58 results on '"Dubensky TW Jr"'
Search Results
2. A Potent and Effective Suicidal Listeria Vaccine Platform.
- Author
-
Hanson WG, Benanti EL, Lemmens EE, Liu W, Skoble J, Leong ML, Rae CS, Fassò M, Brockstedt DG, Chen C, Portnoy DA, Dubensky TW Jr, and Lauer P
- Subjects
- Animals, Female, Immunotherapy, Listeria monocytogenes pathogenicity, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, T-Lymphocytes immunology, Vaccines, Attenuated immunology, Virulence, Bacterial Vaccines immunology, Listeria monocytogenes immunology
- Abstract
Live-attenuated Listeria monocytogenes has shown encouraging potential as an immunotherapy platform in preclinical and clinical settings. However, additional safety measures will enable application across malignant and infectious diseases. Here, we describe a new vaccine platform, termed Lm-RIID ( L. monocytogenes recombinase-induced intracellular death), that induces the deletion of genes required for bacterial viability yet maintains potent T cell responses to encoded antigens. Lm-RIID grows normally in broth but commits suicide inside host cells by inducing Cre recombinase and deleting essential genes flanked by loxP sites, resulting in a self-limiting infection even in immunocompromised mice. Lm-RIID vaccination of mice induces potent CD8
+ T cells and protects against virulent challenges, similar to live L. monocytogenes vaccines. When combined with α-PD-1, Lm-RIID is as effective as live-attenuated L. monocytogenes in a therapeutic tumor model. This impressive efficacy, together with the increased clearance rate, makes Lm-RIID ideal for prophylactic immunization against diseases that require T cells for protection., (Copyright © 2019 Hanson et al.)- Published
- 2019
- Full Text
- View/download PDF
3. Cyclic Dinucleotide-Adjuvanted Dengue Virus Nonstructural Protein 1 Induces Protective Antibody and T Cell Responses.
- Author
-
Espinosa DA, Beatty PR, Reiner GL, Sivick KE, Hix Glickman L, Dubensky TW Jr, and Harris E
- Subjects
- Animals, Mice, Mice, Inbred C57BL, Mice, Knockout, Adjuvants, Immunologic, Antibodies, Viral immunology, Dengue Virus immunology, Nucleotides, Cyclic immunology, T-Lymphocytes immunology, Viral Nonstructural Proteins immunology
- Abstract
Endothelial dysfunction and vascular leak, pathogenic hallmarks of severe dengue disease, are directly triggered by dengue virus (DENV) nonstructural protein 1 (NS1). Previous studies have shown that immunization with NS1, as well as passive transfer of NS1-immune serum or anti-NS1 mAb, prevent NS1-mediated lethality in vivo. In this study, we evaluated the immunogenicity and protective capacity of recombinant DENV NS1 administered with cyclic dinucleotides (CDNs), potent activators of innate immune pathways and highly immunogenic adjuvants. Using both wild-type C57BL/6 mice and IFN-α/β receptor-deficient mice, we show that NS1-CDN immunizations elicit serotype-specific and cross-reactive Ab and T cell responses. Furthermore, NS1-CDN vaccinations conferred significant homotypic and heterotypic protection from DENV2-induced morbidity and mortality. In addition, we demonstrate that high anti-NS1 Ab titers are associated with protection, supporting the role of humoral responses against DENV NS1 as correlates of protection. These findings highlight the potential of CDN-based adjuvants for inducing Ab and T cell responses and validate NS1 as an important candidate for dengue vaccine development., (Copyright © 2019 by The American Association of Immunologists, Inc.)
- Published
- 2019
- Full Text
- View/download PDF
4. Magnitude of Therapeutic STING Activation Determines CD8 + T Cell-Mediated Anti-tumor Immunity.
- Author
-
Sivick KE, Desbien AL, Glickman LH, Reiner GL, Corrales L, Surh NH, Hudson TE, Vu UT, Francica BJ, Banda T, Katibah GE, Kanne DB, Leong JJ, Metchette K, Bruml JR, Ndubaku CO, McKenna JM, Feng Y, Zheng L, Bender SL, Cho CY, Leong ML, van Elsas A, Dubensky TW Jr, and McWhirter SM
- Subjects
- Animals, CTLA-4 Antigen metabolism, Cell Line, Tumor, Cytokines metabolism, Dose-Response Relationship, Immunologic, Drug Resistance, Neoplasm, Hematopoiesis, Mice, Inbred BALB C, Mice, Inbred C57BL, Neoplasms pathology, Programmed Cell Death 1 Receptor metabolism, S100 Proteins administration & dosage, S100 Proteins immunology, CD8-Positive T-Lymphocytes immunology, Immunity, Membrane Proteins metabolism, Neoplasms immunology
- Abstract
Intratumoral (IT) STING activation results in tumor regression in preclinical models, yet factors dictating the balance between innate and adaptive anti-tumor immunity are unclear. Here, clinical candidate STING agonist ADU-S100 (S100) is used in an IT dosing regimen optimized for adaptive immunity to uncover requirements for a T cell-driven response compatible with checkpoint inhibitors (CPIs). In contrast to high-dose tumor ablative regimens that result in systemic S100 distribution, low-dose immunogenic regimens induce local activation of tumor-specific CD8
+ effector T cells that are responsible for durable anti-tumor immunity and can be enhanced with CPIs. Both hematopoietic cell STING expression and signaling through IFNAR are required for tumor-specific T cell activation, and in the context of optimized T cell responses, TNFα is dispensable for tumor control. In a poorly immunogenic model, S100 combined with CPIs generates a survival benefit and durable protection. These results provide fundamental mechanistic insights into STING-induced anti-tumor immunity., (Copyright © 2018 Aduro Biotech, Inc. Published by Elsevier Inc. All rights reserved.)- Published
- 2018
- Full Text
- View/download PDF
5. Combining STING-based neoantigen-targeted vaccine with checkpoint modulators enhances antitumor immunity in murine pancreatic cancer.
- Author
-
Kinkead HL, Hopkins A, Lutz E, Wu AA, Yarchoan M, Cruz K, Woolman S, Vithayathil T, Glickman LH, Ndubaku CO, McWhirter SM, Dubensky TW Jr, Armstrong TD, Jaffee EM, and Zaidi N
- Subjects
- Adenocarcinoma genetics, Adenocarcinoma immunology, Adjuvants, Immunologic administration & dosage, Animals, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, Antineoplastic Agents, Immunological therapeutic use, Cancer Vaccines genetics, Cancer Vaccines immunology, Cell Line, Tumor transplantation, Combined Modality Therapy methods, Disease Models, Animal, Epitopes, T-Lymphocyte genetics, Epitopes, T-Lymphocyte immunology, Humans, Immunogenicity, Vaccine, Membrane Proteins immunology, Mice, Pancreatic Neoplasms genetics, Pancreatic Neoplasms immunology, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor immunology, Receptors, OX40 agonists, Receptors, OX40 immunology, Treatment Outcome, Tumor Escape drug effects, Tumor Escape immunology, Vaccines, Subunit administration & dosage, Vaccines, Subunit genetics, Vaccines, Subunit immunology, Adenocarcinoma therapy, Antineoplastic Agents, Immunological pharmacology, Cancer Vaccines administration & dosage, Immunotherapy methods, Pancreatic Neoplasms therapy
- Abstract
Tumor neoantigens arising from somatic mutations in the cancer genome are less likely to be subject to central immune tolerance and are therefore attractive targets for vaccine immunotherapy. We utilized whole-exome sequencing, RNA sequencing (RNASeq), and an in silico immunogenicity prediction algorithm, NetMHC, to generate a neoantigen-targeted vaccine, PancVAX, which was administered together with the STING adjuvant ADU-V16 to mice bearing pancreatic adenocarcinoma (Panc02) cells. PancVAX activated a neoepitope-specific T cell repertoire within the tumor and caused transient tumor regression. When given in combination with two checkpoint modulators, namely anti-PD-1 and agonist OX40 antibodies, PancVAX resulted in enhanced and more durable tumor regression and a survival benefit. The addition of OX40 to vaccine reduced the coexpression of T cell exhaustion markers, Lag3 and PD-1, and resulted in rejection of tumors upon contralateral rechallenge, suggesting the induction of T cell memory. Together, these data provide the framework for testing personalized neoantigen-based combinatorial vaccine strategies in patients with pancreatic and other nonimmunogenic cancers.
- Published
- 2018
- Full Text
- View/download PDF
6. Recombinant Listeria promotes tumor rejection by CD8 + T cell-dependent remodeling of the tumor microenvironment.
- Author
-
Deng W, Lira V, Hudson TE, Lemmens EE, Hanson WG, Flores R, Barajas G, Katibah GE, Desbien AL, Lauer P, Leong ML, Portnoy DA, and Dubensky TW Jr
- Subjects
- Animals, Antigens, Neoplasm genetics, Antigens, Neoplasm therapeutic use, Cancer Vaccines genetics, Cancer Vaccines therapeutic use, Cell Line, Tumor, Drug Evaluation, Preclinical, Female, Humans, Listeria monocytogenes genetics, Macrophages immunology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Neoplasms immunology, Treatment Outcome, Vaccination methods, Vaccines, Attenuated genetics, Vaccines, Attenuated immunology, Vaccines, Attenuated therapeutic use, Vaccines, DNA genetics, Vaccines, DNA immunology, Vaccines, DNA therapeutic use, Xenograft Model Antitumor Assays, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines immunology, Listeria monocytogenes immunology, Neoplasms therapy, Tumor Microenvironment immunology
- Abstract
Agents that remodel the tumor microenvironment (TME), prime functional tumor-specific T cells, and block inhibitory signaling pathways are essential components of effective immunotherapy. We are evaluating live-attenuated, double-deleted Listeria monocytogenes expressing tumor antigens (LADD-Ag) in the clinic. Here we show in numerous mouse models that while treatment with nonrecombinant LADD induced some changes in the TME, no antitumor efficacy was observed, even when combined with immune checkpoint blockade. In contrast, LADD-Ag promoted tumor rejection by priming tumor-specific KLRG1
+ PD1lo CD62L- CD8+ T cells. These IFNγ-producing effector CD8+ T cells infiltrated the tumor and converted the tumor from an immunosuppressive to an inflamed microenvironment that was characterized by a decrease in regulatory T cells (Treg) levels, a proinflammatory cytokine milieu, and the shift of M2 macrophages to an inducible nitric oxide synthase (iNOS)+ CD206- M1 phenotype. Remarkably, these LADD-Ag-induced tumor-specific T cells persisted for more than 2 months after primary tumor challenge and rapidly controlled secondary tumor challenge. Our results indicate that the striking antitumor efficacy observed in mice with LADD-based immunotherapy stems from TME remodeling which is a direct consequence of eliciting potent, systemic tumor-specific CD8+ T cells., Competing Interests: Conflict of interest statement: W.D., V.L., T.E.H., E.E.L., W.G.H., R.F., G.B., G.E.K., A.L.D., P.L., M.L.L., and T.W.D. are current or former paid employees of Aduro Biotech, and hold stock in the company. D.A.P. was supported by National Institutes of Health Grants 1P01 AI063302 and 1R01 AI027655 and a Grant from Aduro Biotech (IVRI). D.A.P. has a consulting relationship with and a financial interest in Aduro Biotech, and both he and the company stand to benefit from the commercialization of the results of this research., (Copyright © 2018 the Author(s). Published by PNAS.)- Published
- 2018
- Full Text
- View/download PDF
7. STING-Activating Adjuvants Elicit a Th17 Immune Response and Protect against Mycobacterium tuberculosis Infection.
- Author
-
Van Dis E, Sogi KM, Rae CS, Sivick KE, Surh NH, Leong ML, Kanne DB, Metchette K, Leong JJ, Bruml JR, Chen V, Heydari K, Cadieux N, Evans T, McWhirter SM, Dubensky TW Jr, Portnoy DA, and Stanley SA
- Subjects
- Animals, BCG Vaccine immunology, Disease Models, Animal, Immunity, Cellular drug effects, Mice, Mice, Knockout, Th1 Cells immunology, Th1 Cells pathology, Th17 Cells pathology, Tuberculosis, Pulmonary immunology, Tuberculosis, Pulmonary pathology, Vaccines, Subunit immunology, Vaccines, Subunit pharmacokinetics, Adjuvants, Immunologic pharmacology, BCG Vaccine pharmacology, Membrane Proteins immunology, Mycobacterium tuberculosis immunology, Th17 Cells immunology, Tuberculosis, Pulmonary prevention & control
- Abstract
There are a limited number of adjuvants that elicit effective cell-based immunity required for protection against intracellular bacterial pathogens. Here, we report that STING-activating cyclic dinucleotides (CDNs) formulated in a protein subunit vaccine elicit long-lasting protective immunity to Mycobacterium tuberculosis in the mouse model. Subcutaneous administration of this vaccine provides equivalent protection to that of the live attenuated vaccine strain Bacille Calmette-Guérin (BCG). Protection is STING dependent but type I IFN independent and correlates with an increased frequency of a recently described subset of CXCR3-expressing T cells that localize to the lung parenchyma. Intranasal delivery results in superior protection compared with BCG, significantly boosts BCG-based immunity, and elicits both Th1 and Th17 immune responses, the latter of which correlates with enhanced protection. Thus, a CDN-adjuvanted protein subunit vaccine has the capability of eliciting a multi-faceted immune response that results in protection from infection by an intracellular pathogen., (Copyright © 2018 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
8. TNFα and Radioresistant Stromal Cells Are Essential for Therapeutic Efficacy of Cyclic Dinucleotide STING Agonists in Nonimmunogenic Tumors.
