Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 78
Filter
1.
Nat Immunol ; 25(3): 537-551, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38337035

ABSTRACT

A nasally delivered chimpanzee adenoviral-vectored severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine (ChAd-SARS-CoV-2-S) is currently used in India (iNCOVACC). Here, we update this vaccine by creating ChAd-SARS-CoV-2-BA.5-S, which encodes a prefusion-stabilized BA.5 spike protein. Whereas serum neutralizing antibody responses induced by monovalent or bivalent adenoviral vaccines were poor against the antigenically distant XBB.1.5 strain and insufficient to protect in passive transfer experiments, mucosal antibody and cross-reactive memory T cell responses were robust, and protection was evident against WA1/2020 D614G and Omicron variants BQ.1.1 and XBB.1.5 in mice and hamsters. However, depletion of memory CD8+ T cells before XBB.1.5 challenge resulted in loss of protection against upper and lower respiratory tract infection. Thus, nasally delivered vaccines stimulate mucosal immunity against emerging SARS-CoV-2 strains, and cross-reactive memory CD8+ T cells mediate protection against lung infection by antigenically distant strains in the setting of low serum levels of cross-reactive neutralizing antibodies.


Subject(s)
COVID-19 , Respiratory Tract Infections , Vaccines , Cricetinae , Animals , Mice , CD8-Positive T-Lymphocytes , SARS-CoV-2 , COVID-19/prevention & control , Antibodies, Neutralizing , Broadly Neutralizing Antibodies , Pan troglodytes
2.
Nat Immunol ; 25(10): 1913-1927, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39227514

ABSTRACT

A mucosal route of vaccination could prevent severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) replication at the site of infection and limit transmission. We compared protection against heterologous XBB.1.16 challenge in nonhuman primates (NHPs) ~5 months following intramuscular boosting with bivalent mRNA encoding WA1 and BA.5 spike proteins or mucosal boosting with a WA1-BA.5 bivalent chimpanzee adenoviral-vectored vaccine delivered by intranasal or aerosol device. NHPs boosted by either mucosal route had minimal virus replication in the nose and lungs, respectively. By contrast, protection by intramuscular mRNA was limited to the lower airways. The mucosally delivered vaccine elicited durable airway IgG and IgA responses and, unlike the intramuscular mRNA vaccine, induced spike-specific B cells in the lungs. IgG, IgA and T cell responses correlated with protection in the lungs, whereas mucosal IgA alone correlated with upper airway protection. This study highlights differential mucosal and serum correlates of protection and how mucosal vaccines can durably prevent infection against SARS-CoV-2.


Subject(s)
Antibodies, Viral , COVID-19 Vaccines , COVID-19 , Immunization, Secondary , Immunoglobulin A , SARS-CoV-2 , Animals , Immunoglobulin A/immunology , SARS-CoV-2/immunology , COVID-19/prevention & control , COVID-19/immunology , COVID-19/virology , Antibodies, Viral/immunology , Antibodies, Viral/blood , COVID-19 Vaccines/immunology , COVID-19 Vaccines/administration & dosage , Spike Glycoprotein, Coronavirus/immunology , Spike Glycoprotein, Coronavirus/genetics , Macaca mulatta , Adenoviridae/immunology , Adenoviridae/genetics , Immunity, Mucosal , Adenovirus Vaccines/immunology , Adenovirus Vaccines/administration & dosage , Female , Lung/virology , Lung/immunology , B-Lymphocytes/immunology , Immunoglobulin G/immunology , Immunoglobulin G/blood , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/blood , Administration, Intranasal , Vaccination/methods , Humans
3.
Cell ; 183(1): 169-184.e13, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32931734

ABSTRACT

The coronavirus disease 2019 pandemic has made deployment of an effective vaccine a global health priority. We evaluated the protective activity of a chimpanzee adenovirus-vectored vaccine encoding a prefusion stabilized spike protein (ChAd-SARS-CoV-2-S) in challenge studies with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mice expressing the human angiotensin-converting enzyme 2 receptor. Intramuscular dosing of ChAd-SARS-CoV-2-S induces robust systemic humoral and cell-mediated immune responses and protects against lung infection, inflammation, and pathology but does not confer sterilizing immunity, as evidenced by detection of viral RNA and induction of anti-nucleoprotein antibodies after SARS-CoV-2 challenge. In contrast, a single intranasal dose of ChAd-SARS-CoV-2-S induces high levels of neutralizing antibodies, promotes systemic and mucosal immunoglobulin A (IgA) and T cell responses, and almost entirely prevents SARS-CoV-2 infection in both the upper and lower respiratory tracts. Intranasal administration of ChAd-SARS-CoV-2-S is a candidate for preventing SARS-CoV-2 infection and transmission and curtailing pandemic spread.


Subject(s)
Coronavirus Infections/immunology , Immunogenicity, Vaccine , Pneumonia, Viral/immunology , Viral Vaccines/immunology , Adenoviridae/genetics , Administration, Intranasal , Animals , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , COVID-19 , COVID-19 Vaccines , Chlorocebus aethiops , Coronavirus Infections/pathology , Coronavirus Infections/prevention & control , Female , HEK293 Cells , Humans , Injections, Intramuscular , Mice , Mice, Inbred BALB C , Pandemics , Pneumonia, Viral/pathology , Respiratory Mucosa/immunology , Respiratory Mucosa/pathology , Respiratory Mucosa/virology , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/immunology , Vero Cells , Viral Vaccines/administration & dosage
5.
Mol Ther ; 31(9): 2600-2611, 2023 09 06.
Article in English | MEDLINE | ID: mdl-37452494

ABSTRACT

B cells are the antibody-producing arm of the adaptive immune system and play a critical role in controlling pathogens. Several groups have now demonstrated the feasibility of using engineered B cells as a therapy, including infectious disease control and gene therapy of serum deficiencies. These studies have largely utilized ex vivo modification of the cells. Direct in vivo engineering would be of utility to the field, particularly in infectious disease control where the infrastructure needs of ex vivo cell modification would make a broad vaccination campaign highly challenging. In this study we demonstrate that engineered adenoviral vectors are capable of efficiently transducing murine and human primary B cells both ex vivo and in vivo. We found that unmodified human adenovirus C5 was capable of infecting B cells in vivo, likely due to interactions between the virus penton base protein and integrins. We further describe vector modification with B cell-specific gene promoters and successfully restrict transgene expression to B cells, resulting in a strong reduction in gene expression from the liver, the main site of human adenovirus C5 infection in vivo.


Subject(s)
Adenoviridae , Communicable Diseases , Mice , Humans , Animals , Adenoviridae/genetics , Genetic Vectors/genetics , Genetic Therapy/methods , Viral Proteins/genetics , B-Lymphocytes
6.
J Am Chem Soc ; 145(46): 25098-25102, 2023 Nov 22.
Article in English | MEDLINE | ID: mdl-37947488

ABSTRACT

Olefins are widely available at low costs, which explains the usefulness of developing new methods for their functionalization. Here we report a simple protocol that uses a photoredox catalyst and an inexpensive thiol catalyst to stitch together two olefins, forming a new C-C bond. Specifically, an electron-poor olefin is reduced by the photoredox catalyst to generate, upon protonation, a carbon radical, which is then captured by a neutral olefin. This intermolecular cross-coupling process provides a tool for rapidly synthesizing sp3-dense molecules from olefins using an unconventional disconnection.

7.
J Am Chem Soc ; 145(1): 47-52, 2023 01 11.
Article in English | MEDLINE | ID: mdl-36574031

ABSTRACT

We report a photochemical method for the functionalization of pyridines with radicals derived from allylic C-H bonds. Overall, two substrates undergo C-H functionalization to form a new C(sp2)-C(sp3) bond. The chemistry harnesses the unique reactivity of pyridinyl radicals, generated upon single-electron reduction of pyridinium ions, which undergo effective coupling with allylic radicals. This novel mechanism enables distinct positional selectivity for pyridine functionalization that diverges from classical Minisci chemistry. Crucial was the identification of a dithiophosphoric acid that masters three catalytic tasks, sequentially acting as a Brønsted acid for pyridine protonation, a single electron transfer (SET) reductant for pyridinium ion reduction, and a hydrogen atom abstractor for the activation of allylic C(sp3)-H bonds. The resulting pyridinyl and allylic radicals then couple with high regioselectivity.


