70 results on '"Sourindra Maiti"'
Search Results
2. Data from A New Approach to Simultaneously Quantify Both TCR α- and β-Chain Diversity after Adoptive Immunotherapy
- Author
-
Laurence J.N. Cooper, Laszlo Radvanyi, Patrick Hwu, Luis M. Vence, Richard E. Champlin, Brian Rabinovich, Helen Huls, Chantale Bernatchez, Sourindra Maiti, and Minying Zhang
- Abstract
Purpose: T-cell receptor (TCR) variable Vα and Vβ gene diversity is a surrogate biomarker for the therapeutic potential of adoptive immunotherapy and cellular immunity. Therefore, creating a straightforward, rapid, sensitive, and reliable method to view the global changes of both TCRVα and Vβ transcripts in heterogeneous populations of T cells is appealing.Experimental Design: We designed a “direct TCR expression assay” (DTEA) using a panel of customized bar-coded probes that simultaneously detects and quantifies 45 Vα and 46 Vβ transcripts in a nonenzymatic digital multiplexed assay from a small number of cells (104 cells) or as little as 100 ng of total RNA.Results: We evaluated DTEA on total RNA samples of tumor-infiltrating lymphocytes and peripheral blood obtained from patients with melanoma after adoptive T-cell therapy. DTEA detected a similar spectrum of the dominant patterns of TCRVβ gene usage as sequencing cloned TCRVβ CDR3 regions. However, DTEA was rapid, achieved a level of sensitivity to identify rare T-cell populations, and simultaneously tracked the full array of Vα and Vβ transcripts.Conclusions: DTEA can rapidly and sensitively track changes in TCRVα and Vβ gene usages in T-cell pools following immune interventions, such as adoptive T-cell transfer, and may also be used to assess impact of vaccination or reconstitution of T-cell compartment after hematopoietic stem cell transplantation. Clin Cancer Res; 18(17); 4733–42. ©2012 AACR.
- Published
- 2023
3. Supplementary Figures 1 - 7 from A New Approach to Simultaneously Quantify Both TCR α- and β-Chain Diversity after Adoptive Immunotherapy
- Author
-
Laurence J.N. Cooper, Laszlo Radvanyi, Patrick Hwu, Luis M. Vence, Richard E. Champlin, Brian Rabinovich, Helen Huls, Chantale Bernatchez, Sourindra Maiti, and Minying Zhang
- Abstract
PDF file, 264K, Supplementary Fig. S1. TCR Va and Vb gene expression in total RNA from TILs patient 303 and patient 232. Supplementary Fig S2. Global measurement of TCR Va (A-C) and TCR Vb (D-F) gene expression in patient 231 including in vitro rapid expanded TIL (A and D) and PBMC (B-C and E-F) collected after TIL infusion. Supplementary Fig S3. Global measurement of TCR Va (A-D) and TCR Vb (E-H) gene expression in patient 228 including in vitro rapid expanded TIL (A and E) and PBMC (B-D) and (F-H) collected after TIL infusion. Supplementary Fig S4. Global measurement of TCR Va (A-D) and TCR Vb (E-H) gene expression in patient 106 including in vitro rapid expanded TIL (A and E) and PBMC (B-D) and (F-H) collected after TIL infusion. Supplementary Fig S5. Global measurement of TCR Va (A-D) and TCR Vb (E-H) gene expression in patient 172 including in vitro rapid expanded TIL (A and E) and PBMC (B-D) and (F-H) collected after TIL infusion. Supplementary Fig S6. Global measurement of TCR Va (A-D) and TCR Vb (E-H) gene expression in patient 152 including in vitro rapid expanded TIL (A and E) and PBMC (B-D) and (F-H) collected after TIL infusion. Supplementary Fig S7. Global measurement of TCR Va (A-D) and TCR Vb (E-H) gene expression in patient 188 including in vitro rapid expanded TIL (A and E) and PBMC (B-D) and (F-H) collected after TIL infusion.
- Published
- 2023
4. Supplementary Figure 1 from Reprogramming CD19-Specific T Cells with IL-21 Signaling Can Improve Adoptive Immunotherapy of B-Lineage Malignancies
- Author
-
Laurence J.N. Cooper, Richard E. Champlin, Dean A. Lee, Partow Kebriaei, Sourindra Maiti, Tiejuan Mi, Kirsten Switzer, Simon Olivares, Helen Huls, Margaret J. Dawson, Matthew J. Figliola, and Harjeet Singh
- Abstract
Supplementary Figure 1 from Reprogramming CD19-Specific T Cells with IL-21 Signaling Can Improve Adoptive Immunotherapy of B-Lineage Malignancies
- Published
- 2023
5. Monitoring malignant T‐cell clones by direct TCR expression assay in patients with leukemic cutaneous T‐cell lymphoma during extracorporeal photopheresis
- Author
-
Xiao Ni, Pedram Bijani, Madeleine Duvic, Alissa Redko, and Sourindra Maiti
- Subjects
Skin Neoplasms ,T-Lymphocytes ,T cell ,Immunology ,Receptors, Antigen, T-Cell ,chemical and pharmacologic phenomena ,Dermatology ,Peripheral blood mononuclear cell ,Extracorporeal Photopheresis ,medicine ,Humans ,Immunology and Allergy ,Radiology, Nuclear Medicine and imaging ,Mycosis fungoides ,business.industry ,T-cell receptor ,Cutaneous T-cell lymphoma ,hemic and immune systems ,General Medicine ,medicine.disease ,Clone Cells ,Lymphoma, T-Cell, Cutaneous ,Lymphoma ,Non-Hodgkin's lymphoma ,medicine.anatomical_structure ,Photopheresis ,Leukocytes, Mononuclear ,business - Abstract
BACKGROUND/PURPOSE Accurate assessment of malignant T-cell clones in patients with leukemic cutaneous T-cell lymphoma (L-CTCL) is crucial for diagnosis, treatment, and monitoring disease. Although multiple approaches to quantitate malignant T-cell clones have been reported, a cost-effective assay with broad coverage is not available. We report a NanoString-nCounter-Technology-based direct TCR expression assay (DTEA) that was previously developed to quantify both TCR-Vα and TCR-Vβ usages after adoptive immunotherapy. This study was performed to test the effectiveness of DTEA in assessing malignant T-cell clones in L-CTCL patients. METHODS Total RNAs extracted from peripheral blood mononuclear cells of patients before starting extracorporeal photopheresis (ECP) (n = 15) and during therapy at 3 months and 6 months (n = 12) were used for DTEA, with customized probes for 45 TCR-Vα and 46 TCR-Vβ family members. RESULTS At baseline, DTEA detected TCR-Vβ clones in all 15 patients (100%) compared to flow cytometry that detected TCR-Vβ clones in 9 of 13 patients (69.2%). In addition to predominant TCR-Vβ clones, DTEA also detected additional TCR-Vβ clones in 8 of 15 patients (53.3%). Furthermore, DTEA simultaneously identified clonal TCR-Vα usages, which allowed us to pair TCR-Vα and TCRVβ usages by malignant T-cells and identify diversified clonotypes. Changes in the relative frequencies of clonal TCR-Vβ and TCRVα usages over therapy were consistent with patients' clinical responses. CONCLUSIONS Our results indicate that DTEA can effectively assess malignant T-cell clones by detecting clonal TCR-Vα and TCR-Vβ usages. By providing a global view of TCR repertoires, DTEA may also help us understand the origin(s) of malignant T-cells and pathogenesis of CTCL.
- Published
- 2021
6. Aurora kinase inhibition sensitizes melanoma cells to T-cell-mediated cytotoxicity
- Author
-
Cara Haymaker, Jason Roszik, Jie Qing Chen, Jahan Khalili, Zhe Wang, Nikunj Satani, Rina M. Mbofung, Laurence J.N. Cooper, Marie-Andree Forget, Willem W. Overwijk, Chunyu Xu, Leila Williams, Weiyi Peng, Chengwen Liu, Deborah A. Silverman, Simone Punt, Sourindra Maiti, Florian L. Muller, Elien M Doorduijn, Chantale Bernatchez, Trang N. Tieu, Ana Lucia Dominguez, Soraya Zorro Manrique, Patrick Hwu, Shruti Malu, Emily Ashkin, Jodi A. McKenzie, Rodabe N. Amaria, and Timothy P. Heffernan
- Subjects
Cancer Research ,High-throughput screen ,medicine.medical_treatment ,Immunology ,Apoptosis ,Mice ,03 medical and health sciences ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Aurora kinase ,In vivo ,Tumor Cells, Cultured ,Tumor Microenvironment ,medicine ,Animals ,Aurora Kinase B ,Humans ,Immunology and Allergy ,Cytotoxicity ,Melanoma ,Aurora Kinase A ,Cell Proliferation ,030304 developmental biology ,0303 health sciences ,Tumor microenvironment ,Chemistry ,T-cell cytotoxicity ,Immunotherapy ,Prognosis ,medicine.disease ,Xenograft Model Antitumor Assays ,Survival Rate ,Oncology ,Drug Resistance, Neoplasm ,030220 oncology & carcinogenesis ,Cancer research ,Original Article ,Female ,T cell mediated cytotoxicity ,Immune checkpoint blockade ,T-Lymphocytes, Cytotoxic - Abstract
Although immunotherapy has achieved impressive durable clinical responses, many cancers respond only temporarily or not at all to immunotherapy. To find novel, targetable mechanisms of resistance to immunotherapy, patient-derived melanoma cell lines were transduced with 576 open reading frames, or exposed to arrayed libraries of 850 bioactive compounds, prior to co-culture with autologous tumor-infiltrating lymphocytes (TILs). The synergy between the targets and TILs to induce apoptosis, and the mechanisms of inhibiting resistance to TILs were interrogated. Gene expression analyses were performed on tumor samples from patients undergoing immunotherapy for metastatic melanoma. Finally, the effect of inhibiting the top targets on the efficacy of immunotherapy was investigated in multiple preclinical models. Aurora kinase was identified as a mediator of melanoma cell resistance to T-cell-mediated cytotoxicity in both complementary screens. Aurora kinase inhibitors were validated to synergize with T-cell-mediated cytotoxicity in vitro. The Aurora kinase inhibition-mediated sensitivity to T-cell cytotoxicity was shown to be partially driven by p21-mediated induction of cellular senescence. The expression levels of Aurora kinase and related proteins were inversely correlated with immune infiltration, response to immunotherapy and survival in melanoma patients. Aurora kinase inhibition showed variable responses in combination with immunotherapy in vivo, suggesting its activity is modified by other factors in the tumor microenvironment. These data suggest that Aurora kinase inhibition enhances T-cell cytotoxicity in vitro and can potentiate antitumor immunity in vivo in some but not all settings. Further studies are required to determine the mechanism of primary resistance to this therapeutic intervention. Electronic supplementary material The online version of this article (10.1007/s00262-020-02748-9) contains supplementary material, which is available to authorized users.
- Published
- 2020
7. Genetic Engineering of T Cells to Target HERV-K, an Ancient Retrovirus on Melanoma
- Author
-
Sourindra Maiti, Harjeet Singh, Laurence J.N. Cooper, Feng Wang-Johanning, Pappanaicken R. Kumaresan, Simon Olivares, Kirsten Switzer, Brian Rabinovich, Janani Krishnamurthy, Joshua B. Plummer, Helen Huls, and Tiejuan Mi
- Subjects
Cancer Research ,T-Lymphocytes ,viruses ,medicine.medical_treatment ,T cell ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,Biology ,Immunotherapy, Adoptive ,Mice ,Viral Proteins ,Interleukin 21 ,Antigen ,Mice, Inbred NOD ,medicine ,Animals ,Humans ,Cytotoxic T cell ,IL-2 receptor ,Antigen-presenting cell ,Melanoma ,Genetic Therapy ,Immunotherapy ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Virology ,Chimeric antigen receptor ,medicine.anatomical_structure ,Oncology ,embryonic structures ,Cancer research ,Genetic Engineering - Abstract
Purpose: The human endogenous retrovirus (HERV-K) envelope (env) protein is a tumor-associated antigen (TAA) expressed on melanoma but not normal cells. This study was designed to engineer a chimeric antigen receptor (CAR) on T-cell surface, such that they target tumors in advanced stages of melanoma. Experimental Design: Expression of HERV-K protein was analyzed in 220 melanoma samples (with various stages of disease) and 139 normal organ donor tissues using immunohistochemical (IHC) analysis. HERV-K env–specific CAR derived from mouse monoclonal antibody was introduced into T cells using the transposon-based Sleeping Beauty (SB) system. HERV-K env–specific CAR+ T cells were expanded ex vivo on activating and propagating cells (AaPC) and characterized for CAR expression and specificity. This includes evaluating the HERV-K–specific CAR+ T cells for their ability to kill A375-SM metastasized tumors in a mouse xenograft model. Results: We detected HERV-K env protein on melanoma but not in normal tissues. After electroporation of T cells and selection on HERV-K+ AaPC, more than 95% of genetically modified T cells expressed the CAR with an effector memory phenotype and lysed HERV-K env+ tumor targets in an antigen-specific manner. Even though there is apparent shedding of this TAA from tumor cells that can be recognized by HERV-K env–specific CAR+ T cells, we observed a significant antitumor effect. Conclusions: Adoptive cellular immunotherapy with HERV-K env–specific CAR+ T cells represents a clinically appealing treatment strategy for advanced-stage melanoma and provides an approach for targeting this TAA on other solid tumors. Clin Cancer Res; 21(14); 3241–51. ©2015 AACR.
