Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Clin Transl Sci ; 5(1): e148, 2021.
Article in English | MEDLINE | ID: mdl-34527288

ABSTRACT

Commercializing biomedical discoveries is a challenging process for many reasons. However, Academic Medical Centers (AMC) that have teaching, patient care, research, and service engrained in their mission are well poised to host these discoveries. These academic discoveries can lead to improvement in patient health and economic development if supported to cross the "valley of death" through institutional assistance, by providing guidance, gap funding and product development expertise. Colorado has a vibrant local startup ecosystem, state support for commercialization and entrepreneurship as well as critical mass of product development expertise. University of Colorado Anschutz Medical Campus, as a major AMC, is an engine for growth for the region. This article discusses innovation efforts at the University of Colorado Anschutz Medical Campus as a case study, which is built around two major efforts: the CCTSI and CU Innovations. I-Corps at CCTSI and the SPARK|REACH program of CU Innovations have been instrumental in fostering innovation, commercialization, and entrepreneurship on the campus.

2.
Mol Ther ; 20(12): 2234-43, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22850679

ABSTRACT

Fas ligand (FasL) gene therapy for cancer has shown promise in rodents; however, its efficacy in higher mammals remains unknown. Here, we used intratumoral FasL gene therapy delivered in an adenovirus vector (Ad-FasL) as neoadjuvant to standard of care in 56 dogs with osteosarcoma. Tumors from treated dogs had greater inflammation, necrosis, apoptosis, and fibrosis at day 10 (amputation) compared to pretreatment biopsies or to tumors from dogs that did not receive Ad-FasL. Survival improvement was apparent in dogs with inflammation or lymphocyte-infiltration scores >1 (in a 3-point scale), as well as in dogs that had apoptosis scores in the top 50th percentile (determined by cleaved caspase-3). Survival was no different than that expected from standard of care alone in dogs with inflammation scores ≤1 or apoptosis scores in the bottom 50th percentile. Reduced Fas expression by tumor cells was associated with prognostically advantageous inflammation, and this was seen only in dogs that received Ad-FasL. Together, the data suggest that Ad-FasL gene therapy improves survival in a subset of large animals with naturally occurring tumors, and that at least in some tumor types like osteosarcoma, it is most effective when tumor cells fail to express Fas.


Subject(s)
Bone Neoplasms/therapy , Fas Ligand Protein/genetics , Genetic Therapy/methods , Animals , Apoptosis/genetics , Apoptosis/physiology , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , Dogs , Necrosis , Osteosarcoma/genetics , Osteosarcoma/metabolism , Osteosarcoma/therapy
3.
Vaccine ; 28(37): 6028-35, 2010 Aug 23.
Article in English | MEDLINE | ID: mdl-20619375

ABSTRACT

Chromosomal translocations generating the BCR-ABL oncogene cause chronic myeloid leukemia (CML) and a subset of acute lymphoblastic leukemia. The BCR-ABL(T315I) mutation confers drug resistance to FDA-approved targeted therapeutics imatinib mesylate, dasatinib, and nilotinib. We tested the ability of a recombinant yeast-based vaccine expressing the T315I-mutated BCR-ABL antigen to stimulate an anti-BCR-ABL(T315I) immune response. The yeast-based immunotherapy significantly reduced or eliminated BCR-ABL(T315I) leukemia cells from the peripheral blood of immunized animals and extended leukemia-free survival in a murine model of BCR-ABL(+) leukemia compared to animals receiving sham injection or yeast expressing ovalbumin. With immunization, leukemic cells harboring BCR-ABL(T315I) were selectively eliminated after challenge with a mixed population of BCR-ABL and BCR-ABL(T315I) leukemias. In summary, yeast-based immunotherapy represents a novel approach against the emergence of cancer drug resistance by the pre-emptive targeted ablation of tumor escape mutants.


Subject(s)
Cancer Vaccines/immunology , Fusion Proteins, bcr-abl/immunology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Amino Acid Sequence , Animals , Disease-Free Survival , Genes, MHC Class I , Immunotherapy , Leukemia, Experimental/immunology , Leukemia, Experimental/therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Molecular , Molecular Sequence Data , Protein Structure, Tertiary , Saccharomyces cerevisiae , Vaccines, Synthetic/immunology
4.
Vaccine ; 25(8): 1452-63, 2007 Feb 09.
Article in English | MEDLINE | ID: mdl-17098335

