Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Vaccines (Basel) ; 7(2)2019 May 22.
Article in English | MEDLINE | ID: mdl-31121939

ABSTRACT

Identification of novel molecular adjuvants which can boost and enhance vaccine-mediated immunity and provide dose-sparing potential against complex infectious diseases and for immunotherapy in cancer is likely to play a critical role in the next generation of vaccines. Given the number of challenging targets for which no or only partial vaccine options exist, adjuvants that can address some of these concerns are in high demand. Here, we report that a designed truncated Interleukin-36 gamma (IL-36 gamma) encoded plasmid can act as a potent adjuvant for several DNA-encoded vaccine targets including human immunodeficiency virus (HIV), influenza, and Zika in immunization models. We further show that the truncated IL-36 gamma (opt-36γt) plasmid provides improved dose sparing as it boosts immunity to a suboptimal dose of a Zika DNA vaccine, resulting in potent protection against a lethal Zika challenge.

2.
Oncoimmunology ; 8(1): e1515058, 2019.
Article in English | MEDLINE | ID: mdl-30546956

ABSTRACT

Ovarian cancer is frequently diagnosed as peritoneal carcinomatosis. Unlike other tumor locations, the peritoneal cavity is commonly exposed to gut-breaching and ascending genital microorganisms and has a unique immune environment. IL-33 is a local cytokine that can activate innate and adaptive immunity. We studied the effectiveness of local IL-33 delivery in the treatment of cancer that has metastasized to the peritoneal cavity. Direct peritoneal administration of IL-33 delayed the progression of metastatic peritoneal cancer. Prolongation in survival was not associated with a direct effect of IL-33 on tumor cells, but with major changes in the immune microenvironment of the tumor. IL-33 promoted a significant increase in the leukocyte compartment of the tumor immunoenvironment and an allergic cytokine profile. We observed a substantial increase in the number of activated CD4+ T-cells accompanied by peritoneal eosinophil infiltration, B-cell activation and activation of peritoneal macrophages which displayed tumoricidal capacity. Depletion of CD4+ cells, eosinophils or macrophages reduced the anti-tumor effects of IL-33 but none of these alone were sufficient to completely abrogate its positive benefit. In conclusion, local administration of IL-33 generates an allergic tumor environment resulting in a novel approach for treatment of metastatic peritoneal malignancies, such as advanced ovarian cancer.

3.
J Infect Dis ; 219(4): 544-555, 2019 01 29.
Article in English | MEDLINE | ID: mdl-30304515

ABSTRACT

Background: There remains an important need for prophylactic anti-Ebola virus vaccine candidates that elicit long-lasting immune responses and can be delivered to vulnerable populations that are unable to receive live-attenuated or viral vector vaccines. Methods: We designed novel synthetic anti-Ebola virus glycoprotein (EBOV-GP) DNA vaccines as a strategy to expand protective breadth against diverse EBOV strains and evaluated the impact of vaccine dosing and route of administration on protection against lethal EBOV-Makona challenge in cynomolgus macaques. Long-term immunogenicity was monitored in nonhuman primates for >1 year, followed by a 12-month boost. Results: Multiple-injection regimens of the EBOV-GP DNA vaccine, delivered by intramuscular administration followed by electroporation, were 100% protective against lethal EBOV-Makona challenge. Impressively, 2 injections of a simple, more tolerable, and dose-sparing intradermal administration followed by electroporation generated strong immunogenicity and was 100% protective against lethal challenge. In parallel, we observed that EBOV-GP DNA vaccination induced long-term immune responses in macaques that were detectable for at least 1 year after final vaccination and generated a strong recall response after the final boost. Conclusions: These data support that this simple intradermal-administered, serology-independent approach is likely important for additional study towards the goal of induction of anti-EBOV immunity in multiple at-risk populations.