- Published
- 2015
8. Activating and Propagating Polyclonal Gamma Delta T Cells with Broad Specificity for Malignancies
- Author
-
Robert C. Bast, Tiejuan Mi, Laurence J.N. Cooper, Simon Olivares, Richard E. Champlin, Drew C. Deniger, Dean A. Lee, Sonny Ang, Kirsten Switzer, M. Helen Huls, Brian Rabinovich, Lenka V. Hurton, Vijaya Ramachandran, and Sourindra Maiti
- Subjects
Antigens, Differentiation, T-Lymphocyte ,Cancer Research ,Adoptive cell transfer ,T cell ,Cell Culture Techniques ,Antigen-Presenting Cells ,Gene Expression ,Mice, Transgenic ,T-Cell Antigen Receptor Specificity ,Lymphocyte Activation ,Article ,Interferon-gamma ,Mice ,Antigen ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Neoplasms ,medicine ,Animals ,Humans ,RNA, Messenger ,Antigen-presenting cell ,biology ,T-cell receptor ,Receptors, Antigen, T-Cell, gamma-delta ,NKG2D ,Adoptive Transfer ,Xenograft Model Antitumor Assays ,Molecular biology ,Disease Models, Animal ,medicine.anatomical_structure ,Oncology ,NK Cell Lectin-Like Receptor Subfamily K ,Polyclonal antibodies ,biology.protein ,Cytokines ,Cytokine secretion - Abstract
Purpose: To activate and propagate populations of γδ T cells expressing polyclonal repertoire of γ and δ T-cell receptor (TCR) chains for adoptive immunotherapy of cancer, which has yet to be achieved. Experimental Design: Clinical-grade artificial antigen-presenting cells (aAPC) derived from K562 tumor cells were used as irradiated feeders to activate and expand human γδ T cells to clinical scale. These cells were tested for proliferation, TCR expression, memory phenotype, cytokine secretion, and tumor killing. Results: γδ T-cell proliferation was dependent upon CD137L expression on aAPC and addition of exogenous IL2 and IL21. Propagated γδ T cells were polyclonal as they expressed TRDV1, TRDV2-2, TRDV3, TRDV5, TRDV7, and TRDV8 with TRGV2, TRGV3F, TRGV7, TRGV8, TRGV9*A1, TRGV10*A1, and TRGV11 TCR chains. IFNγ production by Vδ1, Vδ2, and Vδ1negVδ2neg subsets was inhibited by pan-TCRγδ antibody when added to cocultures of polyclonal γδ T cells and tumor cell lines. Polyclonal γδ T cells killed acute and chronic leukemia, colon, pancreatic, and ovarian cancer cell lines, but not healthy autologous or allogeneic normal B cells. Blocking antibodies demonstrated that polyclonal γδ T cells mediated tumor cell lysis through combination of DNAM1, NKG2D, and TCRγδ. The adoptive transfer of activated and propagated γδ T cells expressing polyclonal versus defined Vδ TCR chains imparted a hierarchy (polyclonal>Vδ1>Vδ1negVδ2neg>Vδ2) of survival of mice with ovarian cancer xenografts. Conclusions: Polyclonal γδ T cells can be activated and propagated with clinical-grade aAPCs and demonstrate broad antitumor activities, which will facilitate the implementation of γδ T-cell cancer immunotherapies in humans. Clin Cancer Res; 20(22); 5708–19. ©2014 AACR.
- Published
- 2014
9. Activation and Propagation of Tumor-infiltrating Lymphocytes on Clinical-grade Designer Artificial Antigen-presenting Cells for Adoptive Immunotherapy of Melanoma
- Author
-
Helen Huls, Francesco M. Marincola, Cara Haymaker, Laszlo Radvanyi, Shruti Malu, Sourindra Maiti, Charuta Kale, Patrick Hwu, Christopher Toth, Laurence J.N. Cooper, Marie Andrée Forget, Ena Wang, Chantale Bernatchez, and Hui Liu
- Subjects
Cancer Research ,Skin Neoplasms ,medicine.medical_treatment ,Immunology ,Antigen-Presenting Cells ,chemical and pharmacologic phenomena ,CD8-Positive T-Lymphocytes ,Biology ,Immunotherapy, Adoptive ,Article ,Cell therapy ,Lymphocytes, Tumor-Infiltrating ,Artificial antigen presenting cells ,medicine ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Melanoma ,Pharmacology ,CD86 ,Tumor-infiltrating lymphocytes ,hemic and immune systems ,Immunotherapy ,medicine.disease ,K562 Cells ,Transcriptome ,CD8 - Abstract
Purpose: Adoptive cell therapy with autologous tumor-infiltrating lymphocytes (TIL) is a therapy for metastatic melanoma with response rates of up to 50%. However, the generation of the TIL transfer product is challenging, requiring pooled allogeneic normal donor peripheral blood mononuclear cells (PBMC) used in vitro as “feeders” to support a rapid-expansion protocol. Here, we optimized a platform to propagate TIL to a clinical scale using K562 cells genetically modified to express costimulatory molecules such as CD86, CD137-ligand, and membrane-bound IL-15 to function as artificial antigen-presenting cells (aAPC) as an alternative to using PBMC feeders. Experimental Design: We used aAPC or g-irradiated PBMC feeders to propagate TIL and measured rates of expansion. The activation and differentiation state was evaluated by flow cytometry and differential gene expression analyses. Clonal diversity was assessed on the basis of the pattern of T-cell receptor usage. T-cell effector function was measured by evaluation of cytotoxic granule content and killing of target cells. Results: The aAPC propagated TIL at numbers equivalent to that found with PBMC feeders, whereas increasing the frequency of CD8 + T-cell expansion with a comparable effector-memory phenotype. mRNA profiling revealed an upregulation of genes in the Wnt and stem-cell pathways with the aAPC. The aAPC platform did not skew clonal diversity, and CD8 + T cells showed comparable antitumor function as those expanded with PBMC feeders. Conclusions: TIL can be rapidly expanded with aAPC to clinical scale generating T cells with similar phenotypic and effector profiles as with PBMC feeders. These data support the clinical application of aAPC to manufacture TIL for the treatment of melanoma.
- Published
- 2014
10. Bioengineering T cells to target carbohydrate to treat opportunistic fungal infection
- Author
-
Dean A. Lee, Janani Krishnamurthy, Pappanaicken R. Kumaresan, Amer Najjar, Nathaniel D. Albert, Harjeet Singh, M. Helen Huls, Pallavi R. Manuri, Ling Zhang, Tiejuan Mi, Simon Olivares, Dimitrios P. Kontoyiannis, Richard E. Champlin, Jason Roszik, Brian Rabinovich, Sourindra Maiti, and Laurence J.N. Cooper
- Subjects
T-Lymphocytes ,Antigens, CD19 ,Carbohydrates ,Hyphae ,Receptors, Antigen, T-Cell ,Bioengineering ,Opportunistic Infections ,Biology ,Lymphocyte Activation ,Dexamethasone ,Immunophenotyping ,Microbiology ,Mice ,Antigen ,Animals ,Aspergillosis ,Humans ,Cytotoxic T cell ,Lectins, C-Type ,IL-2 receptor ,Antigen-presenting cell ,Multidisciplinary ,CD28 ,Biological Sciences ,Natural killer T cell ,Chimeric antigen receptor ,Aspergillus ,Interleukin 12 - Abstract
Clinical-grade T cells are genetically modified ex vivo to express chimeric antigen receptors (CARs) to redirect their specificity to target tumor-associated antigens in vivo. We now have developed this molecular strategy to render cytotoxic T cells specific for fungi. We adapted the pattern-recognition receptor Dectin-1 to activate T cells via chimeric CD28 and CD3-ζ (designated "D-CAR") upon binding with carbohydrate in the cell wall of Aspergillus germlings. T cells genetically modified with the Sleeping Beauty system to express D-CAR stably were propagated selectively on artificial activating and propagating cells using an approach similar to that approved by the Food and Drug Administration for manufacturing CD19-specific CAR(+) T cells for clinical trials. The D-CAR(+) T cells exhibited specificity for β-glucan which led to damage and inhibition of hyphal growth of Aspergillus in vitro and in vivo. Treatment of D-CAR(+) T cells with steroids did not compromise antifungal activity significantly. These data support the targeting of carbohydrate antigens by CAR(+) T cells and provide a clinically appealing strategy to enhance immunity for opportunistic fungal infections using T-cell gene therapy.
- Published
- 2014
11. Universal Artificial Antigen Presenting Cells to Selectively Propagate T Cells Expressing Chimeric Antigen Receptor Independent of Specificity
- Author
-
Simon Olivares, David Rushworth, Dean A. Lee, Sourindra Maiti, Jianliang Dai, Neima Briggs, Bipulendu Jena, Srinivas S. Somanchi, and Laurence J.N. Cooper
- Subjects
Cancer Research ,Recombinant Fusion Proteins ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Antigens, CD19 ,Immunology ,Receptors, Antigen, T-Cell ,Antigen-Presenting Cells ,T-Cell Antigen Receptor Specificity ,Biology ,Major histocompatibility complex ,Immunotherapy, Adoptive ,Article ,Mice ,Artificial antigen presenting cells ,Antigen ,Cell Line, Tumor ,parasitic diseases ,medicine ,Animals ,Humans ,Immunology and Allergy ,Pharmacology ,Immunotherapy ,Molecular biology ,Chimeric antigen receptor ,Cell biology ,Receptors, Antigen ,medicine.anatomical_structure ,Cell culture ,biology.protein ,K562 Cells ,human activities ,K562 cells - Abstract
T cells genetically modified to stably express immunoreceptors are being assessed for therapeutic potential in clinical trials. T cells expressing a chimeric antigen receptor (CAR) are endowed with a new specificity to target tumor-associated antigen (TAA) independent of major histocompatibility complex. Our approach to non-viral gene transfer in T cells uses ex vivo numeric expansion of CAR+ T cells on irradiated artificial antigen presenting cells (aAPC) bearing the targeted TAA. The requirement for aAPC to express a desired TAA limits the human application of CARs with multiple specificities when selective expansion through co-culture with feeder cells is sought. As an alternative to expressing individual TAAs on aAPC, we expressed one ligand that could activate CAR+ T cells for sustained proliferation independent of specificity. We expressed a CAR ligand (designated CARL) that binds the conserved IgG4 extracellular domain of CAR and demonstrated CARL+ aAPC propagate CAR+ T cells of multiple specificities. CARL avoids technical issues and costs associated with deploying clinical-grade aAPC for each TAA targeted by a given CAR. Employing CARL enables one aAPC to numerically expand all CAR+ T cells containing the IgG4 domain, and simplifies expansion, testing, and clinical translation of CAR+ T cells of any specificity.
- Published
- 2014
12. Rapid Personalized Manufacture (RPM) of Sleeping Beauty System-Generated NY-ESO-1-Specific TCR-T Cells Co-Expressing Membrane-Bound IL-15 Yields Anti-Tumor Responses
- Author
-
Drew C. Deniger, Harjeet Singh, Misun Kim, Ling Zhang, Sourindra Maiti, Lenka V. Hurton, Matthew J. Figliola, Karishma Bavisi, Cuiping Dai, Shihuang Su, Simon Olivares, Kirsten Switzer, Laurence J.N. Cooper, Vazquez M Lydia, and Tiejuan Mi
- Subjects
Antitumor activity ,Electroporation ,Immunology ,T-cell receptor ,Cell Biology ,Hematology ,Human leukocyte antigen ,Biology ,Biochemistry ,Antigen ,Interleukin 15 ,T cell differentiation ,Cancer research ,NY-ESO-1 - Abstract
CAR-redirected T cells have demonstrated clinical effectiveness in early phase clinical trials, with persistence of adoptively transferred CD19-specific T cells correlated with positive outcomes. Notwithstanding the successes for some hematological malignancies, CAR-T targets a limited number of cell-surface antigens that curtails their appeal for solid tumors. This can be overcome by TCR gene transfer with specificity for intracellular tumor-associated antigens such as NY-ESO-1 expressed by hematologic malignancies and solid tumors. The predominant technologies for both CAR- and TCR-redirection of T cells utilize viral genetic modification as well as require a lengthy period of in vitro propagation with resultant deleterious differentiation to achieve clinically relevant cell numbers. A major impediment with current TCR-T is the high-cost and lengthy time associated with a viral-based manufacture of a library of TCRs that can address a multitude of desired targets and match HLA restriction to meet the need to infuse personalized TCR-T products with multiple specificities in each recipient. The Sleeping Beauty (SB) platform is the most clinically advanced non-viral gene transfer technology and overcomes the issues of scalability with viral based manufacture of TCR-T. We initially showed in pre-clinical models that co-expression of membrane-bound interleukin-15 (mbIL15) enhanced in vivo persistence of CAR-T (PMID: 27849617). This technology has been recently advanced to produce CD19-specific T cells in ≤ 2 days after electro-transfer of DNA plasmids using so-called "rapid personalized manufacture" (RPM). This was based on the SB system to stably co-express CAR and mbIL15 with a kill switch (HER1t). We have now adapted these technologies to address current limitations for T-cell therapy by using the RPM process to very rapidly generate TCR-modified T cells. The rationale for RPM of TCR-T is based on: (i) SB to genetically modify resting T cells thus eliminating the need to propagate cells prior to, or after, genetic modification, (ii) introduction of TCR to redirect T-cell specificity to tumor-associated antigens, (iii) mbIL15-HER1t to support T-cell persistence and enable selective elimination to increase safety, and (iv) manufacture within two days of gene transfer which limits T-cell differentiation and decreases time to manufacture. Mononuclear cells were electroporated with SB-derived DNA plasmids expressing (a) HLA A2-restricted NY-ESO-1-specific TCR or (b) the TCR and mbIL15-HER1t in separate plasmids. Following electroporation, cells were directly (unpropagated) injected into NSG (immunocompromised) mice bearing established HLA A2+ NY-ESO-1+ tumor. Administration of RPM TCR-mbIL15 T cells exhibited superior anti-tumor activity compared with RPM TCR T cells (Figure). Though engraftment of TCR+ T cells was not significantly different between the two groups, the RPM TCR-mbIL15 T cell-treated mice exhibited increased frequency of CD27+TCR+ T cells (p = 0.035, n = 6-7, Mann Whitney test), a phenotype that is correlated with improved therapeutic responses in human subjects. The RPM technology can thus be adapted to co-express TCR with mbIL15 (and HER1t), which can now be scaled to provide a cost-effective approach to manufacturing a multitude of TCR-T products from a library of TCRs with the necessary complexity to manage the range of specificities and HLA restrictions to treat multiple patients. Disclosures Hurton: • M.D. Anderson Cancer Center: Patents & Royalties; Intrexon: Patents & Royalties: US 9,629,877 B2 ; Ziopharm Oncology: Employment, Equity Ownership, Patents & Royalties: US 9,629,877 B2 . Zhang:Intrexon: Patents & Royalties: US 9,629,877 B2; Ziopharm Oncology: Patents & Royalties: US 9,629,877 B2. Deniger:Ziopharm Oncology: Employment, Equity Ownership. Olivares:Ziopharm Oncology: Patents & Royalties: US9629877B2, US20160158285A1, WO2009091826A2, US20190085079A1, US20170158749A1, US20170333480A1, US20190055299A1; Intrexon: Patents & Royalties: US9629877B2, US20160158285A1, WO2009091826A2, US20190085079A1, US20170158749A1, US20170333480A1, US20190055299A1. Cooper:CytoSen: Equity Ownership; Targazyme: Equity Ownership; MD Anderson Cancer Center: Patents & Royalties; Sangamo BioSciences: Patents & Royalties; Immatics: Equity Ownership, Patents & Royalties; City of Hope: Patents & Royalties; Ziopharm Oncology: Employment, Equity Ownership, Other: Contracted research, Patents & Royalties; Secure Transfusion Services: Equity Ownership; CellChorus: Equity Ownership. Singh:Ziopharm Oncology: Patents & Royalties: US9629877B2, US20160096902A1, US20170333480A1, US10125193B2; Intrexon: Patents & Royalties: US9629877B2, US20160096902A1, US20170333480A1, US10125193B2.
