31 research outputs found

    Nanobiopolymer for Direct Targeting and Inhibition of EGFR Expression in Triple Negative Breast Cancer

    Get PDF
    Treatment options for triple negative breast cancer (TNBC) are generally limited to cytotoxic chemotherapy. Recently, anti-epidermal growth factor receptor (EGFR) therapy has been introduced for TNBC patients. We engineered a novel nanobioconjugate based on a poly(β-L-malic acid) (PMLA) nanoplatform for TNBC treatment. The nanobioconjugate carries anti-tumor nucleosome-specific monoclonal antibody (mAb) 2C5 to target breast cancer cells, anti-mouse transferrin receptor (TfR) antibody for drug delivery through the host endothelial system, and Morpholino antisense oligonucleotide (AON) to inhibit EGFR synthesis. The nanobioconjugates variants were: (1) P (BioPolymer) with AON, 2C5 and anti-TfR for tumor endothelial and cancer cell targeting, and EGFR suppression (P/AON/2C5/TfR), and (2) P with AON and 2C5 (P/AON/2C5). Controls included (3) P with 2C5 but without AON (P/2C5), (4) PBS, and (5) P with PEG and leucine ester (LOEt) for endosomal escape (P/mPEG/LOEt). Drugs were injected intravenously to MDA-MB-468 TNBC bearing mice. Tissue accumulation of injected nanobioconjugates labeled with Alexa Fluor 680 was examined by Xenogen IVIS 200 (live imaging) and confocal microscopy of tissue sections. Levels of EGFR, phosphorylated and total Akt in tumor samples were detected by western blotting

    Loss of testosterone impairs anti-tumor neutrophil function.

    No full text

    Polymalic acid nanobioconjugate for simultaneous immunostimulation and inhibition of tumor growth in HER2/neu-positive breast cancer

    Get PDF
    Breast cancer remains the second leading cause of cancer death among women in the United States. Breast cancer prognosis is particularly poor in case of tumors overexpressing the oncoprotein HER2/neu. A new nanobioconjugate of the PolycefinTM family of anti-cancer drugs based on biodegradable and non-toxic polymalic acid (PMLA) was engineered for a multi-pronged attack on HER2/neu-positive breast cancer cells. An antibody-cytokine fusion protein consisting of the immunostimulatory cytokine interleukin-2 (IL-2) genetically fused to an antibody specific for human HER2/neu [anti-HER2/neu IgG3-(IL-2)] was covalently attached to the PMLA backbone to target HER2/neu expressing tumors and ensure the delivery of IL-2 to the tumor microenvironment. Antisense oligonucleotides (AON) were conjugated to the nanodrug to inhibit the expression of vascular tumor protein laminin-411 in order to block tumor angiogenesis. It is shown that the nanobioconjugate was capable of specifically binding human HER2/neu and retained the biological activity of IL-2. We also showed the uptake of the nanobioconjugate into HER2/neu-positive breast cancer cells and enhanced tumor targeting in vivo. The nanobioconjugate exhibited marked anti-tumor activity manifested by significantly longer animal survival and significantly increased anti-HER2/neu immune response in immunocompetent mice bearing D2F2/E2 murine mammary tumors that express human HER2/neu. The combination of laminin-411 AON and antibody-cytokine fusion protein on a single polymeric platform results in a new nanobioconjugate that can act against cancer cells through inhibition of tumor growth and angiogenesis and the orchestration of an immune response against the tumor. The present PolycefinTM variant may be a promising agent for treating HER2/neu expressing tumors and demonstrates the versatility of the PolycefinTM nanobioconjugate platform.Fil: Ding, Hui. Cedars Sinai Medical Center;Fil: Helguera, Gustavo Fernando. University of California at Los Angeles; Estados Unidos. Consejo Nacional de Investigaciones Científicas y Técnicas. Instituto de Biología y Medicina Experimental. Fundación de Instituto de Biología y Medicina Experimental. Instituto de Biología y Medicina Experimental; ArgentinaFil: Rodríguez, José A.. University of California at Los Angeles; Estados UnidosFil: Markman, Janet. Cedars Sinai Medical Center;Fil: Luria Pérez, Rosendo. University of California at Los Angeles; Estados UnidosFil: Gangalum, Pallavi. Cedars Sinai Medical Center;Fil: Portilla Arias, Jose. Cedars Sinai Medical Center;Fil: Inoue, Satoshi. Cedars Sinai Medical Center; . Saga University; JapónFil: Daniels Wells, Tracy R.. University of California at Los Angeles; Estados UnidosFil: Black, Keith. Cedars Sinai Medical Center;Fil: Holler, Eggehard. Cedars Sinai Medical Center;Fil: Penichet, Manuel L.. University of California at Los Angeles; Estados UnidosFil: Ljubimova, Julia Y.. Cedars Sinai Medical Center

    T-Cell-Intrinsic Receptor Interacting Protein 2 Regulates Pathogenic T Helper 17 Cell Differentiation.

