11 results on '"Riddell SR"'
Search Results
2. CBFA2T3-GLIS2 model of pediatric acute megakaryoblastic leukemia identifies FOLR1 as a CAR T cell target.
- Author
-
Le Q, Hadland B, Smith JL, Leonti A, Huang BJ, Ries R, Hylkema TA, Castro S, Tang TT, McKay CN, Perkins L, Pardo L, Sarthy J, Beckman AK, Williams R, Idemmili R, Furlan S, Ishida T, Call L, Srivastava S, Loeb AM, Milano F, Imren S, Morris SM, Pakiam F, Olson JM, Loken MR, Brodersen L, Riddell SR, Tarlock K, Bernstein ID, Loeb KR, and Meshinchi S
- Subjects
- Animals, Child, Child, Preschool, Humans, Infant, Disease Models, Animal, T-Lymphocytes, Transcriptome, Xenograft Model Antitumor Assays, Folate Receptor 1 genetics, Folate Receptor 1 metabolism, Immunotherapy, Adoptive, Leukemia, Megakaryoblastic, Acute genetics, Oncogene Proteins, Fusion genetics, Oncogene Proteins, Fusion metabolism
- Abstract
The CBFA2T3-GLIS2 (C/G) fusion is a product of a cryptic translocation primarily seen in infants and early childhood and is associated with dismal outcome. Here, we demonstrate that the expression of the C/G oncogenic fusion protein promotes the transformation of human cord blood hematopoietic stem and progenitor cells (CB HSPCs) in an endothelial cell coculture system that recapitulates the transcriptome, morphology, and immunophenotype of C/G acute myeloid leukemia (AML) and induces highly aggressive leukemia in xenograft models. Interrogating the transcriptome of C/G-CB cells and primary C/G AML identified a library of C/G-fusion-specific genes that are potential targets for therapy. We developed chimeric antigen receptor (CAR) T cells directed against one of the targets, folate receptor α (FOLR1), and demonstrated their preclinical efficacy against C/G AML using in vitro and xenograft models. FOLR1 is also expressed in renal and pulmonary epithelium, raising concerns for toxicity that must be addressed for the clinical application of this therapy. Our findings underscore the role of the endothelial niche in promoting leukemic transformation of C/G-transduced CB HSPCs. Furthermore, this work has broad implications for studies of leukemogenesis applicable to a variety of oncogenic fusion-driven pediatric leukemias, providing a robust and tractable model system to characterize the molecular mechanisms of leukemogenesis and identify biomarkers for disease diagnosis and targets for therapy.
- Published
- 2022
- Full Text
- View/download PDF
3. A therapeutic cancer vaccine delivers antigens and adjuvants to lymphoid tissues using genetically modified T cells.
- Author
-
Veatch JR, Singhi N, Srivastava S, Szeto JL, Jesernig B, Stull SM, Fitzgibbon M, Sarvothama M, Yechan-Gunja S, James SE, and Riddell SR
- Subjects
- Adjuvants, Immunologic administration & dosage, Allografts, Animals, Antigen Presentation, Antigens, Neoplasm administration & dosage, Autografts, CD8-Positive T-Lymphocytes transplantation, Cancer Vaccines immunology, Cross Reactions immunology, Dendritic Cells immunology, Female, Humans, Immunologic Memory, Immunotherapy, Adoptive, Interleukin-12 immunology, Lymphoid Tissue immunology, Male, Melanoma, Experimental immunology, Melanoma, Experimental therapy, Mice, Mice, Inbred C57BL, Mice, Knockout, Translational Research, Biomedical, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines therapeutic use
- Abstract
Therapeutic vaccines that augment T cell responses to tumor antigens have been limited by poor potency in clinical trials. In contrast, the transfer of T cells modified with foreign transgenes frequently induces potent endogenous T cell responses to epitopes in the transgene product, and these responses are undesirable, because they lead to rejection of the transferred T cells. We sought to harness gene-modified T cells as a vaccine platform and developed cancer vaccines composed of autologous T cells modified with tumor antigens and additional adjuvant signals (Tvax). T cells expressing model antigens and a broad range of tumor neoantigens induced robust and durable T cell responses through cross-presentation of antigens by host DCs. Providing Tvax with signals such as CD80, CD137L, IFN-β, IL-12, GM-CSF, and FLT3L enhanced T cell priming. Coexpression of IL-12 and GM-CSF induced the strongest CD4+ and CD8+ T cell responses through complimentary effects on the recruitment and activation of DCs, mediated by autocrine IL-12 receptor signaling in the Tvax. Therapeutic vaccination with Tvax and adjuvants showed antitumor activity in subcutaneous and metastatic preclinical mouse models. Human T cells modified with neoantigens readily activated specific T cells derived from patients, providing a path for clinical translation of this therapeutic platform in cancer.
