured in the absence of IL-2 as well as (C) in vivo compared to conventional TILs supplemented with IL-2, when engrafted into NSG mice (in vitro: representative data from 1 TIL donor, performed in >3 replicate donors, in vivo: n=5/group, representative of 1 TIL donor, performed in >3 replicate donors). (D) cytoTIL15 (with 200mg/kg ACZ PO QD) demonstrate enhanced anti-tumor efficacy in a xenograft melanoma model as compared to conventional TILs (with 50000 IU IL-2 q8h BID, IP for 5 days) (n=8/group, representative of 1 TIL donor, performed in >2 replicate donors; ACT = adoptive cell transfer).ConclusionsTaken together, the superior persistence and potency of cytoTIL15 in the complete absence of IL-2 highlights the clinical potential of cytoTIL15 as a novel TIL product with enhanced safety and efficacy for patients with melanomas, and other solid tumors.AcknowledgementsThe authors wish to acknowledge the Cooperative Human Tissue Network for the their supply of human tumor tissue, and the MD Anderson Cancer Center for technical support; schematic created with BioRender.com.ReferencesChandran SS, Somerville RPT, Yang JC, Sherry RM, Klebanoff CA, Goff SL, Wunderlich JR, Danforth DN, Zlott D, Paria BC, Sabesan AC, Srivastava AK, Xi L, Pham TH, Raffeld M, White DE, Toomey MA, Rosenberg SA, Kammula US. Treatment of metastatic uveal melanoma with adoptive transfer of tumour-infiltrating lymphocytes: a single-centre, two-stage, single-arm, phase 2 study. Lancet Oncol 2017 Jun;18(6):792–802. doi: 10.1016/S1470-2045(17)30251-6. Epub 2017 Apr 7. PMID: 28395880; PMCID: PMC5490083.Yang JC. Toxicities associated with adoptive T-cell transfer for Cancer. Cancer J 2015;21:506–9.Schwartz RN, Stover L, Dutcher JP. Managing toxicities of high-dose interleukin-2. Oncology (Williston Park) 2002 Nov;16(11 Suppl 13):11–20. PMID: 12469935.
- Published
- 2021
9. A small-molecule SUMOylation inhibitor activates antitumor immune responses and potentiates immune therapies in preclinical models
- Author
-
Keli Song, James Minissale, Richard A. Klinghoffer, James Garnsey, Katherine Galvin, Kristina Xega, Sai M Pulukuri, James M. Gavin, Xiaofeng Yang, Pooja Shah, Gary Shapiro, Cong Li, Vaishali Shinde, Eric S. Lightcap, Dennis Huszar, Zhen Lu, Stephen Grossman, Erik Koenig, Pengfei Yu, Steve Langston, Hisashi Imaichi, Serge Y. Fuchs, Mithun Khattar, Beryl A. Hatton, Yu Fu, Hongru Zhang, Dylan England, Michael Milhollen, Jessica Riceberg, and Xingyue He
- Subjects
Immune system ,Chemistry ,Interferon ,Immunity ,Cancer research ,medicine ,SUMO protein ,Sumoylation ,General Medicine ,Small molecule ,Article ,Immune therapy ,medicine.drug - Abstract
SUMOylation, the covalent conjugation of small ubiquitin-like modifier (SUMO) proteins to protein substrates, has been reported to suppress type I interferon (IFN1) responses. TAK-981, a selective small molecule inhibitor of SUMOylation, pharmacologically reactivates IFN1 signaling and immune responses against cancers. In vivo treatment of wild type mice with TAK-981 upregulated IFN1 gene expression in blood cells and splenocytes. Ex vivo treatment of mouse and human dendritic cells promoted their IFN1-dependent activation, and vaccination studies in mice demonstrated stimulation of antigen cross-presentation and T cell priming in vivo. TAK-981 also directly stimulated T cell activation, driving enhanced T cell sensitivity and response to antigen ex vivo. Consistent with these observations, TAK-981 inhibited growth of syngeneic A20 and MC38 tumors in mice, dependent upon IFN1 signaling and CD8(+) T cells, and associated with increased intratumoral T and NK cell number and activation. Combination of TAK-981 with anti-PD1 or anti-CTLA4 antibodies improved the survival of mice bearing syngeneic CT26 and MC38 tumors. In conclusion, TAK-981 is a first in class SUMOylation inhibitor that promotes anti-tumor immune responses by activation of IFN1 signaling. TAK-981 is currently being studied in phase I clinical trials (NCT03648372, NCT04074330, NCT04776018 and NCT04381650) for the treatment of patients with solid tumors and lymphomas.
- Published
- 2021
- Full Text
- View/download PDF
10. Abstract LB212: Allogeneic, IL-2-independent tumor-infiltrating lymphocytes expressing membrane-bound IL-15 (cytoTIL15࣪) eradicate tumors in a melanoma PDX model through recognition of shared tumor antigens
- Author
-
Jeremy H. Tchaicha, Scott Lajoie, Rachel Burga, Theresa Ross, Benjamin Primack, Meghan Langley, Violet Young, Alonso Villasmil Ocando, Kyle Pedro, Jack Tremblay, Gauri Kulkarni, Mithun Khattar, Dhruv Sethi, Michelle Ols, Gabriel Helmlinger, Gary Vanasse, Shyam Subramanian, and Jan ter Meulen
- Subjects
Cancer Research ,Oncology - Abstract
Standard tumor-infiltrating lymphocyte (TIL) therapy requires IL-2 administration to support TIL expansion and survival, but this cytokine is associated with T cell exhaustion and can result in severe toxicities that limit patient eligibility (1). To this end, we genetically engineered TIL to express membrane-bound IL-15 (mbIL15) under the control of Obsidian’s cytoDRIVE® technology (cytoTIL15࣪), which allows regulation of protein expression via a drug-responsive domain upon acetazolamide (ACZ) administration. IL-15 is a preferred cytokine over IL-2 to mediate TIL activation and expansion, because it does not result in CD8 T cell exhaustion or stimulate regulatory CD4 T cells, and enhances development of a memory T-cell phenotype. We have previously demonstrated IL-2-independent, 3-6-fold increased cytoTIL15 persistence in an antigen-independent setting relative to unengineered TIL therapy with IL-2 (uTIL) (2). Due to the challenge of generating autologous tumor/TIL-matched pairs and most importantly, to assess cytoTIL15 cell’s functional impact on anti-tumor growth across multiple donors, we developed an allogeneic patient-derived xenograft (PDX) model. To establish the model, different melanoma tumor digests were co-incubated in vitro with select HLA-A*02-matched, allogeneic melanoma TIL donors to assess their reactivity. Tumors were screened for expression of shared antigens, such as gp100 and MART1, and TIL donor TCRs were screened with tetramers. Once established, serially passaged tumor fragments were grown, measured, and randomized into groups to receive intravenous transfer of TIL (n=8/cohort). Mice receiving uTIL were treated with four saturating doses of recombinant IL-2, and mice receiving cytoTIL15 cells received either vehicle or oral 200 mg/kg ACZ daily for the entire study, without any IL-2. Three of four cytoTIL15 cell preparations from different donors dosed with ACZ achieved significant tumor growth inhibition compared to uTIL. Four mice developed complete responses as early as 17 days post cytoTIL15 cell transfer. The level of anti-tumor response was associated with increased frequency of MART1-reactive cytoTIL15 cells. On day 20 after TIL transfer, tumors and secondary lymphoid organs were collected (n=4/cohort). Tumors treated with cytoTIL15 cells + ACZ showed an 8-10-fold increased TIL infiltration compared to uTIL or cytoTIL15 cells + vehicle. Moreover, enhanced cytoTIL15 cell infiltration and anti-tumor activity was associated with increases in pro-inflammatory cytokines (e.g., IFNγ). Taken together, these data clearly demonstrate the superiority of cytoTIL15 cells over uTIL for controlling or eradicating melanoma tumor outgrowth and the utility of an allogeneic PDX model for comparative evaluation of tumor-antigen specific TIL reactivity. References: 1. Yang JC. Toxicities associated with adoptive T-cell transfer for Cancer. Cancer J. 2015. 2. Burga R. et al Genetically engineered tumor-infiltrating lymphocytes (cytoTIL15) exhibit IL-2-independent persistence and anti-tumor efficacy against melanoma in vivo. SITC 36th annual meeting 2021. Citation Format: Jeremy H. Tchaicha, Scott Lajoie, Rachel Burga, Theresa Ross, Benjamin Primack, Meghan Langley, Violet Young, Alonso Villasmil Ocando, Kyle Pedro, Jack Tremblay, Gauri Kulkarni, Mithun Khattar, Dhruv Sethi, Michelle Ols, Gabriel Helmlinger, Gary Vanasse, Shyam Subramanian, Jan ter Meulen. Allogeneic, IL-2-independent tumor-infiltrating lymphocytes expressing membrane-bound IL-15 (cytoTIL15࣪) eradicate tumors in a melanoma PDX model through recognition of shared tumor antigens [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr LB212.
