Your email was sent successfully. Check your inbox.

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

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. CT541A-CT541A
    Abstract: The combination of chimeric antigen receptor (CAR) T cell therapy, which delivers large numbers of tumor reactive T cells, and oncolytic viral therapy, causing activation of host immune responses, is an attractive approach for improving outcomes for patients with glioblastoma (GBM). Here we present data from two independent phase I clinical trials evaluating IL13Rα2-targeted CAR therapy (NCT02208362) and C134 oncolytic viral (OV) therapy (NCT03657576) for the treatment of recurrent GBM (rGBM), along with preclinical studies supporting the utility of combining these two therapies. For NCT02208362, locoregional delivery of IL13Rα2-targeted CAR T cells were evaluated in heavily pretreated patients with rGBM. Interrogating biomarkers of clinical response revealed that levels of intratumoral T cells prior to treatment were positively associated with overall survival; furthermore, two patients who achieved a complete response had the highest levels of intratumoral CD3+ T cells pre-therapy. These findings suggest that therapeutic strategies which increase endogenous immune infiltrates could augment CAR T cell mediated responses. For NCT03657576, intratumoral delivery of C134, a herpes simplex virus (HSV-1) that has been genetically engineered to safely replicate and kill glioma tumor cells, is also being evaluated for treatment of rGBM. We report findings from a patient treated intratumorally with 1 × 106 pfu of C134. At 6-7 weeks post treatment this patient had MRI changes that suggested possible recurrence or pseudoprogression, and therefore underwent resection with biopsy assessment. Evaluation of virus-treated areas showed increased immune infiltrates as compared to untreated tumor sites, suggesting that C134 activated host immune responses. These clinical findings provide the rationale for evaluating a combination therapy of C134 OV and IL13Rα2-CAR T cells to potentially reshape the tumor microenvironment (TME) and enhance CAR therapy. In orthotopic GBM models in nude mice, we show that co-treatment with the two agents gave no adverse reaction, and more notably pre-treatment with C134 re-shaped the TME by increasing immune cell infiltrates and enhanced the efficacy of sub-therapeutic doses of CAR T cell therapy delivered either intraventricularly or intratumorally. Ongoing preclinical studies aim to provide detailed phenotypic analysis, as well as a mechanistic understanding of this combination approach to support the potential benefit of a soon to be opened combination trial evaluating C134 and IL13Rα2-CAR T cells. In this clinical trial in patients with IL13Rα2+ rGBM and anaplastic astrocytoma, increasing doses of intratumorally administered C134 will be followed by dual intracranial intratumoral and intraventricular administration of IL13Rα2-targeted CAR T cell therapy. Citation Format: Christine E. Brown, Agata Xella, Jonathan C. Hibbard, Vanessa Salvary, Brenda Aguilar, Jamie Wagner, Bruce Dezube, Knut Niss, Lynn Bayless, James Edinger, Jianmei Leavenworth, Stephen J. Forman, Behnam Badie, James M. Markert, Kevin A. Cassady. Oncolytic viral reshaping of the tumor microenvironment to promote CAR T cell therapy for glioblastoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr CT541A.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A133-A133
    Abstract: Meditope is a small cyclic peptide that was identified to bind to cetuximab within the Fab region. The meditope binding site can be grafted onto any Fab framework, creating a platform to uniquely and specifically target monoclonal antibodies. Here we demonstrate that the meditope binding site can be grafted onto chimeric antigen receptors (CARs) and utilized to regulate and extend CAR T cell function. We demonstrate that the platform can be used to overcome key barriers to CAR T cell therapy, including T cell exhaustion and antigen escape. Methods Meditope-enabled CARs (meCARs) were generated by amino acid substitutions to create binding sites for meditope peptide (meP) within the Fab tumor targeting domain of the CAR. meCAR expression was validated by anti-Fc FITC or meP-Alexa 647 probes. In vitro and in vivo assays were performed and compared to standard scFv CAR T cells. For meCAR T cell proliferation and dual-targeting assays, the meditope peptide (meP) was conjugated to recombinant human IL15 fused to the CD215 sushi domain (meP-IL15:sushi) and anti-CD20 monoclonal antibody rituximab (meP-rituximab). Results We generated meCAR T cells targeting HER2, CD19 and HER1/3 and demonstrate the selective specific binding of the meditope peptide along with potent meCAR T cell effector function. We next demonstrated the utility of a meP-IL15:sushi for enhancing meCAR T cell proliferation in vitro and in vivo. Proliferation and persistence of meCAR T cells was dose dependent, establishing the ability to regulate CAR T cell expansion using the meditope platform. We also demonstrate the ability to redirect meCAR T cells tumor killing using meP-antibody adaptors. As proof-of-concept, meHER2-CAR T cells were redirected to target CD20+ Raji tumors, establishing the potential of the meditope platform to alter the CAR specificity and overcome tumor heterogeneity. Conclusions Our studies show the utility of the meCAR platform for overcoming key challenges for CAR T cell therapy by specifically regulating CAR T cell functionality. Specifically, the meP-IL15:sushi enhanced meCAR T cell persistence and proliferation following adoptive transfer in vivo and protects against T cell exhaustion. Further, meP-ritiuximab can redirect meCAR T cells to target CD20-tumors, showing the versatility of this platform to address the tumor antigen escape variants. Future studies are focused on conferring additional ‘add-on’ functionalities to meCAR T cells to potentiate the therapeutic effectiveness of CAR T cell therapy.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Proceedings of the National Academy of Sciences, Proceedings of the National Academy of Sciences, Vol. 119, No. 33 ( 2022-08-16)
    Abstract: IL13Rα2 is an attractive target due to its overexpression in a variety of cancers and rare expression in healthy tissue, motivating expansion of interleukin 13 (IL13)–based chimeric antigen receptor (CAR) T cell therapy from glioblastoma into systemic malignancies. IL13Rα1, the other binding partner of IL13, is ubiquitously expressed in healthy tissue, raising concerns about the therapeutic window of systemic administration. IL13 mutants with diminished binding affinity to IL13Rα1 were previously generated by structure-guided protein engineering. In this study, two such variants, termed C4 and D7, are characterized for their ability to mediate IL13Rα2-specific response as binding domains for CAR T cells. Despite IL13Rα1 and IL13Rα2 sharing similar binding interfaces on IL13, mutations to IL13 that decrease binding affinity for IL13Rα1 did not drastically change binding affinity for IL13Rα2. Micromolar affinity to IL13Rα1 was sufficient to pacify IL13-mutein CAR T cells in the presence of IL13Rα1-overexpressing cells in vitro. Interestingly, effector activity of D7 CAR T cells, but not C4 CAR T cells, was demonstrated when cocultured with IL13Rα1/IL4Rα-coexpressing cancer cells. While low-affinity interactions with IL13Rα1 did not result in observable toxicities in mice, in vivo biodistribution studies demonstrated that C4 and D7 CAR T cells were better able to traffic away from IL13Rα1+ lung tissue than were wild-type (WT) CAR T cells. These results demonstrate the utility of structure-guided engineering of ligand-based binding domains with appropriate selectivity while validating IL13-mutein CARs with improved selectivity for application to systemic IL13Rα2-expressing malignancies.
    Type of Medium: Online Resource
    ISSN: 0027-8424 , 1091-6490
    RVK:
    RVK:
    Language: English
    Publisher: Proceedings of the National Academy of Sciences
    Publication Date: 2022
    detail.hit.zdb_id: 209104-5
    detail.hit.zdb_id: 1461794-8
    SSG: 11
    SSG: 12
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages