Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
medrxiv; 2022.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2022.01.17.22269242

ABSTRACT

BackgroundSeveral COVID-19 vaccines are currently being deployed but supply constraints, concerns over durability of immune responses, solidifying vaccine hesitancy/resistance and vaccine efficacy in the face of emerging variants mean that new vaccines continue to be needed to fight the ongoing pandemic. The vaccine described here is an enveloped, coronavirus-like particle produced in plants (CoVLP) that displays the prefusion-stabilized spike (S) glycoprotein of SARS-CoV-2 (ancestral Wuhan strain) and is adjuvanted with AS03 (CoVLP+AS03). MethodsThis Phase 3 randomized, observer-blind, placebo-controlled trial was conducted at 85 centers in Argentina, Brazil, Canada, Mexico, the UK, and the USA. Adults [≥]18 years of age including those at high risk for COVID-19 complications were randomly assigned 1:1 to receive two intramuscular injections of CoVLP (3.75 g) adjuvanted with AS03 or placebo, 21 days apart. The primary efficacy endpoint was prevention of symptomatic ([≥] 1 symptom), PCR-confirmed SARS-CoV-2 infection with onset at least 7 days after the second injection and was triggered by the identification of [≥]160 virologically-confirmed cases. Tolerability and safety of CoVLP+AS03 were also determined. ResultsA total of 24,141 volunteers were randomly assigned 1:1 to receive vaccine or placebo (N= 12,074 and 12,067, respectively: median age 29, range 18 to 86 years). Overall, 83% received both doses. 14.8% were SARS-CoV-2 seropositive at baseline. Symptomatic SARS-CoV-2 infection was confirmed in 165 study participants in the intention to treat (ITT) set and 157 in the per-protocol population (PP) set. Of the 157 in the PP set, 118 COVID-19 cases were in the placebo group and 39 COVID-19 cases were in the CoVLP+AS03 group for an overall vaccine efficacy (VE) of 71.0% (95% confidence interval (CI) 58.6, 80.0). Moderate-to-severe COVID-19 occurred in 8 and 32 participants in the CoVLP+AS03 and placebo groups, respectively: VE 78.1% (95% CI: 53.9, 90.5) in the PP set overall and 84.5% (95% CI: 62.0, 94.7) in those seronegative at recruitment. To date, 100% of the sequenced strains (122/165 cases: 73.39%) were variants, dominated by Delta (45.9%) and Gamma (43.4%) strains. Vaccine efficacy by variant was 75.3% (95% CI 52.8, 87.9) against Delta and 88.6% (95% CI 74.6, 95.6) against Gamma. Cross-protection was also observed against Alpha, Lambda and Mu variants; although fewer cases were identified, all were in the placebo group. At diagnosis, viral loads in the CoVLP+AS03 breakthrough cases were >100-fold lower than in the placebo cases. Reactogenicity data for solicited adverse events (AEs) was analysed for a subset (N=4,136 in vaccine arm and N=3,683 for placebo) of participants. Reactogenicity was mostly mild to moderate, and transient, and occurred more frequently in the CoVLP+AS03 group. The safety analysis set used for unsolicited AE assessment comprised 24,076 participants who received at least one study injection: 12,036 received CoVLP+AS03 and 12,040 received placebo. All serious adverse events were assessed as unrelated, except two events reported in the same subject in the placebo group. No significant imbalance or safety concern was noted in medically attended AEs (MAAEs), adverse event of special interest (AESIs), AEs leading to withdrawal, deaths, or adverse events potentially associated with currently authorized vaccines. ConclusionsThe CoVLP+AS03 vaccine candidate conferred an efficacy of 71.0% in preventing symptomatic SARS-CoV-2 infection caused by a spectrum of variants. Vaccine efficacy of 78.1% was observed against moderate and severe disease, while variant-specific efficacy ranged from 75.3% to 100%. Markedly lower viral loads in the CoVLP+AS03 group at the time of diagnosis suggests a significant virologic impact of vaccination even in the breakthrough cases. CoVLP+AS03 vaccine candidate was well tolerated, and no safety concerns were identified during the study. If approved by regulators, this more traditional protein+adjuvant vaccine produced using the novel plant-based platform may be able to make an important contribution to the global struggle against the increasingly complex family of SARS-CoV-2 viruses (Funded by Medicago with grants from the governments of Quebec and Canada; NCT04636697).