- Published
- 2021
- Full Text
- View/download PDF
4. Tumor-infiltrating BRAFV600E-specific CD4+ T cells correlated with complete clinical response in melanoma.
- Author
-
Veatch JR, Lee SM, Fitzgibbon M, Chow IT, Jesernig B, Schmitt T, Kong YY, Kargl J, Houghton AM, Thompson JA, McIntosh M, Kwok WW, and Riddell SR
- Subjects
- Amino Acid Substitution, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes pathology, Humans, Male, Middle Aged, Receptors, Chimeric Antigen genetics, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes pathology, CD4-Positive T-Lymphocytes transplantation, Immunotherapy, Adoptive, Melanoma genetics, Melanoma immunology, Melanoma pathology, Melanoma therapy, Mutation, Missense, Proto-Oncogene Proteins B-raf genetics, Proto-Oncogene Proteins B-raf immunology, Receptors, Chimeric Antigen immunology
- Abstract
T cells specific for neoantigens encoded by mutated genes in cancers are increasingly recognized as mediators of tumor destruction after immune checkpoint inhibitor therapy or adoptive cell transfer. Unfortunately, most neoantigens result from random mutations and are patient specific, and some cancers contain few mutations to serve as potential antigens. We describe a patient with stage IV acral melanoma who achieved a complete response following adoptive transfer of tumor-infiltrating lymphocytes (TILs). Tumor exome sequencing surprisingly revealed fewer than 30 nonsynonymous somatic mutations, including oncogenic BRAFV600E. Analysis of the specificity of TILs identified rare CD4+ T cells specific for BRAFV600E and diverse CD8+ T cells reactive to nonmutated self-antigens. These specificities increased in blood after TIL transfer and persisted long-term, suggesting they contributed to the effective antitumor immune response. Gene transfer of the BRAFV600E-specific T cell receptor (TCR) conferred recognition of class II MHC-positive cells expressing the BRAF mutation. Therapy with TCR-engineered BRAFV600E-specific CD4+ T cells may have direct antitumor effects and augment CD8+ T cell responses to self- and/or mutated tumor antigens in patients with BRAF-mutated cancers.
- Published
- 2018
- Full Text
- View/download PDF
5. Targeted antibody-mediated depletion of murine CD19 CAR T cells permanently reverses B cell aplasia.
- Author
-
Paszkiewicz PJ, Fräßle SP, Srivastava S, Sommermeyer D, Hudecek M, Drexler I, Sadelain M, Liu L, Jensen MC, Riddell SR, and Busch DH
- Subjects
- Agammaglobulinemia immunology, Agammaglobulinemia pathology, Animals, B-Lymphocytes pathology, Female, Mice, T-Lymphocytes pathology, Agammaglobulinemia drug therapy, Antigens, CD19 immunology, B-Lymphocytes immunology, Cetuximab pharmacology, Lymphocyte Depletion, T-Lymphocytes immunology
- Abstract
The adoptive transfer of T cells that have been genetically modified to express a CD19-specific chimeric antigen receptor (CAR) is effective for treating human B cell malignancies. However, the persistence of functional CD19 CAR T cells causes sustained depletion of endogenous CD19+ B cells and hypogammaglobulinemia. Thus, there is a need for a mechanism to ablate transferred T cells after tumor eradication is complete to allow recovery of normal B cells. Previously, we developed a truncated version of the epidermal growth factor receptor (EGFRt) that is coexpressed with the CAR on the T cell surface. Here, we show that targeting EGFRt with the IgG1 monoclonal antibody cetuximab eliminates CD19 CAR T cells both early and late after adoptive transfer in mice, resulting in complete and permanent recovery of normal functional B cells, without tumor relapse. EGFRt can be incorporated into many clinical applications to regulate the survival of gene-engineered cells. These results support the concept that EGFRt represents a promising approach to improve safety of cell-based therapies.
