GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Science Translational Medicine, American Association for the Advancement of Science (AAAS), Vol. 13, No. 586 ( 2021-03-24)
    Abstract: A substantial number of patients with leukemia and lymphoma treated with anti-CD19 or anti-CD22 monoCAR-T cell therapy relapse because of antigen loss or down-regulation. We hypothesized that B cell tumor antigen escape may be overcome by a chimeric antigen receptor (CAR) design that simultaneously targets three B cell leukemia antigens. We engineered trispecific duoCAR-T cells with lentiviral vectors encoding two CAR open reading frames that target CD19, CD20, and CD22. The duoCARs were composed of a CAR with a tandem CD19- and CD20-targeting binder, linked by the P2A self-cleaving peptide to a second CAR targeting CD22. Multiple combinations of intracellular T cell signaling motifs were evaluated. The most potent duoCAR architectures included those with ICOS, OX40, or CD27 signaling domains rather than those from CD28 or 4-1BB. We identified four optimal binder and signaling combinations that potently rejected xenografted leukemia and lymphoma tumors in vivo. Moreover, in mice bearing a mixture of B cell lymphoma lines composed of parental triple-positive cells, CD19-negative, CD20-negative, and CD22-negative variants, only the trispecific duoCAR-T cells rapidly and efficiently rejected the tumors. Each of the monoCAR-T cells failed to prevent tumor progression. Analysis of intracellular signaling profiles demonstrates that the distinct signaling of the intracellular domains used may contribute to these differential effects. Multispecific duoCAR-T cells are a promising strategy to prevent antigen loss–mediated relapse or the down-regulation of target antigen in patients with B cell malignancies.
    Type of Medium: Online Resource
    ISSN: 1946-6234 , 1946-6242
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2021
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Frontiers Media SA ; 2022
    In:  Frontiers in Immunology Vol. 13 ( 2022-2-9)
    In: Frontiers in Immunology, Frontiers Media SA, Vol. 13 ( 2022-2-9)
    Abstract: CAR T-cell therapies targeting the B-cell maturation antigen eliminate tumors in relapsed/refractory multiple myeloma patients, however durable remissions remain difficult to attain. Transforming growth factor beta (TGF-β) is a multifunctional cytokine abundantly expressed in the multiple myeloma bone marrow niche, where it promotes an immunosuppressive tumor microenvironment. We hypothesized that BCMA CAR T-cells armored to resist the suppressive effects of TGF-β will provide an advantage in treating multiple myeloma. The armored B2ARM CAR T cells, co-expressing BCMA targeting CAR with TGF-β dominant-negative receptor II, were generated by lentiviral transduction of primary human CD4+ and CD8+ T cells. The B2ARM CAR T cells eliminated MM.1S multiple myeloma targets in long-term cytotoxicity assays, even under TGF-β-high conditions, whereas cytotoxic function of the non-armored B2 CAR -T cells was inhibited by TGF-β. Concordantly, after long-term exposure to targets in the presence of TGF-β, the B2ARM CAR T cells were enriched for Granzyme B, CD107a, Ki67 and polyfunctional cells T-cells (double or triple-positive for IFN-γ, IL-2 and/or TNF-α), as determined by flow cytometry. In addition, the B2ARM CAR T-cells, but not the conventional B2 CAR T-cells, resisted the TGF-β-mediated suppression of activation (CD25), exhaustion (PD-1, LAG3), and differentiation to T effectors (CD45RA+ CD45RO-CD62L-). In NSG mice bearing RPMI-8226 tumors overexpressing TGF-β, the B2ARM CAR mediated 100% tumor rejection and survival, superior infiltration of tumors on day 7 post CAR T treatment (%CD3+CAR+), and greater expression of IFN-γ, TNF-α, Ki67, Granzyme B, and PD-1, as compared to tumor-infiltrating non-armored B2 CAR T-cells. In NSG RPMI-8226 xenograft model in which tumors were additionally supplemented with TGF-β injections on days -1 through 11 of CAR T treatment, the B2ARM CAR T cells rejected tumors faster than the non-armored B2 CARs, and showed greater numbers of CD3+ and CD3+CAR+, central memory (CD45RO+CD62L+) and effector memory (CD45RO+CD62L-) T cells in the peripheral blood 18 days after treatment. In summary, the armored B2ARM CAR T cells mediate superior persistence, proliferation, multi-functionality, effector differentiation and anti-tumor function in pre-clinical models of multiple myeloma, while abrogating TGF-β-mediated suppression.