- Published
- 2022
- Full Text
- View/download PDF
11. PD-1-dependent restoration of self-tolerance in the NOD mouse model of diabetes after transient anti-TCRβ mAb therapy
- Author
-
Mithun Khattar, Yoshihiro Miyahara, Shravan Muralidharan, Paul M. Schroder, Caitlin E. Baum, Stanislaw M. Stepkowski, Wenhao Chen, Beata Mierzejewska, and Rohit Vyas
- Subjects
Blood Glucose ,CD4-Positive T-Lymphocytes ,medicine.drug_class ,Receptors, Antigen, T-Cell, alpha-beta ,Endocrinology, Diabetes and Metabolism ,T cell ,medicine.medical_treatment ,Programmed Cell Death 1 Receptor ,CD8-Positive T-Lymphocytes ,Monoclonal antibody ,medicine.disease_cause ,Diabetes Mellitus, Experimental ,Autoimmunity ,Mice ,Receptors, CCR ,Antigen ,Mice, Inbred NOD ,Internal Medicine ,Animals ,Medicine ,NOD mice ,Inflammation ,Mice, Inbred BALB C ,biology ,business.industry ,T-cell receptor ,Antibodies, Monoclonal ,Immunotherapy ,Glucose Tolerance Test ,Allografts ,Mice, Inbred C57BL ,Diabetes Mellitus, Type 1 ,medicine.anatomical_structure ,Immunology ,biology.protein ,Cytokines ,Female ,Antibody ,business - Abstract
T cells play a major role in the pathogenesis of type 1 diabetes, and there is great interest in developing curative immunotherapies targeting these cells. In this study, a monoclonal antibody (mAb) targeting the T cell receptor β-chain (TCRβ) was investigated for its ability to prevent and reverse disease in mouse models of diabetes.RIP-OVA(hi) (C57BL/6-Tg(Ins2-OVA)59Wehi/WehiJ) mice adoptively transferred with ovalbumin-specific T cells (an induced model of diabetes) and NOD mice (a spontaneous model of diabetes) were used to test anti-TCRβ mAb therapy as a means of preventing and reversing type 1 diabetes.A single dose of anti-TCRβ completely prevented disease in RIP-OVA(hi) mice without inducing the release of inflammatory cytokines. Transient anti-TCRβ therapy prevented diabetes in 90% of NOD mice and reversed the disease after its onset in 73% of NOD mice. Long after the remission of type 1 diabetes, the anti-TCRβ treated mice were able to reject BALB/c skin allografts with normal kinetics while maintaining normoglycaemia. Treatment did not cause significant reductions in lymphocyte numbers in the spleen or pancreatic lymph nodes, but did result in a decreased percentage of chemokine receptor 9 (CCR9) positive, CD8(+) T cells. Notably, anti-TCRβ therapy increased the expression of programmed death 1 (PD-1) on the surface of the T cells; PD-1 expression is important for maintaining anti-TCRβ-induced self-tolerance, as type 1 diabetes recurs in mice following a blockade of PD-1 signalling.Anti-TCRβ mAb is a safe and effective immunotherapy that results in reduced numbers of CCR9(+) T cells, an increased expression of PD-1 on T cells and the restoration of self-tolerance in NOD mice.
- Published
- 2015
- Full Text
- View/download PDF
12. TCR stimulation without co-stimulatory signals induces expression of 'tolerogenic' genes in memory CD4 T cells but does not compromise cell proliferation
- Author
-
Aini Xie, Xiong Zheng, Jiahong Xia, Paul M. Schroder, Stanislaw M. Stepkowski, Wenhao Chen, and Mithun Khattar
- Subjects
CD4-Positive T-Lymphocytes ,CD3 Complex ,Immunology ,Receptors, Antigen, T-Cell ,Biology ,Interleukin 21 ,Immune Tolerance ,STAT5 Transcription Factor ,Animals ,Cytotoxic T cell ,IL-2 receptor ,Phosphorylation ,Antigen-presenting cell ,Molecular Biology ,Early Growth Response Protein 2 ,Cell Proliferation ,Interleukin 3 ,Mice, Inbred BALB C ,ZAP70 ,Antibodies, Monoclonal ,CD28 ,Natural killer T cell ,Cell biology ,Mice, Inbred C57BL ,Proto-Oncogene Proteins c-bcl-2 ,Positive Regulatory Domain I-Binding Factor 1 ,Immunologic Memory ,Signal Transduction ,Transcription Factors - Abstract
Memory T cells resist co-stimulatory blockade and present a unique therapeutic challenge in transplantation and autoimmune diseases. Herein, we determined whether memory T cells express less "tolerogenic" genes than naïve T cells to reinforce a proliferative response under the deprivation of co-stimulatory signals. The expression of ∼40 tolerogenic genes in memory and naïve CD4(+) T cells was thus assessed during an in vitro TCR stimulation without co-stimulation. Briefly, upon TCR stimulation with an anti-CD3 mAb alone, memory CD4(+) T cells exhibited more proliferation than naïve CD4(+) T cells. To our surprise, at 24h upon anti-CD3 mAb stimulation, memory CD4(+) T cells expressed more than a 5-fold higher level of the transcription factor Egr2 and a 20-fold higher level of the transmembrane E3 ubiquitin ligase GRAIL than those in naïve T cells. Hence, the high-level expression of tolerogenic genes, Egr2 and GRAIL, in memory CD4(+) T cells does not prevent cell proliferation. Importantly, anti-CD3 mAb-stimulated memory CD4(+) T cells expressed high protein/gene levels of phosphorylated STAT5, Nedd4, Bcl-2, and Bcl-XL. Therefore, co-stimulation-independent proliferation of memory CD4(+) T cells may be due to elevated expression of molecules that support cell proliferation and survival, but not lack of tolerogenic molecules.
- Published
- 2015
- Full Text
- View/download PDF
13. Interleukin 21 (IL-21) regulates chronic allograft vasculopathy (CAV) in murine heart allograft rejection
- Author
-
Joshua D. Breidenbach, Stanislaw M. Stepkowski, Wenhao Chen, Mithun Khattar, Steven T. Haller, Paul M. Schroder, and Caitlin E. Baum
- Subjects
Graft Rejection ,0301 basic medicine ,B Cells ,Cardiovascular Procedures ,T-Lymphocytes ,medicine.medical_treatment ,Gene Expression ,Biochemistry ,White Blood Cells ,Mice ,0302 clinical medicine ,Animal Cells ,BATF ,Medicine and Health Sciences ,Medicine ,Mice, Knockout ,Heart transplantation ,Mice, Inbred BALB C ,Multidisciplinary ,T Cells ,Interleukin ,Heart ,Skin Transplantation ,Cardiac Transplantation ,Transplant rejection ,Basic-Leucine Zipper Transcription Factors ,Cytokine ,cardiovascular system ,Skin grafting ,Receptors, Interleukin-21 ,Anatomy ,Cellular Types ,Plastic Surgery and Reconstructive Techniques ,Research Article ,Science ,Immune Cells ,Recombinant Fusion Proteins ,Immunology ,Surgical and Invasive Medical Procedures ,Interferon-gamma ,03 medical and health sciences ,Transplantation Immunology ,DNA-binding proteins ,Genetics ,Animals ,Transplantation, Homologous ,Gene Regulation ,Vascular Diseases ,Antibody-Producing Cells ,Transplantation ,Blood Cells ,business.industry ,Interleukins ,Transplant Rejection ,Histocompatibility Antigens Class II ,Biology and Life Sciences ,Proteins ,Organ Transplantation ,Cell Biology ,medicine.disease ,Skin Grafting ,Regulatory Proteins ,Blockade ,Mice, Inbred C57BL ,030104 developmental biology ,Cardiovascular Anatomy ,Heart Transplantation ,Clinical Immunology ,Clinical Medicine ,business ,Transcription Factors ,030215 immunology - Abstract
IL-21 is the most recently discovered common gamma-chain cytokine that promotes persistent T-cell responses in chronic infections, autoimmunity and cancer. However, the therapeutic potential of inhibiting the IL-21-BATF signaling axis, particularly in transplant rejection, remains unclear. We used heart transplant models to examine the effects of IL-21 blockade in prevention of chronic cardiac allograft vasculopathy (CAV) using genetic knock-out and therapeutic approaches. Both wild-type C57BL/6 and IL-21-/- strains acutely rejected Balb/c skin grafts and once immunized with this skin graft, rejected Balb/c heart allografts in an accelerated fashion. However, when transplanted with heart grafts from the class-II major histocompatibility complex mutant, B6bm12 mice; wild-type recipients developed CAV, while IL-21-/- recipients were protected, even at day 100 post-transplant. Similarly, BATF-/- recipients, lacking the transcription factor BATF responsible for IL-21 production, did not develop CAV in B6-bm12 heart allografts. Strikingly, in a transient treatment protocol, the development of CAV in wild-type recipients of B6-bm12 hearts allografts was blocked by the administration of IL-21 receptor fusion protein (R-Fc). Thus, we demonstrate that CAV is regulated at least in part by IL-21 signaling and its blockade by genetic approaches or therapy with IL-21R-Fc prevents CAV in mice.
