Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Blood ; 129(1): 114-125, 2017 01 05.
Article in English | MEDLINE | ID: mdl-27760761

ABSTRACT

Attenuated poxvirus modified vaccinia Ankara (MVA) is a useful viral-based vaccine for clinical investigation, because of its excellent safety profile and property of inducing potent immune responses against recombinant (r) antigens. We developed Triplex by constructing an rMVA encoding 3 immunodominant cytomegalovirus (CMV) antigens, which stimulates a host antiviral response: UL83 (pp65), UL123 (IE1-exon4), and UL122 (IE2-exon5). We completed the first clinical evaluation of the Triplex vaccine in 24 healthy adults, with or without immunity to CMV and vaccinia virus (previous DryVax smallpox vaccination). Three escalating dose levels (DL) were administered IM in 8 subjects/DL, with an identical booster injection 28 days later and 1-year follow-up. Vaccinations at all DL were safe with no dose-limiting toxicities. No vaccine-related serious adverse events were documented. Local and systemic reactogenicity was transient and self-limiting. Robust, functional, and durable Triplex-driven expansions of CMV-specific T cells were detected by measuring T-cell surface levels of 4-1BB (CD137), binding to CMV-specific HLA multimers, and interferon-γ production. Marked and durable CMV-specific T-cell responses were also detected in Triplex-vaccinated CMV-seronegatives, and in DryVax-vaccinated subjects. Long-lived memory effector phenotype, associated with viral control during CMV primary infection, was predominantly found on the membrane of CMV-specific and functional T cells, whereas off-target vaccine responses activating memory T cells from the related herpesvirus Epstein-Barr virus remained undetectable. Combined safety and immunogenicity results of MVA in allogeneic hematopoietic stem cell transplant (HCT) recipients and Triplex in healthy adults motivated the initiation of a placebo-controlled multicenter trial of Triplex in HCT patients. This trial was registered at www.clinicaltrials.gov as #NCT02506933.


Subject(s)
Antigens, Viral/immunology , Cytomegalovirus Vaccines/immunology , Lymphocyte Activation/immunology , T-Lymphocytes/immunology , Adult , Cytomegalovirus , Cytomegalovirus Vaccines/adverse effects , Female , Humans , Immediate-Early Proteins/immunology , Male , Middle Aged , Phosphoproteins/immunology , Trans-Activators/immunology , Vaccines, DNA , Viral Matrix Proteins/immunology , Viral Vaccines , Young Adult
2.
Cancer Immunol Res ; 3(9): 1096-107, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26134178

ABSTRACT

Bacterial-based therapies are emerging as effective cancer treatments and hold promise for refractory neoplasms, such as pancreatic ductal adenocarcinoma (PDAC), which has not shown significant improvement in therapy for more than 25 years. Using a novel combination of shIDO-ST, a Salmonella-based therapy targeting the immunosuppressive molecule indoleamine 2,3-dioxygenase (IDO), with an enzyme, PEGPH20, which depletes extracellular matrix hyaluronan, we observed extended survival with frequent total regression of autochthonous and orthotopic PDAC tumors. This observation was associated with migration and accumulation of activated polymorphonuclear neutrophils (PMN) from spleens into tumors, which was not seen using a scrambled control (shScr-ST). Purified splenic PMNs from PEGPH20/shIDO-ST-treated mice exhibited significant IDO knockdown and were able to kill tumor targets ex vivo through mechanisms involving FasL and serine proteases. In addition, CD8(+) T cells were observed to contribute to late control of pancreatic tumors. Collectively, our data demonstrate that entry of shIDO-ST and PMNs into otherwise impermeable desmoplastic tumors is facilitated by PEGPH20-mediated HA removal, further highlighting an important component of effective treatment for PDAC.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Pancreatic Ductal/therapy , Cell Adhesion Molecules/therapeutic use , Hyaluronic Acid/deficiency , Hyaluronoglucosaminidase/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Pancreatic Neoplasms/therapy , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Combined Modality Therapy , Gene Knockdown Techniques/methods , Genetic Therapy/methods , Hyaluronic Acid/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Mice, Inbred C57BL , Molecular Targeted Therapy/methods , Neoplasm Transplantation , Neutrophils/immunology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Recombinant Proteins/therapeutic use , Pancreatic Neoplasms
3.
Cancer Res ; 72(24): 6447-56, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23090116