- Published
- 2016
- Full Text
- View/download PDF
6. CD19 CAR-T cells of defined CD4+:CD8+ composition in adult B cell ALL patients.
- Author
-
Turtle CJ, Hanafi LA, Berger C, Gooley TA, Cherian S, Hudecek M, Sommermeyer D, Melville K, Pender B, Budiarto TM, Robinson E, Steevens NN, Chaney C, Soma L, Chen X, Yeung C, Wood B, Li D, Cao J, Heimfeld S, Jensen MC, Riddell SR, and Maloney DG
- Subjects
- Adult, Aged, CD4-CD8 Ratio, Disease-Free Survival, Humans, Immunotherapy, Adoptive adverse effects, Lymphocyte Depletion methods, Middle Aged, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma pathology, T-Lymphocyte Subsets transplantation, Tumor Burden immunology, Young Adult, Immunotherapy, Adoptive methods, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma immunology, Precursor B-Cell Lymphoblastic Leukemia-Lymphoma therapy, Receptors, Antigen, T-Cell immunology, T-Lymphocyte Subsets immunology
- Abstract
Background: T cells that have been modified to express a CD19-specific chimeric antigen receptor (CAR) have antitumor activity in B cell malignancies; however, identification of the factors that determine toxicity and efficacy of these T cells has been challenging in prior studies in which phenotypically heterogeneous CAR-T cell products were prepared from unselected T cells., Methods: We conducted a clinical trial to evaluate CD19 CAR-T cells that were manufactured from defined CD4+ and CD8+ T cell subsets and administered in a defined CD4+:CD8+ composition to adults with B cell acute lymphoblastic leukemia after lymphodepletion chemotherapy., Results: The defined composition product was remarkably potent, as 27 of 29 patients (93%) achieved BM remission, as determined by flow cytometry. We established that high CAR-T cell doses and tumor burden increase the risks of severe cytokine release syndrome and neurotoxicity. Moreover, we identified serum biomarkers that allow testing of early intervention strategies in patients at the highest risk of toxicity. Risk-stratified CAR-T cell dosing based on BM disease burden decreased toxicity. CD8+ T cell-mediated anti-CAR transgene product immune responses developed after CAR-T cell infusion in some patients, limited CAR-T cell persistence, and increased relapse risk. Addition of fludarabine to the lymphodepletion regimen improved CAR-T cell persistence and disease-free survival., Conclusion: Immunotherapy with a CAR-T cell product of defined composition enabled identification of factors that correlated with CAR-T cell expansion, persistence, and toxicity and facilitated design of lymphodepletion and CAR-T cell dosing strategies that mitigated toxicity and improved disease-free survival., Trial Registration: ClinicalTrials.gov NCT01865617., Funding: R01-CA136551; Life Science Development Fund; Juno Therapeutics; Bezos Family Foundation.
- Published
- 2016
- Full Text
- View/download PDF
7. Outcomes of acute leukemia patients transplanted with naive T cell-depleted stem cell grafts.
- Author
-
Bleakley M, Heimfeld S, Loeb KR, Jones LA, Chaney C, Seropian S, Gooley TA, Sommermeyer F, Riddell SR, and Shlomchik WD
- Subjects
- Acute Disease, Adolescent, Adrenal Cortex Hormones therapeutic use, Adult, Animals, Chronic Disease, Disease-Free Survival, Female, Graft vs Host Disease drug therapy, Graft vs Host Disease etiology, Graft vs Host Disease prevention & control, Hematopoietic Stem Cell Transplantation adverse effects, Histocompatibility Testing, Humans, Kaplan-Meier Estimate, Leukemia immunology, Lymphocyte Depletion methods, Male, Mice, Middle Aged, Myelodysplastic Syndromes immunology, Myelodysplastic Syndromes therapy, T-Lymphocyte Subsets immunology, Tissue Donors, Transplantation Conditioning, Transplantation, Homologous, Young Adult, Hematopoietic Stem Cell Transplantation methods, Leukemia therapy
- Abstract
Background: Graft-versus-host disease (GVHD) is a major cause of morbidity and mortality following allogeneic hematopoietic stem cell transplantation (HCT). In mice, naive T cells (TN) cause more severe GVHD than memory T cells (TM). We hypothesized that selective depletion of TN from human allogeneic peripheral blood stem cell (PBSC) grafts would reduce GVHD and provide sufficient numbers of hematopoietic stem cells and TM to permit hematopoietic engraftment and the transfer of pathogen-specific T cells from donor to recipient, respectively., Methods: In a single-arm clinical trial, we transplanted 35 patients with high-risk leukemia with TN-depleted PBSC grafts following conditioning with total body irradiation, thiotepa, and fludarabine. GVHD prophylactic management was with tacrolimus immunosuppression alone. Subjects received CD34-selected PBSCs and a defined dose of TM purged of CD45RA+ TN. Primary and secondary objectives included engraftment, acute and chronic GVHD, and immune reconstitution., Results: All recipients of TN-depleted PBSCs engrafted. The incidence of acute GVHD was not reduced; however, GVHD in these patients was universally corticosteroid responsive. Chronic GVHD was remarkably infrequent (9%; median follow-up 932 days) compared with historical rates of approximately 50% with T cell-replete grafts. TM in the graft resulted in rapid T cell recovery and transfer of protective virus-specific immunity. Excessive rates of infection or relapse did not occur and overall survival was 78% at 2 years., Conclusion: Depletion of TN from stem cell allografts reduces the incidence of chronic GVHD, while preserving the transfer of functional T cell memory., Trial Registration: ClinicalTrials.gov (NCT 00914940).