- Author
-
Francica BJ, Ghasemzadeh A, Desbien AL, Theodros D, Sivick KE, Reiner GL, Hix Glickman L, Marciscano AE, Sharabi AB, Leong ML, McWhirter SM, Dubensky TW Jr, Pardoll DM, and Drake CG
- Subjects
- Animals, Antigen-Presenting Cells immunology, Antigen-Presenting Cells metabolism, Bone Marrow metabolism, Cell Line, Tumor, Cytokines metabolism, Disease Models, Animal, Female, Humans, Immunity, Innate, Interferon-beta metabolism, Melanoma, Experimental, Mice, Mice, Knockout, Necrosis metabolism, Necrosis pathology, Neoplasms drug therapy, Neoplasms pathology, Signal Transduction drug effects, Stromal Cells pathology, Stromal Cells radiation effects, Tumor Burden drug effects, Tumor Microenvironment immunology, Antineoplastic Agents pharmacology, Membrane Proteins agonists, Neoplasms etiology, Neoplasms metabolism, Nucleotides, Cyclic pharmacology, Radiation Tolerance drug effects, Radiation Tolerance genetics, Stromal Cells metabolism, Tumor Necrosis Factor-alpha metabolism
- Abstract
The cGAS-STING cytosolic DNA sensing pathway may play an integral role in the initiation of antitumor immune responses. Studies evaluating the immunogenicity of various cyclic dinucleotide (CDN) STING agonists administered by intratumoral (i.t.) injection showed potent induction of inflammation, tumor necrosis, and, in some cases, durable tumor-specific adaptive immunity. However, the specific immune mechanisms underlying these responses remain incompletely defined. The majority of these studies have focused on the effect of CDNs on immune cells but have not conclusively interrogated the role of stromal cells in the acute rejection of the CDN-injected tumor. Here, we revealed a mechanism of STING agonist-mediated tumor response that relied on both stromal and immune cells to achieve tumor regression and clearance. Using knockout and bone marrow chimeric mice, we showed that although bone marrow-derived TNFα was necessary for CDN-induced necrosis, STING signaling in radioresistant stromal cells was also essential for CDN-mediated tumor rejection. These results provide evidence for crosstalk between stromal and hematopoietic cells during CDN-mediated tumor collapse after i.t. administration. These mechanistic insights may prove critical in the clinical development of STING agonists. Cancer Immunol Res; 6(4); 422-33. ©2018 AACR ., (©2018 American Association for Cancer Research.)
- Published
- 2018
- Full Text
- View/download PDF
9. The VirAB ABC Transporter Is Required for VirR Regulation of Listeria monocytogenes Virulence and Resistance to Nisin.
- Author
-
Grubaugh D, Regeimbal JM, Ghosh P, Zhou Y, Lauer P, Dubensky TW Jr.,, and Higgins DE
- Subjects
- ATP-Binding Cassette Transporters genetics, Animals, Bacterial Proteins genetics, Drug Resistance, Bacterial, Female, Gene Expression Regulation, Bacterial, Humans, Listeria monocytogenes drug effects, Listeria monocytogenes genetics, Mice, Inbred BALB C, Microbial Sensitivity Tests, Regulon, Transcription Factors metabolism, Virulence, ATP-Binding Cassette Transporters metabolism, Anti-Bacterial Agents pharmacology, Bacterial Proteins metabolism, Listeria monocytogenes metabolism, Listeria monocytogenes pathogenicity, Listeriosis microbiology, Nisin pharmacology, Transcription Factors genetics
- Abstract
Listeria monocytogenes is a Gram-positive intracellular pathogen that causes a severe invasive disease. Upon infecting a host cell, L. monocytogenes upregulates the transcription of numerous factors necessary for productive infection. VirR is the response regulator component of a two-component regulatory system in L. monocytogenes In this report, we have identified the putative ABC transporter encoded by genes lmo1746-lmo1747 as necessary for VirR function. We have designated lmo1746-lmo1747 virAB We constructed an in-frame deletion of virAB and determined that the Δ virAB mutant exhibited reduced transcription of VirR-regulated genes. The Δ virAB mutant also showed defects in in vitro plaque formation and in vivo virulence that were similar to those of a Δ virR deletion mutant. Since VirR is important for innate resistance to antimicrobial agents, we determined the MICs of nisin and bacitracin for Δ virAB bacteria. We found that VirAB expression was necessary for nisin resistance but was dispensable for resistance to bacitracin. This result suggested a VirAB-independent mechanism of VirR regulation in response to bacitracin. Lastly, we found that the Δ virR and Δ virAB mutants had no deficiency in growth in broth culture, intracellular replication, or production of the ActA surface protein, which facilitates actin-based motility and cell-to-cell spread. However, the Δ virR and Δ virAB mutants produced shorter actin tails during intracellular infection, which suggested that these mutants have a reduced ability to move and spread via actin-based motility. These findings have demonstrated that L. monocytogenes VirAB functions in a pathway with VirR to regulate the expression of genes necessary for virulence and resistance to antimicrobial agents., (Copyright © 2018 American Society for Microbiology.)
- Published
- 2018
- Full Text
- View/download PDF
10. New Cancer Immunotherapy Agents in Development: a report from an associated program of the 31 st Annual Meeting of the Society for Immunotherapy of Cancer, 2016.
- Author
-
Adusumilli PS, Cha E, Cornfeld M, Davis T, Diab A, Dubensky TW Jr, Evans E, Grogan JL, Irving BA, Leidner RS, Olwill SA, Soon-Shiong P, Triebel F, Tuck D, Bot A, Dansey RD, Drake CG, Freeman GJ, Ibrahim R, Patel S, and Chen DS
- Subjects
- Cancer Vaccines immunology, Humans, Neoplasms immunology, Tumor Microenvironment drug effects, Cancer Vaccines therapeutic use, Immunotherapy, Neoplasms drug therapy, Tumor Microenvironment immunology
- Abstract
This report is a summary of 'New Cancer Immunotherapy Agents in Development' program, which took place in association with the 31st Annual Meeting of the Society for Immunotherapy of Cancer (SITC), on November 9, 2016 in National Harbor, Maryland. Presenters gave brief overviews of emerging clinical and pre-clinical immune-based agents and combinations, before participating in an extended panel discussion with multidisciplinary leaders, including members of the FDA, leading academic institutions and industrial drug developers, to consider topics relevant to the future of cancer immunotherapy.
- Published
- 2017
- Full Text
- View/download PDF
11. A STING Agonist Given with OX40 Receptor and PD-L1 Modulators Primes Immunity and Reduces Tumor Growth in Tolerized Mice.
- Author
-
Foote JB, Kok M, Leatherman JM, Armstrong TD, Marcinkowski BC, Ojalvo LS, Kanne DB, Jaffee EM, Dubensky TW Jr, and Emens LA
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Lymphocytes, Tumor-Infiltrating immunology, Mice, Transgenic, Neoplasms pathology, Tumor Burden, B7-H1 Antigen immunology, Membrane Proteins agonists, Neoplasms immunology, Receptors, OX40 immunology
- Abstract
Stimulator of interferon genes (STING) signaling induces IFNβ production by intratumoral dendritic cells (DC), driving T-cell priming and recruitment into the tumor microenvironment (TME). We examined to what extent preexisting antigen-specific tolerance influenced the efficacy of in situ delivery of a potent STING-activating cyclic dinucleotide (CDN), ADU S-100, against established HER-2
+ breast tumors. ADU S-100 induced HER-2-specific CD8+ T-cell priming and durable tumor clearance in 100% of nontolerant parental FVB/N mice. In contrast, ADU S-100 did not sufficiently prime HER-2-specific CD8+ T cells in tolerant neu/N mice, resulting in only delayed tumor growth and tumor clearance in 10% of the mice. No differences in IFNβ production, DC priming, or HER-2-specific CD8+ T-cell trafficking were detected between FVB/N and neu/N mice. However, activation and expansion of HER-2-specific CD8+ T cells were defective in neu/N mice. Immune cell infiltrates of untreated tumor-bearing neu/N mice expressed high numbers of PD1 and OX40 receptors on their CD8+ T cells, and PD-L1 was highly expressed on both myeloid and tumor cells. Modulating PD-L1 and OX40 receptor signaling combined with intratumoral ADU S-100 administration enhanced HER-2-specific CD8+ T-cell activity, clearing tumors in 40% of neu/N mice. Thus, intratumoral STING agonists could potently prime tumor antigen-specific CD8+ T-cell responses, and adding PD-L1 blockade and OX40 receptor activation can overcome antigen-enforced immune tolerance to induce tumor regression. Cancer Immunol Res; 5(6); 468-79. ©2017 AACR ., (©2017 American Association for Cancer Research.)- Published
- 2017
- Full Text
- View/download PDF
12. Comment on "The Common R71H-G230A-R293Q Human TMEM173 Is a Null Allele".
- Author
-
Sivick KE, Surh NH, Desbien AL, Grewal EP, Katibah GE, McWhirter SM, and Dubensky TW Jr
- Subjects
- Humans, Alleles, Gene Frequency
- Published
- 2017
- Full Text
- View/download PDF
13. The host STING pathway at the interface of cancer and immunity.
- Author
-
Corrales L, McWhirter SM, Dubensky TW Jr, and Gajewski TF
- Subjects
- Adaptive Immunity, Animals, Antigen-Presenting Cells immunology, Antineoplastic Agents pharmacology, CD8-Positive T-Lymphocytes cytology, Cytosol metabolism, DNA, Neoplasm analysis, Dendritic Cells immunology, Humans, Immunity, Innate, Interferon-beta immunology, Mice, Tumor Microenvironment immunology, Interferons immunology, Neoplasms immunology, Neoplasms metabolism
- Abstract
A major subset of human cancers shows evidence for spontaneous adaptive immunity, which is reflected by the presence of infiltrating CD8+ T cells specific for tumor antigens within the tumor microenvironment. This observation has raised the question of which innate immune sensing pathway might detect the presence of cancer and lead to a natural adaptive antitumor immune response in the absence of exogenous infectious pathogens. Evidence for a critical functional role for type I IFNs led to interrogation of candidate innate immune sensing pathways that might be triggered by tumor presence and induce type I IFN production. Such analyses have revealed a major role for the stimulator of IFN genes pathway (STING pathway), which senses cytosolic tumor-derived DNA within the cytosol of tumor-infiltrating DCs. Activation of this pathway is correlated with IFN-β production and induction of antitumor T cells. Based on the biology of this natural immune response, pharmacologic agonists of the STING pathway are being developed to augment and optimize STING activation as a cancer therapy. Intratumoral administration of STING agonists results in remarkable therapeutic activity in mouse models, and STING agonists are being carried forward into phase I clinical testing.
