Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
1.
Age Ageing ; 53(8)2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39141078

RESUMEN

BACKGROUND: Molnupiravir and nirmatrelvir-ritonavir are orally administered pharmacotherapies for mild to moderate COVID-19. However, the effectiveness of these drugs among very old (≥80 years), hospitalised patients remains unclear, limiting the risk-benefit assessment of these antivirals in this specific group. This study investigates the effectiveness of these antivirals in reducing mortality among this group of hospitalised patients with COVID-19. METHODS: Using a territory-wide public healthcare database in Hong Kong, a target trial emulation study was conducted with data from 13 642 eligible participants for the molnupiravir trial and 9553 for the nirmatrelvir-ritonavir trial. The primary outcome was all-cause mortality. Immortal time and confounding bias was minimised using cloning-censoring-weighting approach. Mortality odds ratios were estimated by pooled logistic regression after adjusting confounding biases by stabilised inverse probability weights. RESULTS: Both molnupiravir (HR: 0.895, 95% CI: 0.826-0.970) and nirmatrelvir-ritonavir (HR: 0.804, 95% CI: 0.678-0.955) demonstrated moderate mortality risk reduction among oldest-old hospitalised patients. No significant interaction was observed between oral antiviral treatment and vaccination status. The 28-day risk of mortality was lower in initiators than non-initiators for both molnupiravir (risk difference: -1.09%, 95% CI: -2.29, 0.11) and nirmatrelvir-ritonavir (risk difference: -1.71%, 95% CI: -3.30, -0.16) trials. The effectiveness of these medications was observed regardless of the patients' prior vaccination status. CONCLUSIONS: Molnupiravir and nirmatrelvir-ritonavir are moderately effective in reducing mortality risk among hospitalised oldest-old patients with COVID-19, regardless of their vaccination status.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Hospitalización , SARS-CoV-2 , Humanos , Antivirales/administración & dosificación , Antivirales/uso terapéutico , Masculino , Femenino , Anciano de 80 o más Años , Hospitalización/estadística & datos numéricos , Hong Kong/epidemiología , Administración Oral , Ritonavir/uso terapéutico , Ritonavir/administración & dosificación , COVID-19/mortalidad , COVID-19/epidemiología , Hidroxilaminas/administración & dosificación , Hidroxilaminas/uso terapéutico , Resultado del Tratamiento , Citidina/análogos & derivados , Citidina/administración & dosificación , Citidina/uso terapéutico , Leucina/análogos & derivados
2.
Expert Opin Investig Drugs ; 33(8): 867-876, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38988285

RESUMEN

BACKGROUND: Considering the rise of new SARS-CoV-2 variants that have reduced the efficacy of COVID-19 vaccines, the development of new antiviral medications for the disease has become increasingly necessary. In this study, ASC10, a novel antiviral prodrug, was studied in a phase 1 trial in healthy Chinese participants. RESEARCH DESIGN AND METHODS: Part 1 involved 60 participants, receiving 50-800 mg ASC10 or placebo twice daily for 5.5 days. Part 2, with 12 participants, explored ASC10 dosing in the fed/fasting states. RESULTS: ASC10-A, the main pharmacologically active metabolite, rapidly appeared in plasma (Tmax: 1.00-2.00 h) and decreased (t1/2: 1.10-3.04 h) without accumulation. The Cmax and area under the plasma concentration - time curve (AUC) of ASC10-A increased dose-dependently (50-800 mg BID) over 5.5 days, with no accumulation. The Tmax was slightly delayed in the fed state; however, the Cmax and AUC were similar between the fed and fasting states. Adverse events (AEs) were comparable (ASC10/placebo, 66.7%) and mostly mild (95%). CONCLUSION: ASC10 was demonstrated to be safe and well tolerated and exhibited dose-proportional exposure and minimal food effects. CLINICAL TRIAL REGISTRATION: www.clinicaltrials.gov identifier is NCT05523141.