- Published
- 2015
- Full Text
- View/download PDF
8. Adoptive transfer of effector CD8+ T cells derived from central memory cells establishes persistent T cell memory in primates.
- Author
-
Berger C, Jensen MC, Lansdorp PM, Gough M, Elliott C, and Riddell SR
- Subjects
- Animals, Cell Culture Techniques, Cell Survival immunology, Cells, Cultured, Macaca nemestrina, Neoplasms immunology, Neoplasms therapy, Time Factors, Adoptive Transfer, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes transplantation, Immunologic Memory
- Abstract
The adoptive transfer of antigen-specific T cells that have been expanded ex vivo is being actively pursued to treat infections and malignancy in humans. The T cell populations that are available for adoptive immunotherapy include both effector memory and central memory cells, and these differ in phenotype, function, and homing. The efficacy of adoptive immunotherapy requires that transferred T cells persist in vivo, but identifying T cells that can reproducibly survive in vivo after they have been numerically expanded by in vitro culture has proven difficult. Here we show that in macaques, antigen-specific CD8(+) T cell clones derived from central memory T cells, but not effector memory T cells, persisted long-term in vivo, reacquired phenotypic and functional properties of memory T cells, and occupied memory T cell niches. These results demonstrate that clonally derived CD8+ T cells isolated from central memory T cells are distinct from those derived from effector memory T cells and retain an intrinsic capacity that enables them to survive after adoptive transfer and revert to the memory cell pool. These results could have significant implications for the selection of T cells to expand or to engineer for adoptive immunotherapy of human infections or malignancy.
- Published
- 2008
- Full Text
- View/download PDF
9. Expression of cutaneous lymphocyte-associated antigen by CD8(+) T cells specific for a skin-tropic virus.
- Author
-
Koelle DM, Liu Z, McClurkan CM, Topp MS, Riddell SR, Pamer EG, Johnson AS, Wald A, and Corey L
- Subjects
- Animals, Antigens, Differentiation, T-Lymphocyte, Antigens, Neoplasm, CD8-Positive T-Lymphocytes classification, CHO Cells, Cell Line, Transformed, Cell Movement, Cells, Cultured, Cricetinae, Herpes Genitalis diagnosis, Herpes Genitalis pathology, Humans, Immunologic Memory, Immunophenotyping, Ligands, Lymphocyte Activation, Skin virology, CD8-Positive T-Lymphocytes immunology, Herpes Genitalis immunology, Herpesvirus 2, Human immunology, Membrane Glycoproteins metabolism, Skin immunology
- Abstract
Virus-specific CD8(+) T cells traffic to infected tissues to promote clearance of infection. We used herpes simplex virus type 2 (HSV-2) as a model system to investigate CD8(+) T cell trafficking to the skin in humans. Using human leukocyte antigen (HLA) class I tetramers, we observed that HSV-specific CD8(+) T cells in the peripheral blood expressed high levels of cutaneous lymphocyte-associated antigen (CLA). In contrast, CD8(+) T cells specific for non-skin-tropic herpesviruses lacked CLA expression. CLA-positive HSV-2-specific CD8(+) T cells had the characteristics of central memory cells, expressing CCR7, CD62L, and CD28, and they proliferated briskly in response to antigen. CLA is related to a functional E-selectin ligand, and both E-selectin and CLA-positive cells were detected in HSV-2-infected skin. HSV-2-specific T cells adhered to cells transfected with E-selectin. A higher proportion of HSV-specific CD8(+) T cells recovered from herpes lesions express CLA compared with blood, consistent with a role for CLA in skin homing. To our knowledge, this is the first report of expression of tissue-specific adhesion-associated molecules by virus-specific CD8(+) T cells. The evaluation of vaccines for skin and mucosal pathogens should include study of the induction of appropriate tissue-specific homing molecules.