Subject(s)
Hydrogen , Pyridines , Pyridines/chemistry , Hydrogen/chemistry , Electron Transport , Reducing Agents , Catalysis
8.
J Virol ; 94(10)2020 05 04.
Article in English | MEDLINE | ID: mdl-32102889

ABSTRACT

Human adenoviruses have many attractive features for gene therapy applications. However, the high prevalence of preexisting immunity against these viruses in general populations worldwide has greatly limited their clinical utility. In addition, the most commonly used human adenovirus, human adenovirus subgroup C serotype 5 (HAd5), when systemically administered, triggers systemic inflammation and toxicity, with the liver being the most severely affected organ. Here, we evaluated the utility and safety of a new low-seroprevalence gorilla adenovirus (GAd; GC46) as a gene transfer vector in mice. Biodistribution studies revealed that systemically administered GAd had a selective and robust lung endothelial cell (EC) tropism with minimal vector expression throughout many other organs and tissues. Administration of a high dose of GAd accomplished extensive transgene expression in the lung yet elicited no detectable inflammatory histopathology in this organ. Furthermore, GAd, unlike HAd5, did not exhibit hepatotropism or induce liver inflammatory toxicity in mice, demonstrating the exceptional safety profile of the vector vis-à-vis systemic utility. We further demonstrated that the GAd capsid fiber shared the flexibility of the HAd5 equivalent for permitting genetic modification; GAd with the pan-EC-targeting ligand myeloid cell-binding peptide (MBP) incorporated in the capsid displayed a reduced lung tropism and efficiently retargeted gene expression to vascular beds in other organs.IMPORTANCE In the aggregate, our mouse studies suggest that GAd is a promising gene therapy vector that utilizes lung ECs as a source of therapeutic payload production and a highly desirable toxicity profile. Further genetic engineering of the GAd capsid holds the promise of in vivo vector tropism modification and targeting.


Subject(s)
Adenoviridae/genetics , Capsid/metabolism , Genetic Vectors , Gorilla gorilla/virology , Lung/metabolism , Tropism/immunology , Adenoviridae Infections/pathology , Adenoviridae Infections/virology , Adenoviruses, Human/genetics , Animals , Capsid Proteins/genetics , Endothelial Cells , Gene Expression , Genetic Therapy , Liver , Lung/pathology , Lung/virology , Mice , Mice, Inbred C57BL , Seroepidemiologic Studies , Transduction, Genetic , Transgenes , Virion
10.
Beilstein J Org Chem ; 16: 3104-3108, 2020.
Article in English | MEDLINE | ID: mdl-33437323

ABSTRACT

A method for the one-step construction of 3,3,4,4-tetrafluorinated piperidines from nitrones and readily accessible tetrafluorinated iodobromobutane is described. The reaction requires an excess amount of ascorbic acid as the terminal reductant and is performed in the presence of an iridium photocatalyst activated by blue light. The annelation is a result of a radical addition at the nitrone, intramolecular nucleophilic substitution, and reduction of the N-O bond.

12.
J Immunol ; 197(7): 2748-61, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27574299

ABSTRACT

An ideal malaria vaccine should target several stages of the parasite life cycle and induce antiparasite and antidisease immunity. We have reported a Plasmodium yoelii chimeric multistage recombinant protein (P. yoelii linear peptide chimera/recombinant modular chimera), engineered to express several autologous T cell epitopes and sequences derived from the circumsporozoite protein and the merozoite surface protein 1. This chimeric protein elicits protective immunity, mediated by CD4(+) T cells and neutralizing Abs. However, experimental evidence, from pre-erythrocytic vaccine candidates and irradiated sporozoites, has shown that CD8(+) T cells play a significant role in protection. Recombinant viral vectors have been used as a vaccine platform to elicit effective CD8(+) T cell responses. The human adenovirus (Ad) serotype 5 has been tested in malaria vaccine clinical trials with excellent safety profile. Nevertheless, a major concern for the use of Ad5 is the high prevalence of anti-vector neutralizing Abs in humans, hampering its immunogenicity. To minimize the impact of anti-vector pre-existing immunity, we developed a chimeric Ad5/3 vector in which the knob region of Ad5 was replaced with that of Ad3, conferring partial resistance to anti-Ad5 neutralizing Abs. Furthermore, we implemented heterologous Ad/protein immunization regimens that include a single immunization with recombinant Ad vectors. Our data show that immunization with the recombinant Ad5/3 vector induces protective efficacy indistinguishable from that elicited by Ad5. Our study also demonstrates that the dose of the Ad vectors has an impact on the memory profile and protective efficacy. The results support further studies with Ad5/3 for malaria vaccine development.


Subject(s)
Adenoviruses, Human/genetics , Antigens, Protozoan/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors/genetics , Immunity, Cellular/immunology , Malaria Vaccines/immunology , Plasmodium yoelii/immunology , Animals , Antigens, Protozoan/genetics , Female , HEK293 Cells , Humans , Malaria Vaccines/genetics , Mice , Mice, Transgenic , Recombinant Proteins/genetics , Recombinant Proteins/immunology
13.
Infect Immun ; 83(1): 286-91, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25368111

ABSTRACT

Hemolytic-uremic syndrome (HUS), caused by Shiga toxin (Stx)-producing Escherichia coli (STEC), remains untreatable. Production of human monoclonal antibodies against Stx, which are highly effective in preventing Stx sequelae in animal models, is languishing due to cost and logistics. We reported previously that the production and evaluation of a camelid heavy-chain-only VH domain (VHH)-based neutralizing agent (VNA) targeting Stx1 and Stx2 (VNA-Stx) protected mice from Stx1 and Stx2 intoxication. Here we report that a single intramuscular (i.m.) injection of a nonreplicating adenovirus (Ad) vector carrying a secretory transgene of VNA-Stx (Ad/VNA-Stx) protected mice challenged with Stx2 and protected gnotobiotic piglets infected with STEC from fatal systemic intoxication. One i.m. dose of Ad/VNA-Stx prevented fatal central nervous system (CNS) symptoms in 9 of 10 animals when it was given to piglets 24 h after bacterial challenge and in 5 of 9 animals when it was given 48 h after bacterial challenge, just prior to the onset of CNS symptoms. All 6 placebo animals died or were euthanized with severe CNS symptoms. Ad/VNA-Stx treatment had no impact on diarrhea. In conclusion, Ad/VNA-Stx treatment is effective in protecting piglets from fatal Stx2-mediated CNS complications following STEC challenge. With a low production cost and further development, this could presumably be an effective treatment for patients with HUS and/or individuals at high risk of developing HUS due to exposure to STEC.


Subject(s)
Adenoviruses, Human/genetics , Antibodies, Neutralizing/therapeutic use , Escherichia coli Infections/drug therapy , Escherichia coli O157/immunology , Hemolytic-Uremic Syndrome/drug therapy , Shiga Toxin 1/antagonists & inhibitors , Shiga Toxin 2/antagonists & inhibitors , Animals , Antibodies, Neutralizing/genetics , Disease Models, Animal , Drug Carriers/administration & dosage , Escherichia coli Infections/immunology , Escherichia coli Infections/microbiology , Escherichia coli O157/genetics , Female , Genetic Vectors , Hemolytic-Uremic Syndrome/immunology , Hemolytic-Uremic Syndrome/microbiology , Injections, Intramuscular , Mice , Shiga Toxin 1/immunology , Shiga Toxin 2/immunology , Survival Analysis , Swine , Time Factors
14.
J Cell Physiol ; 229(1): 34-43, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23868767

ABSTRACT

Few options are available for treating patients with advanced prostate cancer (PC). As PC is a slow growing disease and accessible by ultrasound, gene therapy could provide a viable option for this neoplasm. Conditionally replication-competent adenoviruses (CRCAs) represent potentially useful reagents for treating PC. We previously constructed a CRCA, cancer terminator virus (CTV), which showed efficacy both in vitro and in vivo for PC. The CTV was generated on a serotype 5-background (Ad.5-CTV) with infectivity depending on Coxsackie-Adenovirus Receptors (CARs). CARs are frequently reduced in many tumor types, including PCs thereby limiting effective Ad-mediated therapy. Using serotype chimerism, a novel CTV (Ad.5/3-CTV) was created by replacing the Ad.5 fiber knob with the Ad.3 fiber knob thereby facilitating infection in a CAR-independent manner. We evaluated Ad.5/3-CTV in comparison with Ad.5-CTV in low CAR human PC cells, demonstrating higher efficiency in inhibiting cell viability in vitro. Moreover, Ad.5/3-CTV potently suppressed in vivo tumor growth in a nude mouse xenograft model and in a spontaneously induced PC that develops in Hi-myc transgenic mice. Considering the significant responses in a Phase I clinical trial of a non-replicating Ad.5-mda-7 in advanced cancers, Ad.5/3-CTV may exert improved therapeutic benefit in a clinical setting.


Subject(s)
Coxsackie and Adenovirus Receptor-Like Membrane Protein/genetics , Genetic Therapy , Oncolytic Viruses/genetics , Prostatic Neoplasms/genetics , Prostatic Neoplasms/therapy , Adenoviridae/genetics , Animals , Cell Line, Tumor , Cell Survival/genetics , Coxsackie and Adenovirus Receptor-Like Membrane Protein/therapeutic use , Gene Transfer Techniques , Humans , Male , Mice , Mice, Nude , Oncolytic Virotherapy , Prostatic Neoplasms/pathology , Recombinant Proteins/therapeutic use , Serotyping , Xenograft Model Antitumor Assays
15.
Mol Imaging ; 132014.
Article in English | MEDLINE | ID: mdl-25249483

ABSTRACT

A significant limiting factor to the human clinical application of conditionally replicative adenovirus (CRAd)-based virotherapy is the inability to noninvasively monitor these agents and their potential persistence. To address this issue, we proposed a novel imaging approach that combines transient expression of the human somatostatin receptor (SSTR) subtype 2 reporter gene with genetic labeling of the viral capsid with mCherry fluorescent protein. To test this dual modality system, we constructed the Ad5/3Δ24pIXcherry/SSTR CRAd and validated its capacity to generate fluorescent and nuclear signals in vitro and following intratumoral injection. Analysis of 64Cu-CB-TE2A-Y3-TATE biodistribution in mice revealed reduced uptake in tumors injected with the imaging CRAd relative to the replication-incompetent, Ad-expressing SSTR2 but significantly greater uptake compared to the negative CRAd control. Optical imaging demonstrated relative correlation of fluorescent signal with virus replication as determined by viral genome quantification in tumors. Positron emission tomography/computed tomography studies demonstrated that we can visualize radioactive uptake in tumors injected with imaging CRAd and the trend for greater uptake by standardized uptake value analysis compared to control CRAd. In the aggregate, the plasticity of our dual imaging approach should provide the technical basis for monitoring CRAd biodistribution and persistence in preclinical studies while offering potential utility for a range of clinical applications.


Subject(s)
Adenoviridae/physiology , Capsid/physiology , Luminescent Agents/metabolism , Luminescent Proteins/metabolism , Ovarian Neoplasms/virology , Receptors, Somatostatin/metabolism , Animals , Capsid/chemistry , Cell Line, Tumor , Coordination Complexes/pharmacokinetics , Female , HEK293 Cells , Humans , Mice , Neoplasm Transplantation , Oncolytic Virotherapy , Oncolytic Viruses/physiology , Peptides/pharmacokinetics , Receptors, Somatostatin/genetics , Virus Replication , Red Fluorescent Protein
16.
Mol Ther ; 21(4): 758-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23319057

ABSTRACT

Hepatocellular carcinoma (HCC) is a highly fatal disease mandating development of novel, targeted therapies to elicit prolonged survival benefit to the patients. Insulin-like growth factor-binding protein-7 (IGFBP7), a secreted protein belonging to the IGFBP family, functions as a potential tumor suppressor for HCC. In the present study, we evaluated the therapeutic efficacy of a replication-incompetent adenovirus expressing IGFBP7 (Ad.IGFBP7) in human HCC. Ad.IGFBP7 profoundly inhibited viability and induced apoptosis in multiple human HCC cell lines by inducing reactive oxygen species (ROS) and activating a DNA damage response (DDR) and p38 MAPK. In orthotopic xenograft models of human HCC in athymic nude mice, intravenous administration of Ad.IGFBP7 profoundly inhibited primary tumor growth and intrahepatic metastasis. In a nude mice subcutaneous model, xenografts from human HCC cells were established in both flanks and only left-sided tumors received intratumoral injection of Ad.IGFBP7. Growth of both left-sided injected tumors and right-sided uninjected tumors were markedly inhibited by Ad.IGFBP7 with profound suppression of angiogenesis. These findings indicate that Ad.IGFBP7 might be a potent therapeutic eradicating both primary HCC and distant metastasis and might be an effective treatment option for terminal HCC patients.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Insulin-Like Growth Factor Binding Proteins/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Adenoviridae/genetics , Animals , Carcinoma, Hepatocellular/genetics , Humans , Insulin-Like Growth Factor Binding Proteins/genetics , Liver Neoplasms/genetics , Male , Mice , Mice, Nude , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
17.
Proc Natl Acad Sci U S A ; 108(21): 8785-90, 2011 May 24.
Article in English | MEDLINE | ID: mdl-21555592

ABSTRACT

Limited options are available for treating patients with advanced prostate cancer (PC). Melanoma differentiation associated gene-7/interleukin-24 (mda-7/IL-24), an IL-10 family cytokine, exhibits pleiotropic anticancer activities without adversely affecting normal cells. We previously demonstrated that suppression of the prosurvival Bcl-2 family member, myeloid cell leukemia-1 (Mcl-1), is required for mda-7/IL-24-mediated apoptosis of prostate carcinomas. Here we demonstrate that pharmacological inhibition of Mcl-1 expression with the unique Apogossypol derivative BI-97C1, also called Sabutoclax, is sufficient to sensitize prostate tumors to mda-7/IL-24-induced apoptosis, whereas ABT-737, which lacks efficacy in inhibiting Mcl-1, does not sensitize mda-7/IL-24-mediated cytotoxicity. A combination regimen of tropism-modified adenovirus delivered mda-7/IL-24 (Ad.5/3-mda-7) and BI-97C1 enhances cytotoxicity in human PC cells, including those resistant to mda-7/IL-24 or BI-97C1 alone. The combination regimen causes autophagy that facilitates NOXA- and Bim-induced and Bak/Bax-mediated mitochondrial apoptosis. Treatment with Ad.5/3-mda-7 and BI-97C1 significantly inhibits the growth of human PC xenografts in nude mice and spontaneously induced PC in Hi-myc transgenic mice. Tumor growth inhibition correlated with increased TUNEL staining and decreased Ki-67 expression in both PC xenografts and prostates of Hi-myc mice. These findings demonstrate that pharmacological inhibition of Mcl-1 with the Apogossypol derivative, BI-97C1, sensitizes human PCs to mda-7/IL-24-mediated cytotoxicity, thus potentially augmenting the therapeutic benefit of this combinatorial approach toward PC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Genetic Therapy/methods , Gossypol/analogs & derivatives , Prostatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Animals , Cell Line, Tumor , Gossypol/pharmacology , Gossypol/therapeutic use , Humans , Interleukins/administration & dosage , Male , Mice , Mice, Nude , Myeloid Cell Leukemia Sequence 1 Protein , Prostatic Neoplasms/pathology , Prostatic Neoplasms/therapy , Transfection , Xenograft Model Antitumor Assays
18.
Vaccines (Basel) ; 12(9)2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39340002

ABSTRACT

Chikungunya virus (CHIKV) is a pathogenic arthritogenic alphavirus responsible for large-scale human epidemics for which a vaccine was recently approved for use. Mayaro virus (MAYV) is a related emerging alphavirus with epidemic potential with circulation overlap potential with CHIKV. We previously reported the ability of a non-replicating human adenovirus (AdV)-vectored vaccine expressing the MAYV structural polyprotein to protect against disease in mice following challenge with MAYV, CHIKV and UNAV. Herein, we evaluated mouse immunity and protective efficacy for an AdV-CHIKV full structural polyprotein vaccine in combination with heterologous AdV-MAYV prime/boost regimens versus vaccine coadministration. Heterologous prime/boost regimens skewed immunity toward the prime vaccine antigen but allowed for a boost of cross-neutralizing antibodies, while vaccine co-administration elicited robust, balanced responses capable of boosting. All immunization strategies protected against disease from homologous virus infection, but reciprocal protective immunity differences were revealed upon challenge with heterologous viruses. In vivo passive transfer experiments reproduced the inequity in reciprocal cross-protection after heterologous MAYV challenge. We detected in vitro antibody-dependent enhancement of MAYV replication, suggesting a potential mechanism for the lack of cross-protection. Our findings provide important insights into rational alphavirus vaccine design that may have important implications for the evolving alphavirus vaccine landscape.