Asunto(s)
Antivirales , Profármacos , Adulto , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Administración Oral , Antivirales/administración & dosificación , Antivirales/efectos adversos , Antivirales/farmacocinética , Área Bajo la Curva , Pueblo Asiatico , China , COVID-19 , Tratamiento Farmacológico de COVID-19 , Citidina/administración & dosificación , Citidina/efectos adversos , Citidina/análogos & derivados , Citidina/farmacocinética , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Voluntarios Sanos , Profármacos/efectos adversos , Profármacos/administración & dosificación
3.
Support Care Cancer ; 32(8): 496, 2024 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-38980433

RESUMEN

PURPOSE: Immunocompromised individuals, such as those diagnosed with cancer, are at a significantly higher risk for severe illness and mortality when infected with SARS-CoV-2 (COVID-19) than the general population. Two oral antiviral treatments are approved for COVID-19: Paxlovid® (nirmatrelvir/ritonavir) and Lagevrio® (molnupiravir). There is a paucity of data regarding the benefit from these antivirals among immunocompromised patients with cancer, and recent studies have questioned their efficacy among vaccinated patients, even those with risk factors for severe COVID-19. METHODS: We evaluated the efficacy and safety of nirmatrelvir/ritonavir and molnupiravir in preventing severe illness and death using our database of 457 patients with cancer and COVID-19 from Brown University-affiliated hospitals. RESULTS: Sixty-seven patients received nirmatrelvir/ritonavir or molnupiravir and were compared to 45 concurrent controls who received no antiviral treatment despite being eligible to receive it. Administration of nirmatrelvir/ritonavir or molnupiravir was associated with improved survival and lower 90-day all-cause and COVID-19-attributed mortality (p < 0.05) and with lower peak O2 requirements (ordinal odds ratio [OR] 1.52, 95% confidence interval [CI] 0.92-2.56). CONCLUSION: Acknowledging the small size of our sample as a limitation, we concluded that early antiviral treatment might be beneficial to immunocompromised individuals, particularly those with cancer, when infected with SARS-CoV-2. Larger-scale, well-stratified studies are needed in this patient population.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Neoplasias , Ritonavir , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/complicaciones , Masculino , Antivirales/uso terapéutico , Antivirales/administración & dosificación , Femenino , Persona de Mediana Edad , Anciano , Ritonavir/uso terapéutico , Ritonavir/administración & dosificación , Administración Oral , Citidina/análogos & derivados , Citidina/uso terapéutico , Citidina/administración & dosificación , Hidroxilaminas/uso terapéutico , Hidroxilaminas/administración & dosificación , COVID-19 , Adulto , Huésped Inmunocomprometido , Leucina/análogos & derivados , Leucina/uso terapéutico , Anciano de 80 o más Años , SARS-CoV-2 , Estudios Retrospectivos
4.
Int J Infect Dis ; 143: 107021, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38561040

RESUMEN

OBJECTIVES: Evaluate and compare the efficacy and safety of molnupiravir and favipiravir in outpatients with mild to moderate COVID-19 and at risk of severe COVID-19. METHODS: In an open-label, parallel-group, multicenter trial in Thailand, participants with moderate COVID-19 and at least one factor associated with severe COVID-19 were randomly assigned 1:1 to receive oral molnupiravir or oral favipiravir (standard of care). Phone calls for remote symptom assessment were made on Days 6, 15, and 29. Participants with worsening symptoms were instructed to return to the hospital. The primary endpoint was pulmonary involvement by Day 29, as evidenced by ≥2 of the following: dyspnea, oxygen saturation <92% or imaging. RESULTS: Nine hundred seventy-seven participants (487 molnupiravir, 490 favipiravir) were enrolled from 8 July 2022 to 19 January 2023. 98% had received ≥1 dose of COVID-19 vaccine and 83% ≥3 doses. By Day 29, pulmonary involvement occurred in 0% (0/483) in molnupiravir arm versus 1% (5/482) in favipiravir arm (-1.0%; Newcombe 95.2% CI: -2.4% to -0.0%; P = 0.021); all-cause death in 0% (0/483) and <1% (1/482); COVID-19 related hospitalization in <1% (1/483) and 1% (3/482); treatment-related adverse event in 1% (5/483) and 1% (4/486); and serious adverse event in 1% (4/483) and 1% (4/486). CONCLUSIONS: Favipiravir and molnupiravir had a similar efficacy and safety profile. Whether either of the two reduced the risk of complications during the omicron era in this population with a low risk of pulmonary involvement and a high vaccine coverage remains unclear. There were no differences in any of the safety endpoints. THAI CLINICAL TRIALS REGISTRY ID: TCTR20230111009.