ABSTRACT

Generating antitumor responses through the inhibition of tumor-derived immune suppression represents a promising strategy in the development of cancer immunotherapeutics. Here, we present a strategy incorporating delivery of the bacterium Salmonella typhimurium (ST), naturally tropic for the hypoxic tumor environment, transformed with a small hairpin RNA (shRNA) plasmid against the immunosuppressive molecule indoleamine 2,3-dioxygenase 1 (shIDO). When systemically delivered into mice, shIDO silences host IDO expression and leads to massive intratumoral cell death that is associated with significant tumor infiltration by polymorphonuclear neutrophils (PMN). shIDO-ST treatment causes tumor cell death independently of host IDO and adaptive immunity, which may have important implications for use in immunosuppressed patients with cancer. Furthermore, shIDO-ST treatment increases reactive oxygen species (ROS) produced by infiltrating PMNs and, conversely, PMN immunodepletion abrogates tumor control. Silencing of host IDO significantly enhances S. typhimurium colonization, suggesting that IDO expression within the tumor controls the immune response to S. typhimurium. In summary, we present a novel approach to cancer treatment that involves the specific silencing of tumor-derived IDO that allows for the recruitment of ROS-producing PMNs, which may act primarily to clear S. typhimurium infection, but in the process also induces apoptosis of surrounding tumor tissue resulting in a vigorous antitumor effect.


Subject(s)
Gene Transfer Techniques , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Neoplasms/therapy , RNA, Small Interfering/administration & dosage , Salmonella typhimurium/growth & development , Salmonella typhimurium/genetics , Transformation, Bacterial , Administration, Intravenous , Animals , Cell Line, Tumor , Cell Proliferation , Colony Count, Microbial , Gene Silencing , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/physiology , HEK293 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/microbiology , Neoplasms/pathology , Organisms, Genetically Modified , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Salmonella typhimurium/immunology , Salmonella typhimurium/physiology , Transformation, Bacterial/physiology
4.
Cancer Res ; 71(12): 4183-91, 2011 Jun 15.
Article in English | MEDLINE | ID: mdl-21527558

ABSTRACT

Cancer vaccine therapies have only achieved limited success when focusing on effector immunity with the goal of eliciting robust tumor-specific T-cell responses. More recently, there is an emerging understanding that effective immunity can only be achieved by coordinate disruption of tumor-derived immunosuppression. Toward that goal, we have developed a potent Salmonella-based vaccine expressing codon-optimized survivin (CO-SVN), referred to as 3342Max. When used alone as a therapeutic vaccine, 3342Max can attenuate growth of aggressive murine melanomas overexpressing SVN. However, under more immunosuppressive conditions, such as those associated with larger tumor volumes, we found that the vaccine was ineffective. Vaccine efficacy could be rescued if tumor-bearing mice were treated initially with Salmonella encoding a short hairpin RNA (shRNA) targeting the tolerogenic molecule STAT3 (YS1646-shSTAT3). In vaccinated mice, silencing STAT3 increased the proliferation and granzyme B levels of intratumoral CD4(+) and CD8(+) T cells. The combined strategy also increased apoptosis in tumors of treated mice, enhancing tumor-specific killing of tumor targets. Interestingly, mice treated with YS1646-shSTAT3 or 3342Max alone were similarly unsuccessful in rejecting established tumors, whereas the combined regimen was highly potent. Our findings establish that a combined strategy of silencing immunosuppressive molecules followed by vaccination can act synergistically to attenuate tumor growth, and they offer a novel translational direction to improve tumor immunotherapy.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy, Active , Inhibitor of Apoptosis Proteins/genetics , Melanoma, Experimental/therapy , RNA, Small Interfering/genetics , Repressor Proteins/genetics , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Apoptosis , Codon , Cytotoxicity, Immunologic , Genetic Vectors , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , STAT3 Transcription Factor/genetics , Survivin , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...