- Published
- 2019
13. Differential T-cell subset representation in cutaneous squamous cell carcinoma arising in immunosuppressed versus immunocompetent individuals
- Author
-
Sourindra Maiti, Richard E. Davis, Emily Y. Chu, Vida Chitsazzadeh, Varun Bansal, Laurence Feldmeyer, Grace Ching, Wencai Ma, Richard R. Jahan-Tigh, Peter Fuller, Harina Vin, Kenneth Y. Tsai, and Laurence J.N. Cooper
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Skin Neoplasms ,Cutaneous squamous cell carcinoma ,CD3 Complex ,T cell ,medicine.medical_treatment ,Dermatology ,Biochemistry ,Immunocompromised Host ,03 medical and health sciences ,0302 clinical medicine ,T-Lymphocyte Subsets ,medicine ,Cluster Analysis ,Humans ,CD40 Antigens ,610 Medicine & health ,Molecular Biology ,Immunosuppression Therapy ,Inflammation ,Forkhead Box Protein O1 ,business.industry ,Gene Expression Profiling ,Immunosuppression ,Immunohistochemistry ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,T cell subset ,Carcinoma, Squamous Cell ,business - Published
- 2015
14. Bispecific T-cells Expressing Polyclonal Repertoire of Endogenous γδ T-cell Receptors and Introduced CD19-specific Chimeric Antigen Receptor
- Author
-
Simon Olivares, Drew C. Deniger, Sourindra Maiti, Kirsten Switzer, Richard E. Champlin, Harjeet Singh, Tiejuan Mi, Laurence J.N. Cooper, Lenka V. Hurton, Dean A. Lee, and Helen Huls
- Subjects
Adoptive cell transfer ,T-Lymphocytes ,T cell ,Antigens, CD19 ,Antigen-Presenting Cells ,Transposases ,Biology ,Lymphocyte Activation ,Immunotherapy, Adoptive ,CD19 ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigen ,Cell Line, Tumor ,Drug Discovery ,Genetics ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Antigen-presenting cell ,Molecular Biology ,030304 developmental biology ,Mice, Knockout ,Pharmacology ,0303 health sciences ,Leukemia ,T-cell receptor ,Receptors, Antigen, T-Cell, gamma-delta ,Molecular biology ,Chimeric antigen receptor ,Electroporation ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Leukocytes, Mononuclear ,biology.protein ,Molecular Medicine ,Original Article - Abstract
Even though other γδ T-cell subsets exhibit antitumor activity, adoptive transfer of γδ Tcells is currently limited to one subset (expressing Vγ9Vδ2 T-cell receptor (TCR)) due to dependence on aminobisphosphonates as the only clinically appealing reagent for propagating γδ T cells. Therefore, we developed an approach to propagate polyclonal γδ T cells and rendered them bispecific through expression of a CD19-specific chimeric antigen receptor (CAR). Peripheral blood mononuclear cells (PBMC) were electroporated with Sleeping Beauty (SB) transposon and transposase to enforce expression of CAR in multiple γδ T-cell subsets. CAR(+)γδ T cells were expanded on CD19(+) artificial antigen-presenting cells (aAPC), which resulted in10(9) CAR(+)γδ T cells from10(6) total cells. Digital multiplex assay detected TCR mRNA coding for Vδ1, Vδ2, and Vδ3 with Vγ2, Vγ7, Vγ8, Vγ9, and Vγ10 alleles. Polyclonal CAR(+)γδ T cells were functional when TCRγδ and CAR were stimulated and displayed enhanced killing of CD19(+) tumor cell lines compared with CAR(neg)γδ T cells. CD19(+) leukemia xenografts in mice were reduced with CAR(+)γδ T cells compared with control mice. Since CAR, SB, and aAPC have been adapted for human application, clinical trials can now focus on the therapeutic potential of polyclonal γδ T cells.
- Published
- 2013
15. Sleeping Beauty System to Redirect T-cell Specificity for Human Applications
- Author
-
Brian Rabinovich, Helen Huls, Sonny Ang, Denise L. Crossland, Laurence J.N. Cooper, Pullavathi Rao, Hiroki Torikai, Dean A. Lee, Margaret J. Dawson, Richard E. Champlin, Harjeet Singh, Matthew J. Figliola, Ling Zhang, Perry B. Hackett, Partow Kebriaei, Sourindra Maiti, Simon Olivares, Ge Yang, Yi Jue Zhao, and Asha S. Multani
- Subjects
Cancer Research ,CD3 Complex ,Recombinant Fusion Proteins ,T-Lymphocytes ,CD3 ,Antigens, CD19 ,Immunology ,Receptors, Antigen, T-Cell ,Transposases ,Biology ,Immunotherapy, Adoptive ,Article ,CD19 ,Mice ,Artificial antigen presenting cells ,Antigen ,Cell Line, Tumor ,Animals ,Humans ,Immunology and Allergy ,Cytotoxic T cell ,Cells, Cultured ,Transposase ,Pharmacology ,T-cell receptor ,Gene Transfer Techniques ,Molecular biology ,Chimeric antigen receptor ,Electroporation ,biology.protein ,Glioblastoma - Abstract
The Sleeping Beauty (SB) transposon/transposase DNA plasmid system is used to genetically modify cells for long-term transgene expression. We adapted the SB system for human application and generated T cells expressing a chimeric antigen receptor (CAR) specific for CD19. Electrotransfer of CD19-specific SB DNA plasmids in peripheral blood mononuclear cells and propagation on CD19 artificial antigen presenting cells was used to numerically expand CD3 T cells expressing CAR. By day 28 of coculture, >90% of expanded CD3 T cells expressed CAR. CAR T cells specifically killed CD19 target cells and consisted of subsets expressing biomarkers consistent with central memory, effector memory, and effector phenotypes. CAR T cells contracted numerically in the absence of the CD19 antigen, did not express SB11 transposase, and maintained a polyclonal TCR Vα and TCR Vβ repertoire. Quantitative fluorescence in situ hybridization revealed that CAR T cells preserved the telomere length. Quantitative polymerase chain reaction and fluorescence in situ hybridization showed CAR transposon integrated on average once per T-cell genome. CAR T cells in peripheral blood can be detected by quantitative polymerase chain reaction at a sensitivity of 0.01%. These findings lay the groundwork as the basis of our first-in-human clinical trials of the nonviral SB system for the investigational treatment of CD19 B-cell malignancies (currently under 3 INDs: 14193, 14577, and 14739).
- Published
- 2013
16. Tethered IL-15 augments antitumor activity and promotes a stem-cell memory subset in tumor-specific T cells
- Author
-
Vrushali Datar, Brian Rabinovich, Lenka V. Hurton, Harjeet Singh, Richard E. Champlin, Amer Najjar, Marie Andrée Forget, Kirsten Switzer, Helen Huls, Dean A. Lee, Laurence J.N. Cooper, Tiejuan Mi, Simon Olivares, Partow Kebriaei, and Sourindra Maiti
- Subjects
0301 basic medicine ,Recombinant Fusion Proteins ,Antigens, CD19 ,Receptors, Antigen, T-Cell ,Biology ,Lymphocyte Activation ,Immunotherapy, Adoptive ,CD19 ,Mice ,03 medical and health sciences ,Interleukin 21 ,T-Lymphocyte Subsets ,medicine ,Animals ,Humans ,Cytotoxic T cell ,IL-2 receptor ,Interleukin-15 ,Precursor Cells, T-Lymphoid ,Multidisciplinary ,Neoplasms, Experimental ,medicine.disease ,Chimeric antigen receptor ,Leukemia ,030104 developmental biology ,Interleukin 15 ,Immunology ,Cancer research ,biology.protein ,Stem cell ,Signal Transduction - Abstract
Adoptive immunotherapy retargeting T cells to CD19 via a chimeric antigen receptor (CAR) is an investigational treatment capable of inducing complete tumor regression of B-cell malignancies when there is sustained survival of infused cells. T-memory stem cells (TSCM) retain superior potential for long-lived persistence, but challenges exist in manufacturing this T-cell subset because they are rare among circulating lymphocytes. We report a clinically relevant approach to generating CAR+ T cells with preserved TSCM potential using the Sleeping Beauty platform. Because IL-15 is fundamental to T-cell memory, we incorporated its costimulatory properties by coexpressing CAR with a membrane-bound chimeric IL-15 (mbIL15). The mbIL15-CAR T cells signaled through signal transducer and activator of transcription 5 to yield improved T-cell persistence independent of CAR signaling, without apparent autonomous growth or transformation, and achieved potent rejection of CD19+ leukemia. Long-lived T cells were CD45ROnegCCR7+CD95+, phenotypically most similar to TSCM, and possessed a memory-like transcriptional profile. Overall, these results demonstrate that CAR+ T cells can develop long-term persistence with a memory stem-cell phenotype sustained by signaling through mbIL15. This observation warrants evaluation in clinical trials.
- Published
- 2016
17. Targeting B-cell malignancies through human B-cell receptor specific CD4
- Author
-
Michael Popescu, Sourindra Maiti, Soung Chul Cha, Sattva S. Neelapu, Dongho Gwak, Larry W. Kwak, Hua Wang, Jinsheng Weng, Kelsey E. Moriarty, Laurence J.N. Cooper, Flavio Egidio Baio, Hiroki Torikai, and Zhiqiang Liu
- Subjects
0301 basic medicine ,Idiotype ,biology ,T cell ,Immunology ,B-cell receptor ,breakpoint cluster region ,Molecular biology ,Epitope ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Oncology ,Perforin ,030220 oncology & carcinogenesis ,biology.protein ,medicine ,Immunology and Allergy ,Cytotoxic T cell ,B cell ,Original Research - Abstract
The B-cell receptor (BCR) expressed by a clonal B cell tumor is a tumor specific antigen (idiotype). However, the T-cell epitopes within human BCRs which stimulate protective immunity still lack detailed characterization. In this study, we identified 17 BCR peptide-specific CD4+ T-cell epitopes derived from BCR heavy and light chain variable region sequences. Detailed analysis revealed these CD4+ T-cell epitopes stimulated normal donors' and patients' Th1 CD4+ T cells to directly recognize the autologous tumors by secretion of IFNγ, indicating the epitopes are processed and presented by tumor cells. One BCR peptide-specific CD4+ T cell line was also cytotoxic and lysed autologous tumor cells through the perforin pathway. Sequence analysis of the epitopes revealed that 10 were shared by multiple primary patients' tumors, and 16 had the capacity to bind to more than one HLA DRB1 allele. T cells stimulated by shared epitopes recognized primary tumors expressing the same sequences on multiple HLA DRB1 alleles. In conclusion, we identified 17 BCR-derived CD4+ T-cell epitopes with promiscuous HLA DRB1 binding affinity that are shared by up to 36% of patients, suggesting a strategy to overcome the requirement for individual preparation of therapeutic agents targeting idiotype.
- Published
- 2016
18. Redirecting T-Cell Specificity to EGFR Using mRNA to Self-limit Expression of Chimeric Antigen Receptor
- Author
-
Helen Huls, Jianliang Dai, Amy B. Heimberger, Hillary G. Caruso, Hiroki Torikai, Harjeet Singh, Dean A. Lee, Sourindra Maiti, Kim Anh Do, Ling Zhang, Laurence J.N. Cooper, and Richard E. Champlin
- Subjects
0301 basic medicine ,Cancer Research ,Immunology ,Receptors, Antigen, T-Cell ,T-Cell Antigen Receptor Specificity ,Biology ,CD8-Positive T-Lymphocytes ,Lymphocyte Activation ,Cancer Vaccines ,Immunotherapy, Adoptive ,Article ,03 medical and health sciences ,Interleukin 21 ,Artificial antigen presenting cells ,Antigen ,Antigens, Neoplasm ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Immunology and Allergy ,Cytotoxic T cell ,Humans ,IL-2 receptor ,RNA, Messenger ,Antigen-presenting cell ,Cell Proliferation ,Pharmacology ,CD40 ,Molecular biology ,Coculture Techniques ,ErbB Receptors ,030104 developmental biology ,biology.protein ,Interleukin 12 ,Genetic Engineering ,Glioblastoma ,Immunologic Memory - Abstract
Potential for on-target, but off-tissue toxicity limits therapeutic application of genetically modified T cells constitutively expressing chimeric antigen receptors (CARs) from tumor-associated antigens expressed in normal tissue, such as epidermal growth factor receptor (EGFR). Curtailing expression of CAR through modification of T cells by in vitro-transcribed mRNA species is one strategy to mitigate such toxicity. We evaluated expression of an EGFR-specific CAR coded from introduced mRNA in human T cells numerically expanded ex vivo to clinically significant numbers through coculture with activating and propagating cells (AaPC) derived from K562 preloaded with anti-CD3 antibody. The density of AaPC could be adjusted to affect phenotype of T cells such that reduced ratio of AaPC resulted in higher proportion of CD8 and central memory T cells that were more conducive to electrotransfer of mRNA than T cells expanded with high ratios of AaPC. RNA-modified CAR T cells produced less cytokine, but demonstrated similar cytolytic capacity as DNA-modified CAR T cells in response to EGFR-expressing glioblastoma cells. Expression of CAR by mRNA transfer was transient and accelerated by stimulation with cytokine and antigen. Loss of CAR abrogated T-cell function in response to tumor and normal cells expressing EGFR. We describe a clinically applicable method to propagate and modify T cells to transiently express EGFR-specific CAR to target EGFR-expressing tumor cells that may be used to limit on-target, off-tissue toxicity to normal tissue.
