Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros












Base de dados
Intervalo de ano de publicação
1.
Front Immunol ; 14: 1188392, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37662899

RESUMO

Introduction: Vaccination is the most effective mechanism to prevent severe COVID-19. However, breakthrough infections and subsequent transmission of SARS-CoV-2 remain a significant problem. Intranasal vaccination has the potential to be more effective in preventing disease and limiting transmission between individuals as it induces potent responses at mucosal sites. Methods: Utilizing a replication-deficient adenovirus serotype 5-vectored vaccine expressing the SARS-CoV-2 RBD (AdCOVID) in homozygous and heterozygous transgenic K18-hACE2, we investigated the impact of the route of administration on vaccine immunogenicity, SARS-CoV-2 transmission, and survival. Results: Mice vaccinated with AdCOVID via the intramuscular or intranasal route and subsequently challenged with SARS-CoV-2 showed that animals vaccinated intranasally had improved cellular and mucosal antibody responses. Additionally, intranasally vaccinated animals had significantly better viremic control, and protection from lethal infection compared to intramuscularly vaccinated animals. Notably, in a novel transmission model, intranasal vaccination reduced viral transmission to naïve co-housed mice compared to intramuscular vaccination. Discussion: Our data provide convincing evidence for the use of intranasal vaccination in protecting against SARS-CoV-2 infection and transmission.


Assuntos
Infecções por Adenoviridae , Vacinas contra Adenovirus , COVID-19 , Vacinas , Animais , Camundongos , Adenoviridae/genética , SARS-CoV-2 , COVID-19/prevenção & controle , Vacinação , Animais Geneticamente Modificados
2.
Hum Vaccin Immunother ; 18(6): 2127292, 2022 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-36194255

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic has illustrated the critical need for effective prophylactic vaccination to prevent the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Intranasal vaccination is an attractive approach for preventing COVID-19 as the nasal mucosa is the site of initial SARS-CoV-2 entry and viral replication prior to aspiration into the lungs. We previously demonstrated that a single intranasal administration of a candidate adenovirus type 5-vectored vaccine encoding the receptor-binding domain of the SARS-CoV-2 spike protein (AdCOVID) induced robust immunity in both the airway mucosa and periphery, and completely protected K18-hACE2 mice from lethal SARS-CoV-2 challenge. Here we show that a single intranasal administration of AdCOVID limits viral replication in the nasal cavity of K18-hACE2 mice. AdCOVID also induces sterilizing immunity in the lungs of mice as reflected by the absence of infectious virus. Finally, AdCOVID prevents SARS-CoV-2 induced pathological damage in the lungs of mice. These data show that AdCOVID not only limits viral replication in the respiratory tract, but it also prevents virus-induced inflammation and immunopathology following SARS-CoV-2 infection.


Assuntos
Vacinas contra COVID-19 , COVID-19 , SARS-CoV-2 , Animais , Humanos , Camundongos , Administração Intranasal , Anticorpos Antivirais , COVID-19/prevenção & controle , Pulmão , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus , Vacinas Virais/administração & dosagem , Vacinas contra COVID-19/administração & dosagem
3.
Vaccines (Basel) ; 9(8)2021 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-34452006

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic has highlighted the urgent need for effective prophylactic vaccination to prevent the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Intranasal vaccination is an attractive strategy to prevent COVID-19 as the nasal mucosa represents the first-line barrier to SARS-CoV-2 entry. The current intramuscular vaccines elicit systemic immunity but not necessarily high-level mucosal immunity. Here, we tested a single intranasal dose of our candidate adenovirus type 5-vectored vaccine encoding the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein (AdCOVID) in inbred, outbred, and transgenic mice. A single intranasal vaccination with AdCOVID elicited a strong and focused immune response against RBD through the induction of mucosal IgA in the respiratory tract, serum neutralizing antibodies, and CD4+ and CD8+ T cells with a Th1-like cytokine expression profile. A single AdCOVID dose resulted in immunity that was sustained for over six months. Moreover, a single intranasal dose completely protected K18-hACE2 mice from lethal SARS-CoV-2 challenge, preventing weight loss and mortality. These data show that AdCOVID promotes concomitant systemic and mucosal immunity and represents a promising vaccine candidate.