- Published
- 2019
- Full Text
- View/download PDF
14. Abstract 3252: TAK-981: A first in class SUMO inhibitor in Phase 1 trials that promotes dendritic cell activation, antigen-presentation, and T cell priming
- Author
-
Kristina Xega, Xingyue He, Neeraja Idamakanti, Mithun Khattar, Stephen Grossman, Keli Song, and Dennis Huszar
- Subjects
0301 basic medicine ,CD86 ,Cancer Research ,CD40 ,biology ,Chemistry ,T cell ,Antigen presentation ,Priming (immunology) ,Dendritic cell ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,biology.protein ,medicine ,Cancer research ,Cytotoxic T cell ,CD80 ,030215 immunology - Abstract
SUMOylation is a reversible post-translational modification that regulates protein function by covalent attachment of the small ubiquitin-like modifier (SUMO) protein to protein substrates. TAK-981 is a novel, selective small molecule inhibitor of the SUMOylation enzymatic cascade, which has demonstrated induction of potent anti-tumor immunity in preclinical models. In this study, we evaluated changes in immune cell composition and states after TAK-981 treatment in preclinical models. In both mouse bone-marrow and human peripheral blood mononuclear cell derived dendritic cells (DCs), TAK-981 treatment ex vivo induced markers of activation and maturation including CD40, CD80 and CD86, as well as increased secretion of inflammatory cytokines like IP-10, MCP1, MIP-1α, MIP1β, IFNα and IFNβ. These effects were reversed by an interferon alpha receptor (IFNAR) blocking antibody, demonstrating dependence of TAK-981 induced pharmacodynamic effects on Type I interferon signaling in DCs. In vivo, a single sub-cutaneous injection of TAK-981 in naïve Balb/c mice at the brachial lymph nodes induced activation of DCs, measured as significant increases in CD40 and CD86 expression. Concomitantly, an increase in expression of the early lymphocyte activation marker CD69 was observed on T cells and NK cells, demonstrating pleiotropic effects of TAK-981 on immune cells. Increased expression of CD69 was also observed in purified human T and NK cells treated with TAK-981 ex vivo. To investigate if enhanced activation of DCs by TAK-981 leads to improved cross-presentation of exogenous antigens, we challenged naïve C57BL/6 mice with chicken-ovalbumin (OVA) protein alone or in combination with TAK-981. Interestingly, an increase in the frequency of Kb-SIINFEKL tetramer positive CD8 T cells was observed in the draining lymph nodes overnight, and in the spleen on day 14, after treatment with TAK-981+OVA, suggesting that TAK-981 promotes cross-presentation of SIINFEKL, the class-I MHC epitope of OVA, to cognate antigen-specific CD8 T cells. We also observed an increase in the frequency of CD8α+ DCs loaded with the peptide ‘SIINFEKL’ on H-2Kb (class-I MHC of C57BL/6 mice), providing direct evidence for enhanced antigen-cross presentation. Similar findings were observed with TAK-981 administered either sub-cutaneously or via the therapeutic route of intra-venous injection in the above OVA immunization model. These results suggest a mechanism by which TAK-981 may promote anti-tumor immune responses in mice via enhanced cross-presentation of exogenous antigens released by dying tumor cells, leading to priming and activation of antigen reactive cytotoxic T cells. Currently, TAK-981 is being evaluated in Phase 1 clinical trials in adult patients with metastatic solid tumors or lymphomas (ClinicalTrials.gov Identifier: NCT03648372). Citation Format: Mithun Khattar, Keli Song, Stephen Grossman, Kristina Xega, Xingyue He, Neeraja Idamakanti, Dennis Huszar. TAK-981: A first in class SUMO inhibitor in Phase 1 trials that promotes dendritic cell activation, antigen-presentation, and T cell priming [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3252.
- Published
- 2019
- Full Text
- View/download PDF
15. Optimizing the use of regulatory T cells in allotransplantation: recent advances and future perspectives
- Author
-
Paul M. Schroder, Stanislaw M. Stepkowski, Caitlin E. Baum, Mithun Khattar, and Beata Mierzejewska
- Subjects
Graft Rejection ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Clonal deletion ,Autoimmunity ,Cell therapy ,Immune system ,medicine ,Animals ,Humans ,Immunology and Allergy ,Immunosuppression Therapy ,Transplantation ,business.industry ,FOXP3 ,hemic and immune systems ,Immunotherapy ,Transplantation Tolerance ,business ,Allotransplantation - Abstract
Apart from clonal deletion of self-reactive T cells in the thymus, Tregs are the major regulators of immune responses in the periphery and maintain a state of self-tolerance free from autoimmune diseases. Due to their inherent suppressive function, Tregs are being explored for their therapeutic potential in preventing autoimmunity and improving survival of allografts. This review provides recent updates on Treg biology and their use in animal as well as clinical models of transplantation. We discuss potential problems that limit the widespread clinical application of Tregs and provide future perspectives on how to optimize their medical use. Special consideration is given to methods by which Tregs should be isolated and expanded in order to facilitate clinical therapies. We also focus on recent discussions of Treg stability and plasticity, with specific insights into preventing the loss of Treg suppression by allotransplant-mediated inflammation.
- Published
- 2013
- Full Text
- View/download PDF
16. A Dynamic Dual Role of IL-2 Signaling in the Two-Step Differentiation Process of Adaptive Regulatory T Cells
- Author
-
Yoshihiro Miyahara, Stanislaw M. Stepkowski, Wenhao Chen, Xiaoshung He, Guohua Wang, Mithun Khattar, Zhiyong Guo, and Paul M. Schroder
- Subjects
CD4-Positive T-Lymphocytes ,Interleukin 2 ,T cell ,Cellular differentiation ,Immunology ,Receptors, Antigen, T-Cell ,Priming (immunology) ,chemical and pharmacologic phenomena ,Biology ,T-Lymphocytes, Regulatory ,Article ,Mice ,Transforming Growth Factor beta ,STAT5 Transcription Factor ,medicine ,Animals ,Immunology and Allergy ,IL-2 receptor ,Phosphorylation ,Mice, Knockout ,Precursor Cells, T-Lymphoid ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,Cell Differentiation ,Forkhead Transcription Factors ,hemic and immune systems ,Transforming growth factor beta ,Colitis ,Molecular biology ,Cell biology ,medicine.anatomical_structure ,CD4 Antigens ,biology.protein ,Interleukin-2 ,Signal transduction ,Signal Transduction ,medicine.drug - Abstract
The molecular mechanism of the extrathymic generation of adaptive, or inducible, CD4+Foxp3+ regulatory T cells (iTregs) remains incompletely defined. We show that exposure of splenic CD4+CD25+Foxp3− cells to IL-2, but not other common γ-chain cytokines, resulted in Stat5 phosphorylation and induced Foxp3 expression in ∼10% of the cells. Thus, IL-2/Stat5 signaling may be critical for Foxp3 induction in peripheral CD4+CD25+Foxp3– iTreg precursors. In this study, to further define the role of IL-2 in the formation of iTreg precursors as well as their subsequent Foxp3 expression, we designed a two-step iTreg differentiation model. During the initial “conditioning” step, CD4+CD25−Foxp3− naive T cells were activated by TCR stimulation. Inhibition of IL-2 signaling via Jak3–Stat5 was required during this step to generate CD4+CD25+Foxp3− cells containing iTreg precursors. During the subsequent Foxp3-induction step driven by cytokines, IL-2 was the most potent cytokine to induce Foxp3 expression in these iTreg precursors. This two-step method generated a large number of iTregs with relatively stable expression of Foxp3, which were able to prevent CD4+CD45RBhigh cell–mediated colitis in Rag1−/− mice. In consideration of this information, whereas initial inhibition of IL-2 signaling upon T cell priming generates iTreg precursors, subsequent activation of IL-2 signaling in these precursors induces the expression of Foxp3. These findings advance the understanding of iTreg differentiation and may facilitate the therapeutic use of iTregs in immune disorders.