- Published
- 2016
- Full Text
- View/download PDF
14. STING Pathway Activation Stimulates Potent Immunity against Acute Myeloid Leukemia.
- Author
-
Curran E, Chen X, Corrales L, Kline DE, Dubensky TW Jr, Duttagupta P, Kortylewski M, and Kline J
- Subjects
- Adaptive Immunity drug effects, Animals, Antigen-Presenting Cells drug effects, Antigen-Presenting Cells metabolism, Antigens, Neoplasm immunology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Disease Models, Animal, Genetic Engineering, Humans, Immunologic Memory drug effects, Interferon Type I metabolism, Leukemia, Myeloid, Acute pathology, Mice, Inbred C57BL, Survival Analysis, Xanthones pharmacology, Immunity, Innate drug effects, Leukemia, Myeloid, Acute immunology, Membrane Proteins metabolism, Signal Transduction drug effects
- Abstract
Type I interferon (IFN), essential for spontaneous T cell priming against solid tumors, is generated through recognition of tumor DNA by STING. Interestingly, we observe that type I IFN is not elicited in animals with disseminated acute myeloid leukemia (AML). Further, survival of leukemia-bearing animals is not diminished in the absence of type I IFN signaling, suggesting that STING may not be triggered by AML. However, the STING agonist, DMXAA, induces expression of IFN-β and other inflammatory cytokines, promotes dendritic cell (DC) maturation, and results in the striking expansion of leukemia-specific T cells. Systemic DMXAA administration significantly extends survival in two AML models. The therapeutic effect of DMXAA is only partially dependent on host type I IFN signaling, suggesting that other cytokines are important. A synthetic cyclic dinucleotide that also activates human STING provided a similar anti-leukemic effect. These data demonstrate that STING is a promising immunotherapeutic target in AML., Competing Interests: of Conflicts of Interest T.W.D is a paid employee of Aduro Biotech, holds stock in the company, and may be an inventor on patent applications that apply to the CDN molecules described in the manuscript., (Copyright © 2016 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
15. Antagonism of the STING Pathway via Activation of the AIM2 Inflammasome by Intracellular DNA.
- Author
-
Corrales L, Woo SR, Williams JB, McWhirter SM, Dubensky TW Jr, and Gajewski TF
- Subjects
- Animals, Antigen-Presenting Cells immunology, Antigen-Presenting Cells metabolism, Caspase 1 metabolism, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, Female, Interferon-gamma biosynthesis, Macrophages immunology, Macrophages metabolism, Mice, Mice, Knockout, Neoplasms genetics, Neoplasms immunology, Neoplasms metabolism, Nucleotidyltransferases metabolism, Pyroptosis genetics, Pyroptosis immunology, DNA immunology, DNA metabolism, DNA-Binding Proteins metabolism, Inflammasomes, Membrane Proteins metabolism, Signal Transduction
- Abstract
Recent evidence has indicated that innate immune sensing of cytosolic DNA in dendritic cells via the host STING pathway is a major mechanism leading to spontaneous T cell responses against tumors. However, the impact of the other major pathway triggered by intracellular DNA, the absent in melanoma 2 (AIM2) inflammasome, on the functional output from the stimulator of IFN genes (STING) pathway is poorly understood. We found that dendritic cells and macrophages deficient in AIM2, apoptosis-associated specklike protein, or caspase-1 produced markedly higher IFN-β in response to DNA. Biochemical analyses showed enhanced generation of cyclic GMP-AMP, STING aggregation, and TANK-binding kinase 1 and IFN regulatory factor 3 phosphorylation in inflammasome-deficient cells. Induction of pyroptosis by the AIM2 inflammasome was a major component of this effect, and inhibition of caspase-1 reduced cell death, augmenting phosphorylation of TANK-binding kinase 1/IFN regulatory factor 3 and production of IFN-β. Our data suggest that in vitro activation of the AIM2 inflammasome in murine macrophages and dendritic cells leads to reduced activation of the STING pathway, in part through promoting caspase-1-dependent cell death., (Copyright © 2016 by The American Association of Immunologists, Inc.)
- Published
- 2016
- Full Text
- View/download PDF
16. Systemic Tolerance Mediated by Melanoma Brain Tumors Is Reversible by Radiotherapy and Vaccination.
- Author
-
Jackson CM, Kochel CM, Nirschl CJ, Durham NM, Ruzevick J, Alme A, Francica BJ, Elias J, Daniels A, Dubensky TW Jr, Lauer P, Brockstedt DG, Baxi EG, Calabresi PA, Taube JM, Pardo CA, Brem H, Pardoll DM, Lim M, and Drake CG
- Subjects
- Animals, Antigens, Neoplasm administration & dosage, Antigens, Neoplasm immunology, Brain Neoplasms blood, Brain Neoplasms immunology, Brain Neoplasms radiotherapy, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes radiation effects, Central Nervous System Neoplasms blood, Central Nervous System Neoplasms immunology, Central Nervous System Neoplasms radiotherapy, Female, Humans, Melanoma, Experimental blood, Melanoma, Experimental immunology, Melanoma, Experimental radiotherapy, Mice, Microglia immunology, Microglia pathology, T-Lymphocytes, Cytotoxic immunology, Transforming Growth Factor beta antagonists & inhibitors, Vaccination, Brain Neoplasms therapy, Central Nervous System Neoplasms therapy, Immune Tolerance, Melanoma, Experimental therapy, Transforming Growth Factor beta blood
- Abstract
Purpose: Immune responses to antigens originating in the central nervous system (CNS) are generally attenuated, as collateral damage can have devastating consequences. The significance of this finding for the efficacy of tumor-targeted immunotherapies is largely unknown., Experimental Design: The B16 murine melanoma model was used to compare cytotoxic responses against established tumors in the CNS and in the periphery. Cytokine analysis of tissues from brain tumor-bearing mice detected elevated TGFβ secretion from microglia and in the serum and TGFβ signaling blockade reversed tolerance of tumor antigen-directed CD8 T cells. In addition, a treatment regimen using focal radiation therapy and recombinant Listeria monocytogenes was evaluated for immunologic activity and efficacy in this model., Results: CNS melanomas were more tolerogenic than equivalently progressed tumors outside the CNS as antigen-specific CD8 T cells were deleted and exhibited impaired cytotoxicity. Tumor-bearing mice had elevated serum levels of TGFβ; however, blocking TGFβ signaling with a small-molecule inhibitor or a monoclonal antibody did not improve survival. Conversely, tumor antigen-specific vaccination in combination with focal radiation therapy reversed tolerance and improved survival. This treatment regimen was associated with increased polyfunctionality of CD8 T cells, elevated T effector to T regulatory cell ratios, and decreased TGFβ secretion from microglia., Conclusions: These data suggest that CNS tumors may impair systemic antitumor immunity and consequently accelerate cancer progression locally as well as outside the CNS, whereas antitumor immunity may be restored by combining vaccination with radiation therapy. These findings are hypothesis-generating and warrant further study in contemporary melanoma models as well as human trials., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
17. Radiotherapy Combined with Novel STING-Targeting Oligonucleotides Results in Regression of Established Tumors.
- Author
-
Baird JR, Friedman D, Cottam B, Dubensky TW Jr, Kanne DB, Bambina S, Bahjat K, Crittenden MR, and Gough MJ
- Subjects
- Animals, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal immunology, Carcinoma, Pancreatic Ductal radiotherapy, Cell Line, Tumor, Combined Modality Therapy, Disease Models, Animal, Membrane Proteins biosynthesis, Membrane Proteins immunology, Mice, Mice, Inbred C3H, Mice, Inbred C57BL, Molecular Targeted Therapy, Oligonucleotides genetics, Pancreatic Neoplasms genetics, Pancreatic Neoplasms immunology, Pancreatic Neoplasms radiotherapy, Random Allocation, Tumor Microenvironment, Carcinoma, Pancreatic Ductal therapy, Membrane Proteins genetics, Oligonucleotides pharmacology, Pancreatic Neoplasms therapy
- Abstract
Cytotoxic therapies prime adaptive immune responses to cancer by stimulating the release of tumor-associated antigens. However, the tumor microenvironment into which these antigens are released is typically immunosuppressed, blunting the ability to initiate immune responses. Recently, activation of the DNA sensor molecule STING by cyclic dinucleotides was shown to stimulate infection-related inflammatory pathways in tumors. In this study, we report that the inflammatory pathways activated by STING ligands generate a powerful adjuvant activity for enhancing adaptive immune responses to tumor antigens released by radiotherapy. In a murine model of pancreatic cancer, we showed that combining CT-guided radiotherapy with a novel ligand of murine and human STING could synergize to control local and distant tumors. Mechanistic investigations revealed T-cell-independent and TNFα-dependent hemorrhagic necrosis at early times, followed by later CD8 T-cell-dependent control of residual disease. Clinically, STING was found to be expressed extensively in human pancreatic tumor and stromal cells. Our findings suggest that this novel STING ligand could offer a potent adjuvant for leveraging radiotherapeutic management of pancreatic cancer., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
18. Direct Activation of STING in the Tumor Microenvironment Leads to Potent and Systemic Tumor Regression and Immunity.
- Author
-
Corrales L, Glickman LH, McWhirter SM, Kanne DB, Sivick KE, Katibah GE, Woo SR, Lemmens E, Banda T, Leong JJ, Metchette K, Dubensky TW Jr, and Gajewski TF
- Subjects
- Animals, Antineoplastic Agents chemical synthesis, Blotting, Western, Cell Line, Tumor, Enzyme-Linked Immunosorbent Assay, Gene Knockout Techniques, Humans, Macrophages, Membrane Proteins immunology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Neoplasms, Experimental drug therapy, Nucleotides, Cyclic chemical synthesis, Polymerase Chain Reaction, Transfection, Xanthones pharmacology, Antineoplastic Agents pharmacology, Membrane Proteins antagonists & inhibitors, Neoplasms, Experimental immunology, Nucleotides, Cyclic pharmacology, Tumor Microenvironment immunology
- Abstract
Spontaneous tumor-initiated T cell priming is dependent on IFN-β production by tumor-resident dendritic cells. On the basis of recent observations indicating that IFN-β expression was dependent upon activation of the host STING pathway, we hypothesized that direct engagement of STING through intratumoral (IT) administration of specific agonists would result in effective anti-tumor therapy. After proof-of-principle studies using the mouse STING agonist DMXAA showed a potent therapeutic effect, we generated synthetic cyclic dinucleotide (CDN) derivatives that activated all human STING alleles as well as murine STING. IT injection of STING agonists induced profound regression of established tumors in mice and generated substantial systemic immune responses capable of rejecting distant metastases and providing long-lived immunologic memory. Synthetic CDNs have high translational potential as a cancer therapeutic., (Copyright © 2015 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
19. STING agonist formulated cancer vaccines can cure established tumors resistant to PD-1 blockade.
- Author
-
Fu J, Kanne DB, Leong M, Glickman LH, McWhirter SM, Lemmens E, Mechette K, Leong JJ, Lauer P, Liu W, Sivick KE, Zeng Q, Soares KC, Zheng L, Portnoy DA, Woodward JJ, Pardoll DM, Dubensky TW Jr, and Kim Y
- Subjects
- Animals, CD8-Positive T-Lymphocytes cytology, Cell Line, Tumor, Cytosol metabolism, Dendritic Cells cytology, Female, Humans, Interferon-gamma metabolism, Ligands, Mice, Mice, Inbred BALB C, Mice, Inbred C3H, Mice, Inbred C57BL, Monocytes cytology, NF-kappa B metabolism, Neoplasm Transplantation, Phosphates chemistry, Protein Serine-Threonine Kinases metabolism, STAT6 Transcription Factor metabolism, Antineoplastic Agents chemistry, Cancer Vaccines chemistry, Membrane Proteins agonists, Programmed Cell Death 1 Receptor metabolism
- Abstract
Stimulator of interferon genes (STING) is a cytosolic receptor that senses both exogenous and endogenous cytosolic cyclic dinucleotides (CDNs), activating TBK1/IRF3 (interferon regulatory factor 3), NF-κB (nuclear factor κB), and STAT6 (signal transducer and activator of transcription 6) signaling pathways to induce robust type I interferon and proinflammatory cytokine responses. CDN ligands were formulated with granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing cellular cancer vaccines--termed STINGVAX--that demonstrated potent in vivo antitumor efficacy in multiple therapeutic models of established cancer. We found that rationally designed synthetic CDN derivative molecules, including one with an Rp,Rp dithio diastereomer and noncanonical c[A(2',5')pA(3',5')p] phosphate bridge structure, enhanced antitumor efficacy of STINGVAX in multiple aggressive therapeutic models of established cancer in mice. Antitumor activity was STING-dependent and correlated with increased activation of dendritic cells and tumor antigen-specific CD8(+) T cells. Tumors from STINGVAX-treated mice demonstrated marked PD-L1 (programmed death ligand 1) up-regulation, which was associated with tumor-infiltrating CD8(+)IFNγ(+) T cells. When combined with PD-1 (programmed death 1) blockade, STINGVAX induced regression of palpable, poorly immunogenic tumors that did not respond to PD-1 blockade alone., (Copyright © 2015, American Association for the Advancement of Science.)