Subject(s)
Ebola Vaccines/immunology , Ebolavirus/immunology , Hemorrhagic Fever, Ebola/prevention & control , Vaccines, DNA/immunology , Animals , Disease Models, Animal , Ebola Vaccines/administration & dosage , Female , Injections, Intramuscular , Macaca fascicularis , Male , Vaccines, DNA/administration & dosage
4.
Cancer Res ; 78(18): 5340-5348, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30026324

ABSTRACT

CCR8 is a chemokine receptor expressed principally on regulatory T cells (Treg) and is known to be critical for CCR8+ Treg-mediated immunosuppression. Recent studies have demonstrated that CCR8 is uniquely upregulated in human tumor-resident Tregs of patients with breast, colon, and lung cancer when compared with normal tissue-resident Tregs. Therefore, CCR8+ tumor-resident Tregs are rational targets for cancer immunotherapy. Here, we demonstrate that mAb therapy targeting CCR8 significantly suppresses tumor growth and improves long-term survival in colorectal tumor mouse models. This antitumor activity correlated with increased tumor-specific T cells, enhanced infiltration of CD4+ and CD8+ T cells, and a significant decrease in the frequency of tumor-resident CD4+CCR8+ Tregs. Tumor-specific CD8+ T cells displayed lower expression of exhaustion markers as well as increased functionality upon restimulation. Treatment with anti-CCR8 mAb prevented de novo induction and suppressive function of Tregs without affecting CD8+ T cells. Initial studies explored a combinatorial regimen using anti-CCR8 mAb therapy and a Listeria monocytogenes-based immunotherapy. Anti-CCR8 mAb therapy synergized with L. monocytogenes-based immunotherapy to significantly delay growth of established tumors and to prolong survival. Collectively, these findings identify CCR8 as a promising new target for tumor immunotherapy and provide a strong rationale for further development of this approach, either as a monotherapy or in combination with other immunotherapies.Significance: Inhibition of CCR8 represents a promising new cancer immunotherapy strategy that modulates tumor-resident regulatory T cells to enhance antitumor immunity and prolong patient survival. Cancer Res; 78(18); 5340-8. ©2018 AACR.


Subject(s)
Cancer Vaccines/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Receptors, CCR8/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Humans , Immune Tolerance , Immunosuppression Therapy , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, CCR8/immunology , T-Lymphocytes, Regulatory/immunology , Treatment Outcome , Tumor Microenvironment/immunology , Up-Regulation
5.
Mol Ther ; 26(2): 435-445, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29249395

ABSTRACT

Immune checkpoint blockade antibodies are setting a new standard of care for cancer patients. It is therefore important to assess any new immune-based therapies in the context of immune checkpoint blockade. Here, we evaluate the impact of combining a synthetic consensus TERT DNA vaccine that has improved capacity to break tolerance with immune checkpoint inhibitors. We observed that blockade of CTLA-4 or, to a lesser extent, PD-1 synergized with TERT vaccine, generating more robust anti-tumor activity compared to checkpoint alone or vaccine alone. Despite this anti-tumor synergy, none of these immune checkpoint therapies showed improvement in TERT antigen-specific immune responses in tumor-bearing mice. αCTLA-4 therapy enhanced the frequency of T-bet+/CD44+ effector CD8+ T cells within the tumor and decreased the frequency of regulatory T cells within the tumor, but not in peripheral blood. CTLA-4 blockade synergized more than Treg depletion with TERT DNA vaccine, suggesting that the effect of CTLA-4 blockade is more likely due to the expansion of effector T cells in the tumor rather than a reduction in the frequency of Tregs. These results suggest that immune checkpoint inhibitors function to alter the immune regulatory environment to synergize with DNA vaccines, rather than boosting antigen-specific responses at the site of vaccination.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/immunology , Neoplasms/genetics , Neoplasms/immunology , Telomerase/immunology , Vaccines, DNA/immunology , Animals , Biomarkers, Tumor , CTLA-4 Antigen/antagonists & inhibitors , Cancer Vaccines/genetics , Cell Line, Tumor , Combined Modality Therapy , Disease Models, Animal , Female , Humans , Immunotherapy , Mice , Neoplasms/pathology , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Telomerase/genetics , Vaccines, DNA/genetics , Xenograft Model Antitumor Assays
6.
Oncotarget ; 8(24): 39117-39130, 2017 Jun 13.
Article in English | MEDLINE | ID: mdl-28388572