Subject(s)
Severe Acute Respiratory Syndrome , Drug-Related Side Effects and Adverse Reactions , Death , COVID-19
2.
medrxiv; 2021.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2021.07.28.21261159

ABSTRACT

Background: BNT162b2 is a lipid nanoparticle-formulated, nucleoside-modified RNA vaccine encoding a prefusion-stabilized, membrane-anchored SARS-CoV-2 full-length spike protein. BNT162b2 is highly efficacious against COVID-19 and is currently authorized for emergency use or conditional approval worldwide. At the time of authorization, data beyond 2 months post-vaccination were unavailable. Methods: In an ongoing, placebo-controlled, observer-blinded, multinational, pivotal efficacy study, 44,165 [≥]16-year-old participants and 2,264 12-15-year-old participants were randomized to receive 2 doses, 21 days apart, of 30 g BNT162b2 or placebo. Study endpoints reported here are vaccine efficacy (VE) against laboratory-confirmed COVID-19 and safety data, both up to 6 months post-vaccination. Results: BNT162b2 continued to be safe and well tolerated. Few participants had adverse events leading to study withdrawal. VE against COVID-19 was 91% (95% CI 89.0-93.2) through up to 6 months of follow-up, among evaluable participants and irrespective of previous SARS-CoV-2 infection. VE of 86%-100% was seen across countries and in populations with diverse characteristics of age, sex, race/ethnicity, and COVID-19 risk factors in participants without evidence of previous SARS-CoV-2 infection. VE against severe disease was 97% % (95% CI 80.3-99.9). In South Africa, where the SARS-CoV-2 variant of concern, B.1.351 (beta), was predominant, 100% (95% CI 53.5, 100.0) VE was observed. Conclusion: With up to 6 months of follow-up and despite a gradually declining trend in vaccine efficacy, BNT162b2 had a favorable safety profile and was highly efficacious in preventing COVID-19. (ClinicalTrials.gov number, NCT04368728)


Subject(s)
COVID-19
3.
Romina Libster; Gonzalo Perez Marc; Diego Wappner; Silvina Coviello; Alejandra Bianchi; Virginia Braem; Ignacio Esteban; Mauricio Tomas Caballero; Cristian J Wood; Mabel Berrueta; Anibal Rondan; Gabriela Lescano; Pablo Cruz; Ivonne Ritou; Valeria Fernandez Vina; Damian Alvarez Paggi; Sebastian Esperante; Adrian Ferretti; Gaston Ofman; Alvaro Ciganda; Rocio Rodriguez; Jorge Lantos; Ricardo Valentini; Nicolas Itcovici; Alejandra Hintze; Laura Oyarvide; Candela Etchegaray; Alejandra Neira; Ivonne Name; Julieta Alfonso; Rocio Lopez Castelo; Gisela Caruso; Sofia Rapelius; Fernando Alvez; Federico Cesar Etchenique; Federico Dimase; Dario Raul Alvarez; Sofia Sol Aranda; Clara Sanchez Yanotti; Julian DeLuca; Sofia Jarez Baglivo; Sofia Lujan Laudanno; Florencia Nowogrodzki; Florencia Izetta; Maria Teresa Paniguetti; Paula Fernandez Estrella; Maria Emilia Gutierrez Meyer; Viviana Dominguez; Marcela Balduzzi; Romina Militerno; Jimena Ochoa; Sebastian Perez Marc; Lucila DiNunzio; Mariano Aizpurua; Romina Zadoff; Carla Marchionatti; Natalia Garcia Escude; Romina Romero; Noelia Iraizos; Emmanuel Ezequiel Valls; Patricia Rearte Carvalho; Jimena Franco; Natali Estrada; Juan Rusconi; Guido Ochoa; Maria Veronica Paz; Patricia Lesch; Maria Fernanda Caracciolo; Maria Eugenia Macaneo; Lia Pocket; Silvana Marquez; Gaston Pellegrino; Jorge Geffner; Rocio Zarlenga; Camila Witteveen; Agustina Venditti; Indira Pichetto Olanda; Juan Mauricio Vargas; Micaela Piani; Daniela Carolina Galnarez; Florencia De la Fuente; Andrea Gamarnik; Maria del Carmen Nigro; Susana Villaroel; Cristina Soler Riera; Leonel Langellotti; Clarisa Taffarel; Jose L Scapellato; Mariano Girasolli; Maximiliano de Zan; Juan Sebastian Riera; Enio Garcia; Mario Rovere; Juan Canela; Agostina Pagella; Cecilia Pampuro; Yanina Miragaya; Silvina Kuperman; Alfonso Raggio; Ramiro Manuel Larrea; Maria Dolores Silveyra; Gabriela Leberzstein; Alejandra Debonis; Juan Molinos; Miguel Gonzalez; Eduardo Perez; Nicolas Kreplak; Susana Pastor Arguello; Luz Gibbons; Fernando Althabe; Eduardo Bergel; Fernando P Polack.
medrxiv; 2020.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2020.11.20.20234013