- Published
- 2013
- Full Text
- View/download PDF
17. Abstract LB-294: Synergy of an anti-HER2 ADC TAK-522 (XMT-1522) in combination with anti-PD1 monoclonal antibody (mAb) in a syngeneic breast cancer model expressing human HER2
- Author
-
Tary Traore and Mithun Khattar
- Subjects
0301 basic medicine ,Cancer Research ,medicine.drug_class ,business.industry ,Cancer ,medicine.disease ,Monoclonal antibody ,Immune checkpoint ,Anti-PD1 Monoclonal Antibody ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Antigen ,SKBR3 ,030220 oncology & carcinogenesis ,Cancer cell ,Cancer research ,medicine ,Immunogenic cell death ,business - Abstract
Antibody-drug conjugates (ADCs) are a highly potent class of drugs that specifically target cancer cells expressing a tumor associated antigen (TAA). The ADC TAK-522 (XMT-1522) consists of a novel human IgG1 anti-HER2 monoclonal antibody and a novel, auristatin-based cytotoxic payload (Auristatin F-hydroxypropylamide, AF-HPA). An average DAR (drug-to-antibody ratio) of twelve AF-HPA molecules is achieved via a biodegradable polymer conjugation platform. We have characterized the ability of both the free payload AF-HPA and the ADC TAK-522 to induce immunogenic cell death (ICD) in vitro in multiple cell lines (NCI-N87, HT-29, SKBR3), as measured by cell surface expression of the ICD marker calreticulin (CRT) using microscopy and flow cytometry. CRT, usually contained in the lumen of the endoplasmic reticulum, translocated to the cell surface within a few hours after treatment with AF-HPA or TAK-522. Furthermore, we developed a novel syngeneic breast cancer (4T1) model expressing human HER2 at a relatively low antigen density. Treatment in this poorly immunogenic tumor model with TAK-522 but not Kadcyla showed significant inhibition of tumor growth in vivo. Importantly, a combination of anti-PD1 mAb and TAK-522 therapy substantially and synergistically enhanced the anti-tumor efficacy, resulting in complete responses in some mice. The frequency of complete responders was further increased when the two drugs were sequentially, rather than concurrently, administered such that TAK-522 administration was followed by anti-PD1 mAb therapy. These results suggest an immunological mechanism involving induction of immunogenic cell death by TAK-522, which in turn may activate the adaptive immune system by releasing tumor specific antigens. TAK-522 is currently being tested in a phase-1b clinical trial in patients with advanced breast, lung and gastric cancer expressing HER2. Based on our data, TAK-522 represents a potential candidate for combination therapies with immune checkpoint modulators in patients with poorly immunogenic HER2 expressing tumors. Citation Format: Tary Traore, Mithun Khattar. Synergy of an anti-HER2 ADC TAK-522 (XMT-1522) in combination with anti-PD1 monoclonal antibody (mAb) in a syngeneic breast cancer model expressing human HER2 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr LB-294.
- Published
- 2018
- Full Text
- View/download PDF
18. Synergy of an anti-HER2 ADC TAK-522 (XMT-1522) in combination with anti-PD1 monoclonal antibody (mAb) in a syngeneic breast cancer model expressing human HER2
- Author
-
Mithun Khattar, Tary Traore, Kristin Horton, Melissa Gallery, Pamela Brauer, Jessica Riceberg, Natalya Bodyak, Marina Protopopova, Qingxiu Zhang, Timothy B. Lowinger, Petter Veiby, Dennis Huszar, and Frank Wang
- Subjects
Immunology ,Immunology and Allergy - Abstract
Antibody-drug conjugates (ADCs) are a highly potent class of drugs that specifically target cancer cells expressing a tumor associated antigen. The ADC TAK-522 consists of a novel human IgG1 anti-HER2 monoclonal antibody and a novel, auristatin-based cytotoxic payload (Auristatin F-hydroxypropylamide, AF-HPA). An average DAR of twelve AF-HPA molecules is achieved via a biodegradable polymer conjugation platform. The free payload AF-HPA and TAK-522 induced immunogenic cell death (ICD) in vitro in multiple cell lines measured by cell surface expression of calreticulin using microscopy and flow cytometry. Furthermore, we developed a novel syngeneic breast cancer model expressing human HER2 at a relatively low antigen density. Treatment in this poorly immunogenic tumor model with TAK-522 but not Kadcyla showed significant inhibition of tumor growth in vivo. Importantly, a combination of anti-PD1 mAb and TAK-522 therapy substantially and synergistically enhanced the anti-tumor efficacy, resulting in complete responses in some mice. The frequency of complete responders was further increased when the two drugs were sequentially, rather than concurrently, administered such that TAK-522 administration was followed by anti-PD1 mAb therapy, suggesting an immunological mechanism with induction of immunogenic cell death by TAK-522, which may activate the adaptive immune system by releasing tumor specific antigens. TAK-522 is currently being tested in a phase-1b clinical trial in patients with advanced breast, lung and gastric cancer expressing HER2. Based on our data, TAK-522 represents a potential candidate for combination therapies with immune checkpoint modulators in patients with poorly immunogenic HER2 expressing tumors.
- Published
- 2018
- Full Text
- View/download PDF
19. Expanding and converting regulatory T cells: a horizon for immunotherapy
- Author
-
Mithun Khattar, Stanislaw M. Stepkowski, and Wenhao Chen
- Subjects
T cell ,medicine.medical_treatment ,Immunology ,chemical and pharmacologic phenomena ,Biology ,T-Lymphocytes, Regulatory ,Immune system ,Antigen ,Antigens, CD ,T-Lymphocyte Subsets ,medicine ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,CTLA-4 Antigen ,IL-2 receptor ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,Cell Differentiation ,Forkhead Transcription Factors ,General Medicine ,Immunotherapy ,Cell biology ,Transplantation ,medicine.anatomical_structure ,Cytokines - Abstract
The human immune system is a myriad of diverse cellular populations, each contributing to maintaining an effective and optimal immune response against infectious agents. It is important to maintain a "self-check" in the immune system so that responses do not go haywire, leading to the development of autoimmune diseases. Regulatory/suppressor T (Treg) cells are a specialized subpopulation of T cells that suppress the activation, expansion, and function of other T cells, thereby maintaining homeostasis through a fine balance between reactivity to foreign and self antigens. Tregs are characterized by surface expression of interleukin (IL)-2 receptor alpha chain (CD25) and intracellular expression of forkhead box protein P3 (FoxP3). There are at least two important functional populations of Treg cells, namely natural Treg (nTreg), which are continuously derived from the thymus, and induced Treg (iTreg), which are converted from naive T cells. The development and function of both nTreg and iTreg cells are regulated by several factors, such as antigen T-cell receptor, co-stimulatory receptors (i.e., cytotoxic T lymphocyte-associated antigen, or CTLA-4), and cytokines (IL-2, IL-10, and tumor growth factor-beta, or TGF-beta). In addition, the TGF-beta inhibitor ALK5, retinoid acid, and rapamycin influence the expansion of nTreg cells and the conversion of iTreg cells in vitro and in vivo. The heightening of Treg expansion may be harnessed to therapeutic methods for the treatment of autoimmune diseases and the induction of transplantation tolerance.