    No full text
    Receptor interacting protein 2 (RIP2) plays a role in sensing intracellular pathogens, but its function in T cells is unclear. We show that RIP2 deficiency in CD4+ T cells resulted in chronic and severe interleukin-17A-mediated inflammation during Chlamydia pneumoniae lung infection, increased T helper 17 (Th17) cell formation in lungs of infected mice, accelerated atherosclerosis, and more severe experimental autoimmune encephalomyelitis. While RIP2 deficiency resulted in reduced conventional Th17 cell differentiation, it led to significantly enhanced differentiation of pathogenic (p)Th17 cells, which was dependent on RORα transcription factor and interleukin-1 but independent of nucleotide oligomerization domain (NOD) 1 and 2. Overexpression of RIP2 resulted in suppression of pTh17 cell differentiation, an effect mediated by its CARD domain, and phenocopied by a cell-permeable RIP2 CARD peptide. Our data suggest that RIP2 has a T cell-intrinsic role in determining the balance between homeostatic and pathogenic Th17 cell responses

    Blood–brain barrier permeable nano immunoconjugates induce local immune responses for glioma therapy

    No full text
    Brain glioma treatment with checkpoint inhibitor antibodies to cytotoxic T-lymphocyte-associated antigen 4 (a-CTLA-4) and programmed cell death-1 (a-PD-1) was largely unsuccessful due to their inability to cross blood-brain barrier (BBB). Here we describe targeted nanoscale immunoconjugates (NICs) on natural biopolymer scaffold, poly(ss-L-malic acid), with covalently attached a-CTLA-4 or a-PD-1 for systemic delivery across the BBB and activation of local brain anti-tumor immune response. NIC treatment of mice bearing intracranial GL261 glioblastoma (GBM) results in an increase of CD8+ T cells, NK cells and macrophages with a decrease of regulatory T cells (Tregs) in the brain tumor area. Survival of GBM-bearing mice treated with NIC combination is significantly longer compared to animals treated with single checkpoint inhibitor-bearing NICs or free a-CTLA-4 and a-PD-1. Our study demonstrates trans-BBB delivery of tumor-targeted polymer-conjugated checkpoint inhibitors as an effective GBM treatment via activation of both systemic and local privileged brain tumor immune response

    Significant accumulation of P/2C5 in MDA-MB-468 breast tumor <i>in vivo</i>.

    No full text
    <p>(3a), An MDA-MB-468 subcutaneous tumor-bearing mouse was administered with P/2C5 intravenously. 24 hours later, the animal showed drug distribution mostly in the tumor, as well as in kidney and liver (drug clearing organs). (3b), Other than in kidney and liver, the drug was seen exclusively in the tumor.</p

    Tumor inhibition in mouse model, and effects on EGFR expression and Akt phosphorylation.

    No full text
    <p>(5a) Tumor growth inhibition in mice. MDA-MB-468 subcutaneous breast tumors treated systemically with P/AON/2C5, or P/AON/2C5/TfR were significantly inhibited compared with PBS (control), P(polymer only), or P/2C5 (without anti-tumor component) (P<0.03). The highest inhibition of tumor growth was observed in mice treated with P/AON/2C5/TfR (p<0.03 vs. controls; p<0.05 vs. P/AON/2C5). Error bars denote SEM. (5b) Expression of EGFR and phosphorylated Akt (p-Akt) after treatment of EGFR-positive tumors <i>in vivo</i>. Western blot analysis showed the decrease in EGFR and p-Akt expression in P/AON/2C5-, or P/AON/2C5/TfR -treated mice compared to controls. The highest inhibition of EGFR and p-Akt was seen upon treatment with P/AON/2C5/TfR where 2 targeting antibodies were combined. GAPDH was an internal control to normalize gel loading. (5c) Histopathological analysis of tumors. Hematoxylin and eosin staining of tumors treated with PBS, P, P/2C5, P/AON/2C5 or the leading drug P/AON/2C5/TfR. Consistent with tumor size reduction data, the leading drug P/AON/2C5/TfR treated tumor showed significant reduction in the number of viable tumor cells as compared to other treatments.</p
    corecore