16 research outputs found

    Immunotherapy Gone Viral: Bortezomib and oHSV Enhance Antitumor NK-Cell Activity

    No full text

    Advances and Hurdles in CAR T Cell Immune Therapy for Solid Tumors

    Get PDF
    Chimeric antigen receptor (CAR) T cells in solid tumors have so far yielded limited results, in terms of therapeutic effects, as compared to the dramatic results observed for hematological malignancies. Many factors involve both the tumor cells and the microenvironment. The lack of specific target antigens and severe, potentially fatal, toxicities caused by on-target off-tumor toxicities constitute major hurdles. Furthermore, the tumor microenvironment is usually characterized by chronic inflammation, the presence of immunosuppressive molecules, and immune cells that can reduce CAR T cell efficacy and facilitate antigen escape. Nonetheless, solid tumors are under investigation as possible targets despite their complexity, which represents a significant challenge. In preclinical mouse models, CAR T cells are able to efficiently recognize and kill several tumor xenografts. Overall, in the next few years, there will be intensive research into optimizing novel cell therapies to improve their effector functions and keep untoward effects in check. In this review, we provide an update on the state-of-the-art CAR T cell therapies in solid tumors, focusing on the preclinical studies and preliminary clinical findings aimed at developing optimal strategies to reduce toxicity and improve efficacy

    EGFRvIII-specific chimeric antigen receptor T cells migrate to and kill tumor deposits infiltrating the brain parenchyma in an invasive xenograft model of glioblastoma.

    No full text
    Glioblastoma (GBM) is the most common primary malignant brain tumor in adults and is uniformly lethal. T-cell-based immunotherapy offers a promising platform for treatment given its potential to specifically target tumor tissue while sparing the normal brain. However, the diffuse and infiltrative nature of these tumors in the brain parenchyma may pose an exceptional hurdle to successful immunotherapy in patients. Areas of invasive tumor are thought to reside behind an intact blood brain barrier, isolating them from effective immunosurveillance and thereby predisposing the development of "immunologically silent" tumor peninsulas. Therefore, it remains unclear if adoptively transferred T cells can migrate to and mediate regression in areas of invasive GBM. One barrier has been the lack of a preclinical mouse model that accurately recapitulates the growth patterns of human GBM in vivo. Here, we demonstrate that D-270 MG xenografts exhibit the classical features of GBM and produce the diffuse and invasive tumors seen in patients. Using this model, we designed experiments to assess whether T cells expressing third-generation chimeric antigen receptors (CARs) targeting the tumor-specific mutation of the epidermal growth factor receptor, EGFRvIII, would localize to and treat invasive intracerebral GBM. EGFRvIII-targeted CAR (EGFRvIII+ CAR) T cells demonstrated in vitro EGFRvIII antigen-specific recognition and reactivity to the D-270 MG cell line, which naturally expresses EGFRvIII. Moreover, when administered systemically, EGFRvIII+ CAR T cells localized to areas of invasive tumor, suppressed tumor growth, and enhanced survival of mice with established intracranial D-270 MG tumors. Together, these data demonstrate that systemically administered T cells are capable of migrating to the invasive edges of GBM to mediate antitumor efficacy and tumor regression

    EGFRvIII<sup>+</sup> CAR T cells effectively migrate to invasive GBM tumors.

    No full text
    <p>1×10<sup>7</sup> extGLuc<sup>+</sup> EGFRvIII<sup>+</sup> CAR T cells were administered systemically to D-270MG<sup>FLuc/GFP</sup> tumor-bearing mice, and T-cell trafficking and/or accumulation near tumor was monitored using BLI on days 2 (<b>a</b>) and 9 (<b>b</b>). To assess the role of antigen-specificity on T-cell localization at the site of tumor, 1×10<sup>7</sup> extGLuc-only T cells were systemically administered to a separate group of tumor-bearing mice and monitored for trafficking and/or accumulation near tumor using BLI on day 7 (<b>c, d</b>). NSG mice treated with extGLuc<sup>+</sup> EGFRvIII<sup>+</sup> CAR T cells (<b>e</b>) or saline (<b>f</b>) were sacrificed on day 9, and brains were harvested, formalin-fixed, and paraffin-embedded. 5 μm coronal sections were immunostained with rabbit anti-human CD3 antibody and counterstained with hematoxylin. Images are representative of tumors obtained and analyzed from four mice (n = 4). Data represent one of two (n = 2) experiments with similar results.</p
    corecore