- Published
- 2009
- Full Text
- View/download PDF
20. Abstract 3654: AGEN1884, an IgG1 anti-CTLA-4 antibody, combines effectively with PD-1 blockade in primary human T cell assays and in a non-human primate pharmacodynamic (PD) model
- Author
-
Jennifer Buell, David Savitsky, John C. Castle, Xia Yang, Jeremy D. Waight, Gerd Ritter, Mithun Khattar, Randi Gombos, Jean-Marie Cuillerot, Ana M. Martín González, John M. Goldberg, Robert Stein, David Schaer, Nicholas S. Wilson, Dhan Chand, Daniel L. Levey, Antoine Tanne, Marc van Dijk, Benjamin Morin, Mark Findeis, Taha Merghoub, and Elise Drouin
- Subjects
CD86 ,Cancer Research ,T cell ,Priming (immunology) ,Pharmacology ,Biology ,medicine.anatomical_structure ,Oncology ,Antigen ,medicine ,biology.protein ,Cytotoxic T cell ,Antibody ,Antigen-presenting cell ,CD80 - Abstract
Cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) play important non-redundant roles in negatively regulating T cell immune responses. Therapeutic blockade of CTLA-4 or PD-1 pathways has been demonstrated to enhance T cell reactivity to tumor-specific antigens, translating to a significant improvement in overall survival. This anti-tumor effect can be further augmented when PD-1 and CTLA-4 antagonist antibodies are co-administered. The therapeutic impact of this combination is exemplified by the approval of this combination for advanced melanoma, as well as clinical benefit of the combination observed in NSCLC, mRCC, and most recently, mUC. AGEN1884, a human IgG1 antibody directed against CTLA-4, potently inhibits CTLA-4 binding to CD80 and CD86, resulting in enhanced T cell responsiveness in vitro, as well as in a vaccination model in non-human primates. A Phase 1 clinical study (NCT02694822) is currently ongoing to evaluate the safety and pharmacokinetic (PK)/pharmacodynamic (PD) relationships in patients with advanced solid tumors. AGEN2034 is a human IgG4 antibody that binds selectively to PD-1 with high affinity and potentiates T cell responsiveness via the blockade of PD-L1 and PD-L2 binding to PD-1. Here we evaluated the pharmacologic effect of combining AGEN1884 with AGEN2034, and other molecules targeting the PD-1/PD-L1 pathway, on primary human T cell immune responses. AGEN1884 combined effectively with AGEN2034, and other antibodies targeting the PD1/PD-L1 pathway, to promote superior T cell immune responses compared to monotherapies. Consistent with these in vitro findings, the co-administration of AGEN1884 with an anti-PD-1 antibody in cynomolgus monkeys (Macaca fascicularis) induced a dynamic PD effect, including a proliferative T cell response in peripheral blood, as compared to animals receiving either antibody alone. Finally, co-administration of an anti-mouse CTLA-4 antibody together with Agenus’ tumor-specific neo-epitope-based vaccine (AutoSynVax™) in mice induced effective amplification of vaccine-driven T cell responses, compared to animals that received the vaccine alone. These data further exemplify the versatility of harnessing antibody-mediated CTLA-4 blockade to influence apical events involved in T cell priming by antigen presenting cells. Taken together, these in vitro and in vivo findings demonstrate that the combination of AGEN1884 with PD-1 pathway blockade or with neo-epitope-based vaccines has the potential to provide therapeutic activity that is superior to that of either checkpoint- or vaccine-based monotherapies. Citation Format: Elise E. Drouin, David Savitsky, Ana M. Gonzalez, Randi Gombos, Dhan Chand, Jeremy Waight, Xia Yang, Mithun Khattar, Benjamin Morin, Mark Findeis, David Schaer, Taha Merghoub, Gerd Ritter, Antoine Tanne, Marc van Dijk, John M. Goldberg, Daniel Levey, John Castle, Jean-Marie Cuillerot, Jennifer S. Buell, Robert Stein, Nicholas S. Wilson. AGEN1884, an IgG1 anti-CTLA-4 antibody, combines effectively with PD-1 blockade in primary human T cell assays and in a non-human primate pharmacodynamic (PD) model [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 3654. doi:10.1158/1538-7445.AM2017-3654
- Published
- 2017
- Full Text
- View/download PDF
21. Abstract 4609: Agenus’ next generation cancer vaccine platforms
- Author
-
Daniel L. Levey, Mohamed Uduman, Antoine Tanne, Paisley T. Myers, Benjamin Morin, Jeffrey Raizer, Dennis J. Underwood, Shiwen Lin, Robert Stein, Mithun Khattar, Donald F. Hunt, Bishnu P. Joshi, Victor H. Engelhard, John C. Castle, Sandra Craig, Nicholas S. Wilson, Armen Karapetyan, Elise Drouin, Mark Findeis, Jennifer Buell, Erin D. Jeffery, John M. Goldberg, Amy Yang, and Mark Cobbold
- Subjects
Cancer Research ,Oncology ,business.industry ,Medicine ,Cancer vaccine ,business ,Virology - Abstract
Most cancer cells carry mutations unique to the patient’s individual tumor and shared biochemical signatures that are not present in healthy cells. Agenus has three vaccine platforms designed to treat cancers based on the unique needs of a given patient. Our vaccine platforms are designed to educate the patient’s immune system to recognize tumor-specific aberrations, or neo-antigens, and mount an anti-tumor immune response. Agenus’ Prophage™ vaccine platform is an individualized vaccine made from the patient’s own tumor tissue. Heat shock proteins (gp-96) that naturally chaperone and bind tumor-derived peptides are extracted from the patient’s tumor and constitute the vaccine. Some of these peptides are neo-antigens. Agenus has completed Phase 2 clinical trials with Prophage™ vaccine in newly diagnosed glioblastoma (ndGBM), and has previously reported that there was improved progression-free and overall survival with Prophage™ vaccine compared to standard of care. Agenus’ AutoSynVax™ vaccines are uniquely designed and manufactured for each patient based on NGS profiling of the patient’s tumor from a biopsy. Leveraging the Agenus Immunogenic Mutation (AIM™) workflow, we are able to generate a synthesis-ready blueprint for an optimal immunogenic and personalized neo-antigen vaccine. The AIM™ platform provides a robust and efficient approach to computational vaccinology designed to deliver a set of likely immunogenic peptides, agnostic to vaccine format, followed by generation of a format-specific blueprint ready for vaccine synthesis and manufacture. The synthesized neo-epitopes are complexed to recombinant heat shock protein 70 (HSC70) and are administered along with our QS-21 Stimulon® adjuvant. HSC70 is known to transport epitopes and play a role in displaying them to T cells. While the first two of Agenus’ vaccine platforms are highly individualized, our PhosphoSynVax™ vaccine is an off-the-shelf vaccine format targeting a novel class of tumor neo-antigens arising from post translational modifications (PTMs). Due to dysregulated cell signaling pathways in cancer, self-peptides can be aberrantly phosphorylated, a number of which are subsequently presented on HLA molecules. Using mass-spectrometry, we have identified a library of over a thousand HLA phospholigands. Many of these are tumor specific and found in multiple patients across multiple indications, enabling pre-manufacture of PhosphoSynVax™ vaccines for ready use. Upon testing the HSP plus synthetic peptide vaccine format in murine models, we have demonstrated effective tumor control in a therapeutic setting and also effective immune memory in a long-term prophylactic setting. Given Agenus’ diverse portfolio we have the opportunity to combine our immune education strategies with immunomodulatory antibodies to increase therapeutic efficacy. Citation Format: Mohamed Uduman, Mithun Khattar, Bishnu Joshi, Antoine Tanne, Benjamin Morin, Armen Karapetyan, Elise Drouin, Sandra Craig, Paisley Myers, Erin Jeffery, Nicholas Wilson, Amy Yang, Victor H. Engelhard, Mark Cobbold, Donald F. Hunt, Dennis Underwood, Shiwen Lin, Mark Findeis, Jeffrey Raizer, John Goldberg, Jennifer S. Buell, Robert Stein, Daniel L. Levey, John Castle. Agenus’ next generation cancer vaccine platforms [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4609. doi:10.1158/1538-7445.AM2017-4609
- Published
- 2017
- Full Text
- View/download PDF
22. Anti-TCR mAb induces peripheral tolerance to alloantigens and delays islet allograft rejection in autoimmune diabetic NOD mice
- Author
-
Aini Xie, Stanislaw M. Stepkowski, Paul M. Schroder, Wenhao Chen, Mithun Khattar, Xiaoshun He, and Ronghai Deng
- Subjects
Graft Rejection ,Isoantigens ,Time Factors ,medicine.medical_treatment ,Islets of Langerhans Transplantation ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Mice, SCID ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Autoimmunity ,Diabetes Mellitus, Experimental ,Mice ,Mice, Inbred NOD ,medicine ,Animals ,Transplantation, Homologous ,Cells, Cultured ,NOD mice ,Transplantation ,geography ,Mice, Inbred BALB C ,Mice, Inbred C3H ,geography.geographical_feature_category ,business.industry ,Graft Survival ,FOXP3 ,Peripheral tolerance ,Antibodies, Monoclonal ,Forkhead Transcription Factors ,Immunotherapy ,Skin Transplantation ,Islet ,Allografts ,Adoptive Transfer ,Mice, Inbred C57BL ,surgical procedures, operative ,Diabetes Mellitus, Type 1 ,Immunology ,Skin grafting ,Transplantation Tolerance ,business - Abstract
BACKGROUND Clinical application of islet transplantation to treat type 1 diabetes has been limited by islet allograft destruction by both allogeneic and autoimmune diabetogenic T-cell responses. The current study aims at determining whether an anti-T-cell receptor (TCR) monoclonal antibody (mAb) has potential as a novel and potent induction immunotherapy for islet transplantation. METHODS We have investigated the therapeutic efficacy and mechanisms of action of anti-TCR therapy in four different murine models, which comprise either allo- or autoimmune responses alone or both together. RESULTS T-cell response to islet allografts was potently abrogated by a brief treatment with an anti-TCRβ mAb (clone H57-597), resulting in long-term survival of BALB/c islet allografts in streptozotocin-induced diabetic B6 mice. Moreover, transient anti-TCR treatment permanently prevented BALB/c skin allograft rejection on Rag1 B6 recipients that were reconstituted with Foxp3 cell-depleted B6 splenocytes, but did not impair the reconstituted cells' ability to reject the later transplanted C3H skin allografts (transplanted at 120 days after BALB/c skin grafting). Transient anti-TCR treatment was also able to completely prevent diabetes onset in NOD.SCID.γc mice that were transferred with lymphocytes from diabetic NOD mice. Next, transient anti-TCR treatment significantly prolonged the survival of transplanted BALB/c islets in overtly diabetic NOD mice, which comprise both allogeneic and autoimmune diabetogenic T-cell responses to the transplanted islets. CONCLUSIONS Overall, anti-TCR mAb induced peripheral tolerance to specific alloantigens even in the absence of Foxp3-expressing natural regulatory T cells. These findings reveal the potential for using TCR-targeting mAbs as induction immunotherapy for islet transplantation.