- Published
- 2015
- Full Text
- View/download PDF
20. Virological and preclinical characterization of a dendritic cell targeting, integration-deficient lentiviral vector for cancer immunotherapy.
- Author
-
Odegard JM, Kelley-Clarke B, Tareen SU, Campbell DJ, Flynn PA, Nicolai CJ, Slough MM, Vin CD, McGowan PJ, Nelson LT, Ter Meulen J, Dubensky TW Jr, and Robbins SH
- Subjects
- Animals, Carcinoma immunology, Cell Adhesion Molecules metabolism, Cell Line, Tumor, Clinical Trials, Phase I as Topic, Colonic Neoplasms immunology, Cytotoxicity, Immunologic, Dendritic Cells transplantation, Dendritic Cells virology, Genetic Engineering, Glycoproteins genetics, Glycoproteins metabolism, Humans, Immunologic Memory, Lectins, C-Type metabolism, Lymphocyte Activation, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Protein Binding, Receptors, Cell Surface metabolism, Receptors, Interleukin-7 metabolism, Viral Proteins genetics, Viral Proteins metabolism, Virus Integration genetics, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines, Carcinoma therapy, Colonic Neoplasms therapy, Dendritic Cells immunology, Genetic Vectors, Immunotherapy, Adoptive, Lentivirus genetics, Sindbis Virus genetics, Vaccinia immunology, Vaccinia virus immunology
- Abstract
Dendritic cells (DCs) are essential antigen-presenting cells for the initiation of cytotoxic T-cell responses and therefore attractive targets for cancer immunotherapy. We have developed an integration-deficient lentiviral vector termed ID-VP02 that is designed to deliver antigen-encoding nucleic acids selectively to human DCs in vivo. ID-VP02 utilizes a genetically and glycobiologically engineered Sindbis virus glycoprotein to target human DCs through the C-type lectin DC-SIGN (CD209) and also binds to the homologue murine receptor SIGNR1. Specificity of ID-VP02 for antigen-presenting cells in the mouse was confirmed through biodistribution studies showing that following subcutaneous administration, transgene expression was only detectable at the injection site and the draining lymph node. A single immunization with ID-VP02 induced a high level of antigen-specific, polyfunctional effector and memory CD8 T-cell responses that fully protected against vaccinia virus challenge. Upon homologous readministration, ID-VP02 induced a level of high-quality secondary effector and memory cells characterized by stable polyfunctionality and expression of IL-7Rα. Importantly, a single injection of ID-VP02 also induced robust cytotoxic responses against an endogenous rejection antigen of CT26 colon carcinoma cells and conferred both prophylactic and therapeutic antitumor efficacy. ID-VP02 is the first lentiviral vector which combines integration deficiency with DC targeting and is currently being investigated in a phase I trial in cancer patients.
- Published
- 2015
- Full Text
- View/download PDF
21. Attenuated Listeria monocytogenes vectors overcome suppressive plasma factors during HIV infection to stimulate myeloid dendritic cells to promote adaptive immunity and reactivation of latent virus.
- Author
-
Miller EA, Spadaccia MR, Norton T, Demmler M, Gopal R, O'Brien M, Landau N, Dubensky TW Jr, Lauer P, Brockstedt DG, and Bhardwaj N
- Subjects
- Adaptive Immunity immunology, CD8-Positive T-Lymphocytes immunology, Cells, Cultured, Coinfection immunology, HIV Infections virology, HIV-1 immunology, Humans, Interleukin-12 metabolism, Interleukin-6 metabolism, Myeloid Cells immunology, Th1 Cells immunology, Tumor Necrosis Factor-alpha metabolism, Virus Latency, gag Gene Products, Human Immunodeficiency Virus immunology, Dendritic Cells immunology, HIV Infections immunology, HIV-1 physiology, Listeria monocytogenes immunology, Virus Activation
- Abstract
HIV-1 infection is characterized by myeloid dendritic cell (DC) dysfunction, which blunts the responsiveness to vaccine adjuvants. We previously showed that nonviral factors in HIV-seropositive plasma are partially responsible for mediating this immune suppression. In this study we investigated recombinant Listeria monocytogenes (Lm) vectors, which naturally infect and potently activate DCs from seronegative donors, as a means to overcome DC dysfunction associated with HIV infection. Monocyte-derived DCs were cocultured with plasma from HIV-infected donors (HIV-moDCs) to induce a dysregulated state and infected with an attenuated, nonreplicative vaccine strain of Lm expressing full length clade B consensus gag (KBMA Lm-gag). Lm infection stimulated cytokine secretion [interleukin (IL)-12p70, tumor necrosis factor (TNF)-α, and IL-6] and Th-1 skewing of allogeneic naive CD4 T cells by HIV-moDCs, in contrast to the suppressive effects observed by HIV plasma on moDCs on toll-like receptor ligand stimulation. Upon coculture of "killed" but metabolically active (KBMA) Lm-gag-infected moDCs from HIV-infected donors with autologous cells, expansion of polyfunctional, gag-specific CD8(+) T cells was observed. Reactivation of latent proviruses by moDCs following Lm infection was also observed in models of HIV latency in a TNF-α-dependent manner. These findings reveal the unique ability of Lm vectors to contend with dysregulation of HIV-moDCs, while simultaneously possessing the capacity to activate latent virus. Concurrent stimulation of innate and adaptive immunity and disruption of latency may be an approach to reduce the pool of latently infected cells during HIV infection. Further study of Lm vectors as part of therapeutic vaccination and eradication strategies may advance this evolving field.
- Published
- 2015
- Full Text
- View/download PDF
22. A Listeria vaccine and depletion of T-regulatory cells activate immunity against early stage pancreatic intraepithelial neoplasms and prolong survival of mice.
- Author
-
Keenan BP, Saenger Y, Kafrouni MI, Leubner A, Lauer P, Maitra A, Rucki AA, Gunderson AJ, Coussens LM, Brockstedt DG, Dubensky TW Jr, Hassan R, Armstrong TD, and Jaffee EM
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, CD11b Antigen metabolism, Cancer Vaccines immunology, Carcinoma in Situ genetics, Carcinoma in Situ immunology, Carcinoma in Situ metabolism, Carcinoma in Situ pathology, Carcinoma, Pancreatic Ductal genetics, Carcinoma, Pancreatic Ductal immunology, Carcinoma, Pancreatic Ductal metabolism, Carcinoma, Pancreatic Ductal pathology, Cyclophosphamide pharmacology, Disease Models, Animal, Disease Progression, Forkhead Transcription Factors metabolism, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Inflammation Mediators metabolism, Integrases genetics, Integrases metabolism, Interferon-gamma metabolism, Interleukin-17 metabolism, Listeria monocytogenes genetics, Listeria monocytogenes metabolism, Mice, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Transgenic, Mutation, Pancreatic Neoplasms genetics, Pancreatic Neoplasms immunology, Pancreatic Neoplasms metabolism, Pancreatic Neoplasms pathology, Proto-Oncogene Proteins p21(ras) genetics, Proto-Oncogene Proteins p21(ras) metabolism, Receptors, Chemokine metabolism, T-Lymphocytes, Regulatory metabolism, Time Factors, Trans-Activators genetics, Trans-Activators metabolism, Tumor Suppressor Protein p53 genetics, Tumor Suppressor Protein p53 metabolism, Cancer Vaccines therapeutic use, Carcinoma in Situ drug therapy, Carcinoma, Pancreatic Ductal drug therapy, Listeria monocytogenes immunology, Pancreatic Neoplasms drug therapy, T-Lymphocytes, Regulatory immunology
- Abstract
Background & Aims: Premalignant lesions and early stage tumors contain immunosuppressive microenvironments that create barriers for cancer vaccines. Kras(G12D/+);Trp53(R172H/+);Pdx-1-Cre (KPC) mice, which express an activated form of Kras in pancreatic tissues, develop pancreatic intraepithelial neoplasms (PanIN) that progress to pancreatic ductal adenocarcinoma (PDA). We used these mice to study immune suppression in PDA., Methods: We immunized KPC and Kras(G12D/+);Pdx-1-Cre mice with attenuated intracellular Listeria monocytogenes (which induces CD4(+) and CD8(+) T-cell immunity) engineered to express Kras(G12D) (LM-Kras). The vaccine was given alone or in sequence with an anti-CD25 antibody (PC61) and cyclophosphamide to deplete T-regulatory (Treg) cells. Survival times were measured; pancreatic and spleen tissues were collected and analyzed by histologic, flow cytometry, and immunohistochemical analyses., Results: Interferon γ-mediated, CD8(+) T-cell responses were observed in KPC and Kras(G12D/+);Pdx-1-Cre mice given LM-Kras, but not in unvaccinated mice. Administration of LM-Kras to KPC mice 4-6 weeks old (with early stage PanINs), depleted of Treg cells, significantly prolonged survival and reduced PanIN progression (median survival, 265 days), compared with unvaccinated mice (median survival, 150 days; P = .002), mice given only LM-Kras (median survival, 150 days; P = .050), and unvaccinated mice depleted of Treg cells (median survival, 170 days; P = .048). In 8- to 12-week-old mice (with late-stage PanINs), LM-Kras, alone or in combination with Treg cell depletion, did not increase survival time or slow PanIN progression. The combination of LM-Kras and Treg cell depletion reduced numbers of Foxp3(+)CD4(+) T cells in pancreatic lymph nodes, increased numbers of CD4(+) T cells that secrete interleukin 17 and interferon γ, and caused CD11b(+)Gr1(+) cells in the pancreas to acquire an immunostimulatory phenotype., Conclusions: Immunization of KPC mice with Listeria monocytogenes engineered to express Kras(G12D), along with depletion of Treg cells, reduces progression of early stage, but not late-stage, PanINs. This approach increases infiltration of the lesion with inflammatory cells. It might be possible to design immunotherapies against premalignant pancreatic lesions to slow or prevent progression to PDA., (Copyright © 2014 AGA Institute. Published by Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
23. Design of a novel integration-deficient lentivector technology that incorporates genetic and posttranslational elements to target human dendritic cells.
- Author
-
Tareen SU, Kelley-Clarke B, Nicolai CJ, Cassiano LA, Nelson LT, Slough MM, Vin CD, Odegard JM, Sloan DD, Van Hoeven N, Allen JM, Dubensky TW Jr, and Robbins SH
- Subjects
- Genetic Vectors administration & dosage, HEK293 Cells, Humans, Immunity, Cellular immunology, Tissue Distribution, Antigens, Viral immunology, CD8-Positive T-Lymphocytes immunology, Dendritic Cells immunology, Lentivirus genetics, Sindbis Virus genetics, Viral Envelope Proteins genetics
- Abstract
As sentinels of the immune system, dendritic cells (DCs) play an essential role in regulating cellular immune responses. One of the main challenges of developing DC-targeted therapies includes the delivery of antigen to DCs in order to promote the activation of antigen-specific effector CD8 T cells. With the goal of creating antigen-directed immunotherapeutics that can be safely administered directly to patients, Immune Design has developed a platform of novel integration-deficient lentiviral vectors that target and deliver antigen-encoding nucleic acids to human DCs. This platform, termed ID-VP02, utilizes a novel genetic variant of a Sindbis virus envelope glycoprotein with posttranslational carbohydrate modifications in combination with Vpx, a SIVmac viral accessory protein, to achieve efficient targeting and transduction of human DCs. In addition, ID-VP02 incorporates safety features in its design that include two redundant mechanisms to render ID-VP02 integration-deficient. Here, we describe the characteristics that allow ID-VP02 to specifically transduce human DCs, and the advances that ID-VP02 brings to conventional third-generation lentiviral vector design as well as demonstrate upstream production yields that will enable manufacturing feasibility studies to be conducted.