- Published
- 2016
19. A miR-192-EGR1-HOXB9 regulatory network controls the angiogenic switch in cancer
- Author
-
Cristian Rodriguez-Aguayo, Gabriel Lopez-Berestein, Fangrong Shen, Shelley M. Herbrich, Kshipra M. Gharpure, Justyna Filant, Prahlad T. Ram, Viviana Vidal-Anaya, Elena G. Seviour, Guillermo N. Armaiz-Pena, Ehsan A. Ehsanipour, Anil K. Sood, Monika Haemmerle, Sourindra Maiti, Rajesha Rupaimoole, Min Zhang, Chad V. Pecot, Sunila Pradeep, Laurence J.N. Cooper, Ji Hoon Kim, Courtney Olsen, Hiroto Hatakeyama, Susan L. Tucker, Archana S. Nagaraja, Hee Dong Han, Elizabeth Pham, Da Yang, Sherry Y. Wu, Rouba Ali-Fehmi, Keith A. Baggerly, Xinna Zhang, Cristina Ivan, Menashe Bar-Eli, Mien Chie Hung, Li Huang, Ju Seog Lee, and Michael McGuire
- Subjects
0301 basic medicine ,Angiogenic Switch ,Angiogenesis ,Science ,Regulator ,Down-Regulation ,General Physics and Astronomy ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Neovascularization ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Cell Line, Tumor ,microRNA ,medicine ,Animals ,Humans ,Gene Regulatory Networks ,Early Growth Response Protein 1 ,Homeodomain Proteins ,Ovarian Neoplasms ,Multidisciplinary ,Neovascularization, Pathologic ,Cancer ,Genetic Therapy ,General Chemistry ,medicine.disease ,Kidney Neoplasms ,Tumor Burden ,3. Good health ,MicroRNAs ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,Cancer cell ,Immunology ,Phosphatidylcholines ,Cancer research ,Female ,Growth inhibition ,medicine.symptom - Abstract
A deeper mechanistic understanding of tumour angiogenesis regulation is needed to improve current anti-angiogenic therapies. Here we present evidence from systems-based miRNA analyses of large-scale patient data sets along with in vitro and in vivo experiments that miR-192 is a key regulator of angiogenesis. The potent anti-angiogenic effect of miR-192 stems from its ability to globally downregulate angiogenic pathways in cancer cells through regulation of EGR1 and HOXB9. Low miR-192 expression in human tumours is predictive of poor clinical outcome in several cancer types. Using 1,2-dioleoyl-sn-glycero-3-phosphatidylcholine (DOPC) nanoliposomes, we show that miR-192 delivery leads to inhibition of tumour angiogenesis in multiple ovarian and renal tumour models, resulting in tumour regression and growth inhibition. This anti-angiogenic and anti-tumour effect is more robust than that observed with an anti-VEGF antibody. Collectively, these data identify miR-192 as a central node in tumour angiogenesis and support the use of miR-192 in an anti-angiogenesis therapy., The formation of blood vessels in tumours, angiogenesis, is a promising target for therapy. Here, the authors show that microRNA192 has anti-angiogenic functions and negatively regulates EGR1 and HOXB9, and that delivery of this microRNA to tumours in vivo can reduce angiogenesis and tumour growth.
- Published
- 2016
20. Measurement of Average Telomere Length in Ex Vivo Expanded Natural Killer Cells by Fluorescence In Situ Hybridization (FISH) and Flow Cytometry
- Author
-
Sourindra Maiti
- Subjects
0301 basic medicine ,Innate immune system ,medicine.medical_treatment ,Immunotherapy ,Biology ,Molecular biology ,Telomere ,Cell biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Telomere Homeostasis ,Flow-FISH ,medicine ,Cytotoxic T cell ,Cytokine secretion ,Ex vivo ,030215 immunology - Abstract
Natural killer (NK) cells are a subset of cytotoxic lymphocytes that play a critical role in innate immune surveillance against infections and tumors through cytokine secretion and target cell lysis. NK cells function without any need for prior antigen exposure. Thus, more recently NK cells are considered a promising source of lymphocytes for adoptive tumor therapy. However, because NK cells represent only a small lymphocyte fraction, expand poorly ex vivo, and have limited life spans, clinical scale generation of NK cells for tumor immunotherapy was a challenging issue. To overcome this challenge, numerous expansion platforms have been developed. However, ex vivo expansion of NK cells could lead to proliferation-induced senescence. Telomeres at the end of chromosomes play a crucial role in maintaining the integrity of the chromosome and are lost at each cell division in somatic cells and have emerged as important cellular elements in aging and cancer. Because telomere length is known to decrease in adult human NK cells and is associated with proliferation-induced senescence, it is important to determine the effect of NK cell expansion systems on telomere length. In this chapter, a detailed protocol is provided to analyze the telomere length of expanded NK cells using a commercially available Flow FISH kit.
- Published
- 2016
21. Author response: Tumor microenvironment derived exosomes pleiotropically modulate cancer cell metabolism
- Author
-
Laurence J.N. Cooper, Deepak Nagrath, Joelle Baddour, Donna M. Peehl, Sourindra Maiti, F. Anthony San Lucas, Héctor M. Alvarez, Hongyun Zhao, Prahlad T. Ram, Abhinav Achreja, Elena G. Seviour, Thavisha Tudawe, Sonal Gupta, Anirban Maitra, Tyler J. Moss, Vincent Bernard, Lifeng Yang, and Juan C. Marini
- Subjects
Tumor microenvironment ,Cancer cell ,Cancer research ,Metabolism ,Biology ,Microvesicles - Published
- 2015
22. Reprogramming CD19-Specific T Cells with IL-21 Signaling Can Improve Adoptive Immunotherapy of B-Lineage Malignancies
- Author
-
Simon Olivares, Tiejuan Mi, Partow Kebriaei, Sourindra Maiti, Matthew J. Figliola, Dean A. Lee, Richard E. Champlin, Margaret J. Dawson, Harjeet Singh, Laurence J.N. Cooper, Helen Huls, and Kirsten Switzer
- Subjects
STAT3 Transcription Factor ,Cancer Research ,Adoptive cell transfer ,T-Lymphocytes ,medicine.medical_treatment ,Antigens, CD19 ,Biology ,Immunotherapy, Adoptive ,Article ,Interferon-gamma ,Mice ,Interleukin 21 ,CD28 Antigens ,Antigen ,Mice, Inbred NOD ,medicine ,Animals ,Humans ,Cell Lineage ,L-Selectin ,B-Lymphocytes ,Interleukins ,CD28 ,T lymphocyte ,Immunotherapy ,Interleukin-12 ,Coculture Techniques ,Chimeric antigen receptor ,Cell biology ,Oncology ,Hematologic Neoplasms ,Immunology ,Interleukin 12 ,K562 Cells ,Signal Transduction - Abstract
Improving the therapeutic efficacy of T cells expressing a chimeric antigen receptor (CAR) represents an important goal in efforts to control B-cell malignancies. Recently an intrinsic strategy has been developed to modify the CAR itself to improve T-cell signaling. Here we report a second extrinsic approach based on altering the culture milieu to numerically expand CAR+ T cells with a desired phenotype, for the addition of interleukin (IL)-21 to tissue culture improves CAR-dependent T-cell effector functions. We used electrotransfer of Sleeping Beauty system to introduce a CAR transposon and selectively propagate CAR+ T cells on CD19+ artificial antigen-presenting cells (aAPC). When IL-21 was present, there was preferential numeric expansion of CD19-specific T cells which lysed and produced IFN-γ in response to CD19. Populations of these numerically expanded CAR+ T cells displayed an early memory surface phenotype characterized as CD62L+CD28+ and a transcriptional profile of naïve T cells. In contrast, T cells propagated with only exogenous IL-2 tended to result in an overgrowth of CD19-specific CD4+ T cells. Furthermore, adoptive transfer of CAR+ T cells cultured with IL-21 exhibited improved control of CD19+ B-cell malignancy in mice. To provide coordinated signaling to propagate CAR+ T cells, we developed a novel mutein of IL-21 bound to the cell surface of aAPC that replaced the need for soluble IL-21. Our findings show that IL-21 can provide an extrinsic reprogramming signal to generate desired CAR+ T cells for effective immunotherapy. Cancer Res; 71(10); 3516–27. ©2011 AACR.
- Published
- 2011
23. The hyperactive Sleeping Beauty transposase SB100X improves the genetic modification of T cells to express a chimeric antigen receptor
- Author
-
Harjeet Singh, Richard E. Champlin, Laurence J.N. Cooper, Simon Olivares, Zeming Jin, Helen Huls, Sourindra Maiti, Zoltán Ivics, Zsuzsanna Izsvák, Lajos Mátés, and Dean A. Lee
- Subjects
Transposition (music) ,P element ,Transposable element ,Plasmid ,Antigen ,Genetics ,Molecular Medicine ,Biology ,Sleeping Beauty transposon system ,Molecular Biology ,Molecular biology ,Transposase ,Transposase activity - Abstract
Sleeping Beauty (SB3) transposon and transposase constitute a DNA plasmid system used for therapeutic human cell genetic engineering. Here we report a comparison of SB100X, a newly developed hyperactive SB transposase, to a previous generation SB11 transposase to achieve stable expression of a CD19-specific chimeric antigen receptor (CAR3) in primary human T cells. The electro-transfer of SB100X expressed from a DNA plasmid or as an introduced mRNA species had superior transposase activity in T cells based on the measurement of excision circles released after transposition and emergence of CAR expression on T cells selectively propagated upon CD19+ artificial antigen-presenting cells. Given that T cells modified with SB100X and SB11 integrate on average one copy of the CAR transposon in each T-cell genome, the improved transposition mediated by SB100X apparently leads to an augmented founder effect of electroporated T cells with durable integration of CAR. In aggregate, SB100X improves SB transposition in primary human T cells and can be titrated with an SB transposon plasmid to improve the generation of CD19-specific CAR+ T cells.
- Published
- 2011
24. Adoptive T-Cell Therapy with TCL1-Specific TCR for B-Cell Lymphomas
- Author
-
Sattva S. Neelapu, Fuliang Chu, Hiroki Torikai, Sourindra Maiti, Rohit Mathur, Zheng Zhang, R. Eric Davis, Laurence J.N. Cooper, Kelsey E. Moriarty, Deepshika Medapalli, Jinsheng Weng, Man Chun John Ma, Swathi Karri, Yong Pan, and Xiaoyun Cheng
- Subjects
Adoptive cell transfer ,business.industry ,T cell ,Immunology ,Follicular lymphoma ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,medicine.anatomical_structure ,Cancer research ,medicine ,Cytotoxic T cell ,Mantle cell lymphoma ,business ,Diffuse large B-cell lymphoma ,B cell ,CD8 - Abstract
Chimeric antigen receptor (CAR)-modified T-cell therapy targeting CD19 induces high response rates in patients with relapsed or refractory B-cell lymphomas. However, about 60% of patients experience primary or secondary resistance after CD19-targeted CAR T-cell therapy and a major of cause of failure appears to be due to loss of CD19 expression on the tumor. Therefore, novel targets for adoptive T-cell therapeutic approaches are needed to further improve clinical outcome in these patients. T-cell leukemia/lymphoma antigen1 (TCL1) is an oncoprotein that is overexpressed in multiple B-cell malignancies including follicular lymphoma (FL), mantle cell lymphoma (MCL), diffuse large B-cell lymphoma (DLBCL), and chronic lymphocytic leukemia (CLL). Importantly, it has restricted expression in only a subset of B cells among normal tissues. We previously identified a TCL1-derived HLA-A2-binding epitope (TCL170-79 SLLPIMWQLY) that can be used to generate TCL1-specific CD8+ T cells from peripheral blood mononuclear cells of both HLA-A2+ normal donors and lymphoma patients. More importantly, we showed that the TCL1-specific CD8+ T cells lysed autologous primary lymphoma cells but not normal B cells (Weng et al. Blood 2012). To translate the above discovery into clinic, we cloned the T-cell receptor (TCR) alpha and beta chains from a TCL1-specific CD8+ T-cell clone and showed that this TCL1-TCR could be transduced into polyclonal donor T cells using a lentiviral system with a transduction efficiency of >40% as determined by TCL170-79 tetramer positive T cells. Furthermore, we demonstrated that the TCL1-TCR-transduced T cells recognized T2 cells pulsed with TCL170-79 peptide producing IFN- γ >8 ng/ml and IL-2 >350 ng/ml but were not reactive to control HIV-Gag peptide (IFN- γ 10 ng/ml) suggesting it has moderate to high avidity. Importantly, TCL1-TCR-transduced T cells lysed HLA-A2+ (up to 43% lysis of Mino and 25% lysis of Jeko-1 at 40:1 Effector:Target ratio) but not HLA-A2- lymphoma cell lines (5.5% lysis of HLA A2- Raji and 2.3% lysis of Daudi at 40:1 Effector:Target ratio). TCL1-TCR-transduced T cells were also cytotoxic to HLA-A2+ primary lymphoma tumor cells (up to 48% lysis of CLL, 43% lysis of FL, 41% lysis of DLBCL, 46% lysis of splenic marginal zone lymphoma, and 11% lysis of MCL at 40:1 Effector:Target ratio) but not normal B cells derived from the same patients. Lastly, TCL1-TCR transduced T cells showed high efficacy in in vivo models. Adoptive transfer of the TCL1-TCR-tranduced T cells significantly reduced lymphoma tumor growth and extended survival in Mino mantle cell lymphoma cell line xenograft model (48% survival in TCL1-TCR-T treated group vs. 12.5% survival in control group at 10 weeks n=7-8 mice/group; P=0.02). Collectively, our data suggest that the high expression in B-cell tumors, restricted expression in normal tissues, and presence of an immunogenic CD8 T-cell epitope, make TCL1 a target for T cell-based therapeutic approaches in multiple B-cell malignancies. Our results also demonstrate that the TCL1-specific TCR-transduced T cells may serve as a novel adoptive immunotherapy approach for the treatment of patients with various B-cell malignancies (including FL, MCL, DLBCL, CLL). Acknowledgments: This study is supported by MD Anderson Moon Shot Program and CPRIT and the National Natural Science Foundation of China Grant (No. 81570189) Disclosures Neelapu: Kite/Gilead: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Celgene: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Cellectis: Research Funding; Poseida: Research Funding; Merck: Consultancy, Membership on an entity's Board of Directors or advisory committees, Research Funding; Acerta: Research Funding; Karus: Research Funding; Bristol-Myers Squibb: Research Funding; Novartis: Membership on an entity's Board of Directors or advisory committees; Unum Therapeutics: Membership on an entity's Board of Directors or advisory committees.
- Published
- 2018
25. Abstract 2566: A TCL1-specific T-cell receptor redirects T cells against B-cell lymphomas and non-hematological tumors
- Author
-
Yong Pan, Hua Wang, Fuliang Chu, Hiroki Torikai, Jinsheng Weng, Sourindra Maiti, Xiaoyun Cheng, Laurence J.N. Cooper, Sattva S. Neelapu, Kelsey E. Moriarty, Deepshika Medapalli, Flavio Egidio Baio, and Swathi Karri
- Subjects
Cancer Research ,Adoptive cell transfer ,medicine.medical_treatment ,T cell ,Immunotherapy ,Biology ,medicine.disease ,Epitope ,Lymphoma ,medicine.anatomical_structure ,Oncology ,medicine ,Cancer research ,Mantle cell lymphoma ,CD8 ,B cell - Abstract
T-cell leukemia/lymphoma Antigen1 (TCL1) is a B-cell oncoprotein that is overexpressed in multiple forms of B-cell lymphoma. In our previous study, we identified a HLA*A0201-restricted T cell epitope from TCL1 that stimulated CD8+ T cells to specifically lyse lymphoma cells. In this study, we have cloned the TCL170-79 peptide-specific T-cell receptor (TCR) gene that after lentivirus transduction into donor T cells can redirect the specificity of T cells to lyse T2 cells pulsed with TCL170-79 but not control peptide. The TCR-transduced CD8+ T cells efficiently lysed HLA-0201+, TCL1-expressing human lymphoma cell lines and primary lymphoma cells, but not normal B cells. Adoptive transfer of TCR-transduced T cells into a mantle cell lymphoma xenograft mouse model inhibited the tumor growth and significantly extended the survival of mice. In addition, we found that TCL1 is expressed in multiple solid tumors. The TCR-transduced T cells lysed colon and liver tumor cells but not normal human cells in vitro as well as in a mouse xenograft model. Collectively, our data demonstrate that TCL1-specific TCR-based immunotherapy is highly effective against human B-cell lymphomas and non-hematological tumors. Citation Format: Jinsheng Weng, Kelsey E. Moriarty, Yong Pan, Flavio Egidio Baio, Hua Wang, Hiroki Torikai, Sourindra N. Maiti, Fuliang Chu, Xiaoyun Cheng, Swathi Karri, Deepshika Medapalli, Laurence J. N. Cooper, Sattva S. Neelapu. A TCL1-specific T-cell receptor redirects T cells against B-cell lymphomas and non-hematological tumors [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 2566.
- Published
- 2018
26. piggyBac Transposon/Transposase System to Generate CD19-Specific T Cells for the Treatment of B-Lineage Malignancies
- Author
-
Pulivarthi H. Rao, Tiejuan Mi, Laurence J.N. Cooper, Pallavi R. Manuri, Joseph M. Kaminski, Richard E. Champlin, Harjeet Singh, Margaret J. Dawson, Stephen Gottschalk, Matthew H. Wilson, Helen Huls, Simon Olivares, Elizabeth J. Shpall, Yozo Nakazawa, Partow Kebriaei, Sourindra Maiti, and Dean A. Lee
- Subjects
Lymphoma, B-Cell ,T-Lymphocytes ,T cell ,Antigens, CD19 ,Genetic Vectors ,Antigen-Presenting Cells ,Transposases ,Biology ,P element ,Antigen ,Cell Line, Tumor ,Genetics ,medicine ,Humans ,Transgenes ,Antigen-presenting cell ,Molecular Biology ,Research Articles ,Cells, Cultured ,Transposase ,Genetic Therapy ,Sleeping Beauty transposon system ,Molecular biology ,Coculture Techniques ,Chimeric antigen receptor ,Receptors, Antigen ,Electroporation ,medicine.anatomical_structure ,PiggyBac Transposon System ,DNA Transposable Elements ,Molecular Medicine ,Glioblastoma ,K562 Cells ,Plasmids - Abstract
Nonviral integrating vectors can be used for expression of therapeutic genes. piggyBac (PB), a transposon/transposase system, has been used to efficiently generate induced pluripotent stems cells from somatic cells, without genetic alteration. In this paper, we apply PB transposition to express a chimeric antigen receptor (CAR) in primary human T cells. We demonstrate that T cells electroporated to introduce the PB transposon and transposase stably express CD19-specific CAR and when cultured on CD19(+) artificial antigen-presenting cells, numerically expand in a CAR-dependent manner, display a phenotype associated with both memory and effector T cell populations, and exhibit CD19-dependent killing of tumor targets. Integration of the PB transposon expressing CAR was not associated with genotoxicity, based on chromosome analysis. PB transposition for generating human T cells with redirected specificity to a desired target such as CD19 is a new genetic approach with therapeutic implications.
- Published
- 2010
27. β-2-Glycoprotein 1-dependent Macrophage Uptake of Apoptotic Cells
- Author
-
Sourindra Maiti, Krishnakumar Balasubramanian, Alan J. Schroit, and Johanna A. Ramoth
- Subjects
Phagocyte ,Protein family ,Phagocytosis ,LRP1B ,Lipoprotein receptor-related protein ,Cell Biology ,Biology ,Biochemistry ,Cell biology ,medicine.anatomical_structure ,medicine ,Macrophage ,Receptor ,Molecular Biology ,Tissue homeostasis - Abstract
The recognition and removal of apoptotic cells is critical to development, tissue homeostasis, and the resolution of inflammation. Many studies have shown that phagocytosis is regulated by signaling mechanisms that involve distinct ligand-receptor interactions that drive the engulfment of apoptotic cells. Studies from our laboratory have shown that the plasma protein β-2-glycoprotein 1 (β2GP1), a member of the short consensus repeat superfamily, binds phosphatidylserine-containing vesicles and apoptotic cells and promotes their bridging and subsequent engulfment by phagocytes. The phagocyte receptor for the protein/apoptotic cell complex, however, is unknown. Here we report that a member of the low density lipoprotein receptor-related protein family on phagocytes binds and facilitates engulfment of β2GP1-phosphatidylserine and β2GP1-apoptotic cell complexes. Using recombinant β2GP1, we also show that β2GP1-dependent uptake is mediated by bridging of the target cell to the phagocyte through the protein C- and N-terminal domains, respectively.
- Published
- 2008
28. Plasmin-Cleaved β-2-Glycoprotein 1 Is an Inhibitor of Angiogenesis
- Author
-
Alan J. Schroit, Taro Sakai, Jyotsna B. Halder, Sourindra Maiti, and Krishnakumar Balasubramanian
- Subjects
Male ,Plasmin ,Angiogenesis ,Molecular Sequence Data ,Angiogenesis Inhibitors ,Biology ,Kringle domain ,Pathology and Forensic Medicine ,Neovascularization ,Mice ,Kringles ,Cell Movement ,In vivo ,medicine ,Animals ,Beta 2-Glycoprotein I ,Amino Acid Sequence ,Fibrinolysin ,Cells, Cultured ,Mice, Inbred BALB C ,Matrigel ,Neovascularization, Pathologic ,Endothelial Cells ,Neoplasms, Experimental ,Gelatin Sponge, Absorbable ,Cell biology ,Mice, Inbred C57BL ,Endothelial stem cell ,Drug Combinations ,Biochemistry ,beta 2-Glycoprotein I ,Proteoglycans ,Collagen ,Endothelium, Vascular ,Laminin ,medicine.symptom ,Neoplasm Transplantation ,Regular Articles ,medicine.drug - Abstract
beta-2-Glycoprotein 1, an abundant plasma glycoprotein, binds anionic cell surfaces and functions as a regulator of thrombosis. Here, we show that cleavage of the kringle domain at Lys317/Thr318 switches its function to a regulator of angiogenesis. In vitro, the cleaved protein specifically inhibited the proliferation and migration of endothelial cells. The protein was without effect on preformed endothelial cell tubes. In vivo, the cleaved protein inhibited neovascularization into subcutaneously implanted Matrigel and Gelfoam sponge implants and the growth of orthotopically injected tumors. Collectively, these data indicate that plasmin-cleaved beta-2-glycoprotein 1 is a potent antiangiogenic and antitumor molecule of potential therapeutic significance.
- Published
- 2007
29. Genetic editing of HLA expression in hematopoietic stem cells to broaden their human application
- Author
-
Philip D. Gregory, Edward J. Rebar, Sourindra Maiti, Gladys P Dulay, Jianliang Dai, Loren Gragert, Hiroki Torikai, Kirsten Switzer, Andreas Reik, Martin Maiers, Elizabeth J. Shpall, Michael C. Holmes, Sonny Ang, Laurence J.N. Cooper, Amer Najjar, Tiejuan Mi, Richard E. Champlin, and Helen Huls
- Subjects
0301 basic medicine ,Transplantation, Heterologous ,Gene Expression ,Histocompatibility Testing ,Human leukocyte antigen ,HLA-C Antigens ,Bioinformatics ,Health Services Accessibility ,Article ,Donor Selection ,03 medical and health sciences ,Mice ,Homologous chromosome ,Medicine ,Animals ,Humans ,Transplantation, Homologous ,Alleles ,Zinc finger ,Multidisciplinary ,Deoxyribonucleases ,HLA-A Antigens ,business.industry ,Racial Groups ,Hematopoietic Stem Cell Transplantation ,Zinc Fingers ,Hematopoietic Stem Cells ,Zinc finger nuclease ,3. Good health ,Transplantation ,Haematopoiesis ,030104 developmental biology ,HLA-B Antigens ,Immunology ,Stem cell ,business ,Genetic Engineering ,Unrelated Donors ,Gene Deletion ,HLA-DRB1 Chains - Abstract
Mismatch of human leukocyte antigens (HLA) adversely impacts the outcome of patients after allogeneic hematopoietic stem-cell transplantation (alloHSCT). This translates into the clinical requirement to timely identify suitable HLA-matched donors which in turn curtails the chances of recipients, especially those from a racial minority, to successfully undergo alloHSCT. We thus sought to broaden the existing pool of registered unrelated donors based on analysis that eliminating the expression of the HLA-A increases the chance for finding a donor matched at HLA-B, -C and -DRB1 regardless of a patient’s race. Elimination of HLA-A expression in HSC was achieved using artificial zinc finger nucleases designed to target HLA-A alleles. Significantly, these engineered HSCs maintain their ability to engraft and reconstitute hematopoiesis in immunocompromised mice. This introduced loss of HLA-A expression decreases the need to recruit large number of donors to match with potential recipients and has particular importance for patients whose HLA repertoire is under-represented in the current donor pool. Furthermore, the genetic engineering of stem cells provides a translational approach to HLA-match a limited number of third-party donors with a wide number of recipients.
- Published
- 2015
30. The Wt1+/R394W Mouse Displays Glomerulosclerosis and Early-Onset Renal Failure Characteristic of Human Denys-Drash Syndrome
- Author
-
Guizhi Sun, Fei Gao, Sourindra Maiti, Jian Min Deng, Richard R. Behringer, Vicki Huff, Nelson G. Ordonez, and Malini Udtha
- Subjects
Male ,medicine.medical_specialty ,Denys–Drash syndrome ,Genes, Wilms Tumor ,Gene Expression ,Kidney development ,Biology ,Mice ,chemistry.chemical_compound ,Species Specificity ,Internal medicine ,Mammalian Genetic Models with Minimal or Complex Phenotypes ,medicine ,Animals ,Humans ,Point Mutation ,Missense mutation ,Renal Insufficiency ,Molecular Biology ,Adaptor Proteins, Signal Transducing ,Genetics ,Proteinuria ,Base Sequence ,Glomerulosclerosis, Focal Segmental ,Intracellular Signaling Peptides and Proteins ,Membrane Proteins ,Proteins ,Glomerulosclerosis ,Heterozygote advantage ,Wilms' tumor ,DNA ,Cell Biology ,Denys-Drash Syndrome ,medicine.disease ,Mice, Mutant Strains ,Mice, Inbred C57BL ,Cytoskeletal Proteins ,Disease Models, Animal ,Microscopy, Electron ,Phenotype ,Endocrinology ,Podocalyxin ,chemistry ,Female ,medicine.symptom ,Cell Division - Abstract
Renal failure is a frequent and costly complication of many chronic diseases, including diabetes and hypertension. One common feature of renal failure is glomerulosclerosis, the pathobiology of which is unclear. To help elucidate this, we generated a mouse strain carrying the missense mutation Wt1 R394W, which predisposes humans to glomerulosclerosis and early-onset renal failure (Denys-Drash syndrome [DDS]). Kidney development was normal in Wt1(+/R394W) heterozygotes. However, by 4 months of age 100% of male heterozygotes displayed proteinuria and glomerulosclerosis characteristic of DDS patients. This phenotype was observed in an MF1 background but not in a mixed B6/129 background, suggestive of the action of a strain-specific modifying gene(s). WT1 encodes a nuclear transcription factor, and the R394W mutation is known to impair this function. Therefore, to investigate the mechanism of Wt1 R394W-induced renal failure, the expression of genes whose deletion leads to glomerulosclerosis (NPHS1, NPHS2, and CD2AP) was quantitated. In mutant kidneys, NPHS1 and NPHS2 were only moderately downregulated (25 to 30%) at birth but not at 2 or 4 months. Expression of CD2AP was not changed at birth but was significantly upregulated at 2 and 4 months. Podocalyxin was downregulated by 20% in newborn kidneys but not in kidneys at later ages. Two other genes implicated in glomerulosclerosis, TGFB1 and IGF1, were upregulated at 2 months and at 2 and 4 months, respectively. It is not clear whether the significant alterations in gene expression are a cause or a consequence of the disease process. However, the data do suggest that Wt1 R394W-induced glomerulosclerosis may be independent of downregulation of the genes for NPHS1, NPHS2, CD2AP, and podocalyxin and may involve other genes yet to be implicated in renal failure. The Wt1(R394W) mouse recapitulates the pathology and disease progression observed in patients carrying the same mutation, and the mutation is completely penetrant in male animals. Thus, it will be a powerful and biologically relevant model for investigating the pathobiology of the earliest events in glomerulosclerosis.