4.
Vaccines (Basel) ; 9(3)2021 Mar 05.
Artigo em Inglês | MEDLINE | ID: mdl-33807649

RESUMO

Annual influenza vaccination greatly reduces morbidity and mortality, but effectiveness remains sub-optimal. Weaknesses of current vaccines include low effectiveness against mismatched strains, lack of mucosal and other effective tissue-resident immune responses, weak cellular immune responses, and insufficiently durable immune responses. The safety and immunogenicity of NasoVAX, a monovalent intranasal influenza vaccine based on a replication-deficient adenovirus type 5 platform, were evaluated in a placebo-controlled single ascending-dose study. Sixty healthy adults (18-49 years) received a single intranasal dose of 1×109 viral particles (vp), 1 × 1010 vp, or 1 × 1011 vp of NasoVAX or placebo. NasoVAX was well-tolerated and elicited robust influenza-specific systemic and mucosal immune responses. The highest NasoVAX dose and the approved Fluzone® influenza vaccine elicited comparable hemagglutination inhibition (HAI) geometric mean titers (152.8 vs. 293.4) and microneutralization (MN) geometric mean titers (142.5 vs. 162.8), with NasoVAX HAI titers maintained more than 1-year on average following a single dose. Hemagglutinin-specific T cells responses were also documented in peripheral mononuclear cell (PBMC) preparations. Consistent with the intranasal route of administration, NasoVAX elicited antigen-specific mucosal IgA responses in the nasopharyngeal cavity with an increase of approximately 2-fold over baseline GMT at the mid- and high-doses. In summary, NasoVAX appeared safe and elicited a broad immune response, including humoral, cellular, and mucosal immunity, with no impact of baseline anti-adenovirus antibody at the most immunogenic dose.

5.
Vaccines (Basel) ; 8(2)2020 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-32272595

RESUMO

Zika virus (ZIKV) is a significant public health concern due to the pathogen's ability to be transmitted by either mosquito bite or sexual transmission, allowing spread to occur throughout the world. The potential consequences of ZIKV infection to human health, specifically neonates, necessitates the development of a safe and effective Zika virus vaccine. Here, we developed an intranasal Zika vaccine based upon the replication-deficient human adenovirus serotype 5 (hAd5) expressing ZIKV pre-membrane and envelope protein (hAd5-ZKV). The hAd5-ZKV vaccine is able to induce both cell-mediated and humoral immune responses to ZIKV epitopes. Importantly, this vaccine generated CD8+ T cells specific for a dominant ZIKV T cell epitope and is shown to be protective against a ZIKV challenge by using a pre-clinical model of ZIKV disease. We also demonstrate that the vaccine expresses pre-membrane and envelope protein in a confirmation recognized by ZIKV experienced individuals. Our studies demonstrate that this adenovirus-based vaccine expressing ZIKV proteins is immunogenic and protective in mice, and it encodes ZIKV proteins in a conformation recognized by the human antibody repertoire.

6.
Clin Vaccine Immunol ; 22(4): 430-9, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25673303

RESUMO

AdVAV is a replication-deficient adenovirus type 5-vectored vaccine expressing the 83-kDa protective antigen (PA83) from Bacillus anthracis that is being developed for the prevention of disease caused by inhalation of aerosolized B. anthracis spores. A noninferiority study comparing the efficacy of AdVAV to the currently licensed Anthrax Vaccine Absorbed (AVA; BioThrax) was performed in New Zealand White rabbits using postchallenge survival as the study endpoint (20% noninferiority margin for survival). Three groups of 32 rabbits were vaccinated with a single intranasal dose of AdVAV (7.5 × 10(7), 1.5 × 10(9), or 3.5 × 10(10) viral particles). Three additional groups of 32 animals received two doses of either intranasal AdVAV (3.5 × 10(10) viral particles) or intramuscular AVA (diluted 1:16 or 1:64) 28 days apart. The placebo group of 16 rabbits received a single intranasal dose of AdVAV formulation buffer. All animals were challenged via the inhalation route with a targeted dose of 200 times the 50% lethal dose (LD50) of aerosolized B. anthracis Ames spores 70 days after the initial vaccination and were followed for 3 weeks. PA83 immunogenicity was evaluated by validated toxin neutralizing antibody and serum anti-PA83 IgG enzyme-linked immunosorbent assays (ELISAs). All animals in the placebo cohort died from the challenge. Three of the four AdVAV dose cohorts tested, including two single-dose cohorts, achieved statistical noninferiority relative to the AVA comparator group, with survival rates between 97% and 100%. Vaccination with AdVAV also produced antibody titers with earlier onset and greater persistence than vaccination with AVA.


Assuntos
Vacinas contra Antraz/administração & dosagem , Vacinas contra Antraz/imunologia , Antraz/prevenção & controle , Antígenos de Bactérias/imunologia , Toxinas Bacterianas/imunologia , Portadores de Fármacos , Mastadenovirus/genética , Infecções Respiratórias/prevenção & controle , Administração Intranasal , Animais , Antraz/imunologia , Vacinas contra Antraz/genética , Anticorpos Antibacterianos/sangue , Anticorpos Neutralizantes/sangue , Antígenos de Bactérias/genética , Antitoxinas/sangue , Toxinas Bacterianas/genética , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Feminino , Vetores Genéticos , Imunoglobulina G/sangue , Masculino , Testes de Neutralização , Coelhos , Infecções Respiratórias/imunologia , Análise de Sobrevida , Vacinação/métodos , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
7.
Curr Cancer Drug Targets ; 7(2): 157-67, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17346107

RESUMO

PV701 is a naturally-attenuated, non-recombinant, oncolytic strain of Newcastle disease virus that displays preclinical intravenous (IV) efficacy. PV701 is selective at killing human cancer cells versus normal human cells based on tumor specific defects in the interferon (IFN)-mediated antiviral response. This oncolytic virus displays a broad spectrum of antitumor activity in vitro and in vivo. Preclinical models successfully predicted key clinical parameters including the mechanism of toxicity, two complementary strategies (desensitization and slow infusion) to reduce toxicity, and the starting dose for phase 1 trials. In three phase 1 trials of 114 patients using IV administration of PV701, Wellstat Biologics Corporation has evaluated the effects of dose, schedule, and infusion rate for PV701. Three general classes of side effects were seen: flu-like symptoms; tumor-site-specific adverse events (AEs); and infusion reactions. The first PV701 dose desensitized the patient to the side effects of further doses, allowing a marked increase in the maximum tolerated dose for subsequent doses compared to the first dose. Tumor responses were first noted at the higher doses achieved using desensitization. In the most recent phase 1 trial of 19 patients at Hamilton, Ontario, that employed desensitization, high repeat doses, and a slower infusion rate (Hamilton Regimen), there were six responses (4 major; 2 minor) and a total of six patients with survival for at least 2 years. In addition, patient tolerability improved using the Hamilton Regimen compared to IV bolus dosing used previously. Phase 2 studies of this novel biologic agent are about to begin.


Assuntos
Neoplasias/terapia , Vírus da Doença de Newcastle , Terapia Viral Oncolítica , Animais , Ensaios Clínicos Fase I como Assunto/métodos , Humanos , Infusões Intravenosas , Injeções Intravenosas , Vírus da Doença de Newcastle/genética , Vírus da Doença de Newcastle/fisiologia , Terapia Viral Oncolítica/efeitos adversos , Projetos de Pesquisa , Resultado do Tratamento , Replicação Viral
8.
Clin Cancer Res ; 13(3): 977-85, 2007 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17289893