    Type of Medium: Online Resource
    ISSN: 1664-3224
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2022
    detail.hit.zdb_id: 2606827-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Frontiers Media SA ; 2018
    In:  Frontiers in Oncology Vol. 8 ( 2018-11-22)
    In: Frontiers in Oncology, Frontiers Media SA, Vol. 8 ( 2018-11-22)
    Type of Medium: Online Resource
    ISSN: 2234-943X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2018
    detail.hit.zdb_id: 2649216-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 37-38
    Abstract: The B-cell maturation antigen (BCMA) is an immunotherapy target selectively expressed on multiple myeloma cells (MM). Despite recent success of experimental BCMA CAR-T cell therapy, clinical remissions in MM are often short, in part due to low persistence of the BCMA CAR-T cells. Additionally, immunosuppressive factors, notably TGF-β, are known to be elevated in the peripheral blood and tumor microenvironment in the bone marrow of MM patients, potentially contributing to the lack of durability of CAR-T cell therapy. We aimed to develop a fully-human BCMA CAR with long-term persistence and functional resistance to the suppressive effects of TGF-β. Initially, two fully human single chain variable fragments (scFv) specific for BCMA, derived by biopanning of a yeast display human scFv library, were characterized in a CAR format. Each of the two scFvs was cloned into a lentiviral vector CAR backbone, comprised of the CD8 hinge and transmembrane domain, 4-1BB co-stimulatory domain and CD3ζ signaling domain, and termed BCMA1 and BCMA2, respectively. BCMA2 and BCMA1 T cells, generated by lentiviral vector transduction of primary human T cells, exhibited high CAR expression at multiplicities of infection 10 to 40 (BCMA1: 64-81%; BCMA2: 84-94%), and consistently demonstrated potent cytotoxicity in an overnight co-culture with BCMA+ MM cell lines RPMI-8226 and MM1.S, but not the BCMA- 293T cells. BCMA2 CAR showed robust proliferative capacity in response to repeated, long-term exposure to MM1.S target cells, concordant with the sustained CD4 T-cell subset and high IL-2 production during the course of repeated exposure to target cells. This is in contrast to BCMA1, that showed precipitous decrease in CD4+ T-cell subset upon co-culture with MM cells, resulting in a CAR T-cell population dominated by CD8+ T-cells. Furthermore, the BCMA2 demonstrated prolonged potency in clearing tumor cells compared to BCMA1, even after continuous 20-day exposure to target cells. This was further confirmed in a mouse intradermal RPMI-8226 xenograft tumor model, in which infusion of BCMA2 T-cells resulted in the rapid and complete eradication of tumors, while BCMA1 showed a slower decline in tumor burden. To further improve the efficacy of the BCMA2, specifically within the TGF-β-rich immunosuppressive tumor microenvironment, we developed an armored BCMA2 CAR variant, which co-expresses the dominant negative TGF-β RII bicistronically via a 2A sequence (BCMA2-TbnegCAR). In a 10 day-long co-culture assay with MM1.S targets in the presence of spiked 10 ng/ml TGF-β, BCMA2-TbnegCAR retained high proliferative capacity and potent cytotoxicity, while the unarmored BCMA2 had diminished proliferation and substantially reduced cytokine and granzyme B production. Consistently, in the in vivo intradermal tumor model that utilizes RPMI-8226, which is a MM cell line that endogenously produces TGF-β, we observed that BCMA2-TbnegCAR treatment resulted in higher T-cell counts in the tumors and an earlier decrease of tumor burden compared to infusion with BCMA2, further demonstrating the increased efficacy and potency of the BCMA2-TbnegCAR. In conclusion, we have designed and characterized a new fully-human BCMA2-TbnegCAR, with a novel scFv that exhibits robust proliferative capacity and persistent cytotoxicity, and remains functionally resistant to the immunosuppressive effects of TGF-β. This novel BCMA CAR can potentially improve the effectiveness and durability of the current BCMA CAR-T cell therapy. Disclosures Alabanza: Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties: CAR-T immunotherapy. Vu:Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties: CAR-T immunotherapy. Wu:Lentigen, a Miltenyi Biotec Company: Current Employment. Zhu:Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties: CAR-T immunotherapy. Dropulic:Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties: CAR-T immunotherapy. Schneider:Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2561-2561
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2561-2561