ABSTRACT

Tumor progression is facilitated immunologically by mechanisms that include low antigen expression, an absence of coimmunostimulatory signals, and the presence of regulatory T cells (Tregs), all of which act to suppress and restrict effector T cells in the tumor. It may be possible to overcome these conditions by a combination of modulatory immunotherapy agents and tumor-antigen targeting to activate and drive effective antitumor T cell responses. Here, we demonstrated that co-administration of aGITR and aPD-1 monoclonal antibodies (mAb) in combination with a peptide vaccine (Vax) in mice bearing established tumors significantly delayed tumor growth and induced complete regression in 50% of the mice. This response was associated with increased expansion and functionality of potent Ag-specific polyfunctional CD8+ T cells, reduced Tregs, and the generation of memory T cells. Tumor regression correlated with the expansion of tumor-infiltrating antigen-specific CD8+ effector memory T cells, as depletion of this cell population significantly reduced the effectiveness of the triple combination Vax/aGITR/aPD-1 therapy. These findings support the concept that dual aGITR/aPD-1 combination with cancer vaccines may be a novel strategy against poorly immunogenic tumors.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Neoplasm/immunology , Cancer Vaccines/administration & dosage , Glucocorticoid-Induced TNFR-Related Protein/antagonists & inhibitors , Melanoma, Experimental/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Female , Immunotherapy , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured , Vaccination
7.
Oncotarget ; 8(65): 109151-109160, 2017 Dec 12.
Article in English | MEDLINE | ID: mdl-29312597

ABSTRACT

Mounting evidence demonstrates that CD8+CD122+ T cells have suppressive properties with the capacity to inhibit T cell responses. Therefore, these cells are rational targets for cancer immunotherapy. Here, we demonstrate that CD122 monoclonal antibody (mAb; aCD122) therapy significantly suppressed tumor growth and improved long-term survival in tumor-bearing mice. This therapeutic effect correlated with enhanced polyfunctional, cytolytic intratumoral CD8+ T cells and a decrease in granulocytic myeloid-derived suppressor cells (G-MDSCs). In addition, aCD122 treatment synergized with a vaccine to augment vaccine-induced antigen (Ag)-specific CD8+ T cell responses, reject established tumors and generate memory T cells. Furthermore, aCD122 mAb synergized with an anti-GITR (aGITR) mAb to confer significant control of tumor growth. These results suggest CD122 might be a promising target for cancer immunotherapy, either as a single agent or in combination with other forms of immunotherapy.

8.
Hum Vaccin Immunother ; 12(10): 2649-2653, 2016 10 02.
Article in English | MEDLINE | ID: mdl-27322875

ABSTRACT

Mycobacterium tuberculosis infects one third of the world's population. Due to variable efficacy of the Bacille Calmette Guerin (BCG) vaccine, development of novel TB vaccines remains a priority. Here, we demonstrate the protective efficacy of a novel multivalent DNA vaccine, which contains 15 synthetic antigens targeting the Mtb ESX secretion system.