ABSTRACT

BackgroundTherapies to interrupt progression of early COVID-19 remain elusive. Among them, convalescent plasma in hospitalized patients was unsuccessful, perhaps because antibody should be administered earlier. We advanced plasma infusions to the first 72 hours of symptoms to arrest COVID-19 progression. MethodsA randomized, double-blind, placebo-controlled trial of convalescent plasma with high IgG titers against SARS-CoV2 in elderly subjects within 72 hours of mild COVID-19 symptoms. The primary endpoint was severe respiratory disease defined as a respiratory rate [≥]30 and/or an O2 sat<93% in room air. The study was interrupted at 76% of its projected sample size, because cases in the region decreased considerably and steady enrollment of study subjects became virtually impossible. Results160 patients underwent randomization. In the intention-to-treat analysis (ITT), 13/80(16.2%) patients receiving plasma vs. 25/80(31.2%) receiving placebo experienced severe respiratory disease [RR(95%CI)= 0.52(0.29,0.94); p=0.026)] with an RRR=48%. A modified ITT analysis, excluding six subjects who experienced the primary endpoint before infusion, showed a larger effect size [RR(95%CI) = 0.40(0.20, 0.81), p=0.007]. High- and low-titer donor analyses, based on a median IgG titer=1:3,200, evidenced a dose-dependent response with an RRR=73.3% for recipients of high-titer plasma (p=0.016) and a number needed to treat (NNT)=4.4. All secondary endpoints exhibited trends towards protection. No solicited adverse events were observed. ConclusionsEarly administration of high-titer convalescent plasma against SARS-CoV2 to mildly ill infected seniors reduced COVID-19 progression. This safe, inexpensive, outpatient intervention facilitates access to treatment from industrialized to LMIC, can decompress demands on hospitals, and may contribute to save lives. Funded by The Bill & Melinda Gates Foundation and The Fundacion INFANT Pandemic Fund. Registered in the Direccion de Sangre y Medicina Transfusional del Ministerio de Salud (PAEPCC19), Plataforma PRIISA (1421), and clinicaltrials.gov (NCT04479163). All authors have completed the ICMJE uniform disclosure form at www.icmje.org/coi_disclosure.pdf and declare: no support from any organization for the submitted work; RL, GPM, DW and FPP are investigators in a phase 3 SARS CoV2 trial from Pfizer; no other relationships or activities that could appear to have influenced the submitted work.


Subject(s)
COVID-19
4.
Rebecca L. Laws; Prabasaj Paul; Emily Mosites; Heather Scobie; Kristie E. N. Clarke; Rachel B. Slayton; Ignacio Esteban; Mauricio Tomas Caballero; Cristian J Wood; Mabel Berrueta; Anibal Rondan; Gabriela Lescano; Pablo Cruz; Ivonne Ritou; Valeria Fernandez Vina; Damian Alvarez Paggi; Sebastian Esperante; Adrian Ferretti; Gaston Ofman; Alvaro Ciganda; Rocio Rodriguez; Jorge Lantos; Ricardo Valentini; Nicolas Itcovici; Alejandra Hintze; Laura Oyarvide; Candela Etchegaray; Alejandra Neira; Ivonne Name; Julieta Alfonso; Rocio Lopez Castelo; Gisela Caruso; Sofia Rapelius; Fernando Alvez; Federico Cesar Etchenique; Federico Dimase; Dario Raul Alvarez; Sofia Sol Aranda; Clara Sanchez Yanotti; Julian DeLuca; Sofia Jarez Baglivo; Sofia Lujan Laudanno; Florencia Nowogrodzki; Florencia Izetta; Maria Teresa Paniguetti; Paula Fernandez Estrella; Maria Emilia Gutierrez Meyer; Viviana Dominguez; Marcela Balduzzi; Romina Militerno; Jimena Ochoa; Sebastian Perez Marc; Lucila DiNunzio; Mariano Aizpurua; Romina Zadoff; Carla Marchionatti; Natalia Garcia Escude; Romina Romero; Noelia Iraizos; Emmanuel Ezequiel Valls; Patricia Rearte Carvalho; Jimena Franco; Natali Estrada; Juan Rusconi; Guido Ochoa; Maria Veronica Paz; Patricia Lesch; Maria Fernanda Caracciolo; Maria Eugenia Macaneo; Lia Pocket; Silvana Marquez; Gaston Pellegrino; Jorge Geffner; Rocio Zarlenga; Camila Witteveen; Agustina Venditti; Indira Pichetto Olanda; Juan Mauricio Vargas; Micaela Piani; Daniela Carolina Galnarez; Florencia De la Fuente; Andrea Gamarnik; Maria del Carmen Nigro; Susana Villaroel; Cristina Soler Riera; Leonel Langellotti; Clarisa Taffarel; Jose L Scapellato; Mariano Girasolli; Maximiliano de Zan; Juan Sebastian Riera; Enio Garcia; Mario Rovere; Juan Canela; Agostina Pagella; Cecilia Pampuro; Yanina Miragaya; Silvina Kuperman; Alfonso Raggio; Ramiro Manuel Larrea; Maria Dolores Silveyra; Gabriela Leberzstein; Alejandra Debonis; Juan Molinos; Miguel Gonzalez; Eduardo Perez; Nicolas Kreplak; Susana Pastor Arguello; Luz Gibbons; Fernando Althabe; Eduardo Bergel; Fernando P Polack.
biorxiv; 2020.
Preprint in English | bioRxiv | ID: ppzbmed-10.1101.2020.11.20.391011