Asunto(s)
Amidas , Antivirales , Tratamiento Farmacológico de COVID-19 , Citidina/análogos & derivados , Pirazinas , SARS-CoV-2 , Humanos , Amidas/uso terapéutico , Masculino , Pirazinas/uso terapéutico , Pirazinas/efectos adversos , Pirazinas/administración & dosificación , Femenino , Tailandia , Antivirales/uso terapéutico , Antivirales/efectos adversos , Antivirales/administración & dosificación , Persona de Mediana Edad , Adulto , Citidina/uso terapéutico , Citidina/efectos adversos , Citidina/administración & dosificación , Hidroxilaminas/uso terapéutico , Hidroxilaminas/efectos adversos , Hidroxilaminas/administración & dosificación , Anciano , Resultado del Tratamiento , COVID-19 , Pacientes Ambulatorios
5.
Nature ; 601(7894): 496, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-35064230

Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , COVID-19/virología , Desarrollo de Medicamentos/tendencias , Farmacorresistencia Viral , Investigadores , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Adenosina Monofosfato/uso terapéutico , Administración Oral , Alanina/administración & dosificación , Alanina/análogos & derivados , Alanina/farmacología , Alanina/uso terapéutico , Antivirales/administración & dosificación , Antivirales/farmacología , Antivirales/provisión & distribución , COVID-19/mortalidad , COVID-19/prevención & control , Vacunas contra la COVID-19/provisión & distribución , Citidina/administración & dosificación , Citidina/análogos & derivados , Citidina/farmacología , Citidina/uso terapéutico , Aprobación de Drogas , Combinación de Medicamentos , Farmacorresistencia Viral/efectos de los fármacos , Farmacorresistencia Viral/genética , Quimioterapia Combinada , Hospitalización/estadística & datos numéricos , Humanos , Hidroxilaminas/administración & dosificación , Hidroxilaminas/farmacología , Hidroxilaminas/uso terapéutico , Lactamas/administración & dosificación , Lactamas/farmacología , Lactamas/uso terapéutico , Leucina/administración & dosificación , Leucina/farmacología , Leucina/uso terapéutico , Cumplimiento de la Medicación , Terapia Molecular Dirigida , Mutagénesis , Nitrilos/administración & dosificación , Nitrilos/farmacología , Nitrilos/uso terapéutico , Prolina/administración & dosificación , Prolina/farmacología , Prolina/uso terapéutico , Asociación entre el Sector Público-Privado/economía , Ritonavir/administración & dosificación , Ritonavir/farmacología , Ritonavir/uso terapéutico , SARS-CoV-2/enzimología , SARS-CoV-2/genética
6.
NEJM Evid ; 1(2): EVIDoa2100043, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-38319179

RESUMEN

BACKGROUND: Safe and effective oral treatments are needed to improve clinical outcomes for nonhospitalized patients with Covid-19. Molnupiravir is an orally administered, small-molecule ribonucleoside prodrug shown to inhibit replication of severe acute respiratory syndrome coronavirus 2 in vitro and in animal models. METHODS: MOVe-OUT is an ongoing, phase 2/3, randomized, placebo-controlled, double-blind study evaluating the safety, efficacy, and pharmacokinetics of molnupiravir in nonhospitalized adults. In the phase 2 component, participants had mild or moderate, laboratory-confirmed Covid-19 with sign/symptom onset up to (and including) 7 days before randomization. Participants were randomly assigned 1:1:1:1 to receive 200, 400, or 800 mg of molnupiravir or placebo twice daily for 5 days, stratified by time since sign/symptom onset and by being at increased risk for severe illness from Covid-19. The primary efficacy end point was the proportion of participants who were hospitalized and/or died through day 29. RESULTS: The phase 2 component randomly assigned 302 participants to treatment; baseline characteristics were comparable across treatment groups. Molnupiravir had no apparent dose-related effect on adverse events, and no clinically meaningful abnormalities in laboratory test results were observed in relation to dose or treatment. Eleven participants were hospitalized or died through day 29. Of 225 participants in the combined molnupiravir group, 7 (3.1%) were hospitalized or died, compared with 4 of 74 participants (5.4%) in the placebo group. Subgroup analyses suggested lower incidences of hospitalization and/or death in the molnupiravir versus placebo groups in participants older than 60 years of age, those with increased risk for severe illness, those with symptom onset up to (and including) 5 days before randomization, and those with both symptom onset up to (and including) 5 days before randomization and increased risk for severe illness. CONCLUSIONS: These interim study results support further evaluation of molnupiravir as a potential treatment to reduce hospitalizations and/or death in nonhospitalized patients with Covid-19. (Funded by Merck Sharp & Dohme Corp., a subsidiary of Merck & Co., Inc.; ClinicalTrials.gov number, NCT04575597.)