Subject(s)
Mycobacterium tuberculosis/immunology , Tuberculosis Vaccines/immunology , Tuberculosis Vaccines/isolation & purification , Tuberculosis/prevention & control , Animals , Disease Models, Animal , Humans , Mice, Inbred C57BL , Tuberculosis/immunology
9.
Methods Mol Biol ; 1403: 355-61, 2016.
Article in English | MEDLINE | ID: mdl-27076140

ABSTRACT

Multivalent DNA vaccines that are delivered by electroporation (EP) through muscle tissue provide a novel method for eliciting immunity against tuberculosis (TB) as well as a broad range of diseases including HIV and cancers. Proper plasmid construction containing suitable protective TB antigens capable of evoking desired vaccine-induced responses would lead to the appropriate induction of both humoral and cellular immunity. DNA vaccines are safe and of low cost in comparison to traditional vaccines while also providing potentially effective prophylactic or therapeutic modalities against currently untreatable diseases. Here, we describe the steps for developing a rational multivalent TB DNA vaccine delivered with intramuscular EP in mice.


Subject(s)
Tuberculosis Vaccines/immunology , Vaccines, DNA/immunology , Animals , Humans , Tuberculosis/immunology , Tuberculosis/prevention & control , Tuberculosis Vaccines/economics , Vaccines, DNA/economics
10.
Sci Transl Med ; 7(301): 301ra132, 2015 Aug 19.
Article in English | MEDLINE | ID: mdl-26290414

ABSTRACT

First identified in 2012, Middle East respiratory syndrome (MERS) is caused by an emerging human coronavirus, which is distinct from the severe acute respiratory syndrome coronavirus (SARS-CoV), and represents a novel member of the lineage C betacoronoviruses. Since its identification, MERS coronavirus (MERS-CoV) has been linked to more than 1372 infections manifesting with severe morbidity and, often, mortality (about 495 deaths) in the Arabian Peninsula, Europe, and, most recently, the United States. Human-to-human transmission has been documented, with nosocomial transmission appearing to be an important route of infection. The recent increase in cases of MERS in the Middle East coupled with the lack of approved antiviral therapies or vaccines to treat or prevent this infection are causes for concern. We report on the development of a synthetic DNA vaccine against MERS-CoV. An optimized DNA vaccine encoding the MERS spike protein induced potent cellular immunity and antigen-specific neutralizing antibodies in mice, macaques, and camels. Vaccinated rhesus macaques seroconverted rapidly and exhibited high levels of virus-neutralizing activity. Upon MERS viral challenge, all of the monkeys in the control-vaccinated group developed characteristic disease, including pneumonia. Vaccinated macaques were protected and failed to demonstrate any clinical or radiographic signs of pneumonia. These studies demonstrate that a consensus MERS spike protein synthetic DNA vaccine can induce protective responses against viral challenge, indicating that this strategy may have value as a possible vaccine modality against this emerging pathogen.


Subject(s)
Middle East Respiratory Syndrome Coronavirus/immunology , Vaccines, DNA/therapeutic use , Animals , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Camelus , Macaca mulatta , Mice
11.
Mol Ther ; 23(10): 1653-62, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26122932

ABSTRACT

ISG15 is an ubiquitin-like protein induced by type I interferon associated with antiviral activity. ISG15 is also secreted and known to function as an immunomodulatory molecule. However, ISG15's role in influencing the adaptive CD8 T-cell responses has not been studied. Here, we demonstrate the efficacy of ISG15 as a vaccine adjuvant, inducing human papilloma virus (HPV) E7-specific IFNγ responses as well as the percentage of polyfunctional, cytolytic, and effector CD8 T-cell responses. Vaccination with ISG15 conferred remarkable control and/or regression of established HPV-associated tumor-bearing mice. T-cell depletion coupled with adoptive transfer experiments revealed that ISG15 protective efficacy was CD8 T-cell mediated. Importantly, we demonstrate that ISG15 vaccine-induced responses could be generated independent of ISGylation, suggesting that responses were mostly influenced by free ISG15. Our results provide more insight into the immunomodulatory properties of ISG15 and its potential to serve as an effective immune adjuvant in a therapeutic tumor or infectious disease setting.