- Published
- 2014
- Full Text
- View/download PDF
24. Rationale, progress and development of vaccines utilizing STING-activating cyclic dinucleotide adjuvants.
- Author
-
Dubensky TW Jr, Kanne DB, and Leong ML
- Abstract
A principal barrier to the development of effective vaccines is the availability of adjuvants and formulations that can elicit both effector and long-lived memory CD4 and CD8 T cells. Cellular immunity is the presumptive immune correlate of protection against intracellular pathogens: a group composed of bacteria, viruses and protozoans that is responsible for a staggering level of morbidity and mortality on a global scale. T-cell immunity is also correlated with clinical benefit in cancer, and the development of therapeutic strategies to harness the immune system to treat diverse malignancies is currently undergoing a renaissance. Cyclic dinucleotides (CDNs) are ubiquitous small molecule second messengers synthesized by bacteria that regulate diverse processes and are a relatively new class of adjuvants that have been shown to increase vaccine potency. CDNs activate innate immunity by directly binding the endoplasmic reticulum-resident receptor STING (stimulator of interferon genes), activating a signaling pathway that induces the expression of interferon-β (IFN-β) and also nuclear factor-κB (NF-κB) dependent inflammatory cytokines. The STING signaling pathway has emerged as a central Toll-like receptor (TLR) independent mediator of host innate defense in response to sensing cytosolic nucleic acids, either through direct binding of CDNs secreted by bacteria, or, as shown recently, through binding of a structurally distinct CDN produced by a host cell receptor in response to binding cytosolic double-stranded (ds)DNA. Although this relatively new class of adjuvants has to date only been evaluated in mice, newly available CDN-STING cocrystal structures will likely intensify efforts in this field towards further development and evaluation in human trials both in preventive vaccine and immunotherapy settings.
- Published
- 2013
- Full Text
- View/download PDF
25. Killed but metabolically active vaccines.
- Author
-
Dubensky TW Jr, Skoble J, Lauer P, and Brockstedt DG
- Subjects
- Animals, Cancer Vaccines immunology, Clinical Trials as Topic, DNA Repair, Ficusin, Humans, Leishmania immunology, Listeria monocytogenes genetics, Listeria monocytogenes immunology, Ultraviolet Rays, Vaccines, Attenuated immunology, Vaccines, Inactivated radiation effects, Vaccines, Inactivated supply & distribution, Vaccines, Synthetic genetics, Vaccines, Synthetic immunology, Vaccines, Inactivated immunology
- Abstract
Beginning in the 20th century and continuing into the new millennia, vaccines against numerous diseases have had an unquestioned principal role of both enhancing the quality of life and increasing life expectancy (Rappuoli R, Mandl CW, Black S, De Gregorio E: Vaccines for the twenty-first century society. Nat Rev Immunol 2011, 11:865-872). Despite this success and the development of sophisticated new vaccine technologies, there remain multiple infectious diseases including tuberculosis, malaria and AIDS that await an effective prophylactic vaccine. In addition, there have been recent clinical successes among individuals with cancer using vaccine treatment strategies-so-called therapeutic vaccines-that stimulate tumor specific immunity and increase survival (Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC, Dreicer R, Sims RB, et al.: Sipuleucel-T immunotherapy for castration-resistant prostate cancer. New Engl J Med 2010, 363:411-422). Here we summarize a new class of vaccines termed Killed But Metabolically Active (KBMA). KBMA vaccines are whole pathogenic or attenuated organisms killed through photochemical inactivation and cannot cause disease, yet retain sufficient metabolic activity to initiate a potent immune response. KBMA vaccines have two broad applications. First, recombinant KBMA vaccines encoding selected antigens relevant to infectious disease or cancer can be used to elicit a desired immune response. In the second application, KBMA vaccines can be derived from attenuated forms of a targeted pathogen, allowing for the presentation of the entire antigenic repertoire to the immune system, of particular importance when the correlates of protection are unknown., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2012
- Full Text
- View/download PDF
26. A live-attenuated Listeria vaccine (ANZ-100) and a live-attenuated Listeria vaccine expressing mesothelin (CRS-207) for advanced cancers: phase I studies of safety and immune induction.
- Author
-
Le DT, Brockstedt DG, Nir-Paz R, Hampl J, Mathur S, Nemunaitis J, Sterman DH, Hassan R, Lutz E, Moyer B, Giedlin M, Louis JL, Sugar EA, Pons A, Cox AL, Levine J, Murphy AL, Illei P, Dubensky TW Jr, Eiden JE, Jaffee EM, and Laheru DA
- Subjects
- Adult, Aged, Bacterial Vaccines adverse effects, Cancer Vaccines adverse effects, Carcinoma secondary, Carcinoma therapy, Cytokines blood, Female, Flow Cytometry, Humans, Immunohistochemistry, Liver Neoplasms immunology, Liver Neoplasms secondary, Lung Neoplasms pathology, Lung Neoplasms therapy, Lymphocyte Activation immunology, Lymphocyte Count, Male, Maximum Tolerated Dose, Mesothelin, Mesothelioma pathology, Mesothelioma therapy, Middle Aged, Ovarian Neoplasms pathology, Ovarian Neoplasms therapy, Pancreatic Neoplasms pathology, Pancreatic Neoplasms therapy, Vaccines, Attenuated administration & dosage, Vaccines, Attenuated adverse effects, Vaccines, Attenuated immunology, Bacterial Vaccines administration & dosage, Bacterial Vaccines immunology, Cancer Vaccines administration & dosage, Cancer Vaccines immunology, GPI-Linked Proteins immunology, Listeria monocytogenes immunology, Liver Neoplasms therapy
- Abstract
Purpose: Listeria monocytogenes (Lm)-based vaccines stimulate both innate and adaptive immunity. ANZ-100 is a live-attenuated Lm strain (Lm ΔactA/ΔinlB). Uptake by phagocytes in the liver results in local inflammatory responses and activation and recruitment of natural killer (NK) and T cells, in association with increased survival of mice bearing hepatic metastases. The Lm ΔactA/ΔinlB strain, engineered to express human mesothelin (CRS-207), a tumor-associated antigen expressed by a variety of tumors, induces mesothelin-specific T-cell responses against mesothelin-expressing murine tumors. These two phase I studies test ANZ-100 and CRS-207 in subjects with liver metastases and mesothelin-expressing cancers, respectively., Experimental Design: A single intravenous injection of ANZ-100 was evaluated in a dose escalation study in subjects with liver metastases. Nine subjects received 1 × 10(6), 3 × 10(7), or 3 × 10(8) colony-forming units (cfu). CRS-207 was evaluated in a dose-escalation study in subjects with mesothelioma, lung, pancreatic, or ovarian cancers. Seventeen subjects received up to 4 doses of 1 × 10(8), 3 × 10(8), 1 × 10(9), or 1 × 10(10) cfu., Results: A single infusion of ANZ-100 was well tolerated to the maximum planned dose. Adverse events included transient laboratory abnormalities and symptoms associated with cytokine release. Multiple infusions of CRS-207 were well tolerated up to 1 × 10(9) cfu, the determined maximum tolerated dose. Immune activation was observed for both ANZ-100 and CRS-207 as measured by serum cytokine/chemokine levels and NK cell activation. In the CRS-207 study, listeriolysin O and mesothelin-specific T-cell responses were detected and 37% of subjects lived ≥15 months., Conclusions: ANZ-100 and CRS-207 administration was safe and resulted in immune activation.
- Published
- 2012
- Full Text
- View/download PDF
27. Adjuvants for cancer vaccines.
- Author
-
Dubensky TW Jr and Reed SG
- Subjects
- Clinical Trials as Topic, Female, Humans, Immunity, Innate, Immunotherapy methods, Male, Neoplasms therapy, Saponins administration & dosage, Saponins immunology, Adjuvants, Immunologic administration & dosage, Cancer Vaccines administration & dosage, Cancer Vaccines immunology, Neoplasms immunology
- Abstract
The recent FDA approval of sipuleucel-T (Provenge), a patient-specific immunotherapy for androgen-independent prostate cancer developed by Dendreon Corporation, has provided support for the concept of cellular immunotherapy as an approach to cancer treatment. Adjuvants are compounds that enhance the potency of the antigen-specific immune response and can be an essential component of an efficacious vaccine. Cervarix is a prophylactic vaccine against human papilloma virus (HPV) types 16 and 18, which can cause cervical cancer, and recently received approval from the FDA, due in part to the protective immunity it conferred against not only HPV types contained in the vaccine but in addition to oncogenic HPV strains that were not contained in the vaccine. Cervarix is formulated with MPL (monophosphoryl lipid A), a TLR-4 targeted adjuvant shown to promote immune response broadening. The recent FDA approvals of these pioneering vaccines are landmark events, and will likely usher in renewed interest and investment in the development of new therapeutic cancer vaccine candidates. In this review, we examine new molecularly defined adjuvants and formulations and its application to cancer vaccines under development., (Copyright 2010 Elsevier Ltd. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
28. Priming and activation of human ovarian and breast cancer-specific CD8+ T cells by polyvalent Listeria monocytogenes-based vaccines.
- Author
-
Sinnathamby G, Lauer P, Zerfass J, Hanson B, Karabudak A, Krakover J, Secord AA, Clay TM, Morse MA, Dubensky TW Jr, Brockstedt DG, Philip R, and Giedlin M
- Subjects
- Antigen Presentation, Antigens, Neoplasm genetics, Antigens, Neoplasm metabolism, Breast Neoplasms immunology, Breast Neoplasms pathology, CD8-Positive T-Lymphocytes pathology, Cell Line, Tumor, Chromatography, High Pressure Liquid, Cloning, Molecular, Epitope Mapping, Epitopes, T-Lymphocyte genetics, Epitopes, T-Lymphocyte metabolism, Female, HLA-A2 Antigen metabolism, Humans, Listeriosis immunology, Lymphocyte Activation, Monocytes immunology, Monocytes metabolism, Monocytes microbiology, Ovarian Neoplasms immunology, Ovarian Neoplasms pathology, Breast Neoplasms therapy, CD8-Positive T-Lymphocytes immunology, Cancer Vaccines, Listeria monocytogenes immunology, Ovarian Neoplasms therapy
- Abstract
Immunotherapeutic vaccine is potentially an effective strategy to combat cancer. Essential components of an effective vaccine must include antigens that are processed by the major histocompatibility complex class I pathway, presented by the tumor major histocompatibility complex molecules, and an effective antigen delivery platform that is capable of breaking self-tolerance. In this study, we characterized a set of ovarian cancer-specific T-cell epitopes delivered by live-attenuated recombinant Listeria monocytogenes (Lm DeltaactADeltainlB) as a vaccine vector. We present data that peptide-specific T cells recognize the human monocytic cell line THP-1 infected with recombinant Lm DeltaactADeltainlB encoding the epitopes. Furthermore, we demonstrate that recombinant L. monocytogenes (Lm)-infected antigen-presenting cells can prime and expand epitope-specific CD8 T cells in vitro and such CD8 T cells recognize not only peptide-loaded targets but also ovarian and breast tumor cells presenting endogenous epitopes. Finally, peptide-specific T cells generated using peripheral blood mononuclear cell from ovarian cancer patients recognize target cells infected with recombinant Lm DeltaactADeltainlB encoding the epitopes. Our results demonstrate that live-attenuated recombinant Lm can be used effectively as a vehicle to deliver cancer peptide antigens singly or as a multiepitope construct. Thus, the use of recombinant live-attenuated Lm strains encoding endogenously processed and presented tumor epitopes/antigens represents an attractive strategy for active cancer immunotherapy in a clinical setting.
- Published
- 2009
- Full Text
- View/download PDF
29. Impact of preexisting vector-specific immunity on vaccine potency: characterization of listeria monocytogenes-specific humoral and cellular immunity in humans and modeling studies using recombinant vaccines in mice.