- Published
- 2014
23. Interleukin-21 is a critical regulator of CD4 and CD8 T cell survival during priming under Interleukin-2 deprivation conditions
- Author
-
Paul M. Schroder, Yoshihiro Miyahara, Stanislaw M. Stepkowski, Wenhao Chen, Aini Xie, and Mithun Khattar
- Subjects
CD4-Positive T-Lymphocytes ,Priming (immunology) ,lcsh:Medicine ,CD8-Positive T-Lymphocytes ,Adaptive Immunity ,Interleukin 21 ,Mice ,0302 clinical medicine ,Molecular Cell Biology ,Cytotoxic T cell ,IL-2 receptor ,lcsh:Science ,Immune Response ,0303 health sciences ,Multidisciplinary ,Cell Death ,T Cells ,ZAP70 ,CD28 ,Antibodies, Monoclonal ,3. Good health ,Cell biology ,medicine.anatomical_structure ,Proto-Oncogene Proteins c-bcl-2 ,Cytokines ,Immunotherapy ,Research Article ,Cell Survival ,T cell ,Immune Cells ,Immunology ,Biology ,Cell Growth ,Immune Activation ,Immunomodulation ,03 medical and health sciences ,Cross-Priming ,medicine ,Animals ,030304 developmental biology ,Interleukin 3 ,Interleukins ,lcsh:R ,Immunity ,Immunoregulation ,Antibodies, Neutralizing ,Coculture Techniques ,Mice, Inbred C57BL ,Immune System ,Interleukin-2 ,lcsh:Q ,030215 immunology - Abstract
Optimal T cell activation and expansion require binding of the common gamma-chain (γc) cytokine Interleukin-2 (IL-2) to its cognate receptor that in turn engages a γc/Janus tyrosine kinase (Jak)3 signaling pathway. Because of its restricted expression by antigen-activated T cells and its obligatory role in promoting their survival and proliferation, IL-2 has been considered as a selective therapeutic target for preventing T cell mediated diseases. However, in order to further explore IL-2 targeted therapy, it is critical to precisely understand its role during early events of T cell activation. In this study, we delineate the role of IL-2 and other γc cytokines in promoting the survival of CD4 and CD8 T cells during early phases of priming. Under IL-2 inhibitory conditions (by neutralizing anti-IL-2 mAbs), the survival of activated CD8+ T cells was reduced, whereas CD4+ T cells remained much more resistant. These results correlated with reduced Bcl-2 expression, and mitochondrial membrane potential in CD8+ T cells in comparison to CD4+ T cells. However, using transwell co-culture assays we have found that CD4+ T cells could rescue the survival of CD8+ T cells even under IL-2 deprived conditions via secretion of soluble factors. A cytokine screen performed on CD8+ T cells cultured alone revealed that IL-21, another γc cytokine, was capable of rescuing their survival under IL-2 deprivation. Indeed, blocking the IL-21 signaling pathway along with IL-2 neutralization resulted in significantly reduced survival of both CD4+ and CD8+ T cells. Taken together, we have shown that under IL-2 deprivation conditions, IL-21 may act as the major survival factor promoting T cell immune responses. Thus, investigation of IL-2 targeted therapies may need to be revisited to consider blockade of the IL-21 signaling pathways as an adjunct to provide more effective control of T cell immune responses.
- Published
- 2014
24. Transient Combination Therapy Targeting the Immune Synapse Abrogates T Cell Responses and Prolongs Allograft Survival in Mice
- Author
-
Stanislaw M. Stepkowski, Wenhao Chen, Mithun Khattar, Aini Xie, Ronghai Deng, and Paul M. Schroder
- Subjects
Graft Rejection ,Adoptive cell transfer ,Mouse ,Lymphocyte ,Receptors, Antigen, T-Cell, alpha-beta ,T-Lymphocytes ,lcsh:Medicine ,Adaptive Immunity ,Lymphocyte Activation ,Immunological synapse ,Mice ,0302 clinical medicine ,Cytotoxic T cell ,Lymphocyte function-associated antigen 1 ,lcsh:Science ,0303 health sciences ,Mice, Inbred BALB C ,Multidisciplinary ,T Cells ,Graft Survival ,Antibodies, Monoclonal ,Animal Models ,Skin Transplantation ,Allografts ,Flow Cytometry ,Lymphocyte Function-Associated Antigen-1 ,3. Good health ,medicine.anatomical_structure ,Medicine ,Immunotherapy ,Research Article ,T cell ,Immune Cells ,Immunology ,Biology ,03 medical and health sciences ,Immune system ,Model Organisms ,Antigen ,medicine ,Animals ,030304 developmental biology ,Transplantation ,lcsh:R ,Immunity ,Immunologic Subspecialties ,Mice, Inbred C57BL ,lcsh:Q ,Clinical Immunology ,030215 immunology - Abstract
T cells play a major role in allograft rejection, which occurs after T cell activation by the engagement of several functional molecules to form an immune synapse with alloantigen presenting cells. In this study, the immune synapse was targeted using mAbs directed to the TCR beta-chain (TCRβ) and lymphocyte function-associated antigen-1 (LFA1) to induce long-term allograft survival. Evaluation of antigen-specific T cell responses was performed by adoptively transferring CFSE labeled transgenic OT-II cells into wild-type mice and providing OVA peptide by intravenous injection. Graft survival studies were performed in mice by transplanting BALB/c ear skins onto the flanks of C57BL/6 recipients. The anti-TCRβ plus anti-LFA1 mAb combination (but not either mAb alone) abrogated antigen-specific T cell responses invitro and invivo. Transient combination therapy with these agents resulted in significantly prolonged skin allograft survival in mice (51±10 days; p
- Published
- 2013
25. Novel sphingosine-1-phosphate receptor modulator KRP203 combined with locally delivered regulatory T cells induces permanent acceptance of pancreatic islet allografts
- Author
-
Lynne P. Rutzky, Stanislaw M. Stepkowski, Barry D. Kahan, Tammy Phan, Ronghai Deng, Mithun Khattar, and Paul M. Schroder
- Subjects
Graft Rejection ,Adoptive cell transfer ,Time Factors ,Green Fluorescent Proteins ,Islets of Langerhans Transplantation ,Mice, Transgenic ,Pharmacology ,T-Lymphocytes, Regulatory ,Article ,Mice ,Medicine ,Animals ,IL-2 receptor ,Sulfhydryl Compounds ,Receptor ,Sphingosine-1-Phosphate Receptors ,Cells, Cultured ,S1PR3 ,Transplantation ,geography ,Mice, Inbred BALB C ,Mice, Inbred C3H ,geography.geographical_feature_category ,Dose-Response Relationship, Drug ,business.industry ,Graft Survival ,Interleukin-2 Receptor alpha Subunit ,FOXP3 ,Forkhead Transcription Factors ,Islet ,Adoptive Transfer ,Combined Modality Therapy ,Mice, Inbred C57BL ,Dose–response relationship ,Receptors, Lysosphingolipid ,Immunology ,Transplantation Tolerance ,business ,Biomarkers ,Immunosuppressive Agents - Abstract
Background KRP203, a structural FTY720 analogue, has 5-fold greater selectivity for binding to sphingosine-1-phosphate receptor (S1PR) 1 (S1PR(1)) versus S1PR3 and 100-fold greater selectivity over S1PR(2) and S1PR(5). Although the immunoregulatory effects of FTY720 have been tested in clinical and experimental research, the therapeutic efficacy of KRP203 in allograft models remains elusive. In this study, we investigated the potential of KRP203 alone and in combination with intragraft injection of CD4(+)CD25(+)FoxP3(+) regulatory T cells (Tregs) to induce islet allograft tolerance. Methods BALB/c (H-2(d)) mice received transplants of fresh C57BL/10 (H-2(b)) islet allografts under the kidney capsule and were treated for 7 days with 0.3, 1.0, or 3.0 mg/kg KRP203 alone or in combination with intragraft-infused Tregs. Results Untreated BALB/c mice acutely rejected C57BL/10 islet allografts at a mean survival time of 13.8 ± 2.7 days (n=5). A 7-day dosing of 0.3 or 1.0 mg/kg KRP203 produced long-term islet allograft survival (9200 days) in one of five and two of seven recipients, respectively. A 3 mg/kg KRP203 dose resulted in islet graft survival for more than 200 days in 5 of 12 recipients. Whereas recipients that received 500 allogeneic islets admixed with 5 x 10(5) - 7 x 10(5) Tregssurvived 83.6 ± 67.2 days, addition of transient 3 mg/kg KRP203 therapy induced prolonged drug-free graft survival (9200 days) in all recipients. Conclusions A brief treatment with KRP203 significantly prolonged islet allograft survival, whereas additional intragraft delivery of Tregs induced tolerogenic effects selective to islet alloantigens.