- Published
- 2004
31. 278. Next-Generation Non-Viral Gene Transfer to Redirect T-Cell Specificity
- Author
-
Mary Helen Huls, Simon Olivares, Navin Varadarajan, Lenka V. Hurton, Gabrielle Romain, Tiejuan Mi, Partow Kebriaei, Laurence J.N. Cooper, Sourindra Maiti, Shihuang Su, Matthew J. Figliola, Harjeet Singh, and Ling Zhang
- Subjects
Pharmacology ,biology ,Chemistry ,CD28 ,medicine.disease ,Molecular biology ,Minimal residual disease ,CD19 ,Chimeric antigen receptor ,Leukemia ,In vivo ,Drug Discovery ,Immunology ,Genetics ,medicine ,biology.protein ,Extracellular ,Molecular Medicine ,Receptor ,Molecular Biology - Abstract
Non-viral gene transfer using the Sleeping Beauty (SB) transposon/transposase system has been successfully tested in humans to express a chimeric antigen receptor (CAR) to redirect T-cell specificity to CD19. This system has been modified to (i) improve the design of the CD19-specific CAR and (ii) reduce the time in culture to 14 days. Our previous clinical trials infused T cells expressing a 2nd generation CAR (designated CD19RCD28) with an IgG4-Fc stalk that activated via chimeric CD28 and CD3ζ. To evaluate the length of extracellular domain on function, we tested four CD19-specific CARs with two long [IgG4-Fc (CD19RCD28) and EQ (L235E and N297Q) mutant IgG4-Fc (CD19R*CD28)], medium (CD8α hinge, CD19RCD8CD28) and short (12aa IgG1 hinge, CD19R12aaCD28) stalks which all signaled through chimeric CD28 and CD3ζ endodomains. Generation of our T cells is based on electro-transfer of CARs coded by the SB system and antigen-specific stimulation through activating and K562-derived propagating cells (AaPC) in the presence of exogenous cytokines. After electro-transfer of SB-derived DNA plasmids, T cells were selectively propagated with either a new two-weekly (2x) or standard four-weekly (4x) additions of AaPC. All genetically modified T cells were capable of specific lysis of CD19+ tumor targets and producing IFN-γ in response to CD19+ stimulator cells. Serial killing was tested using massively parallel microscopy to observe single T cells and we observed that CDl9RCD8CD28+ T cells exhibited superior ability to partake in multiple killing events. CAR+ T cells were further tested in vivo for their ability to control CD19+ leukemia in a mouse model of minimal residual disease as well as established disease (Figure A and BFigure A and B). We found that T cells expressing modified CARs (CD19R*CD28, CD19RCD8CD28, CD19R12aaCD28) with reduced ability to bind to Fc gamma receptors (FcγR) were able to control leukemia more efficiently in mice compared to T cells expressing CD19RCD28. The CD19RCD8CD28 CAR was superior in controlling disease in the model of minimal residual disease compared with the CAR design evaluated in our prior clinical trials. T cells expressing CD19R*CD28 and CD19RCD8CD28 were then evaluated in 2x stimulation cycle. Both the 4x CAR+ T cells had similar CAR expression (>70%) whereas the 2x CAR+ T cells exhibited reduced CAR expression (~40%). The 2x CAR+ T cells expressed markers associated with less differentiated state of naive-like and memory T cells when compared to 4x CAR+ T cells, which was supported by measurement of mRNA species using bar-coded probes. The efficacy of the CAR+ T cells was tested in mice bearing established CD19+ leukemia and we observed superior survival in mice receiving the 2x CAR+ T cells compared with the 4x CAR+ T cells (Figure CFigure C). These data depict that length of extracellular domain and its associated binding to FcγR improves T-cell effector functions and that decreasing the time in culture can improve control of leukemia in vivo. These data support the use of cDl9RCD8CD28 testing in a next-generation clinical trial (IND# 16474).View Large Image | Download PowerPoint Slide
- Published
- 2016
32. 654 Monitoring malignant T-cell clones by direct TCR expression assay in patients with leukemic cutaneous T-cell lymphoma over extracorporeal photopheresis
- Author
-
Xiao Ni, Sourindra Maiti, Madeleine Duvic, T. Langridge, and Laurence J.N. Cooper
- Subjects
business.industry ,T cell ,T-cell receptor ,Cutaneous T-cell lymphoma ,Cell Biology ,Dermatology ,medicine.disease ,Biochemistry ,medicine.anatomical_structure ,Immunology ,Extracorporeal Photopheresis ,medicine ,In patient ,business ,Molecular Biology - Published
- 2017
33. Plasma circulating-microRNA profiles are useful for assessing prognosis in patients with cytogenetically normal myelodysplastic syndromes
- Author
-
Sourindra Maiti, Carlos E. Bueso-Ramos, Zhuang Zuo, Hagop M. Kantarjian, Sanam Loghavi, L. Jeffrey Medeiros, Guillermo Garcia-Manero, Shimin Hu, Laurence J.N. Cooper, and George A. Calin
- Subjects
Adult ,Male ,medicine.medical_specialty ,Pathology ,Kaplan-Meier Estimate ,Gene mutation ,Biology ,Lower risk ,Pathology and Forensic Medicine ,Surgical pathology ,medicine ,Humans ,Aged ,Aged, 80 and over ,Myelodysplastic syndromes ,High-Throughput Nucleotide Sequencing ,Anatomical pathology ,Middle Aged ,medicine.disease ,Prognosis ,Circulating MicroRNA ,MicroRNAs ,medicine.anatomical_structure ,Myelodysplastic Syndromes ,Cytogenetic Analysis ,Female ,Bone marrow ,Hematopathology ,Biomarkers - Abstract
Myelodysplastic syndromes are a heterogeneous group of clonal bone marrow hematopoietic stem cell disorders characterized by ineffective hematopoiesis and peripheral cytopenias. Chromosomal abnormalities and gene mutations have been shown to have essential roles in pathogenesis and correlate with prognosis. Molecular markers, however, are not integrated into currently used prognostic systems. The goal of this study is to identify plasma microRNAs useful for classification and risk stratification of myelodysplastic syndromes. We applied a novel, high-throughput digital quantification technology (NanoString) to profile microRNA expression in plasma samples of 72 patients with myelodysplastic syndromes and 12 healthy individuals. We correlated these results with overall survival. In patients with myelodysplastic syndromes associated with a diploid karyotype, we identified and validated a 7-microRNA signature as an independent predictor of survival with a predictive power of 75% accuracy (P=0.008), better than those of the International Prognostic Scoring Systems and the MD Anderson Prognostic Lower Risk Prognostic Model. We also identified differentially expressed plasma microRNAs in patients with myelodysplastic syndromes versus healthy individuals and between patients with myelodysplastic syndromes associated with different cytogenetic features. These results validate the utility of circulating-microRNA levels as noninvasive biomarkers that can inform the management of patients with myelodysplastic syndromes. Our findings also shed light on interactions of gene regulation pathways that are likely involved in the pathogenesis of myelodysplastic syndromes.
- Published
- 2014
34. Very Rapid Production of CAR+ T-Cells upon Non-Viral Gene Transfer Using the Sleeping Beauty System
- Author
-
Amer Najjar, Lenka V. Hurton, Partow Kebriaei, Sourindra Maiti, Laurence J.N. Cooper, M. H. Huls, Tiejuan Mi, Shihuang Su, Aaron F. Orozco, Harjeet Singh, Kirsten Switzer, Richard E. Champlin, and Leo G. Flores
- Subjects
Business administration ,Immunology ,Equity (finance) ,Tissue membrane ,Gene transfer ,Cell Biology ,Hematology ,Biochemistry ,Viral gene ,Cyclic gmp ,Time frame ,Production (economics) ,Business ,Car t cells - Abstract
T cells are genetically modified ex vivo to express chimeric antigen receptors (CARs) for in vivo clinical applications. CAR-modified T cells have demonstrated redirected specificity and, in several clinical trials, potent anti-tumor activity. Manufacture, to date, is based upon gene transfer in cycling T cells followed by a period of tissue culture to achieve stable expression of introduced CARs. In contrast, we have adapted the non-viral-based Sleeping Beauty (SB) system to avoid the need for (i) T-cell activation and (ii) extended ex vivo tissue culture; thereby developing an approach whereby T cells can be both manufactured and delivered at multiple points-of-care (POC). This shortened culture decreases the time frame for manufacturing CAR+ T cells compared with current protocols for viral- or non-viral-based methodologies and is a foundation of our POC technology. Furthermore, reducing the ex vivo culture time preserves the memory and sustained persistence of CAR+ T cells by avoiding the differentiation programming induced by activation events typically required before or after gene transfer. We have previously demonstrated that co-expressing a membrane-bound version of interleukin-15 (mbIL15) significantly enhances the in vivo persistence of CAR+ T cells that are generated following 28-day culture after electro-transfer of SB derived DNA plasmids. Herein, we incorporated mbIL15 to generate POC CD19-specific CAR+ T cells. Peripheral blood mononuclear cells were genetically modified with mbIL15 and 2nd generation CAR coded from individual SB DNA plasmids and placed in culture for less than 2 days prior to adoptive transfer. NSG mice burdened by established and disseminated CD19+ leukemia were intravenously injected with just 7.5 x 105 CAR+ T cells, or an equivalent total T-cell dose of CARneg (unmodified or mock-treated) T cells. The mbIL15-CAR T-cell infusion yielded excellent disease-free survival, anti-tumor activity (Figure), and T-cell persistence. This approach to expediting the generation of genetically modified T cells enables the administration of CAR-modified naïve T cells and demonstrates that POC T cells have potent anti-tumor effects, even at a reduced CAR+ T-cell dose. This improvement to non-viral gene transfer and T-cell production reduces the requirement for tissue culture and thus time to manufacture within a GMP facility which translates to improvements in scalability and reduced costs. In summary, these data provide a translational pathway to undertake clinical trials by rapidly infusing T cells after genetic modification using the SB system. Disclosures Hurton: Intrexon: Equity Ownership, Patents & Royalties; Ziopharm Oncology: Equity Ownership, Patents & Royalties. Singh:Immatics: Equity Ownership, Patents & Royalties; Ziopharm Oncology: Equity Ownership, Patents & Royalties; Intrexon: Equity Ownership, Patents & Royalties. Switzer:Intrexon: Equity Ownership, Patents & Royalties; Ziopharm Oncology: Equity Ownership, Patents & Royalties. Mi:Intrexon: Equity Ownership, Patents & Royalties; Ziopharm Oncology: Equity Ownership, Patents & Royalties. Maiti:Ziopharm Oncology: Equity Ownership, Patents & Royalties; Intrexon: Equity Ownership, Patents & Royalties. Su:Ziopharm Oncology: Equity Ownership, Patents & Royalties; Intrexon: Equity Ownership, Patents & Royalties. Huls:Ziopharm Oncology: Equity Ownership, Patents & Royalties; Intrexon: Employment, Equity Ownership, Patents & Royalties. Champlin:Ziopharm Oncology: Equity Ownership, Patents & Royalties; Intrexon: Equity Ownership, Patents & Royalties. Cooper:Immatics: Equity Ownership; City of Hope: Patents & Royalties; Targazyme, Inc.: Equity Ownership; Sangamo BioSciences: Patents & Royalties; Intrexon: Equity Ownership; Ziopharm Oncology: Employment, Equity Ownership, Patents & Royalties; MD Anderson Cancer Center: Employment; Miltenyi Biotec: Honoraria.
- Published
- 2016
35. Recurrent ESR1-CCDC170 rearrangements in an aggressive subset of oestrogen receptor-positive breast cancers
- Author
-
Jin-Ah Kim, Dean P. Edwards, Gary C. Chamness, Xi Xi Cao, Xian Wang, Ying Tan, Rachel Schiff, Eric C. Chang, Jamunarani Veeraraghavan, Alejandro Contreras, Laurence J.N. Cooper, Sourindra Maiti, Susan G. Hilsenbeck, and Xiaosong Wang
- Subjects
General Physics and Astronomy ,Mice, Nude ,Breast Neoplasms ,Cell Separation ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Article ,Flow cytometry ,Mice ,Open Reading Frames ,Cell Movement ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,RNA, Small Interfering ,skin and connective tissue diseases ,Gene ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,Gene Rearrangement ,Multidisciplinary ,medicine.diagnostic_test ,Cell growth ,Sequence Analysis, RNA ,Gene Expression Profiling ,Estrogen Receptor alpha ,General Chemistry ,Gene rearrangement ,Flow Cytometry ,Phenotype ,3. Good health ,Gene expression profiling ,Gene Expression Regulation, Neoplastic ,Ki-67 Antigen ,Immunology ,Cancer research ,Intercellular Signaling Peptides and Proteins ,Female ,Signal transduction ,Carrier Proteins ,Estrogen receptor alpha ,Neoplasm Transplantation ,Signal Transduction - Abstract
Characterizing the genetic alterations leading to the more aggressive forms of oestrogen receptor-positive (ER+) breast cancers is of critical significance in breast cancer management. Here we identify recurrent rearrangements between the oestrogen receptor gene ESR1 and its neighbour CCDC170, which are enriched in the more aggressive and endocrine-resistant luminal B tumours, through large-scale analyses of breast cancer transcriptome and copy number alterations. Further screening of 200 ER+ breast cancers identifies eight ESR1-CCDC170-positive tumours. These fusions encode amino-terminally truncated CCDC170 proteins (ΔCCDC170). When introduced into ER+ breast cancer cells, ΔCCDC170 leads to markedly increased cell motility and anchorage-independent growth, reduced endocrine sensitivity and enhanced xenograft tumour formation. Mechanistic studies suggest that ΔCCDC170 engages Gab1 signalosome to potentiate growth factor signalling and enhance cell motility. Together, this study identifies neoplastic ESR1-CCDC170 fusions in a more aggressive subset of ER+ breast cancer, which suggests a new concept of ER pathobiology in breast cancer.
- Published
- 2013
36. Splenic NK1.1-Negative, TCRαβ Intermediate CD4+ T Cells Exist in Naive NK1.1 Allelic Positive and Negative Mice, with the Capacity to Rapidly Secrete Large Amounts of IL-4 and IFN-γ Upon Primary TCR Stimulation
- Author
-
Angus M. Moodycliffe, Sourindra Maiti, and Stephen E. Ullrich
- Subjects
Immunology ,Immunology and Allergy - Abstract
Splenic NK1.1+CD4+ T cells that express intermediate levels of TCRαβ molecules (TCRint) and the DX5 Ag (believed to identify an equivalent population in NK1.1 allelic negative mice) possess the ability to rapidly produce high quantities of immunomodulatory cytokines, notably IL-4 and IFN-γ, upon primary TCR activation in vivo. Indeed, only T cells expressing the NK1.1 Ag appear to be capable of this function. In this study, we demonstrate that splenic NK1.1-negative TCRintCD4+ T cells, identified on the basis of FcγR expression, exist in naive NK1.1 allelic positive (C57BL/6) and negative (C3H/HeN) mice with the capacity to produce large amounts of IL-4 and IFN-γ after only 8 h of primary CD3 stimulation in vitro. Furthermore, a comparison of the amounts of early cytokines produced by FcγR+CD4+TCRint T cells with NK1.1+CD4+ or DX5+CD4+TCRint T cells, simultaneously isolated from C57BL/6 or C3H/HeN mice, revealed strain and population differences. Thus, FcγR defines another subpopulation of splenic CD4+TCRint cells that can rapidly produce large concentrations of immunomodulatory cytokines, suggesting that CD4+TCRint T cells themselves may represent a unique family of immunoregulatory CD4+ T cells whose members include FcγR+CD4+ and NK1.1/DX5+CD4+ T cells.