    Abstract: CD33 antigen is a promising target expressed on non-solid cancers, including acute myeloid leukemia (AML). Present investigative approaches to treatment of CD33-positive AML include antibody drug conjugates (My96, Mylotarg®) and CART cells incorporating CD33-targeting domains derived from a humanized scFv. Here, we designed a chimeric antigen receptor utilizing targeting domain derived from a fully human CD33 fragment variable heavy chain sequence, termed CAR33VH, and examined its in vitro and in vivo potency against AML. Primary human CD4+ CD8+ T cells derived from three healthy donors were transduced with lentiviral constructs (LV) encoding CAR33VH, or control CAR construct based on scFv My96 (My96CAR). Flow cytometric analysis revealed expression of CAR33VH at 32%-45%, and expression of My96CAR at 77% - 86%. When challenged with CD33+ AML tumor lines HL60 and MOLM-14 in vitro, both constructs demonstrated efficient target killing. CD33- tumor lines K562 and Reh were not sensitive to CAR killing, underscoring CAR specificity to CD33 antigen. Pro-inflammatory cytokines IFN gamma, TNF alpha and IL-2 in culture supernatants of CART cells incubated with CD33+ HL-60 and MOLM-14 tumors, but not with CD33- K562 cells overnight, were induced, as measured by ELISA. Long-term co-incubation assay of CART cells with HL-60 leukemia at E:T ratios 1:5 to 1: 0.04, suggested similar killing potency and persistence of CAR33VH to the positive control scFv-based CAR. In in vivo AML model, NSG mice engrafted with MOLM-14 cells stably expressing firefly luciferase, both CAR33VH, and My96CAR control were equally efficient in tumor elimination. In conclusion, CAR33VH, comprised of a fully human heavy-chain variable fragment only antigen binding domain, was efficient in tumor killing in vitro and in vivo, and may be used clinically for treatment of CD33+ hematologic malignancies. To our knowledge, this is one of the first instances demonstrating the feasibility of employing heavy chain only binder sequence in CART design. Citation Format: Dina Schneider, Ying Xiong, Weizao Chen, Zhongyu Zhu, Darong Wu, Jennifer Hwang, Dimiter S. Dimitrov, Boro Dropulic, Rimas J. Orentas. Fully human immunoglobulin heavy chain only-derived CD33 CAR for the treatment of acute myeloid leukemia [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 2561.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Blood, American Society of Hematology, Vol. 136, No. Supplement 1 ( 2020-11-5), p. 12-13
    Abstract: In the treatment of B cell leukemia, relapse due to antigen to loss or downregulation remains a major challenge. Tumor antigen escape may be mitigated by multi-targeting CAR T cells redirected to CD19 and CD22, and possessing a superior sensitivity to low-density antigens. Using lentiviral transduction of primary T cells, flow cytometry, cell-based assays, and xenograft mouse models, we systematically optimized 22-19 CAR architecture and co-stimulatory domains for best functionality. Fully-human tandem 22-19 CARs with co-stimulatory domains derived from 4-1BB, CD28, ICOS, OX40 or CD27, and hinge and transmembrane domains derived from CD8, CD28, or OX40 were evaluated. The tandem targeting ScFv domain orientation 22-19 was selected based on greater expression and cytotoxicity vs 19-22. All CARs achieved high T cell expression (mean 50-90%), and efficient dose-dependent killing of RajiCD19+CD22+, 293TCD19+, 293TCD22+, but not 293TCD19-CD22- target cells, and elaborated IL-2, IFN-γ, and TNF-a in antigen-dependent manner. CARs' potency in vitro varied by co-stimulatory domain: 4-1BB & lt; OX40, ICOS, CD27 & lt;CD28, 4-1BB-CD28. The 22-19 tandem CAR with hinge and transmembrane domains (H/TM) derived from CD8 (CD8/CD8) performed as well as the CAR with H/TM CD28/CD28 configuration, whereas configuration OX40/OX40 was superior to CD8/OX40. In RajiCD19+CD22+ mouse xenografts, the potency of tumor rejection by the 2nd generation tandem 22-19 CARs was also dependent on co-stimulatory domain, ranking 4-1 BB & lt;CD27, ICOS & lt;OX40 & lt;CD28. Tandem 3rd generation CAR 22-19, combining CD28 and 4-1BB co-stimulation, achieved the greatest anti-tumor effect. In antigen-heterogeneous Raji xenografts of a mixture of CD19-CD22+, CD19+CD22-, and CD19+CD22+ clones, the 22-19 tandem CARs with CD28, OX40 or ICOS co-stimulation mediated rapid and complete tumor rejection, whereas the conventional single-targeting CAR19 or CAR22 enabled tumor progression due to antigen escape. Low antigen density Raji clones were generated by CRISPR-Cas9-mediated disruption of both CD19 and CD22 expression, followed by lentiviral transduction to express a limited number of antigen molecules on the cell surface. When challenged with Raji CD22 low clone in vitro, 2nd generation tandem CARs with CD28 and ICOS co-stimulation, and the 3rd generation tandem CAR combining CD28 and 4-1BB co-stimulatory domains were more effective than 4-1BB-, CD27-, or OX40-containing tandem CARs. Against RajiCD19 low clone, CARs with CD27 and OX40 domains were more effective than CARs with 4-1BB, and CARs with ICOS or CD28 co-stimulation were the most potent. In summary, the fully-human tandem 22-19 CARs incorporating ICOS and CD28 co-stimulatory domains mitigate tumor antigen escape, exhibit robust anti-tumor function in pre-clinical models, enable superior lysis of CD22low and CD19low tumor clones, and may help improve clinical outcomes. Disclosures Hu: Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties: CAR-T immunotherapy. Xiong:Lentigen, a Miltenyi Biotec Company: Current Employment. Wu:Lentigen, a Miltenyi Biotec Company: Current Employment. Zhu:Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties: CAR-T immunotherapy. Dropulic:Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties: CAR-T immunotherapy. Schneider:Lentigen, a Miltenyi Biotec Company: Current Employment, Patents & Royalties.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2020
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...