RESUMO

PURPOSE: Previous phase 1 trials of i.v.-administered PV701 have shown this virus to be well-tolerated with toxicity primarily associated with the first dose. Our hypothesis, based on preclinical evidence, was that patient tolerability could be improved by slowing the i.v. infusion rate, and that this approach would allow for the safe administration of higher doses. Additionally, this phase 1 trial was the first to measure PV701 clearance. EXPERIMENTAL DESIGN: For the first dose, a 3-h infusion was used compared with the 10- and 30-min infusions administered in the two previous trials. Subsequent doses were infused over 1 h. Six doses were given per 3-week cycle. Escalation of the first dose was done separately from the escalation of doses 2 to 6. Viral clearance was determined using whole blood reverse transcription-PCR. RESULTS: Eighteen patients with advanced chemorefractory cancer were enrolled. The first dose was safely escalated to 24x10(9) plaque-forming units/m2 and doses 2 to 6 were safely escalated to 120x10(9) plaque-forming units/m2. Tolerability was improved compared with the rapid bolus dosing used previously with the elimination of severe flu-like symptoms. Furthermore, infusion reactions were markedly decreased in this trial compared with previous PV701 trials. The presence of neutralizing antibodies did not significantly affect PV701 clearance. Four major and two minor tumor responses were observed. CONCLUSIONS: Using slow infusion, patient tolerability was improved, while the first dose was safely escalated relative to two previous PV701 trials. Based on improved tolerability and encouraging signs of activity, this slow infusion regimen was selected for further PV701 clinical development.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/metabolismo , Adulto , Anticorpos/química , Protocolos Clínicos , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia Viral Oncolítica/efeitos adversos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Tempo , Resultado do Tratamento
9.
Curr Opin Mol Ther ; 8(4): 314-21, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16955694

RESUMO

Naturally oncolytic viruses are replication-competent viruses that have an innate ability to selectively infect and kill tumor cells. Despite being used in the original attempts to treat cancer with live viruses five decades ago, interest in naturally oncolytic viruses has lagged behind the support for engineered adenoviruses and herpesviruses as cancer therapeutics. Recently, however, there has been renewed interest in the high potency and selectivity of these naturally occurring agents. In this review, the mechanism of selectivity as well as the preclinical and clinical activity of naturally oncolytic viruses will be discussed.


Assuntos
Neoplasias/terapia , Terapia Viral Oncolítica , Vírus Oncolíticos , Animais , Vetores Genéticos , Humanos , Proteína Cofatora de Membrana/metabolismo , Neoplasias/patologia , Vírus Oncolíticos/genética , Vírus Oncolíticos/metabolismo , Replicação Viral
10.
Clin Cancer Res ; 12(8): 2555-62, 2006 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-16638865

RESUMO

PURPOSE: In a previous phase 1 study, adverse events, especially flu-like symptoms, were observed mainly following the first i.v. bolus dose of PV701, an oncolytic Newcastle disease virus. Desensitization to adverse events of subsequent doses occurred, allowing a 10-fold increase in the maximum tolerated dose for these doses. Although one-step desensitization (a single desensitizing dose with higher subsequent doses) addressed the tolerability of high repeat doses, additional testing was required to further improve tolerability of the initial dose. This study tested the hypothesis that two-step desensitization, using two dose increments before high repeat doses, would be well tolerated. EXPERIMENTAL DESIGN: Sixteen adults with incurable solid tumors were enrolled. Cycles consisted of six PV701 doses over 2 weeks followed by a 1-week rest. Doses 1 to 2 were 1 and 12 x 10(9) plaque-forming units (pfu)/m(2), respectively, whereas doses 3 to 6 were escalated by cohort from 24 to 120 x 10(9) pfu/m(2). RESULTS: No dose-limiting toxicities were observed, permitting dose escalation through cohort 4 (1, 12, 120, 120, 120, 120 x 10(9) pfu/m(2)). Mild flu-like symptoms were common following the first infusion, diminished with repeated dosing, and were less pronounced than those seen previously. Tumor regression was observed in a patient with anal carcinoma who enrolled with stable disease following palliative radiotherapy. Four patients with clearly progressing cancer before enrollment had disease stabilization of >/=6 months. CONCLUSIONS: This novel two-step desensitization improved patient tolerability compared with the previous regimen. Toxicities were predictable and manageable. PV701, the first oncolytic virus to enter phase 1 i.v. testing, continues to show single-agent activity, warranting planned phase 2 trials.