- Published
- 1999
37. 212. Of States and Fates: Predicting T-Cell Immunity By the Numbers
- Author
-
Laurence J.N. Cooper, Luay Nakhleh, Sourindra Maiti, Jianrong Dong, Colleen M. O'Connor, and Sonny Ang
- Subjects
Pharmacology ,medicine.medical_treatment ,Rand index ,Gene regulatory network ,Immunotherapy ,CHOP ,Biology ,medicine.disease ,Chemotherapy regimen ,Pearson product-moment correlation coefficient ,Lymphoma ,symbols.namesake ,Immune system ,Immunology ,Drug Discovery ,symbols ,medicine ,Genetics ,Molecular Medicine ,Molecular Biology - Abstract
The immune system is a complex network of checks and balances in constant flux. Quantitative characterization of the system dynamics, sampled via constituent components such as T cells, combined with mathematical modeling enabled us to obtain “statistical pattern recognition” of immune states and transitions over time. Here we show a statistical framework to characterize immune states for adoptive immunotherapy using serial infusions of activated polyclonal T cells into companion canines diagnosed with B-cell non-Hodgkin Lymphoma (NHL) post CHOP chemotherapy regimen as a model for human disease. We applied multiplexed gene profiling techniques to assess changes in gene expression data from 10 companion canine patient clinical samples and gene regulatory networks (GRN) information to build an “immune landscape” that predicts immune states, state transitions, and plasticity for immunomodulation (“reprogrammability”) over the course of immunotherapy regimens. The potency of immune modulation and immune surveillance, two critical parameters of efficacy through inferred state transitions, were evaluated by measuring distortions and shifts in the immune landscape of canine patients undergoing treatment through system dynamics, Adjusted Rand Index, Pearson Correlation, and ANOVA analyses. Adjust Rand Index validated hierarchical clustering analyses. Pearson correlations isolated genes that were up- or down-regulated with regard to single genes of interest. ANOVA analyses indicated gene expression difference between temporal samples. Applying such analyses to adoptive T-cell immunotherapy, allowed us to examine clinical/tumor remissions through the lens of immune state transitions, affording real-time improvements to therapeutic regimens. In developing a formal framework to capture the effects of molecular signatures and GRN over time to describe distinct immune states, we enhanced the clinical cancer immunologist's analytical arsenal (with a measuring tool/sensor) and helped bridge the gap between bench and bedside.
- Published
- 2015
- Full Text
- View/download PDF
38. Genetically modified artificial antigen-presenting cells (aAPC) for expansion of melanoma tumor infiltrating lymphocytes with optimal properties for adoptive cell therapy
- Author
-
Helen Huls, Charuta Kale, Hiu Liu, Christopher Toth, Laurence J.N. Cooper, Sourindra Maiti, Laszlo Radvanyi, Patrick Hwu, Ena Wang, Chantale Bernatchez, Marie-Andree Forget, and Shruti Malu
- Subjects
Pharmacology ,Cancer Research ,Metastatic melanoma ,Tumor-infiltrating lymphocytes ,Melanoma ,Immunology ,Salvage therapy ,hemic and immune systems ,chemical and pharmacologic phenomena ,Biology ,medicine.disease ,Genetically modified organism ,Cell therapy ,Artificial antigen presenting cells ,Oncology ,Poster Presentation ,medicine ,Molecular Medicine ,Immunology and Allergy ,Autologous tumor - Abstract
Meeting abstracts Adoptive cell therapy (ACT) with expanded autologous tumor infiltrating lymphocytes (TIL) has emerged to be a powerful salvage therapy for metastatic melanoma with response rates up to 50%. However, the generation of the TIL ACT product is technically challenging with current
- Published
- 2013
39. The Homeobox Gene
- Author
-
J.Suzanne Lindsey, Ron P. Nhim, Miles F. Wilkinson, Shulin Li, Jessica Doskow, and Sourindra Maiti
- Subjects
Messenger RNA ,Exon ,Alternative splicing ,Intron ,Homeobox ,Promoter ,Cell Biology ,Biology ,Molecular Biology ,Biochemistry ,Transcription factor ,Gene ,Molecular biology - Abstract
The Pem gene encodes an atypical homeodomain protein, distantly related to Prd/Pax family members, that we demonstrate is regulated in a complex transcriptional and post-transcriptional manner. We show that the rat Pem genomic structure includes three 5′-untranslated (5′-UT) exons and four coding exons, three of which encode the homeodomain. Several alternatively spliced transcripts were identified, including one that skips an internal coding exon, enabling this mRNA to express a novel form of the Pem protein. Other alternatively spliced mRNAs were characterized that possess different 5′-UT regions, including a muscle-specific transcript. The different 5′-UT termini present in Pem transcripts conferred different levels of translatability in vitro. Two promoters containing multiple transcription initiation sites were identified: a distal promoter (Pd) in the first 5′-UT exon and a proximal promoter (Pp) located in the “intron” upstream of the first coding exon. The Pd was active in placenta, ovary, tumor cell lines, and to a lesser extent in skeletal muscle. In contrast, transcripts from the Pp were only detectable in testis and epididymis and were only expressed in epididymis in the presence of testosterone. To our knowledge no transcription factors have previously been identified that exhibit androgen-dependent expression in the epididymis.
- Published
- 1996
40. ThePemHomeobox Gene: Rapid Evolution of the Homeodomain, X Chromosomal Localization, and Expression in Reproductive Tissue
- Author
-
Karin Klinga Levan, Jessica Doskow, Sourindra Maiti, J.Suzanne Lindsey, David A. Lawlor, Ronald P. Nhim, Keith A. Sutton, and Miles F. Wilkinson
- Subjects
X Chromosome ,Molecular Sequence Data ,EMX2 ,Homeobox A1 ,Biology ,Polymerase Chain Reaction ,Cell Line ,Homeobox protein Nkx-2.5 ,NKX2-3 ,Mice ,Gene mapping ,Sequence Homology, Nucleic Acid ,Consensus Sequence ,Genetics ,Animals ,Humans ,Amino Acid Sequence ,Cloning, Molecular ,Conserved Sequence ,DNA Primers ,Homeodomain Proteins ,Regulation of gene expression ,Mice, Inbred BALB C ,Genome ,Base Sequence ,Sequence Homology, Amino Acid ,Genes, Homeobox ,Chromosome Mapping ,Exons ,Introns ,Recombinant Proteins ,Rats ,DNA-Binding Proteins ,Alternative Splicing ,Homeobox ,Homeotic gene ,Pseudogenes ,Transcription Factors - Abstract
A hallmark of homeobox genes is their high degree of sequence conservation in distantly related species. Here, we report the chromosomal localization, sequence, and expression pattern of an orphan homeobox gene, Pem, that encodes a homeodomain (HD) that has undergone a surprisingly high rate of evolutionary change. The N-terminal portion of the Pem HD, which includes the first two alpha-helices, exhibits only 44% sequence identity between rat Pem (r.Pem) and mouse Pem (m.Pem). This N-terminal subdomain exhibited an extremely high frequency of nonsynonymous substitutions, severalfold higher than other regions of the Pem protein. In contrast, the third helix, which is known to confer most of the base-specific contacts of HDs with DNA, was almost identical in r. Pem and m.Pem. Several lines of evidence suggested that the rat and mouse genes that we identified as Pem genes are true homologues: (1) the r.Pem and m.Pem genes both reside on the X chromosome; (2) they possess identical exon/intron splice junctions; (3) they both encode a distinctive motif upstream of the HD that is unique to Pem; and (4) the only m.Pem-like gene we were able to identify in the rat genome other than r.Pem was a pseudogene, r.Pem-ps, whose sequence and chromosomal localization indicated that it was derived by reverse transcription and reinsertion into the genome. The functional r.Pem gene is selectively expressed in placenta, testis, epididymis, and ovary. This expression pattern is of interest since other genes transcribed in reproductive tissue have also been shown to undergo high rates of sequence divergence. The high rate of amino acid substitutions in the N-terminal region of the Pem HD suggests the possibility of species-specific directional selection.
- Published
- 1996
41. TMIC-04. GLIOBLASTOMA-ASSOCIATED MYELOID CELLS DISPLAY NONPOLARIZED M0 MACROPHAGE PHENOTYPE
- Author
-
Nasser K. Yaghi, Luke M. Healy, Konrad Gabrusiewicz, Janet M. Bruner, Neal Huang, Yuuri Hashimoto, Brandon D. Liebelt, Benjamin Rodriguez, Sourindra Maiti, Laurence J.N. Cooper, Ravesanker Ezhilarasan, Ginu Thomas, Michael A. Curran, Ganesh Rao, Krishna P. Bhat, Jack P. Antel, Sujit S. Prabhu, Jun Wei, Rivka R. Colen, Shouhao Zhou, Amy B. Heimberger, Erik P. Sulman, Qianghu Wang, Jeffrey S. Weinberg, Wei Li, Amit Bar-Or, Ahmed Elakkad, Gregory N. Fuller, Raymond Sawaya, and Lauren A. Langford
- Subjects
Cancer Research ,Oncology ,Myeloid cells ,medicine ,Cancer research ,Macrophage ,Neurology (clinical) ,Biology ,medicine.disease ,Phenotype ,Glioblastoma - Published
- 2016
42. Redirecting Specificity of T cells Using the Sleeping Beauty System to Express Chimeric Antigen Receptors by Mix-and-Matching of VL and VH Domains Targeting CD123+ Tumors
- Author
-
Drew C. Deniger, Laurence J.N. Cooper, Tamara Laskowski, Sourindra Maiti, Radhika Thokala, Harjeet Singh, Tiejuan Mi, Hiroki Torikai, Simon Olivares, Richard E. Champlin, George McNamara, and Helen Huls
- Subjects
Cytotoxicity, Immunologic ,0301 basic medicine ,Myeloid ,T-Lymphocytes ,medicine.medical_treatment ,Cancer Treatment ,lcsh:Medicine ,Gene Expression ,Mice, SCID ,Immunotherapy, Adoptive ,Memory T cells ,Hematologic Cancers and Related Disorders ,White Blood Cells ,Mice ,Spectrum Analysis Techniques ,0302 clinical medicine ,Cancer immunotherapy ,Animal Cells ,Mice, Inbred NOD ,Basic Cancer Research ,Medicine and Health Sciences ,Cytotoxic T cell ,Molecular Targeted Therapy ,lcsh:Science ,B-Lymphocytes ,Multidisciplinary ,biology ,T Cells ,CD28 ,Animal Models ,Hematology ,Precursor Cell Lymphoblastic Leukemia-Lymphoma ,Myeloid Leukemia ,Flow Cytometry ,Caspase 9 ,3. Good health ,Leukemia, Myeloid, Acute ,medicine.anatomical_structure ,Oncology ,Spectrophotometry ,030220 oncology & carcinogenesis ,Cytophotometry ,Immunotherapy ,Cellular Types ,Genetic Engineering ,Research Article ,Plasmids ,Acute Myeloid Leukemia ,Immune Cells ,Recombinant Fusion Proteins ,Immunology ,Interleukin-3 Receptor alpha Subunit ,Mouse Models ,Research and Analysis Methods ,Transfection ,Cancer Immunotherapy ,CD19 ,Tumor Necrosis Factor Receptor Superfamily, Member 9 ,03 medical and health sciences ,Model Organisms ,CD28 Antigens ,Antigen ,Leukemias ,medicine ,Animals ,Humans ,Blood Cells ,lcsh:R ,Biology and Life Sciences ,Cancers and Neoplasms ,Cell Biology ,Single-Domain Antibodies ,Hematopoietic Stem Cells ,Chimeric antigen receptor ,Disease Models, Animal ,030104 developmental biology ,Cancer research ,biology.protein ,lcsh:Q ,Clinical Immunology ,Clinical Medicine - Abstract
Adoptive immunotherapy infusing T cells with engineered specificity for CD19 expressed on B- cell malignancies is generating enthusiasm to extend this approach to other hematological malignancies, such as acute myelogenous leukemia (AML). CD123, or interleukin 3 receptor alpha, is overexpressed on most AML and some lymphoid malignancies, such as acute lymphocytic leukemia (ALL), and has been an effective target for T cells expressing chimeric antigen receptors (CARs). The prototypical CAR encodes a VH and VL from one monoclonal antibody (mAb), coupled to a transmembrane domain and one or more cytoplasmic signaling domains. Previous studies showed that treatment of an experimental AML model with CD123-specific CAR T cells was therapeutic, but at the cost of impaired myelopoiesis, highlighting the need for systems to define the antigen threshold for CAR recognition. Here, we show that CARs can be engineered using VH and VL chains derived from different CD123-specific mAbs to generate a panel of CAR+ T cells. While all CARs exhibited specificity to CD123, one VH and VL combination had reduced lysis of normal hematopoietic stem cells. This CAR's in vivo anti-tumor activity was similar whether signaling occurred via chimeric CD28 or CD137, prolonging survival in both AML and ALL models. Co-expression of inducible caspase 9 eliminated CAR+ T cells. These data help support the use of CD123-specific CARs for treatment of CD123+ hematologic malignancies.
- Published
- 2016
43. Unassisted Production of Clinical-Grade Viral-Specific T Cells: Bringing Production to the Bedside
- Author
-
Pappanaicken R Kumar, Dimitrios P. Kontoyiannis, Richard E. Champlin, Laurence J.N. Cooper, Matthew J. Figliola, N. Kaltz, Priti Tewari, Anne Richter, Sourindra Maiti, and M. H. Huls
- Subjects
Oncology ,Cancer Research ,Transplantation ,medicine.medical_specialty ,business.industry ,Immunology ,Clinical grade ,Cell Biology ,Internal medicine ,medicine ,Immunology and Allergy ,Production (economics) ,business ,Genetics (clinical) - Published
- 2016
44. Erratum: Functional proteomics identifies miRNAs to target a p27/Myc/phospho-Rb signature in breast and ovarian cancer
- Author
-
R. Azencot, Chad V. Pecot, Rajesha Roopaimoole, Sourindra Maiti, Zijun Luo, Elena G. Seviour, Anil K. Sood, Joe W. Gray, Fanmao Zhang, Youtao Lu, Steven M. Hill, Prahlad T. Ram, Gordon B. Mills, Gabriel Lopez-Berestein, Cristian Rodriguez-Aguayo, Laurence J.N. Cooper, Sach Mukherjee, Wenbin Liu, Vasudha Sehgal, and Tyler J. Moss
- Subjects
0301 basic medicine ,Cancer Research ,Oncogene ,Biology ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,Functional proteomics ,microRNA ,Genetics ,medicine ,Cancer research ,Ovarian cancer ,Molecular Biology - Published
- 2016
45. Abstract B028: Transcriptional and epigenetic signatures of ex vivo propagated three distinct TCR Vδ1, TCR Vδ2 and TCR Vδ3 cell subtypes with broad specificity for malignancies
- Author
-
Jianliang Dai, Laurence J.N. Cooper, Drew C. Deniger, and Sourindra Maiti
- Subjects
Cancer Research ,medicine.medical_treatment ,T cell ,Immunology ,T-cell receptor ,Immunotherapy ,Biology ,Major histocompatibility complex ,Artificial antigen presenting cells ,medicine.anatomical_structure ,Immune system ,Cancer immunotherapy ,medicine ,biology.protein ,Cytotoxic T cell - Abstract
Functionally pleiotropic human γδ T cells are a unique set of immune cells that exhibit qualities of both innate and adaptive immune responses. In contrast to αβ T cells, γδ T cells recognize their ligands independent of MHC and are infrequent (1%–5% of T cells) in peripheral blood. Studies have shown that γδ T cells exhibit endogenous cytotoxicity towards tumor cells by directly recognizing a diverse array of Tumor Associated Antigen (TAA) and have the ability to present TAA to elicit an antitumor response. This broad recognition of antitumor activity is achieved because these cells express a diverse TCR γδ repertoire (combination of Vδ1, Vδ2, or Vδ3 with one of fourteen Vγ chains). Because of their characteristics γδ T cells are attractive targets for clinical manipulation and tumor immunotherapy. To obtain clinically significant numbers of cells for immunotherapy current ex vivo γδ T cells expansion protocol uses Zoledronic acid, an aminobisphosphonate that results only in selective propagation of Vγ9Vδ2. These cells however, showed clinical response against both solid and hematologic tumors. We have previously shown a novel ex vivo expansion protocol using artificial antigen presenting cells (aAPC), IL2 and IL21 assist aAPC in activating and expanding polyclonal γδ T-cells (Vδ1, Vδ2 and Vδ1neg Vδ2neg T cell subtypes) to clinically significant numbers. These expanded cell subsets secrete proinflammatory cytokines, lysed a broad range of malignancies and improved survival in ovarian cancer xenograft model. To better define the role of the subsets and their application in immunotherapy here we present high-resolution transcriptome, microRNA and secreted cytokine-chemokine signatures of ex vivo expanded subsets. Our data provide an atlas of transcriptional, epigenetic and secretome of three defined subsets, and subset-specific signatures. The data presented here will have a broader impact in understanding the immunobiology of γδ T cell subsets and facilitate improving the human application of these cells in cancer immunotherapy. Citation Format: Sourindra Maiti, Drew Deniger, Jianliang Dai, Laurence Cooper. Transcriptional and epigenetic signatures of ex vivo propagated three distinct TCR Vδ1, TCR Vδ2 and TCR Vδ3 cell subtypes with broad specificity for malignancies. [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr B028.