- Author
-
Leong ML, Hampl J, Liu W, Mathur S, Bahjat KS, Luckett W, Dubensky TW Jr, and Brockstedt DG
- Subjects
- Adult, Animals, Bacterial Toxins immunology, Cell Line, Female, Genetic Vectors, Heat-Shock Proteins immunology, Hemolysin Proteins immunology, Humans, Interleukin-2 biosynthesis, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, T-Lymphocytes immunology, Vaccines, Attenuated immunology, Antibodies, Bacterial blood, Bacterial Vaccines immunology, Listeria monocytogenes immunology, Vaccines, Synthetic immunology
- Abstract
Recombinant live-attenuated Listeria monocytogenes is currently being developed as a vaccine platform for treatment or prevention of malignant and infectious diseases. The effectiveness of complex biologic vaccines, such as recombinant viral and bacterial vectors, can be limited by either preexisting or vaccine-induced vector-specific immunity. We characterized the level of L. monocytogenes-specific cellular and humoral immunity present in more than 70 healthy adult subjects as a first step to understanding its possible impact on the efficacy of L. monocytogenes-based vaccines being evaluated in early-phase clinical trials. Significant L. monocytogenes-specific humoral immunity was not measured in humans, consistent with a lack of antibodies in mice immunized with wild-type L. monocytogenes. Cellular immune responses specific for listeriolysin O, a secreted bacterial protein required for potency of L. monocytogenes-derived vaccines, were detected in approximately 60% of human donors tested. In mice, while wild-type L. monocytogenes did not induce significant humoral immunity, attenuated L. monocytogenes vaccine strains induced high-titer L. monocytogenes-specific antibodies when given at high doses used for immunization. Passive transfer of L. monocytogenes-specific antiserum to naïve mice had no impact on priming antigen-specific immunity in mice immunized with a recombinant L. monocytogenes vaccine. In mice with preexisting L. monocytogenes-specific immunity, priming of naïve T cells was not prevented, and antigen-specific responses could be boosted by additional vaccinations. For the first time, our findings establish the level of L. monocytogenes-specific cellular immunity in healthy adults, and, together with modeling studies performed with mice, they support the scientific rationale for repeated L. monocytogenes vaccine immunization regimens to elicit a desired therapeutic effect.
- Published
- 2009
- Full Text
- View/download PDF
30. Killed but metabolically active Bacillus anthracis vaccines induce broad and protective immunity against anthrax.
- Author
-
Skoble J, Beaber JW, Gao Y, Lovchik JA, Sower LE, Liu W, Luckett W, Peterson JW, Calendar R, Portnoy DA, Lyons CR, and Dubensky TW Jr
- Subjects
- Animals, Anthrax microbiology, Anthrax prevention & control, Anthrax Vaccines administration & dosage, Anthrax Vaccines genetics, Antigens, Bacterial immunology, Female, Furocoumarins, Guinea Pigs, Immunity, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Inbred DBA, Mutation, Rabbits, Spores, Bacterial genetics, Ultraviolet Rays, Vaccination, Vaccines, Inactivated administration & dosage, Vaccines, Inactivated genetics, Virulence, Anthrax immunology, Anthrax Vaccines immunology, Antibodies, Bacterial blood, Bacillus anthracis genetics, Bacillus anthracis immunology, Bacillus anthracis pathogenicity, Bacillus anthracis radiation effects, Vaccines, Inactivated immunology
- Abstract
Bacillus anthracis is the causative agent of anthrax. We have developed a novel whole-bacterial-cell anthrax vaccine utilizing B. anthracis that is killed but metabolically active (KBMA). Vaccine strains that are asporogenic and nucleotide excision repair deficient were engineered by deleting the spoIIE and uvrAB genes, rendering B. anthracis extremely sensitive to photochemical inactivation with S-59 psoralen and UV light. We also introduced point mutations into the lef and cya genes, which allowed inactive but immunogenic toxins to be produced. Photochemically inactivated vaccine strains maintained a high degree of metabolic activity and secreted protective antigen (PA), lethal factor, and edema factor. KBMA B. anthracis vaccines were avirulent in mice and induced less injection site inflammation than recombinant PA adsorbed to aluminum hydroxide gel. KBMA B. anthracis-vaccinated animals produced antibodies against numerous anthrax antigens, including high levels of anti-PA and toxin-neutralizing antibodies. Vaccination with KBMA B. anthracis fully protected mice against challenge with lethal doses of toxinogenic unencapsulated Sterne 7702 spores and rabbits against challenge with lethal pneumonic doses of fully virulent Ames strain spores. Guinea pigs vaccinated with KBMA B. anthracis were partially protected against lethal Ames spore challenge, which was comparable to vaccination with the licensed vaccine anthrax vaccine adsorbed. These data demonstrate that KBMA anthrax vaccines are well tolerated and elicit potent protective immune responses. The use of KBMA vaccines may be broadly applicable to bacterial pathogens, especially those for which the correlates of protective immunity are unknown.
- Published
- 2009
- Full Text
- View/download PDF
31. Constitutive Activation of the PrfA regulon enhances the potency of vaccines based on live-attenuated and killed but metabolically active Listeria monocytogenes strains.
- Author
-
Lauer P, Hanson B, Lemmens EE, Liu W, Luckett WS, Leong ML, Allen HE, Skoble J, Bahjat KS, Freitag NE, Brockstedt DG, and Dubensky TW Jr
- Subjects
- Amino Acid Substitution genetics, Animals, Antigens genetics, Antigens, Bacterial biosynthesis, Antigens, Bacterial immunology, Bacterial Vaccines genetics, Female, Immunization, Secondary, Injections, Intramuscular, Injections, Intravenous, Lethal Dose 50, Listeria monocytogenes genetics, Listeria monocytogenes pathogenicity, Listeriosis prevention & control, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mutation, Missense, Recombinant Proteins biosynthesis, Recombinant Proteins genetics, Recombinant Proteins immunology, Regulon, Vaccinia prevention & control, Virulence, Virulence Factors biosynthesis, Virulence Factors immunology, Antigens biosynthesis, Antigens immunology, Bacterial Vaccines immunology, Listeria monocytogenes immunology, Peptide Termination Factors genetics
- Abstract
Recombinant vaccines derived from the facultative intracellular bacterium Listeria monocytogenes are presently undergoing early-stage clinical evaluation in oncology treatment settings. This effort has been stimulated in part due to preclinical results that illustrate potent activation of innate and adaptive immune effectors by L. monocytogenes vaccines, combined with efficacy in rigorous animal models of malignant and infectious disease. Here, we evaluated the immunologic potency of a panel of isogenic vaccine strains that varied only in prfA. PrfA is an intracellularly activated transcription factor that induces expression of virulence genes and encoded heterologous antigens (Ags) in appropriately engineered vaccine strains. Mutant strains with PrfA locked into a constitutively active state are known as PrfA* mutants. We assessed the impacts of three PrfA* mutants, G145S, G155S, and Y63C, on the immunologic potencies of live-attenuated and photochemically inactivated nucleotide excision repair mutant (killed but metabolically active [KBMA]) vaccines. While PrfA* substantially increased Ag expression in strains grown in broth culture, Ag expression levels were equivalent in infected macrophage and dendritic cell lines, conditions that more closely parallel those in the immunized host. However, only the prfA(G155S) allele conferred significantly enhanced vaccine potency to KBMA vaccines. In the KBMA vaccine background, we show that PrfA*(G155S) enhanced functional cellular immunity following an intravenous or intramuscular prime-boost immunization regimen. These results form the basis of a rationale for including the prfA(G155S) allele in future live-attenuated or KBMA L. monocytogenes vaccines advanced to the clinical setting.
- Published
- 2008
- Full Text
- View/download PDF
32. Listeria monocytogenes multidrug resistance transporters activate a cytosolic surveillance pathway of innate immunity.
- Author
-
Crimmins GT, Herskovits AA, Rehder K, Sivick KE, Lauer P, Dubensky TW Jr, and Portnoy DA
- Subjects
- ATP Binding Cassette Transporter, Subfamily B genetics, Animals, Bacterial Proteins genetics, Cytosol immunology, Genes, Bacterial, Genes, MDR, Interferon-beta biosynthesis, Listeria monocytogenes genetics, Listeria monocytogenes pathogenicity, Macrophages immunology, Macrophages microbiology, Mice, Mice, Inbred C57BL, Mice, Knockout, Mutation, Receptor, Interferon alpha-beta deficiency, Receptor, Interferon alpha-beta genetics, ATP Binding Cassette Transporter, Subfamily B immunology, Bacterial Proteins immunology, Immunity, Innate, Listeria monocytogenes immunology
- Abstract
To gain insight into the interaction of intracellular pathogens with host innate immune pathways, we performed an unbiased genetic screen of Listeria monocytogenes mutants that induced an enhanced or diminished host innate immune response. Here, we show that the major facilitator superfamily of bacterial multidrug resistance transporters (MDRs) controlled the magnitude of a host cytosolic surveillance pathway, leading to the production of several cytokines, including type I IFN. Mutations mapping to repressors of MDRs resulted in ectopic expression of their cognate transporters, leading to host responses that were increased up to 20-fold over wild-type bacteria, and a 20-fold decrease in bacterial growth in vivo. Mutation of one of the MDRs, MdrM, led to a 3-fold reduction in the IFN-beta response to L. monocytogenes infection, indicating a pivotal role for MdrM in activation of the host cytosolic surveillance system. Bacterial MDRs had previously been associated with resistance to antibiotics and other toxic compounds. This report links bacterial MDRs and host immunity. Understanding the mechanisms through which live pathogens activate innate immune signaling pathways should lead to the discovery of adjuvants, vaccines, and perhaps new classes of therapeutics. Indeed, we show that the mutants identified in this screen induced vastly altered type I IFN response in vivo as well.
- Published
- 2008
- Full Text
- View/download PDF
33. Activation of immature hepatic NK cells as immunotherapy for liver metastatic disease.
- Author
-
Bahjat KS, Prell RA, Allen HE, Liu W, Lemmens EE, Leong ML, Portnoy DA, Dubensky TW Jr, Brockstedt DG, and Giedlin MA
- Subjects
- Animals, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Cell Differentiation immunology, Cell Line, Tumor, Disease Models, Animal, Immunity, Cellular, Immunity, Innate, Interferon-gamma biosynthesis, Ligands, Listeriosis immunology, Liver Neoplasms secondary, Liver Neoplasms therapy, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, NK Cell Lectin-Like Receptor Subfamily K, Receptors, Immunologic immunology, Receptors, Immunologic therapeutic use, Receptors, Natural Killer Cell, Immunotherapy methods, Killer Cells, Natural immunology, Listeria monocytogenes immunology, Liver Neoplasms immunology
- Abstract
NK cells can identify and eliminate emerging tumors due to altered expression of activating and inhibitory ligands on aberrant cells, a process that is greatly enhanced following NK cell activation. As a principal site of both tumor metastases and immature NK cells, the liver represents a unique anatomic location in which activation of the innate immune system could provide substantial therapeutic benefit. We describe here the NK cell-dependent destruction of a primary hepatic tumor following infection with an attenuated intracellular bacterium derived from Listeria monocytogenes. NK cell-mediated immunity correlated with the ordered migration and maturation of NK cells within the liver. Cytolytic activity was partially dependent on NKG2D-mediated tumor cell recognition, but surprisingly was still effective in the absence of type I IFN. Significantly, NK cell-mediated destruction of a primary hepatic tumor in infected mice led to long-lived CD4- and CD8 T cell-dependent tumor-specific adaptive immunity. These findings establish that activation and differentiation of immature NK cells using complex microbial stimuli can elicit potent anti-tumor activity within the liver, promote cross-presentation of tumor-derived Ags leading to long-lived systemic anti-tumor immunity, and suggests a paradigm for clinical intervention of liver metastatic carcinoma.
- Published
- 2007
- Full Text
- View/download PDF
34. Role of PD-1 and its ligand, B7-H1, in early fate decisions of CD8 T cells.
- Author
-
Goldberg MV, Maris CH, Hipkiss EL, Flies AS, Zhen L, Tuder RM, Grosso JF, Harris TJ, Getnet D, Whartenby KA, Brockstedt DG, Dubensky TW Jr, Chen L, Pardoll DM, and Drake CG
- Subjects
- Animals, Autoantigens, B7-H1 Antigen, Cell Differentiation, Mice, Programmed Cell Death 1 Ligand 2 Protein, Programmed Cell Death 1 Receptor, Protein Binding, Self Tolerance, T-Lymphocytes, Cytotoxic, Antigens, Surface physiology, Apoptosis Regulatory Proteins physiology, B7-1 Antigen physiology, CD8-Positive T-Lymphocytes immunology, Membrane Glycoproteins physiology, Peptides physiology
- Abstract
Expression of the PD-1 receptor on T cells has been shown to provide an important inhibitory signal that down-modulates peripheral effector responses in normal tissues and tumors. Furthermore, PD-1 up-regulation on chronically activated T cells can maintain them in a partially reversible inactive state. The function of PD-1 in the very early stages of T-cell response to antigen in vivo has not been fully explored. In this study, we evaluate the role of PD-1 and its 2 B7 family ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), in early fate decisions of CD8 T cells. We show that CD8 T cells specific for influenza hemagglutinin (HA) expressed as a self-antigen become functionally tolerized and express high levels of surface PD-1 by the time of their first cell division. Blockade of PD-1 or B7-H1, but not B7-DC, at the time of self-antigen encounter mitigates tolerance induction and results in CD8 T-cell differentiation into functional cytolytic T lymphocytes (CTLs). These findings demonstrate that, in addition to modulating effector functions in the periphery, B7-H1:PD-1 interactions regulate early T-cell-fate decisions.