- Published
- 2002
- Full Text
- View/download PDF
10. HIV-specific cytotoxic T lymphocytes traffic to lymph nodes and localize at sites of HIV replication and cell death.
- Author
-
Brodie SJ, Patterson BK, Lewinsohn DA, Diem K, Spach D, Greenberg PD, Riddell SR, and Corey L
- Subjects
- Adoptive Transfer, Base Sequence, Cell Death, Cell Movement, DNA Primers genetics, Gene Transfer Techniques, HIV genetics, HIV Seropositivity immunology, HIV Seropositivity pathology, HIV Seropositivity virology, Humans, In Situ Hybridization, Lymph Nodes immunology, Lymph Nodes pathology, Polymerase Chain Reaction, Virus Replication, HIV immunology, HIV physiology, T-Lymphocytes, Cytotoxic immunology
- Abstract
We have tracked the in vivo migration and have identified in vivo correlates of cytotoxic T-lymphocyte (CTL) activity in HIV-seropositive subjects infused with autologous gene-marked CD8(+) HIV-specific CTL. The number of circulating gene-marked CTL ranged from 1.6 to 3.5% shortly after infusion to less than 0.5% 2 weeks later. Gene-marked CTL were present in the lymph node at 4.5- to 11-fold excess and colocalized within parafollicular regions of the lymph node adjacent to cells expressing HIV tat fusion transcripts, a correlate of virus replication. The CTL clones expressed the CCR5 receptor and localized among HIV-infected cells expressing the ligands MIP-1alpha and MIP-1beta, CC-chemokines produced at sites of virus replication. Aggregates of apoptotic cells and cells expressing granzyme-B localized within these same sites. In contrast, lymph node sections from untreated HIV-seropositive subjects, all with significant viral burden (> 50,000 HIV RNA copies/mL plasma), showed no CC-chemokine expression and exhibited only sporadic and randomly distributed cells expressing granzymes and/or apoptotic cells. These studies show that the infused CTL specifically migrate to sites of HIV replication and retain their antigen-specific cytolytic potential. Moreover, these studies provide a methodology that will facilitate studies of both the magnitude and functional phenotype of Ag-specific CD8(+) T cells in vivo.
- Published
- 2000
- Full Text
- View/download PDF
11. Herpes simplex virus infection of human fibroblasts and keratinocytes inhibits recognition by cloned CD8+ cytotoxic T lymphocytes.
- Author
-
Koelle DM, Tigges MA, Burke RL, Symington FW, Riddell SR, Abbo H, and Corey L
- Subjects
- Animals, Cell Line, Dogs, Fibroblasts immunology, Humans, Kidney, Kinetics, Simplexvirus immunology, T-Lymphocyte Subsets immunology, Vero Cells, CD8 Antigens analysis, Cell Transformation, Viral immunology, Cytotoxicity, Immunologic, Keratinocytes immunology, Simplexvirus genetics, T-Lymphocytes, Cytotoxic immunology
- Abstract
CD8+ cytotoxic T lymphocytes (CTL) clones with specificity for herpes simplex virus (HSV) were derived from two donors with genital HSV-2 infection. These CTL clones specifically lysed HSV-infected autologous B lymphoblastoid cells, but not HSV-infected fibroblasts. Exogenous peptide loading sensitized both cell types to lysis by an HSV-specific CTL clone of known specificity. HSV infection rendered fibroblasts refractory to peptide sensitization. HSV infection also rendered fibroblasts and keratinocytes insensitive to lysis by allospecific CD8+ CTL clones. Lysis of B lymphoblastoid cells in this system was only slightly reduced by HSV infection. Reduction of fibroblast allospecific lysis was dose and time dependent and was blocked by acyclovir, indicating the involvement of a late HSV gene product. HSV caused a reduction of fibroblast cell surface HLA class I antigen, at least in part due to reduction of synthesis of heavy chain-beta 2 microglobulin heterodimers. These results suggest that HSV-induced blockade of antigen presentation by cutaneous cells to CD8+ CTL may be a mechanism by which HSV limits or evades the immune response of the host.
- Published
- 1993
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.