- Published
- 2013
26. Abstract A037: The Agenus Immunogenic Mutation platform (AIM™) generates synthesis-ready blueprints for the AutoSynVax™ vaccine patient-specific neo-antigen vaccine
- Author
-
Benjamin Morin, R.B. Stein, Armen Karapetyan, Bishnu P. Joshi, Mohamed Uduman, Daniel Levey, Jeffrey Raizer, Amy Yang, Nicholas S. Wilson, Antoine Tanne, Shiwen Shiwen, Elise Drouin, John C. Castle, Sandra Craig, Jennifer Buell, Mithun Khattar, John M. Goldberg, Justin Zelin, and Mark Findeis
- Subjects
Cancer Research ,Synthetic vaccine ,business.industry ,medicine.medical_treatment ,Immunogenicity ,Immunology ,Cancer ,Context (language use) ,Human leukocyte antigen ,medicine.disease ,Virology ,QS21 ,Cancer immunotherapy ,medicine ,business ,Adjuvant - Abstract
Agenus is clinically testing Prophage™, an individualized tumor vaccine which uses heat-shock proteins endogenously complexed to neo-antigens harvested from the patient's own tumor. Agenus' genomics and bioinformatics unit rapidly identifies potentially immunogenic neo-antigens in the patient's tumor for use in immunomonitoring. However, in cases such as when the tumor mass is insufficient to generate the Prophage vaccine, we plan to treat patients with an individualized synthetic neo-antigen vaccine, AutoSynVax™ vaccine, combined with QS21 Stimulon® adjuvant and optionally immunomodulatory antibodies. In a murine model, we have demonstrated tumor control after treatment with our synthetic vaccine. The majority of mutations in human tumors are unique to the individual tumor, necessitating the rapid identification of mutations using next generation sequencing and bioinformatics. Further, the number of candidate immunogens presented on a tumor can range from a handful to hundreds. Accurately identifying tumor mutations; determining their impact, expression and translation; and predicting ability to bind patient-specific HLA class I and II molecules and elicit an immune response (immunogenicity) is complex, particularly in the context of clinical trials. The AutoSynVax™ vaccine platform leverages AIM™, the Agenus Immunogenic Mutation workflow, to generate a synthesis-ready blueprint for optimal immunogenic patient-specific neo-antigen vaccines. AIM™ comprises a robust and efficient approach to computational vaccinology designed to generate an immunogenic candidate blueprint, agnostic to vaccine format, followed by a format-specific blueprint ready for vaccine synthesis and manufacture. Citation Format: Mohamed Uduman, Armen Karapetyan, Mithun Khattar, Antoine Tanne, Benjamin Morin, Justin Zelin, Sandra Craig, Shiwen Shiwen, Bishnu Joshi, Mark A. Findeis, Nicholas Wilson, Elise Drouin, Amy Yang, Jeffrey Raizer, John Goldberg, Jennifer Buell, Robert Stein, John Castle, Daniel L. Levey. The Agenus Immunogenic Mutation platform (AIM™) generates synthesis-ready blueprints for the AutoSynVax™ vaccine patient-specific neo-antigen vaccine [abstract]. In: Proceedings of the Second CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; 2016 Sept 25-28; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(11 Suppl):Abstract nr A037.
- Published
- 2016
- Full Text
- View/download PDF
27. Anti-TCRβ mAb induces long-term allograft survival by reducing antigen-reactive T cells and sparing regulatory T cells
- Author
-
Paul M. Schroder, Wayne W. Hancock, Yoshihiro Miyahara, Beata Mierzejewska, Ronghai Deng, Rongxiang Han, Mithun Khattar, Stanislaw M. Stepkowski, and Wenhao Chen
- Subjects
T cell ,Receptors, Antigen, T-Cell, alpha-beta ,chemical and pharmacologic phenomena ,Mice, Inbred Strains ,Biology ,T-Lymphocytes, Regulatory ,Article ,Interleukin 21 ,Mice ,Antigen ,Transplantation Immunology ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Humans ,Pharmacology (medical) ,IL-2 receptor ,Antigen-presenting cell ,Transplantation ,Graft Survival ,FOXP3 ,Antibodies, Monoclonal ,hemic and immune systems ,Immunohistochemistry ,medicine.anatomical_structure ,Immunology ,Lymphocyte Culture Test, Mixed ,CD8 - Abstract
TCR specific antibodies may modulate the TCR engagement with antigen-MHC complexes, and in turn regulate in vivo T cell responses to alloantigens. Herein, we found that in vivo administration of mAbs specific for mouse TCRβ (H57-597), TCRα or CD3 promptly reduced the number of CD4(+) and CD8(+) T cells in normal mice, but H57-597 mAb most potently increased the frequency of CD4(+) Foxp3(+) Treg cells. When mice were injected with staphylococcal enterotoxin B (SEB) superantigen and H57-597 mAb, the expansion of SEB-reactive Vβ8(+) T cells was completely abrogated while SEB-nonreactive Vβ2(+) T cells remained unaffected. More importantly, transient H57-597 mAb treatment exerted long-lasting effect in preventing T cell responses to alloantigens, and produced long-term cardiac allograft survival (100 days) in 10 out of 11 recipients. While Treg cells were involved in maintaining donor-specific long-term graft survival, T cell homeostasis recovered over time and immunity was retained against third party allografts. Moreover, transient H57-597 mAb treatment significantly prolonged survival of skin allografts in naïve recipients as well as heart allografts in skin-sensitized recipients. Thus, transient modulation of the TCRβ chain by H57-597 mAb exhibits potent, long-lasting therapeutic effects to control alloimmune responses.
- Published
- 2012
28. IL-21 Is a Key Regulator Coordinating Multiple Immune Responses in Chronic Allograft Rejection
- Author
-
W. Baldwin, Mithun Khattar, Stanislaw M. Stepkowski, Wenhao Chen, Paul M. Schroder, and Caitlin E. Baum
- Subjects
Transplantation ,Immune system ,business.industry ,Allograft rejection ,Immunology ,Key (cryptography) ,Regulator ,Medicine ,business - Published
- 2014
- Full Text
- View/download PDF
29. Anti-TCR mAb Induces Peripheral Tolerance to Alloantigens and Delays Islet Allograft Rejection in Autoimmune Diabetic NOD Mice
- Author
-
Paul M. Schroder, Mithun Khattar, Ronghai Deng, Stanislaw M. Stepkowski, Wenhao Chen, and Aini Xie
- Subjects
Transplantation ,geography ,geography.geographical_feature_category ,business.industry ,medicine.drug_class ,T-cell receptor ,Peripheral tolerance ,Islet ,Monoclonal antibody ,Allograft rejection ,Immunology ,Medicine ,business ,NOD mice - Published
- 2014
- Full Text
- View/download PDF
30. 'Default' generation of neonatal regulatory T cells
- Author
-
Yoshihiro Miyahara, Guohua Wang, Stanislaw M. Stepkowski, Wenhao Chen, Mithun Khattar, and Zhiyong Guo
- Subjects
Male ,Adoptive cell transfer ,Immunology ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Mice, Transgenic ,Biology ,T-Lymphocytes, Regulatory ,Interleukin 21 ,Mice ,Immune system ,Antigen ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,IL-2 receptor ,Cells, Cultured ,Cellular Senescence ,Mice, Knockout ,Mice, Inbred BALB C ,Protein Stability ,T-cell receptor ,FOXP3 ,hemic and immune systems ,Cell Differentiation ,Forkhead Transcription Factors ,Adoptive Transfer ,Mice, Inbred C57BL ,Animals, Newborn ,CD4 Antigens ,Female - Abstract
CD4+Foxp3+ regulatory T (Treg) cells were shown to control all aspects of immune responses. How these Treg cells develop is not fully defined, especially in neonates during development of the immune system. We studied the induction of Treg cells from neonatal T cells with various TCR stimulatory conditions, because TCR stimulation is required for Treg cell generation. Independent of the types of TCR stimulus and without the addition of exogenous TGF-β, up to 70% of neonatal CD4+Foxp3− T cells became CD4+Foxp3+ Treg cells, whereas generally 3-fold increase in Treg cells in neonates but not in adults. Furthermore, neonatal or adult Foxp3− T cells were adoptively transferred into Rag1−/− mice. Twelve days later, the frequency of CD4+Foxp3+ T cells converted from neonatal cells was 6-fold higher than that converted from adult cells. Taken together, neonatal CD4+ T cells have an intrinsic “default” mechanism to become Treg cells in response to TCR stimulations. This finding provides intriguing implications about neonatal immunity, Treg cell generation, and tolerance establishment early in life.