- Published
- 2007
- Full Text
- View/download PDF
35. Inactivation of parvovirus B19 in human platelet concentrates by treatment with amotosalen and ultraviolet A illumination.
- Author
-
Sawyer L, Hanson D, Castro G, Luckett W, Dubensky TW Jr, and Stassinopoulos A
- Subjects
- DNA, Viral analysis, DNA, Viral genetics, Erythema Infectiosum prevention & control, Erythroid Precursor Cells virology, Furocoumarins pharmacology, Genome, Viral genetics, Humans, Immunoassay, Photochemistry, Blood Platelets virology, Parvovirus B19, Human genetics, Ultraviolet Rays, Virus Inactivation drug effects, Virus Inactivation radiation effects
- Abstract
Background: The human erythrovirus B19 (B19) is a small (18- to 26-nm) nonenveloped virus with a single-stranded DNA genome of 5.6 kb. B19 is clinically significant and is also generally resistant to pathogen inactivation methods. Photochemical treatment (PCT) with amotosalen and ultraviolet A (UVA) inactivates viruses, bacteria, and protozoa in platelets (PLTs) and plasma prepared for transfusion. In this study, the capacity of PCT to inactivate B19 in human PLT concentrates was evaluated., Study Design and Methods: B19 inactivation was measured by a novel enzyme-linked immunosorbent spot (ELISPOT) erythroid progenitor cell infectivity assay and by inhibition of long-range (up to 4.3 kb) polymerase chain reaction (PCR), under conditions where the whole coding region of the viral genome was amplified. B19-infected plasma was used to test whether incubation of amotosalen with virus before PCT enhanced inactivation compared to immediate PCT., Results: Inactivation of up to 5.8 log of B19 as measured by the infectivity assay, or up to 6 logs as measured by PCR inhibition can be achieved under non-limiting conditions. Inactivation efficacy was found to increase with incubation prior to UVA illumination. Without incubation prior to illumination 2.1 +0.4 log was inactivated as determined by infectivity assay. When measured by PCR inhibition, inactivation varied inversely with amplicon size. When primers that spanned the entire coding region of the B19 genome were used, maximum inhibition of PCR amplification was demonstrated., Conclusion: Under defined conditions, PCT with amotosalen combined with UVA light can be used to inactivate B19, a clinically significant virus that can be transmitted through blood transfusion, and heretofore has been demonstrated to be refractory to inactivation.
- Published
- 2007
- Full Text
- View/download PDF
36. Cytosolic entry controls CD8+-T-cell potency during bacterial infection.
- Author
-
Bahjat KS, Liu W, Lemmens EE, Schoenberger SP, Portnoy DA, Dubensky TW Jr, and Brockstedt DG
- Subjects
- Animals, Bacterial Toxins genetics, Dose-Response Relationship, Immunologic, Female, Heat-Shock Proteins deficiency, Heat-Shock Proteins genetics, Hemolysin Proteins deficiency, Hemolysin Proteins genetics, Listeria monocytogenes genetics, Listeriosis immunology, Listeriosis microbiology, Listeriosis prevention & control, Lymphocyte Activation genetics, Mice, Mice, Inbred C57BL, Bacterial Toxins immunology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes microbiology, Cytosol immunology, Cytosol microbiology, Heat-Shock Proteins immunology, Hemolysin Proteins immunology, Listeria monocytogenes immunology
- Abstract
Interaction with host immunoreceptors during microbial infection directly impacts the magnitude of the ensuing innate immune response. How these signals affect the quality of the adaptive T-cell response remains poorly understood. Utilizing an engineered strain of the intracellular pathogen Listeria monocytogenes that infects cells but fails to escape from the phagosome, we demonstrate the induction of long-lived memory T cells that are capable of secondary expansion and effector function but are incapable of providing protective immunity. We demonstrate that microbial invasion of the cytosol is required for dendritic cell activation and integration of CD40 signaling, ultimately determining the ability of the elicited CD8+-T-cell pool to protect against lethal wild-type L. monocytogenes challenge. These results reveal a crucial role for phagosomal escape, not for delivery of antigen to the class I major histocompatibility complex pathway but for establishing the appropriate cellular context during CD8+-T-cell priming.
- Published
- 2006
- Full Text
- View/download PDF
37. Killed but metabolically active microbes: a new vaccine paradigm for eliciting effector T-cell responses and protective immunity.
- Author
-
Brockstedt DG, Bahjat KS, Giedlin MA, Liu W, Leong M, Luckett W, Gao Y, Schnupf P, Kapadia D, Castro G, Lim JY, Sampson-Johannes A, Herskovits AA, Stassinopoulos A, Bouwer HG, Hearst JE, Portnoy DA, Cook DN, and Dubensky TW Jr
- Subjects
- Animals, Carbon Radioisotopes, DNA Repair genetics, Dendritic Cells, Endodeoxyribonucleases genetics, Escherichia coli Proteins genetics, Ficusin, Flow Cytometry, Listeria monocytogenes genetics, Mice, Mice, Inbred C57BL, Ultraviolet Rays, Bacterial Vaccines immunology, Immunity, Cellular immunology, Listeria monocytogenes immunology, Vaccination methods
- Abstract
We developed a new class of vaccines, based on killed but metabolically active (KBMA) bacteria, that simultaneously takes advantage of the potency of live vaccines and the safety of killed vaccines. We removed genes required for nucleotide excision repair (uvrAB), rendering microbial-based vaccines exquisitely sensitive to photochemical inactivation with psoralen and long-wavelength ultraviolet light. Colony formation of the nucleotide excision repair mutants was blocked by infrequent, randomly distributed psoralen crosslinks, but the bacterial population was able to express its genes, synthesize and secrete proteins. Using the intracellular pathogen Listeria monocytogenes as a model platform, recombinant psoralen-inactivated Lm DeltauvrAB vaccines induced potent CD4(+) and CD8(+) T-cell responses and protected mice against virus challenge in an infectious disease model and provided therapeutic benefit in a mouse cancer model. Microbial KBMA vaccines used either as a recombinant vaccine platform or as a modified form of the pathogen itself may have broad use for the treatment of infectious disease and cancer.
- Published
- 2005
- Full Text
- View/download PDF
38. Listeria-based cancer vaccines that segregate immunogenicity from toxicity.
- Author
-
Brockstedt DG, Giedlin MA, Leong ML, Bahjat KS, Gao Y, Luckett W, Liu W, Cook DN, Portnoy DA, and Dubensky TW Jr
- Subjects
- Animals, Bacterial Proteins genetics, Bacterial Proteins immunology, Cell Line, Cell Survival drug effects, Cells, Cultured, Female, Gene Deletion, Hepatocytes drug effects, Humans, Immunologic Memory, Listeria monocytogenes genetics, Lung Neoplasms secondary, Membrane Proteins deficiency, Membrane Proteins genetics, Membrane Proteins immunology, Mice, Mice, Inbred BALB C, Monocytes drug effects, RNA, Bacterial genetics, RNA, Transfer, Arg genetics, T-Lymphocytes drug effects, Cancer Vaccines immunology, Cancer Vaccines toxicity, Colonic Neoplasms immunology, Hepatocytes immunology, Listeria monocytogenes immunology, Lung Neoplasms immunology, Monocytes immunology, T-Lymphocytes immunology
- Abstract
The facultative intracellular bacterium Listeria monocytogenes is being developed as a cancer vaccine platform because of its ability to induce potent innate and adaptive immunity. For successful clinical application, it is essential to develop a Listeria platform strain that is safe yet retains the potency of vaccines based on wild-type bacteria. Here, we report the development of a recombinant live-attenuated vaccine platform strain that retains the potency of the fully virulent pathogen, combined with a >1,000-fold reduction in toxicity, as compared with wild-type Listeria. By selectively deleting two virulence factors, ActA (DeltaactA) and Internalin B (DeltainlB), the immunopotency of Listeria was maintained and its toxicity was diminished in vivo, largely by blocking the direct internalin B-mediated infection of nonphagocytic cells, such as hepatocytes, and the indirect ActA-mediated infection by cell-to-cell spread from adjacent phagocytic cells. In contrast, infection of phagocytic cells was not affected, leaving intact the ability of Listeria to stimulate innate immunity and to induce antigenspecific cellular responses. Listeria DeltaactA/DeltainlB-based vaccines were rapidly cleared from mice after immunization and induced potent and durable effector and memory T-cell responses with no measurable liver toxicity. Therapeutic vaccination of BALB/c mice bearing murine CT26 colon tumor lung metastases or palpable s.c. tumors (>100 mm(3)) with recombinant Listeria DeltaactA/DeltainlB expressing an endogenous tumor antigen resulted in breaking of self-tolerance and long-term survival. We propose that recombinant Listeria DeltaactA/DeltainlB expressing human tumor-associated antigens represents an attractive therapeutic strategy for further development and testing in human clinical trials.
- Published
- 2004
- Full Text
- View/download PDF
39. Vesicular stomatitis virus: an exciting new therapeutic oncolytic virus candidate for cancer or just another chapter from Field's Virology?
- Author
-
Giedlin MA, Cook DN, and Dubensky TW Jr
- Subjects
- Animals, Humans, Interferon-beta antagonists & inhibitors, Mutation, Neoplasms therapy, Neoplasms virology, RNA Virus Infections virology, Signal Transduction, Interferon-beta metabolism, RNA Virus Infections therapy, Vesicular stomatitis Indiana virus physiology, Viruses metabolism
- Abstract
Selected mutant strains of vesicular stomatitis virus (VSV) are described that are unable to combat endogenous IFN-beta signaling within infected normal cells and as a result are dramatically more selective for productive growth in tumor cells having a defective antiviral response. The VSV mutants may have the potential to be used clinically as a systemic oncolytic agent for the treatment of distal and metastatic cancers.
- Published
- 2003
- Full Text
- View/download PDF
40. An alphavirus replicon particle chimera derived from venezuelan equine encephalitis and sindbis viruses is a potent gene-based vaccine delivery vector.
- Author
-
Perri S, Greer CE, Thudium K, Doe B, Legg H, Liu H, Romero RE, Tang Z, Bin Q, Dubensky TW Jr, Vajdy M, Otten GR, and Polo JM
- Subjects
- Amino Acid Sequence, Animals, CD8-Positive T-Lymphocytes immunology, Chimera, Cricetinae, Gene Products, gag genetics, Gene Products, gag immunology, Interferons pharmacology, Mice, Mice, Inbred BALB C, Molecular Sequence Data, Protein Precursors genetics, Protein Precursors immunology, RNA, Viral biosynthesis, Virus Assembly, AIDS Vaccines immunology, Encephalitis Virus, Venezuelan Equine genetics, Genetic Vectors, Replicon, Sindbis Virus genetics, Vaccines, Synthetic immunology
- Abstract
Alphavirus replicon particle-based vaccine vectors derived from Sindbis virus (SIN), Semliki Forest virus, and Venezuelan equine encephalitis virus (VEE) have been shown to induce robust antigen-specific cellular, humoral, and mucosal immune responses in many animal models of infectious disease and cancer. However, since little is known about the relative potencies among these different vectors, we compared the immunogenicity of replicon particle vectors derived from two very different parental alphaviruses, VEE and SIN, expressing a human immunodeficiency virus type 1 p55(Gag) antigen. Moreover, to explore the potential benefits of combining elements from different alphaviruses, we generated replicon particle chimeras of SIN and VEE. Two distinct strategies were used to produce particles with VEE-p55(gag) replicon RNA packaged within SIN envelope glycoproteins and SIN-p55(gag) replicon RNA within VEE envelope glycoproteins. Each replicon particle configuration induced Gag-specific CD8(+) T-cell responses in murine models when administered alone or after priming with DNA. However, Gag-specific responses varied dramatically, with the strongest responses to this particular antigen correlating with the VEE replicon RNA, irrespective of the source of envelope glycoproteins. Comparing the replicons with respect to heterologous gene expression levels and sensitivity to alpha/beta interferon in cultured cells indicated that each might contribute to potency differences. This work shows that combining desirable elements from VEE and SIN into a replicon particle chimera may be a valuable approach toward the goal of developing vaccine vectors with optimal in vivo potency, ease of production, and safety.