ABSTRACT

BackgroundCongregate settings are at risk for coronavirus disease 2019 (COVID-19) outbreaks. Diagnostic testing can be used as a tool in these settings to identify outbreaks and to control transmission. MethodsWe used transmission modeling to estimate the minimum number of persons to test and the optimal frequency to detect small outbreaks of COVID-19 in a congregate facility. We also estimated the frequency of testing needed to interrupt transmission within a facility. ResultsThe number of people to test and frequency of testing needed depended on turnaround time, facility size, and test characteristics. Parameters are calculated for a variety of scenarios. In a facility of 100 people, 26 randomly selected individuals would need to be tested at least every 6 days to identify a true underlying prevalence of at least 5%, with test sensitivity of 85%, and greater than 95% outbreak detection sensitivity. Disease transmission could be interrupted with universal, facility-wide testing with rapid turnaround every three days. ConclusionsTesting a subset of individuals in congregate settings can improve early detection of small outbreaks of COVID-19. Frequent universal diagnostic testing can be used to interrupt transmission within a facility, but its efficacy is reliant on rapid turnaround of results for isolation of infected individuals.


Subject(s)
COVID-19
5.
medrxiv; 2020.
Preprint in English | medRxiv | ID: ppzbmed-10.1101.2020.06.01.20116608

ABSTRACT

Covid-19 death has a different relationship with age than is the case for other severe respiratory pathogens. The Covid-19 death rate increases exponentially with age, and the main risk factors are age itself, as well as having underlying conditions such as hypertension, diabetes, cardiovascular disease, severe chronic respiratory disease and cancer. Furthermore, the almost complete lack of deaths in children suggests that infection alone is not sufficient to cause death; rather, one must have gone through a number of changes, either as a result of undefined aspects of aging, or as a result of chronic disease. These characteristics of Covid-19 death are consistent with the multistep model of disease, a model which has primarily been used for cancer, and more recently for amyotrophic lateral sclerosis (ALS). We applied the multi-step model to data on Covid-19 case fatality rates (CFRs) from China, South Korea, Italy, Spain and Japan. In all countries we found that a plot of ln (CFR) against ln (age) was approximately linear with a slope of about 5. As a comparison, we also conducted similar analyses for selected other respiratory diseases. SARS showed a similar log-log age-pattern to that of Covid-19, albeit with a lower slope, whereas seasonal and pandemic influenza showed quite different age-patterns. Thus, death from Covid-19 and SARS appears to follow a distinct age-pattern, consistent with a multistep model of disease that in the case of Covid-19 is probably defined by comorbidities and age producing immune-related susceptibility. Identification of these steps would be potentially important for prevention and therapy for SARS-COV-2 infection.


Subject(s)
Respiratory Tract Diseases , Cardiovascular Diseases , Diabetes Mellitus , Neoplasms , Chronic Disease , Hypertension , Death , COVID-19 , Amyotrophic Lateral Sclerosis
SELECTION OF CITATIONS
SEARCH DETAIL