- Published
- 2010
31. IL-7, but not thymic stromal lymphopoietin (TSLP), during priming enhances the generation of memory CD4+ T cells
- Author
-
Xiaoshun He, Yiqing Pan, Mithun Khattar, Zhiyong Guo, Changxi Wang, Sean P. Linkes, Jiahong Xia, Stanislaw M. Stepkowski, Wenhao Chen, Yoshihiro Miyahara, and Guohua Wang
- Subjects
CD4-Positive T-Lymphocytes ,Thymic stromal lymphopoietin ,Interleukin-7 ,Immunology ,CD28 ,Priming (immunology) ,Biology ,Lymphocyte Activation ,Molecular biology ,Mice, Inbred C57BL ,Interleukin 21 ,Mice ,Mice, Congenic ,Thymic Stromal Lymphopoietin ,Immunology and Allergy ,Cytotoxic T cell ,Animals ,Cytokines ,IL-2 receptor ,Antigen-presenting cell ,Immunologic Memory ,Cells, Cultured ,Interleukin 3 - Abstract
Multiple activation signals (including antigen, co-stimulation, and cytokines) during T-cell priming affect the subsequent generation of memory T cells, whose survival is maintained by IL-7 and IL-15. Since the IL-7 receptor is highly expressed not only on the surface of memory T cells but also on naive T cells, we propose that early exposure to IL-7 during priming of naive T cells may promote their survival, and thus enhances the generation of memory cells. To test this hypothesis, TCR transgenic OT-II CD4(+) T cells were stimulated in vitro with OVA(323-339) peptide presented by syngeneic antigen-presenting cells (APCs). IL-7 or an IL-7 like cytokine, thymic stromal lymphopoietin (TSLP), was added at the initial 2-day cultivation stage. We found that a short exposure to IL-7 or TSLP during priming did not affect activation, proliferation, and glucose uptake by CD4(+) T cells compared to controls when examined on culture day 6. However, the same 6-day cultures showed that IL-7 (but not TSLP) significantly decreased the frequency of apoptotic CD4(+) T cells compared to controls. More importantly, an adoptive transfer of the 6-day primed OT-II T cells into CD45.1(+) congenic mice demonstrated that IL-7 (but not TSLP) elevated by 3- and 4-fold the number of transferred CD4(+) T cells in spleen (p
- Published
- 2009
32. Dissociation of Anergic Phenotype From the Expression of Anergy-Related Genes in Memory CD4 T Cells
- Author
-
Aini Xie, Mithun Khattar, X. Li, Stanislaw M. Stepkowski, Wenhao Chen, and Paul M. Schroder
- Subjects
Transplantation ,Chemistry ,Gene ,Phenotype ,Dissociation (chemistry) ,Cell biology - Published
- 2014
- Full Text
- View/download PDF
33. IL21 is a critical regulator of chronic/persistent but not acute/accelerated auto-immune and allo-immune responses
- Author
-
Mithun, Khattar, primary, Wenhao, Chen, primary, and Stanislaw, Stepkowski, primary
- Published
- 2013
- Full Text
- View/download PDF
34. IL21 is a critical regulator of chronic/persistent auto-immune and allo-immune responses (P5162)
- Author
-
Mithun Khattar, Paul Schroder, Caitlin Baum, Ronghai Deng, Wenhao Chen, and Stanislaw Stepkowski
- Subjects
Immunology ,Immunology and Allergy - Abstract
IL21 is critical for T1D development in NOD mice, but its role in sustaining T cell responses against islets during T1D progression remains unexplored. Our studies show that diabetic NOD mice have high levels of serum IL21. We thus hypothesized that IL21 produced by activated CD4 T cells in spontaneously diabetic NOD mice sustains T cell responses to islets. Indeed, IL21 is required for maintaining the function of auto-reactive immune cells during T1D progression as transient therapy with IL21 receptor fusion protein (IL21RFc) was effective in reversing the onset of T1D in NOD mice (n=11) and inducing long-term T1D remission for up to 20 weeks in at least 3 mice. Pancreatic infiltrates of IL21RFc-treated NOD mice contain mostly CD4 T cells that lost surface ICOS expression while containing fewer CD8 T cells, suggesting that IL21RFc inhibited CD4 T cell help to CD8 T cells. In a parallel study, we have shown that IL21 deficient mice were protected from chronic allograft vasculopathy in a MHC class-II mismatched cardiac allograft model. However, IL21 deficient mice acutely rejected fully mismatched cardiac allografts similar to wild type. Thus, we have demonstrated a novel and critical role for IL21 signaling in driving chronic immune responses against auto and allo-antigens. Based on our findings, IL21 has the potential to act as a serum biomarker for early T1D detection and a therapeutic target to abrogate persistent T cell responses in T1D and chronic allograft rejection.
- Published
- 2013
- Full Text
- View/download PDF
35. Transient TCR engagement with an anti-TCRβ monoclonal antibody induces tolerogenic effects to allografts as well as prevents the development and reverses the onset of type 1 diabetes (169.1)
- Author
-
Mithun Khattar, Yoshihiro Miyahara, Paul Schroder, Beata Mierzejewska, Wenhao Chen, and Stanislaw Stepkowski
- Subjects
Immunology ,Immunology and Allergy - Abstract
In the current study, we investigated the immune regulatory effects of TCR engagement simultaneously by an anti-TCRβ chain mAb (H57-597) without or with antigens. We found that administration of anti-mouse TCRβ mAb resulted in a preferential reduction of antigen-reactive T cells with simultaneous enrichment (~3-fold increase) of CD4+Foxp3+ Treg cells. Moreover, administration of H57-597 mAb did not produce high levels of TNF-α, IFN-γ, IL-2 and IL-6 cytokines in serum in contrast to anti-CD3 mAb (145-2C11). Strikingly, a single injection of H57-597 mAb into RIP-OVAhi mice completely inhibited the development of type 1 diabetes (T1D) that was induced by adoptively transferred OVA-specific T cells. In NOD mice, a short course treatment with H57-597, not only prevented development of T1D (90% disease free treated at 8-weeks), but also induced remission in 7 of 7 mice treated at onset. In a transplantation model, transient H57-597 mAb treatment alone produced long-term Balb/c cardiac allograft survivals (>100 days; n=9) in C57BL/6 mice. Adoptive transfer of splenocytes from the heart allograft-accepting recipients to Rag1-/- recipients (B6 background, heart grafted) showed significantly extended survival of donor-specific (Balb/c) but not third-party (C3H) heart allografts. Thus, transient modulation of TCRβ chain during an ongoing immune response exhibits potent tolerogenic effects applicable for the therapy of T cell-mediated diseases and transplant rejection.
- Published
- 2011
- Full Text
- View/download PDF
36. Characterizing the T-cell survival and differentiation during priming with downregulated γ-c cytokine/Jak3 signals (50.34)
- Author
-
Mithun Khattar, Guohua Wang, Yoshihiro Miyahara, Zhiyong Guo, Wenhao Chen, and Stanislaw Stepkowski
- Subjects
Immunology ,Immunology and Allergy - Abstract
T-cells play a central role in cell mediated immune responses to foreign antigens. Optimal T-cell activation and expansion requires antigen recognition via cognate T-cell receptors (TCRs) along with co-stimulatory and cytokine signals. The cytokine signals that lead to T-cell activation and proliferation, involve binding of common gamma-chain cytokines to their cognate receptors, that in turn activates Janus tyrosine kinase (Jak) 3, inducing cytokine gene transcription via the Jak/Stat (Signal Transducers and Activators of Transcription) pathway. However, it is currently unknown as to whether these paradigms of signaling during T-cell priming are universal to all T-cell subsets and lineages or unique to each. In the current study, we have examined the role of γ-c cytokine/Jak3 axis of signaling in survival and differentiation of different T-cell subsets during early phases of priming. We show highly reduced survival of CD8+ T-cells under cytokine/Jak3 inhibiting conditions during activation, whereas CD4+ T-cells were fairly more resistant. Loss of Jak3 signaling also inhibited the Treg differentiation while promoting pro-inflammatory Th17 cells thus, suggesting discrete and specific roles of this signaling pathway in different T-cell subsets and lineages. These findings contribute to our understanding of T-cell activation in different T-cell lineages as well as their differentiation into subsets, and are important for designing therapeutic approaches of the immune responses.
- Published
- 2010
- Full Text
- View/download PDF
37. Distinct regulatory roles for Janus Tyrosine Kinase 3 (Jak3) in generation of T-helper 17 (TH17) and inducible regulatory T cells (Tregs) (47.10)
- Author
-
Mithun Khattar, Wenhao Chen, and Stanislaw Stepkowski
- Subjects
Immunology ,Immunology and Allergy - Abstract
The development of inducible Foxp3+ regulatory T (iTreg) cells is reciprocally related to that of pro-inflammatory T helper (Th) 17 cells. Both processes depend on transforming growth factor (TGF)-β. Recent studies indicated that interleukin (IL)-2, a Janus kinase 3 (Jak3)-dependent cytokine, is required for the development of iTreg cells, while exhibiting an inhibitory effect on Th17 differentiation. This raises the question about distinct roles of Jak3 in iTreg vs. Th17 differentiation. To address this question, we cultured naïve CD4+ T cells with TGF-β alone for iTreg cell polarization or TGF-β with IL-6 for Th17 cell polarization without or with selective Jak3 inhibitor, NC1153. We found that a dose-dependent inhibition of Jak3 reduced the TGF-β-mediated iTreg differentiation. In contrast, similar NC1153 concentrations under Th17 polarizing condition increased the percentage of Th17 cells. When we examined the proportion of regulatory T (Treg) cells within the CD4+ T-cell population from Jak3-dificient mice, we found that these mice lack Treg cells. Thus, inhibition of Jak3 has differential effects on iTreg/Th17 balance with preferential generation of pro-inflammatory Th17 cells. This work was supported by grants from NIH (grant number HL 69723)
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.