ABSTRACT

Control of primary infection with hepatitis C virus (HCV) is associated with robust and broad T cell immunity. In contrast, chronic infection is characterized by weak T cell responses suggesting that an approach that boosts these responses could be a therapeutic advance. Saccharomyces cerevisiae is an effective inducer of innate and adaptive cellular immunity and we have generated recombinant yeast cells (GI-5005) that produce an HCV NS3-Core fusion protein. Pre-clinical studies in mice showed that GI-5005 induced potent antigen-specific proliferative and cytotoxic T cell responses that were associated with Th1-type cytokine secretion. In studies in which GI-5005 was administered up to 13 times, no detectable vector neutralization or induction of tolerance was observed. Prophylactic as well as therapeutic administration of GI-5005 in mice led to eradication of tumor cells expressing HCV NS3 protein. Immunotherapy with GI-5005 is being evaluated in chronic HCV infected individuals in a Phase 1 clinical trial.


Subject(s)
Hepacivirus/immunology , Immunotherapy/methods , Recombinant Fusion Proteins/immunology , Saccharomyces cerevisiae/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Viral Core Proteins/immunology , Viral Hepatitis Vaccines/immunology , Viral Nonstructural Proteins/immunology , Animals , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , HeLa Cells , Hepacivirus/genetics , Hepatitis C/immunology , Hepatitis C/prevention & control , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Recombinant Fusion Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Viral Core Proteins/biosynthesis , Viral Core Proteins/genetics , Viral Hepatitis Vaccines/genetics , Viral Nonstructural Proteins/biosynthesis , Viral Nonstructural Proteins/genetics
5.
Expert Opin Biol Ther ; 5(4): 565-75, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15934834

ABSTRACT

Immunotherapy for cancer represents an attractive therapeutic target because of its specificity and lack of toxicity, but products investigated so far have been limited by neutralisation, complexity of manufacturing and requirement for patient-specific products. Recombinant yeast cells are capable of stimulating the immune system to produce highly specific and potent cellular responses against target protein antigens with little toxicity. Data from animal models suggest that Tarmogens (yeast-based immunotherapeutics) can elicit protective immunity against xenografted and chemically induced tumours. This concept is now being tested in a Phase I trial in patients with colorectal, pancreatic and non-small cell lung cancers.


Subject(s)
Antigens, Neoplasm/immunology , Immunotherapy/methods , Neoplasms/immunology , Yeasts/immunology , Animals , Antigens, Neoplasm/genetics , Antigens, Neoplasm/therapeutic use , Humans , Neoplasms/genetics , Neoplasms/therapy , Yeasts/genetics
6.
Cancer Res ; 64(15): 5084-8, 2004 Aug 01.
Article in English | MEDLINE | ID: mdl-15289309

ABSTRACT

Activating mutations in Ras oncoproteins represent attractive targets for cancer immunotherapy, but few vectors capable of generating immune responses required for tumor killing without vector neutralization have been described. Whole recombinant yeast heterologously expressing mammalian mutant Ras proteins were used to immunize mice in a carcinogen-induced lung tumor model. Therapeutic immunization with the whole recombinant yeast caused complete regression of established Ras mutation-bearing lung tumors in a dose-dependent, antigen-specific manner. In combination with the genomic sequencing of tumors in patients, the yeast-based immunotherapeutic approach could be applied to treat Ras mutation-bearing human cancers.


Subject(s)
Immunotherapy/methods , Lung Neoplasms/prevention & control , Neoplasms, Experimental/immunology , Neoplasms, Experimental/prevention & control , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/immunology , Adenoma/chemically induced , Adenoma/immunology , Adenoma/prevention & control , Animals , DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/immunology , Male , Mice , Mice, Inbred Strains , Mutation , Neoplasms, Experimental/chemically induced , Proto-Oncogene Proteins p21(ras)/pharmacology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , Saccharomyces cerevisiae/physiology , Urethane
7.
Clin Immunol ; 112(1): 54-65, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15207782