19.
Gynecol Oncol ; 130(3): 518-24, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23756180

ABSTRACT

OBJECTIVE: The conditionally replicative adenovirus Ad5/3-Δ24 has a type-3 knob incorporated into the type-5 fiber that facilitates enhanced ovarian cancer infectivity. Preclinical studies have shown that Ad5/3-Δ24 achieves significant oncolysis and anti-tumor activity in ovarian cancer models. The purpose of this study was to evaluate in a phase I trial the feasibility and safety of intraperitoneal (IP) Ad5/3-Δ24 in recurrent ovarian cancer patients. METHODS: Eligible patients were treated with IP Ad5/3-Δ24 for 3 consecutive days in one of three dose cohorts ranging 1 × 10(10)-1 × 10(12)vp. Toxicity was assessed utilizing CTC grading and efficacy with RECIST. Ascites, serum, and other samples were obtained to evaluate gene transfer, generation of wildtype virus, viral shedding, and antibody response. RESULTS: Nine of 10 patients completed treatment per protocol. A total of 15 vector-related adverse events were experienced in 5 patients. These events included fever or chills, nausea, fatigue, and myalgia. All were grades 1-2 in nature, transient, and medically managed. Of the 8 treated patients evaluable for response, six patients had stable disease and 2 patients had progressive disease. Three patients had decreased CA-125 from pretreatment levels one month after treatment. Ancillary biologic studies indicated Ad5/3-Δ24 replication in patients in the higher dose cohorts. All patients experienced an anti-adenoviral neutralizing antibody effect. CONCLUSIONS: This study suggests the feasibility and safety of a serotype chimeric infectivity-enhanced CRAd, Ad5/3-Δ24, as a potential therapeutic option for recurrent ovarian cancer patients.


Subject(s)
Adenoviridae , Antibodies, Viral/blood , Oncolytic Virotherapy , Oncolytic Viruses , Ovarian Neoplasms/therapy , Adenoviridae/genetics , Adenoviridae/physiology , Aged , Aged, 80 and over , Ascites/virology , CA-125 Antigen/blood , Chills/virology , Disease Progression , Fatigue/virology , Female , Fever/virology , Gene Expression , Humans , Maximum Tolerated Dose , Middle Aged , Nausea/virology , Oncolytic Virotherapy/adverse effects , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Ovarian Neoplasms/blood , Ovarian Neoplasms/virology , Tomography, X-Ray Computed , Virus Replication , Virus Shedding
20.
Viruses ; 15(11)2023 Nov 20.
Article in English | MEDLINE | ID: mdl-38005953

ABSTRACT

mRNA vaccines have attracted widespread research attention with clear advantages in terms of molecular flexibility, rapid development, and potential for personalization. However, current mRNA vaccine platforms have not been optimized for induction of CD4/CD8 T cell responses. In addition, the mucosal administration of mRNA based on lipid nanoparticle technology faces challenges in clinical translation. In contrast, adenovirus-based vaccines induce strong T cell responses and have been approved for intranasal delivery. To leverage the inherent strengths of both the mRNA and adenovirus platforms, we developed a novel modular adenoviral mRNA delivery platform based on Tag/Catcher bioconjugation. Specifically, we engineered adenoviral vectors integrating Tag/Catcher proteins at specific locales on the Ad capsid proteins, allowing us to anchor mRNA to the surface of engineered Ad viruses. In proof-of-concept studies, the Ad-mRNA platform successfully mediated mRNA delivery and could be optimized via the highly flexible modular design of both the Ad-mRNA and protein bioconjugation systems.


Subject(s)
Adenoviridae , Genetic Vectors , mRNA Vaccines , Adenoviridae/genetics , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Genetic Vectors/genetics , Genetic Engineering
SELECTION OF CITATIONS
SEARCH DETAIL