- Published
- 2016
46. Abstract A193: Bioengineered Dectin-1 CAR+ T cells to control invasive fungal infection
- Author
-
Tiejuan Mi, Sourindra Maiti, Simon Olivares, Helen Huls, Dimitrios P. Kontoyiannis, Richard E. Champlin, Laurence J.N. Cooper, Pappanaicken R. Kumaresan, Harjeet Singh, and Nathaniel D. Albert
- Subjects
0301 basic medicine ,Cancer Research ,Immunology ,CD28 ,Biology ,Natural killer T cell ,03 medical and health sciences ,Interleukin 21 ,030104 developmental biology ,Immune system ,Granzyme ,Cancer research ,biology.protein ,Cytotoxic T cell ,IL-2 receptor ,Interleukin 3 - Abstract
Myelosuppressive and ablative regimens are used to deplete lymphocytes in some patients before adoptive cell therapy. CD19-specific engineered T-cells have been used successfully to eliminate B-cells in patients with B-cell leukemias and lymphomas. Prolonged immunosuppression, including B-cell aplasia is associated with increased risk of invasive fungal infection (IFI). Thus, patients will benefit if bio-engineered T cells can control IFI as well as kill tumor cells. The Sleeping Beauty gene transfer system was used to render T cells to express a chimeric antigen receptor (CAR) on the cell surface to redirect specificity via Dectin-1. Upon binding with the β-1,3-glucan sugar moiety found in IFI, Dectin-1 CAR+ T cells signal via chimeric CD28 and CD3-ζ signaling domain. The activated T cells secrete perforin, granzyme and granulysin to damage hyphae and thus inhibit the hyphal growth of Aspergillus and Candida. The T cells also secrete IFN-γ to activate other immune cells to target IFI. To ready the Dectin-1 CAR-modified T cells for the human application, we have modified the Fc domain of the extracellular stalk to alleviate deleterious uptake by Fcγ receptors and thus in vivo loss of infused T cells. We have shown that the Dectin-1 CAR+ T cells do not kill the yeast form of Candida so normal commensals in the gut microbiota should not be affected. We have also demonstrated that Dectin-1 CAR+ T cells can control Aspergillus infection in the presence of immunosuppressive drugs at physiological concentrations. Finally, we have generated dual CAR+ T cells by co-expressing a CD19-specific CAR with Dectin-1 CAR rendering the resultant genetically modified T cells able to target both B-cell malignant cells as well as fungal hyphae. These dual CAR-T cells can thus control both leukemia and invasive fungal infections. These data provide a path forward for clinical trials testing Dectin-1 CAR+ T cells. Citation Format: Pappanaicken R. Kumaresan, Nathaniel Albert, Harjeet Singh, Simon Olivares, Sourindra N. Maiti, Tiejuan Mi, Helen Huls, Richard E. Champlin, Dimitrios P. Kontoyiannis, Laurence J.N. Cooper. Bioengineered Dectin-1 CAR+ T cells to control invasive fungal infection . [abstract]. In: Proceedings of the CRI-CIMT-EATI-AACR Inaugural International Cancer Immunotherapy Conference: Translating Science into Survival; September 16-19, 2015; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2016;4(1 Suppl):Abstract nr A193.
- Published
- 2016
47. A new approach to simultaneously quantify both TCR α- and β-chain diversity after adoptive immunotherapy
- Author
-
Richard E. Champlin, Sourindra Maiti, Chantale Bernatchez, Helen Huls, Patrick Hwu, Minying Zhang, Laszlo Radvanyi, Luis M Vence, Laurence J.N. Cooper, and Brian Rabinovich
- Subjects
Cancer Research ,Cellular immunity ,Melanoma ,medicine.medical_treatment ,T cell ,Receptors, Antigen, T-Cell, alpha-beta ,T-cell receptor ,Immunotherapy ,Biology ,medicine.disease ,Immunotherapy, Adoptive ,Biomarkers, Pharmacological ,Article ,Immune system ,medicine.anatomical_structure ,Lymphocytes, Tumor-Infiltrating ,Oncology ,Antigen ,Immunology ,medicine ,Humans ,RNA ,Gene - Abstract
Purpose: T-cell receptor (TCR) variable Vα and Vβ gene diversity is a surrogate biomarker for the therapeutic potential of adoptive immunotherapy and cellular immunity. Therefore, creating a straightforward, rapid, sensitive, and reliable method to view the global changes of both TCRVα and Vβ transcripts in heterogeneous populations of T cells is appealing. Experimental Design: We designed a “direct TCR expression assay” (DTEA) using a panel of customized bar-coded probes that simultaneously detects and quantifies 45 Vα and 46 Vβ transcripts in a nonenzymatic digital multiplexed assay from a small number of cells (104 cells) or as little as 100 ng of total RNA. Results: We evaluated DTEA on total RNA samples of tumor-infiltrating lymphocytes and peripheral blood obtained from patients with melanoma after adoptive T-cell therapy. DTEA detected a similar spectrum of the dominant patterns of TCRVβ gene usage as sequencing cloned TCRVβ CDR3 regions. However, DTEA was rapid, achieved a level of sensitivity to identify rare T-cell populations, and simultaneously tracked the full array of Vα and Vβ transcripts. Conclusions: DTEA can rapidly and sensitively track changes in TCRVα and Vβ gene usages in T-cell pools following immune interventions, such as adoptive T-cell transfer, and may also be used to assess impact of vaccination or reconstitution of T-cell compartment after hematopoietic stem cell transplantation. Clin Cancer Res; 18(17); 4733–42. ©2012 AACR.
- Published
- 2012
48. Adoptive T-cell therapy improves treatment of canine non–Hodgkin lymphoma post chemotherapy
- Author
-
Helen Huls, Richard E. Champlin, R. Eric Davis, Mark C. Johnson, Heather L. Wilson, Shana L. Palla, Laurence J.N. Cooper, Wencai Ma, Colleen M. O'Connor, Dean A. Lee, Suzanne Craig, Cassie A. Hartline, Sourindra Maiti, and Sabina Sheppard
- Subjects
Vincristine ,Adoptive cell transfer ,Cyclophosphamide ,040301 veterinary sciences ,T-Lymphocytes ,medicine.medical_treatment ,T cell ,Gene Expression ,CHOP ,Article ,Immunophenotyping ,0403 veterinary science ,03 medical and health sciences ,Dogs ,Artificial antigen presenting cells ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Dog Diseases ,030304 developmental biology ,0303 health sciences ,Multidisciplinary ,business.industry ,Lymphoma, Non-Hodgkin ,04 agricultural and veterinary sciences ,Immunotherapy ,medicine.disease ,Adoptive Transfer ,Combined Modality Therapy ,3. Good health ,Lymphoma ,medicine.anatomical_structure ,Doxorubicin ,Immunology ,Cancer research ,Prednisone ,business ,medicine.drug - Abstract
Clinical observations reveal that an augmented pace of T-cell recovery after chemotherapy correlates with improved tumor-free survival, suggesting the add-back of T cells after chemotherapy may improve outcomes. To evaluate adoptive immunotherapy treatment for B-lineage non-Hodgkin lymphoma (NHL), we expanded T cells from client-owned canines diagnosed with NHL on artificial antigen presenting cells (aAPC) in the presence of human interleukin (IL)-2 and IL-21. Graded doses of autologous T cells were infused after CHOP chemotherapy and persisted for 49 days, homed to tumor, and significantly improved survival. Serum thymidine kinase changes predicted T-cell engraftment, while anti-tumor effects correlated with neutrophil-to-lymphocyte ratios and granzyme B expression in manufactured T cells. Therefore, chemotherapy can be used to modulate infused T-cell responses to enhance anti-tumor effects. The companion canine model has translational implications for human immunotherapy which can be readily exploited since clinical-grade canine and human T cells are propagated using identical approaches.
- Published
- 2012
49. Membrane-bound IL-21 promotes sustained ex vivo proliferation of human natural killer cells
- Author
-
Harjeet Singh, Vladimir Senyukov, Sourindra Maiti, Dean A. Lee, Jennifer L. Johnson, Richard E. Champlin, Laurence J.N. Cooper, Lenka V. Hurton, Cecele J. Denman, Srinivas S. Somanchi, Lisa M. Kopp, M. Helen Huls, and Prasad V. Phatarpekar
- Subjects
Anatomy and Physiology ,T-Lymphocytes ,Cancer Treatment ,lcsh:Medicine ,Immunotherapy, Adoptive ,Interleukin 21 ,0302 clinical medicine ,Receptors, KIR ,Immune Physiology ,Molecular Cell Biology ,lcsh:Science ,Cells, Cultured ,Interleukin-15 ,0303 health sciences ,Multidisciplinary ,Chromosome Biology ,U937 Cells ,Telomere ,Flow Cytometry ,3. Good health ,Cell biology ,Killer Cells, Natural ,Oncology ,Interleukin 15 ,Interleukin 12 ,Medicine ,Research Article ,Immune Cells ,Immunology ,Antigen-Presenting Cells ,Biology ,Cell Line ,Immunophenotyping ,03 medical and health sciences ,Cell Line, Tumor ,Humans ,Antigen-presenting cell ,030304 developmental biology ,Cell Proliferation ,Lymphokine-activated killer cell ,Gene Expression Profiling ,Interleukins ,lcsh:R ,Antibody-Dependent Cell Cytotoxicity ,Membrane Proteins ,Molecular Development ,Immunologic Subspecialties ,NKG2D ,Coculture Techniques ,Cell culture ,Immune System ,lcsh:Q ,Cytokine secretion ,Artificial Cells ,Clinical Immunology ,K562 Cells ,030215 immunology ,Developmental Biology - Abstract
NK cells have therapeutic potential for a wide variety of human malignancies. However, because NK cells expand poorly in vitro, have limited life spans in vivo, and represent a small fraction of peripheral white blood cells, obtaining sufficient cell numbers is the major obstacle for NK-cell immunotherapy. Genetically-engineered artificial antigen-presenting cells (aAPCs) expressing membrane-bound IL-15 (mbIL15) have been used to propagate clinical-grade NK cells for human trials of adoptive immunotherapy, but ex vivo proliferation has been limited by telomere shortening. We developed K562-based aAPCs with membrane-bound IL-21 (mbIL21) and assessed their ability to support human NK-cell proliferation. In contrast to mbIL15, mbIL21-expressing aAPCs promoted log-phase NK cell expansion without evidence of senescence for up to 6 weeks of culture. By day 21, parallel expansion of NK cells from 22 donors demonstrated a mean 47,967-fold expansion (median 31,747) when co-cultured with aAPCs expressing mbIL21 compared to 825-fold expansion (median 325) with mbIL15. Despite the significant increase in proliferation, mbIL21-expanded NK cells also showed a significant increase in telomere length compared to freshly obtained NK cells, suggesting a possible mechanism for their sustained proliferation. NK cells expanded with mbIL21 were similar in phenotype and cytotoxicity to those expanded with mbIL15, with retained donor KIR repertoires and high expression of NCRs, CD16, and NKG2D, but had superior cytokine secretion. The mbIL21-expanded NK cells showed increased transcription of the activating receptor CD160, but otherwise had remarkably similar mRNA expression profiles of the 96 genes assessed. mbIL21-expanded NK cells had significant cytotoxicity against all tumor cell lines tested, retained responsiveness to inhibitory KIR ligands, and demonstrated enhanced killing via antibody-dependent cell cytotoxicity. Thus, aAPCs expressing mbIL21 promote improved proliferation of human NK cells with longer telomeres and less senescence, supporting their clinical use in propagating NK cells for adoptive immunotherapy.
- Published
- 2011
50. Large scale generation of genetically modified T-cells using micro-electroporators for cancer treatments
- Author
-
Partow Kebriaei, Sourindra Maiti, Elizabeth J. Shpall, Yoonsu Choi, Richard E. Champlin, Laurence J.N. Cooper, Sibani Lisa Biswal, Sorin Lupascu, Robert M. Raphael, Dean A. Lee, Carrie Yuen, Helen Huls, Daniel Joshua Stark, and Thomas Killian
- Subjects
Messenger RNA ,Immune system ,Electroporation ,Genetic enhancement ,biology.protein ,medicine ,Biology ,medicine.disease_cause ,Molecular biology ,CD19 ,Chimeric antigen receptor ,Genotoxicity ,Genetically modified organism - Abstract
To address B-lineage acute lymphoblastic leukemia (B-ALL), we use a bioengineering approach to achieve high-efficient non-viral gene transfer of tumor-specific CAR+ T cells for on-demand use. Specifically, we have developed CD19-specific chimeric antigen receptors (CARs) to redirect the specificity of T cells for B-cell malignancies. We introduce CARs via a non-viral gene transfer: electroporation. We have fabricated and tested a series of high throughput micro-electroporation devices (HitMeDs) with the end result that a HitMeD can electroporate a large number (≫109) human T cells within short time period (≪4 hours). We connect the HitMeDs to a BTX pulse generator and a monitoring system. We have adapted HitMeDs to the electro-transfer of CAR mRNA which avoids the potential genotoxicity associated with vector integration. After electroporation, 70% to 80% of the T cells express CD19 -specific CAR.
- Published
- 2009
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.