- Published
- 2003
- Full Text
- View/download PDF
41. Generation of retroviral packaging and producer cell lines for large-scale vector production with improved safety and titer.
- Author
-
Dubensky TW Jr and Sauter SL
- Subjects
- Animals, Cell Line, Gene Products, env genetics, Gene Products, env metabolism, Gene Products, gag genetics, Gene Products, gag metabolism, Gene Products, pol genetics, Gene Products, pol metabolism, Gene Transfer Techniques, Genetic Therapy methods, Humans, Membrane Glycoproteins genetics, Membrane Glycoproteins metabolism, Mice, Retroviridae physiology, Viral Envelope Proteins genetics, Viral Envelope Proteins metabolism, Virus Cultivation, Virus Replication, Genetic Vectors, Moloney murine leukemia virus genetics, Retroviridae genetics, Virus Assembly
- Published
- 2003
- Full Text
- View/download PDF
42. A highly efficient gene delivery system derived from feline immunodeficiency virus (FIV).
- Author
-
Sauter SL, Gasmi M, and Dubensky TW Jr
- Subjects
- Animals, Cats, Cell Line, Genetic Therapy methods, Genome, Viral, Humans, Leukemia Virus, Feline physiology, Plasmids genetics, Plasmids metabolism, Recombination, Genetic, Response Elements genetics, Retroviridae Proteins genetics, Retroviridae Proteins metabolism, Terminal Repeat Sequences, Virus Assembly, Virus Replication, Gene Transfer Techniques, Genetic Vectors, Leukemia Virus, Feline genetics, Retroviridae Infections, Tumor Virus Infections
- Published
- 2003
- Full Text
- View/download PDF
43. Virus-based vectors for human vaccine applications.
- Author
-
Polo JM and Dubensky TW Jr
- Subjects
- Humans, Poliovirus genetics, Vaccines genetics, Genetic Vectors, Vaccines therapeutic use, Viruses
- Abstract
Vaccinology has experienced a dramatic resurgence recently, as traditional methodologies of using attenuated live pathogens or inactivated whole pathogens have been either ineffective or are not an acceptable risk for several disease targets, including HIV and Hepatitis C. Gene-based vaccines can stimulate potent humoral and cellular immune responses, and viral vectors might be an efficient strategy for both delivery of antigen-encoding genes, as well as facilitating and enhancing antigen presentation. Vectors derived from diverse viruses with distinct tropism and gene expression strategies have been developed, and are being evaluated in preclinical and clinical vaccine studies. Virus-based vaccines represent a promising approach for vaccines against infectious and malignant disease.
- Published
- 2002
- Full Text
- View/download PDF
44. (Re-)Engineering tumor cell-selective replicating adenoviruses: a step in the right direction toward systemic therapy for metastatic disease.
- Author
-
Dubensky TW Jr
- Subjects
- Animals, Defective Viruses, E2F Transcription Factors, Humans, Retinoblastoma Protein metabolism, Transcription Factors genetics, Transcription Factors metabolism, Virus Replication, Adenoviridae genetics, Cell Cycle Proteins, DNA-Binding Proteins, Genetic Engineering methods, Neoplasms therapy
- Abstract
An approach combining redundant controls to restrict the productive infection of adenoviruses to cells that are disrupted in the pRb pathway-a hallmark of human cancer-has resulted in a novel oncolytic virus that may be well suited for systemic administration to treat metastatic disease.
- Published
- 2002
- Full Text
- View/download PDF
45. Replicon vectors derived from Sindbis virus and Semliki forest virus that establish persistent replication in host cells.
- Author
-
Perri S, Driver DA, Gardner JP, Sherrill S, Belli BA, Dubensky TW Jr, and Polo JM
- Subjects
- Genetic Vectors, RNA, Viral biosynthesis, Replicon, Semliki forest virus genetics, Sindbis Virus genetics
- Abstract
Alphavirus replicon vectors are well suited for applications where transient, high-level expression of a heterologous gene is required. Replicon vector expression in cells leads to inhibition of host macromolecular synthesis, culminating in eventual cell death by an apoptotic mechanism. For many applications, including gene expression studies in cultured cells, a longer duration of transgene expression without resulting cytopathic effects is useful. Recently, noncytopathic Sindbis virus (SIN) variants were isolated in BHK cells, and the mutations responsible were mapped to the protease domain of nonstructural protein 2 (nsP2). We report here the isolation of additional variants of both SIN and Semliki Forest virus (SFV) replicons encoding the neomycin resistance gene that can establish persistent replication in BHK cells. The SIN and SFV variant replicons resulted from previously undescribed mutations within one of three discrete regions of the nsP2 gene. Differences among the panel of variants were observed in processing of the nonstructural polyprotein and in the ratios of subgenomic to genomic RNAs. Importantly, high-level expression of a heterologous gene was retained with most replicons. Finally, in contrast to previous studies, efficient packaging was obtained with several of the variant replicons. This work expands the utility of noncytopathic replicons and the understanding of how alphavirus replicons establish persistent replication in cultured cells.
- Published
- 2000
- Full Text
- View/download PDF
46. VSV-G pseudotyped lentiviral vector particles produced in human cells are inactivated by human serum.
- Author
-
DePolo NJ, Reed JD, Sheridan PL, Townsend K, Sauter SL, Jolly DJ, and Dubensky TW Jr
- Subjects
- Animals, Cell Line, Cricetinae, Humans, Antiviral Agents, Blood, Genetic Vectors, Lentivirus genetics, Vesicular stomatitis Indiana virus genetics
- Abstract
Lentiviral vectors transduce dividing and postmitotic cells and thus are being developed toward therapies for many diseases affecting diverse tissues. One essential requirement for efficacy will be that vector particles are resistant to inactivation by human serum complement. Most animal studies with lentiviral vectors have utilized VSV-G pseudotyped envelopes. Here we demonstrate that VSV-G pseudotyped HIV and FIV vectors produced in human cells are inactivated by human serum complement, suggesting that alternative envelopes may be required for therapeutic efficacy for many clinical applications of lentiviral vectors.
- Published
- 2000
- Full Text
- View/download PDF
47. Generation of retroviral packaging and producer cell lines for large-scale vector production and clinical application: improved safety and high titer.
- Author
-
Sheridan PL, Bodner M, Lynn A, Phuong TK, DePolo NJ, de la Vega DJ Jr, O'Dea J, Nguyen K, McCormack JE, Driver DA, Townsend K, Ibañez CE, Sajjadi NC, Greengard JS, Moore MD, Respess J, Chang SM, Dubensky TW Jr, Jolly DJ, and Sauter SL
- Subjects
- Base Sequence, Cell Line, DNA Primers, Factor VIII genetics, Hemophilia A therapy, Humans, Genetic Vectors, Retroviridae genetics, Virus Assembly
- Abstract
For many applications, human clinical therapies using retroviral vectors still require many technological improvements in key areas of vector design and production. These improvements include higher unprocessed manufacturing titers, complement-resistant vectors, and minimized potential to generate replication-competent retrovirus (RCR). To address these issues, we have developed a panel of human packaging cell lines (PCLs) with reduced homology between retroviral vector and packaging components. These reduced-homology PCLs allowed for the use of a novel high multiplicity of transduction ("high m.o. t.") method to introduce multiple copies of provector within vector-producing cell lines (VPCLs), resulting in high-titer vector without the generation of RCR. In a distinct approach to increase vector yields, we integrated manufacturing parameters into screening strategies and clone selection for large-scale vector production. Collectively, these improvements have resulted in the development of diverse VPCLs with unprocessed titers exceeding 2 x 10(7) CFU/ml. Using this technology, human Factor VIII VPCLs yielding titers as high as 2 x 10(8) CFU/ml unprocessed supernatant were generated. These cell lines produce complement-resistant vector particles (N. J. DePolo et al., J. Virol. 73: 6708-6714, 1999) and provide the basis for an ongoing Factor VIII gene therapy clinical trial.
- Published
- 2000
- Full Text
- View/download PDF
48. Delivery systems for gene-based vaccines.
- Author
-
Dubensky TW Jr, Liu MA, and Ulmer JB
- Subjects
- Humans, Vaccination, Drug Delivery Systems methods, Vaccines, DNA administration & dosage
- Abstract
Along with the elucidation of the role of cytotoxic T lymphocytes in the immune responses against a number of pathogens and cancer, and with the increased understanding of the cellular processing mechanisms of antigens for generation of these cells, has come an increased focus on vaccines that can generate cellular immunity along with antibodies. Promising approaches based on the delivery of genes, either as plasmid DNA or by viral vectors, have been extensively evaluated pre-clinically and in early-phase clinical trials. Although the first generation of DNA plasmid vaccines were broadly effective in animal disease models, early clinical immunogenicity pointed towards the need for increased potency. This manuscript reviews recent developments for gene-based vaccines, specifically, new approaches for formulating and delivering plasmid DNA and alphaviral replicon vectors, all of which have resulted in increased potency of gene-based vaccines.
- Published
- 2000
49. Transduction of murine cerebellar neurons with recombinant FIV and AAV5 vectors.
- Author
-
Alisky JM, Hughes SM, Sauter SL, Jolly D, Dubensky TW Jr, Staber PD, Chiorini JA, and Davidson BL
- Subjects
- Animals, Biological Transport, Active, Cerebellum cytology, Mice, Mice, Inbred C57BL, Purkinje Cells enzymology, beta-Galactosidase genetics, beta-Galactosidase metabolism, Cerebellum physiology, Dependovirus genetics, Genetic Vectors, Immunodeficiency Virus, Feline genetics, Neurons physiology, Transduction, Genetic, Transgenes
- Abstract
Our data demonstrate that vectors derived from recombinant feline immunodeficiency virus (rFIV) and adeno-associated virus type 5 (rAAV5) transduce cerebellar cells following direct injection into the cerebellar lobules of mice. Both recombinant viruses mediated gene transfer predominantly to neurons, with up to 2500 and 1500 Purkinje cells transduced for rAAV5 or rFIV-based vectors, respectively. The vectors also transduced stellate, basket and Golgi neurons, with occasional transduction of granule cells and deep cerebellar nuclei. rAAV5 also spread outside the cerebellum to the inferior colliculus and ventricular epithelium, while rFIV demonstrated the ability to undergo retrograde transport to the physically close lateral vestibular nuclei. Thus, AAV5 and FIV-based vectors show promise for targeting neurons affected in the hereditary spinocerebellar ataxias. These vectors could be important tools for unraveling the pathophysiology of these disorders, or in testing factors which may promote neuronal survival.
- Published
- 2000
- Full Text
- View/download PDF
50. Alphavirus DNA and particle replicons for vaccines and gene therapy.
- Author
-
Polo JM, Gardner JP, Ji Y, Belli BA, Driver DA, Sherrill S, Perri S, Liu MA, and Dubensky TW Jr
- Subjects
- Animals, Biotechnology, Dendritic Cells immunology, Genetic Vectors, Humans, Primates, Sindbis Virus genetics, Sindbis Virus immunology, Alphavirus genetics, Alphavirus immunology, Genetic Therapy, Replicon, Vaccines, DNA genetics
- Abstract
Alphaviruses have several features that make them attractive as gene delivery platforms, and vectors derived principally from Sindbis virus (SIN), Semliki Forest virus (SFV), and Venezuelan equine encephalitis virus (VEE), are currently being developed as prophylactic and therapeutic vaccines for infectious diseases and cancer. Alphavirus vectors, termed "replicons", retain the nonstructural protein genes encoding the viral replicase, that in turn programme high level cytoplasmic amplification of the vector RNA. We have developed plasmid DNA and recombinant vector particle delivery systems derived from the prototype alphavirus, SIN. Each system uses RNA polymerase II-based expression of alphavirus genome components and both vector formats are highly efficacious towards inducing robust antigen-specific immune responses in vaccinated animals. To increase the potency of SIN vector particles, which are not known to be lymphotropic, the tropism was re-directed for efficient infection of dendritic cells, both in vitro and in vivo.
- Published
- 2000
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.