Subject(s)
Adjuvants, Immunologic , Adoptive Transfer , Amino Acid Sequence , Animals , Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Cell Line, Tumor , Cytokines/chemistry , Cytokines/genetics , Cytokines/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Gene Expression , Genetic Vectors/genetics , Humans , Immunization , Lymphocyte Depletion , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Papillomavirus E7 Proteins/immunology , Sequence Alignment , Ubiquitins/chemistry , Ubiquitins/genetics , Ubiquitins/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology
12.
Hum Vaccin Immunother ; 11(8): 1961-71, 2015.
Article in English | MEDLINE | ID: mdl-26158319

ABSTRACT

Botulinum neurotoxins (BoNTs) are deadly, toxic proteins produced by the bacterium Clostridium botulinum that can cause significant diseases in humans. The use of the toxic substances as potential bioweapons has raised concerns by the Centers for Disease Control and Prevention and the United States Military. Currently, there is no licensed vaccine to prevent botulinum intoxication. Here we present an immunogenicity study to evaluate the efficacy of novel monovalent vaccines and a trivalent cocktail DNA vaccine targeting the heavy chain C-terminal fragments of Clostridium botulinum neurotoxin serotypes A, B, and E. These synthetic DNA vaccines induced robust humoral and polyfunctional CD4(+) T-cell responses which fully protected animals against lethal challenge after just 2 immunizations. In addition, naïve animals administered immunized sera mixed with the lethal neurotoxin were 100% protected against intoxication. The data demonstrate the protective efficacy induced by a combinative synthetic DNA vaccine approach. This study has importance for the development of vaccines that provide protective immunity against C. botulinum neurotoxins and other toxins.


Subject(s)
Antitoxins/blood , Botulinum Toxins, Type A/immunology , Botulinum Toxins/immunology , Botulism/prevention & control , CD4-Positive T-Lymphocytes/immunology , Vaccines, DNA/immunology , Animals , Botulinum Toxins/genetics , Botulinum Toxins, Type A/genetics , Female , Mice, Inbred BALB C , Survival Analysis , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
13.
Hum Vaccin Immunother ; 11(8): 1954-60, 2015.
Article in English | MEDLINE | ID: mdl-26091147

ABSTRACT

Tuberculosis (TB) still remains a major public health issue despite the current available vaccine for TB, Bacille Calmette Guerin (BCG). An effective vaccine against TB remains a top priority in the fight against this pandemic bacterial infection. Adequate protection against TB is associated with the development of TH1-type and CD8(+) T cell responses. One alarmin cytokine, interleukin 33 (IL-33), has now been implicated in the development of both CD4(+) TH1 and CD8(+) T cell immunity. In this study, we determined whether the administration of IL-33 as an adjuvant, encoded in a DNA plasmid, could enhance the immunogenicity of a TB DNA vaccine. We report that the co-immunization of IL-33 with a DNA vaccine expressing the Mycobacterium Tuberculosis (Mtb) antigen 85B (Ag85B) induced robust Ag85B-specific IFNγ responses by ELISpot compared to Ag85B alone. Furthermore, these enhanced responses were characterized by higher frequencies of Ag85B-specific, multifunctional CD4(+) and CD8(+) T cells. Vaccination with IL-33 also increased the ability of the Ag85B-specific CD8(+) T cells to undergo degranulation and to secrete IFNγ and TNFα cytokines. These finding highlights IL-33 as a promising adjuvant to significantly improve the immunogenicity of TB DNA vaccines and support further study of this effective vaccine strategy against TB.


Subject(s)
Acyltransferases/immunology , Adjuvants, Immunologic/metabolism , Antigens, Bacterial/immunology , Bacterial Proteins/immunology , Interleukin-33/metabolism , Th1 Cells/immunology , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Vaccines, DNA/immunology , Acyltransferases/genetics , Adjuvants, Immunologic/genetics , Animals , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , CD8-Positive T-Lymphocytes/immunology , Enzyme-Linked Immunospot Assay , Female , Interferon-gamma/metabolism , Interleukin-33/genetics , Mice, Inbred C57BL , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/genetics , Tumor Necrosis Factor-alpha/metabolism , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
14.
Hum Vaccin Immunother ; 11(8): 1972-82, 2015.
Article in English | MEDLINE | ID: mdl-26091432

ABSTRACT

Nucleic acid-based vaccines (NAVs) are a promising alternative to conventional influenza vaccines with the potential to increase influenza vaccine availability due to their simplicity in design and rapid speed of production. NAVs can also target multiple influenza antigens and control flu variants. Traditionally NAVs have been DNA plasmids however, we are continuing to explore new methods that may enhance vaccine efficacy. Recently new focus has been on RNA cassettes as NAVs. RNA vaccines combine conceptual advantages in that they focus on delivery of only the coding cassette. However, RNA vaccines have a short half-life and cause interferon-induced fevers. Here we describe a new NAV approach where we study delivery of a linear DNA cassette [Doggybone linear closed DNA [(dbDNA)] produced by an enzymatic process that yields an antigen expression cassette comprising a promoter, DNA antigen, poly A tail, and telomeric ends. This focused approach has many of the advantages of plasmid DNA as well as a minimal cassette size similar to RNA strategies. For this study, we characterized the specific CD4(+) and CD8(+) T cell responses and determined the hemagglutination inhibition (HI) titers induced by dbDNA and compared the responses with those of an optimized plasmid DNA (pDNA) vaccine encoding the same H1N1 influenza A/PR/8/34 HA gene. Immunizations with the constructs resulted in similar humoral and cellular immune responses. Both constructs induced high-titer HI antibodies and fully protected animals from lethal viral challenge. The data obtained from this study provides important validation for further development of novel vector approaches.


Subject(s)
Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Plasmids , Vaccines, DNA/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Female , Hemagglutination Inhibition Tests , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/genetics , Mice, Inbred BALB C , Survival Analysis , Vaccines, DNA/administration & dosage , Vaccines, DNA/genetics
15.
Vaccine ; 33(35): 4313-20, 2015 Aug 20.
Article in English | MEDLINE | ID: mdl-25887087

ABSTRACT

Identifying new molecular adjuvants that elicit effective vaccine-induced CD8(+) T cell immunity may be critical for the elimination of many challenging diseases including Tuberculosis, HIV and cancer. Here, we report that co-administration of molecular adjuvant IL-33 during vaccination enhanced the magnitude and function of antigen (Ag)-specific CD8(+) T cells against a model Ag, LCMV NP target protein. These enhanced responses were characterized by higher frequencies of Ag-specific, polyfunctional CD8(+) T cells exhibiting cytotoxic characteristics. Importantly, these cells were capable of robust expansion upon Ag-specific restimulation in vivo and conferred remarkable protection against a high dose lethal LCMV challenge. In addition, we demonstrate the ability of IL-33 to amplifying the frequency of Ag-specific KLRG1(+) effector CD8(+) T cells. These data show that IL-33 is a promising immunoadjuvant at improving T cell immunity in a vaccine setting and suggest further development and understanding of this molecular adjuvant for strategies against many obstinate infectious diseases and cancer.


Subject(s)
Adjuvants, Immunologic , Interleukin-33/immunology , Lymphocytic choriomeningitis virus/immunology , Vaccines, DNA/immunology , AIDS Vaccines/administration & dosage , AIDS Vaccines/immunology , Animals , Arenaviridae Infections/prevention & control , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Immunity, Cellular , Immunologic Memory , Interleukin-33/genetics , Lymphocytic choriomeningitis virus/genetics , Lymphocytic choriomeningitis virus/pathogenicity , Mice, Inbred C57BL
16.
Adv Exp Med Biol ; 848: 131-48, 2015.
Article in English | MEDLINE | ID: mdl-25757619

ABSTRACT

There is no licensed vaccine or cure for human cytomegalovirus (CMV), a ubiquitous ß-herpes virus that infects 60-95 % of adults worldwide. Infection is a major cause of congenital abnormalities in newborns, contributes to development of childhood cerebral palsy and medulloblastoma, can result in severe disease in immunocompromised patients, and is a major impediment during successful organ transplantation. While CMV has been increasingly associated with numerous inflammatory diseases and cancers, only recently has it been correlated with increased risk of heart disease in adults, the number-one killer in the USA. These data, among others, suggest that subclinical CMV infection, or microinfection, in healthy individuals may play more of a causative role than an epiphenomenon in development of CMV-associated pathologies. Due to the myriad of diseases and complications associated with CMV, an efficacious vaccine would be highly valuable in reducing human morbidity and mortality as well as saving billions of dollars in annual health-care costs and disability adjusted life years (DALY) in the developing world. Therefore, the development of a safe efficacious CMV vaccine or immune therapy is paramount to the public health. This review aims to provide a brief overview on aspects of CMV infection and disease and focuses on current vaccine strategies. The use of new synthetic DNA vaccines might offer one such approach to this difficult problem.


Subject(s)
Cloning, Molecular/methods , Cytomegalovirus Infections/therapy , Cytomegalovirus Vaccines/therapeutic use , Immunotherapy, Active/methods , Vaccines, DNA/therapeutic use , Adult , Animals , Cytomegalovirus/genetics , Cytomegalovirus/immunology , Cytomegalovirus Infections/genetics , Cytomegalovirus Infections/immunology , Cytomegalovirus Vaccines/genetics , DNA, Recombinant/genetics , DNA, Recombinant/therapeutic use , Humans , Vaccines, DNA/genetics
17.
Expert Rev Vaccines ; 14(4): 489-92, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25656504

ABSTRACT

The identification and characterization of cytokine isoforms is likely to provide critical important new insight into immunobiology. Cytokine isoforms can provide additional diversity to their complex biological effects that participate in control and protection against different foreign pathogens. Recently, IL-33 has been identified as a proinflammatory cytokine having several different biologically active isoform products. Originally associated with Th2 immunity, new evidence now supports the role of two IL-33 isoforms to facilitate the generation of protective Th1 and CD8 T cell immunity against specific pathogens. Therefore, a better understanding of the IL-33 isoforms will inform us on how to utilize them to facilitate their development as tools as vaccine adjuvants for immune therapy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Interleukins/administration & dosage , Interleukins/genetics , Biomedical Research/trends , Humans , Interleukin-33 , Protein Isoforms/administration & dosage , Protein Isoforms/genetics , Vaccines/administration & dosage , Vaccines/immunology
18.
Hum Vaccin Immunother ; 10(8): 2188-98, 2014.
Article in English | MEDLINE | ID: mdl-25424922

ABSTRACT

Development of a broad-spectrum synthetic vaccine against TB would represent an important advance to the limited vaccine armamentarium against TB. It is believed that the esx family of TB antigens may represent important vaccine candidates. However, only 4 esx antigens have been studied as potential vaccine antigens. The challenge remains to develop a vaccine that simultaneously targets all 23 members of the esx family to induce enhanced broad-spectrum cell-mediated immunity. We sought to investigate if broader cellular immune responses could be induced using a multivalent DNA vaccine representing the esx family protein members delivered via electroporation. In this study, 15 designed esx antigens were created to cross target all members of the esx family. They were distributed into groups of 3 self-processing antigens each, resulting in 5 trivalent highly optimized DNA plasmids. Vaccination with all 5 constructs elicited robust antigen-specific IFN-γ responses to all encoded esx antigens and induced multifunctional CD4 Th1 and CD8 T cell responses. Importantly, we show that when all constructs are combined into a cocktail, the RSQ-15 vaccine, elicited substantial broad Ag-specific T cell responses to all esx antigens as compared with vaccination with BCG. Moreover, these vaccine-induced responses were highly cross-reactive with BCG encoded esx family members and were highly immune effective in a BCG DNA prime-boost format. Furthermore, we demonstrate the vaccine potential and immunopotent profile of several novel esx antigens never previously studied. These data highlight the likely importance of these novel immunogens for study as preventative or therapeutic synthetic TB vaccines in combination or as stand alone antigens.


Subject(s)
Antigens, Bacterial/immunology , Immunity, Cellular , Tuberculosis Vaccines/administration & dosage , Tuberculosis Vaccines/immunology , Tuberculosis/prevention & control , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Animals , Antigens, Bacterial/genetics , Electroporation , Interferon-gamma/metabolism , Leukocytes, Mononuclear/immunology , Mice , T-Lymphocyte Subsets/immunology , Tuberculosis/immunology , Tuberculosis Vaccines/genetics , Vaccination/methods , Vaccines, DNA/genetics , Vaccines, Synthetic/administration & dosage , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology
19.
Curr Opin Immunol ; 28: 102-6, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24762410

ABSTRACT

For many years IL-33 has been widely studied in the context of T helper type 2 (Th2)-driven inflammatory disorders. Interestingly, IL-33 has now emerged as a cytokine with a plethora of pleiotropic properties. Depending on the immune cells targeted by IL-33, it is reported to not only promote Th2 immunity, but also to induce T helper type 1 (Th1) immunity. Furthermore, recent studies have revealed that IL-33 can activate CD8(+) T cells. These new studies provide evidence for its beneficial role in antiviral and antitumor immunity. Here we review the evidence of IL-33 to drive protective T cell immunity plus its potential use as an adjuvant in vaccination and tumor therapy.


Subject(s)
Cytokines/immunology , Interleukins/immunology , Animals , Humans , Neoplasms/immunology , Th1 Cells/immunology , Th2 Cells/immunology
20.
Vaccine ; 32(24): 2833-42, 2014 May 19.
Article in English | MEDLINE | ID: mdl-24631084

ABSTRACT

Despite an intensive vaccine program influenza infections remain a major health problem, due to the viruses' ability to change its envelope glycoprotein hemagglutinin (HA), through shift and drift, permitting influenza to escape protection induced by current vaccines or natural immunity. Recently a new variant, H7N9, has emerged in China causing global concern. First, there have been more than 130 laboratory-confirmed human infections resulting in an alarmingly high death rate (32.3%). Second, genetic changes found in H7N9 appear to be associated with enabling avian influenza viruses to spread more effectively in mammals, thus transmitting infections on a larger scale. Currently, no vaccines or drugs are effectively able to target H7N9. Here, we report the rapid development of a synthetic consensus DNA vaccine (pH7HA) to elicit potent protective immunity against the H7N9 viruses. We show that pH7HA induces broad antibody responses that bind to divergent HAs from multiple new members of the H7N9 family. These antibody responses result in high-titer HAI against H7N9. Simultaneously, this vaccine induces potent polyfunctional effector CD4 and CD8T cell memory responses. Animals vaccinated with pH7HA are completely protected from H7N9 virus infection and any morbidity associated with lethal challenge. This study establishes that this synthetic consensus DNA vaccine represents a new tool for targeting emerging infection, and more importantly, its design, testing and development into seed stock for vaccine production in a few days in the pandemic setting has significant implications for the rapid deployment of vaccines protecting against emerging infectious diseases.


Subject(s)
Influenza A Virus, H7N9 Subtype , Influenza Vaccines/immunology , Orthomyxoviridae Infections/prevention & control , Vaccines, DNA/immunology , Animals , Antibodies, Viral/blood , Antibody Formation , Antibody Specificity , Female , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunity, Cellular , Immunologic Memory , Mice, Inbred BALB C , T-Lymphocytes/immunology , Vaccines, Synthetic/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...