ABSTRACT

Signals transmitted by binding of Fas ligand (FasL) to the Fas receptor (CD95/Apo-1) have pleiotropic effects on cellular function that present opportunities for therapeutic applications. For example, depending on the circumstances, overexpression of FasL can enhance, prevent, or reverse growth of spontaneous or transplantable tumors. Furthermore, local administration of FasL into a single paw in susceptible mice protects from or reduces the severity of collagen-induced arthritis (CIA) in all paws. Here, we define mechanisms that mediate systemic protection induced by locally delivered FasL. Protection is not solely dependent on local interactions between Fas and FasL, but rather requires induction of a paradoxical inflammatory response that not only destroys Fas-resistant tumors, but also recruits motile, activated, Fas-bearing T cells that are Fas sensitive. We demonstrate by following the antigen-specific recruitment and subsequent termination of transgenic T cells that activated T cells, including autoreactive cells responsible for CIA, are eliminated within this inflammatory environment through the overexpressed FasL. The nature of the inflammatory response, which depends on the Fas ligand being cell bound and not soluble, and the magnitude of FasL expression within the inflammatory milieu are essential for this effect, as arthritogenic inflammation alone resulting from CIA induction is insufficient to ameliorate the disease or eliminate antigen-specific T cells, even upon systemic delivery of soluble FasL. These data show that gene delivery of membrane-bound FasL can effectively recruit and eliminate autoreactive T cells.


Subject(s)
Arthritis, Experimental/prevention & control , Autoimmunity/drug effects , Membrane Glycoproteins/pharmacology , T-Lymphocytes/drug effects , Adenoviridae/genetics , Adoptive Transfer , Animals , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Autoimmunity/immunology , Cell Death/immunology , Cytotoxicity, Immunologic , Fas Ligand Protein , Female , Lymphocyte Activation , Male , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Neoplasms, Experimental/immunology , T-Lymphocytes/immunology
9.
Methods Mol Biol ; 282: 43-66, 2004.
Article in English | MEDLINE | ID: mdl-15105556

ABSTRACT

Natural killer (NK) cells are a crucial component of the immune system. For example, the NK cell-mediated killing of tumor cells is a first line of defense against the development of cancer. Detection and quantification of apoptosis induced in target cells by NK cells is a useful tool for studies of the mechanisms of both immunological protection and pathogenesis. This chapter describes how to do this by using techniques with a broad application to both NK cells and cytotoxic T cells.


Subject(s)
Apoptosis/physiology , Biological Assay/methods , Chromatin/metabolism , Killer Cells, Natural/physiology , Chelating Agents/metabolism , Chromatin/chemistry , DNA Fragmentation , Egtazic Acid/metabolism , Fas Ligand Protein , Fluorescent Dyes/metabolism , In Situ Nick-End Labeling , Ionomycin/pharmacology , Ionophores/pharmacology , Killer Cells, Natural/cytology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Membrane Glycoproteins/metabolism , Microscopy/methods , T-Lymphocytes, Cytotoxic/immunology , Tetradecanoylphorbol Acetate/pharmacology , Ultraviolet Rays , fas Receptor/metabolism
12.
J Ark Med Soc ; 100(9): 292-3, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15011792
15.
J Ark Med Soc ; 100(5): 152-3, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14606222
17.
J Ark Med Soc ; 100(3): 80-5, 2003 Sep.
Article in English | MEDLINE | ID: mdl-13677138
20.
Cancer Gene Ther ; 10(9): 726-36, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12944992

ABSTRACT

We examined the feasibility of using tumor apoptosis at accessible sites to enhance antimelanoma immune responses in a model of spontaneous canine melanoma. We show that priming peripheral blood mononuclear cells with apoptotic melanoma cells significantly enhanced autologous and allogeneic lymphokine-activated killing of tumor cells. Since various pathways required for intrinsic apoptosis are often inactivated in melanoma, we used Fas ligand (FasL) overexpression to promote extrinsic apoptosis. FasL induced apoptosis in five of six cell lines. Each of the susceptible lines, but not the resistant one, expressed Fas mRNA. In addition, direct intratumoral administration of FasL DNA to tumor-bearing dogs was safe, with no adverse events reported over 7 days of observation. A reduction of tumor burden was seen in three of five dogs treated. The reduction of tumor volume was correlated with Fas expression by the tumors, although one dog with a Fas-negative tumor survived for 82 weeks after treatment. Our data show that overexpression of FasL is suitable to promote apoptosis of Fas(+) melanomas, and support the notion that priming immune responder cells with apoptotic tumor cells may enhance antitumor responses. The results also suggest that intratumoral administration of FasL offers a safe route for therapeutic gene delivery.


Subject(s)
Apoptosis , Dog Diseases/immunology , Dog Diseases/pathology , Melanoma/therapy , Melanoma/veterinary , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/therapeutic use , Animals , Dog Diseases/genetics , Dog Diseases/therapy , Dogs , Fas Ligand Protein , Genetic Therapy , Immunotherapy , Leukocytes, Mononuclear/immunology , Melanoma/immunology , Melanoma/pathology , Membrane Glycoproteins/adverse effects , Membrane Glycoproteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...