Assuntos
Dessensibilização Imunológica/métodos , Neoplasias/terapia , Terapia Viral Oncolítica/métodos , Vírus Oncolíticos/imunologia , Adulto , Idoso , Calafrios/etiologia , Estudos de Coortes , Fadiga/etiologia , Feminino , Febre/etiologia , Cefaleia/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias/imunologia , Terapia Viral Oncolítica/efeitos adversos , Trombocitopenia/etiologia , Resultado do Tratamento
11.
Discov Med ; 6(36): 217-22, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17250786

RESUMO

The use of naturally occurring viruses in the treatment of cancer is not new, and reports describing the administration of viruses to cancer patients date back more than fifty years. The advent of molecular biology and the arrival of biologics to the cancer therapy arena have now set the stage for a fresh look at the potential of these agents. These viruses have the potential to specifically target tumor cells, and insights into the molecular mechanisms of this selectivity are beginning to emerge.


Assuntos
Neoplasias/terapia , Neoplasias/virologia , Terapia Viral Oncolítica , Vírus Oncolíticos/fisiologia , Animais , Humanos , Vírus Oncolíticos/genética
12.
Curr Opin Mol Ther ; 5(6): 618-24, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14755888

RESUMO

PV701 is an attenuated, non-recombinant, oncolytic strain of Newcastle disease virus that displays preclinical intravenous (i.v.) efficacy. PV701 selectively lyses tumor cells versus normal cells based on tumor-specific defects in the interferon-mediated antiviral response. In three phase I trials in 113 patients, the effects of dose, schedule and i.v. infusion rate were evaluated. Three types of adverse events were seen: flu-like, tumor-site-specific and those occurring during infusion. The first PV701 dose desensitized the patient to the side effects of further doses, allowing a 5- to 10-fold increase in the maximum-tolerated dose for subsequent doses compared with the first dose. Tumor responses were first noted at the higher doses achieved using desensitization. In 95 evaluable patients, there were ten responses (six major and four minor), with five of these responses occurring in the most recent trial of 18 patients that employed desensitization, high repeat doses and a slower infusion rate. Phase II studies are planned.


Assuntos
Antineoplásicos/uso terapêutico , Terapia Genética/métodos , Infusões Intravenosas , Vírus da Doença de Newcastle/metabolismo , Vírus/metabolismo , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/efeitos adversos , Ensaios Clínicos Fase I como Assunto , Relação Dose-Resposta a Droga , Humanos , Vírus da Doença de Newcastle/genética , Vírus/genética
13.
J Clin Oncol ; 20(9): 2251-66, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11980996

RESUMO

PURPOSE: PV701, a replication-competent strain of Newcastle disease virus, causes regression of tumor xenografts after intravenous administration. This phase I study was designed to define the maximum-tolerated dose (MTD) and safety of single and multiple intravenous doses of PV701 as a single agent in patients with cancer. PATIENTS AND METHODS: Seventy-nine patients with advanced solid cancers that were unresponsive to standard therapy were enrolled. Four PV701 intravenous dosing regimens were evaluated: (1) single dose: one dose every 28 days; (2) repeat dose: three doses in 1 week every 28 days; (3) desensitizing: one lower dose followed by two higher doses in 1 week every 28 days; and (4) two week: one lower dose followed by five higher doses over 2 weeks every 21 days. RESULTS: A 100-fold dose intensification was achieved over 195 cycles. A first-dose MTD of 12 x 10(9) plaque-forming units (PFU)/m(2) was established for outpatient dosing. After an initial dose of 12 x 10(9) PFU/m(2), patients tolerated an MTD for subsequent doses of 120 x 10(9) PFU/m(2). The most common adverse events were flu-like symptoms that occurred principally after the first dose and were decreased in number and severity with each subsequent dose. Tumor site-specific adverse events and acute dosing reactions were also observed but not cumulative toxicity. Objective responses occurred at higher dose levels, and progression-free survival ranged from 4 to 31 months. Tumor tissue from one patient was obtained after 11 months of therapy and showed evidence of PV701 particles budding from the tumor cell membrane by electron microscopy and a pronounced lymphoplasmacytic infiltrate by histologic examination. CONCLUSION: PV701 warrants further study as a novel therapeutic agent for cancer patients.


Assuntos
Terapia Genética/métodos , Neoplasias/terapia , Vírus da Doença de Newcastle/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Feminino , Humanos , Infusões Intravenosas , Masculino , Pessoa de Meia-Idade , Neoplasias/genética , Neoplasias/patologia , Resultado do Tratamento , Replicação Viral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...