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Citidina , Hidroxilaminas , Humanos , Masculino , Femenino , Persona de Mediana Edad , Método Doble Ciego , Hidroxilaminas/uso terapéutico , Hidroxilaminas/farmacocinética , Citidina/análogos & derivados , Citidina/uso terapéutico , Citidina/administración & dosificación , Antivirales/uso terapéutico , Antivirales/administración & dosificación , Antivirales/efectos adversos , Antivirales/farmacocinética , Adulto , Anciano , COVID-19 , Resultado del Tratamiento , Uridina/análogos & derivados , Uridina/uso terapéutico , Uridina/administración & dosificación , SARS-CoV-2/efectos de los fármacos
10.
Molecules ; 26(19)2021 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-34641339

RESUMEN

The COVID-19 pandemic needs no introduction at present. Only a few treatments are available for this disease, including remdesivir and favipiravir. Accordingly, the pharmaceutical industry is striving to develop new treatments for COVID-19. Molnupiravir, an orally active RdRp inhibitor, is in a phase 3 clinical trial against COVID-19. The objective of this review article is to enlighten the researchers working on COVID-19 about the discovery, recent developments, and patents related to molnupiravir. Molnupiravir was originally developed for the treatment of influenza at Emory University, USA. However, this drug has also demonstrated activity against a variety of viruses, including SARS-CoV-2. Now it is being jointly developed by Emory University, Ridgeback Biotherapeutics, and Merck to treat COVID-19. The published clinical data indicate a good safety profile, tolerability, and oral bioavailability of molnupiravir in humans. The patient-compliant oral dosage form of molnupiravir may hit the market in the first or second quarter of 2022. The patent data of molnupiravir revealed its granted compound patent and process-related patent applications. We also anticipate patent filing related to oral dosage forms, inhalers, and a combination of molnupiravir with marketed drugs like remdesivir, favipiravir, and baricitinib. The current pandemic demands a patient compliant, safe, tolerable, and orally effective COVID-19 treatment. The authors believe that molnupiravir meets these requirements and is a breakthrough COVID-19 treatment.


Asunto(s)
Antivirales/uso terapéutico , Tratamiento Farmacológico de COVID-19 , Citidina/análogos & derivados , Descubrimiento de Drogas , Hidroxilaminas/uso terapéutico , SARS-CoV-2/efectos de los fármacos , Administración Oral , Animales , Antivirales/administración & dosificación , Antivirales/química , Ensayos Clínicos como Asunto , Citidina/administración & dosificación , Citidina/química , Citidina/uso terapéutico , Humanos , Hidroxilaminas/administración & dosificación , Hidroxilaminas/química , Patentes como Asunto , ADN Polimerasa Dirigida por ARN/metabolismo , Inhibidores de la Transcriptasa Inversa/administración & dosificación , Inhibidores de la Transcriptasa Inversa/química , Inhibidores de la Transcriptasa Inversa/uso terapéutico , SARS-CoV-2/enzimología , Proteínas Virales/antagonistas & inhibidores , Proteínas Virales/metabolismo
11.
Mol Pharmacol ; 100(6): 548-557, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34503974

RESUMEN

Equilibrative nucleoside transporters (ENTs) are present at the blood-testis barrier (BTB), where they can facilitate antiviral drug disposition to eliminate a sanctuary site for viruses detectable in semen. The purpose of this study was to investigate ENT-drug interactions with three nucleoside analogs, remdesivir, molnupiravir, and molnupiravir's active metabolite, ß-d-N4-hydroxycytidine (EIDD-1931), and four non-nucleoside molecules repurposed as antivirals for coronavirus disease 2019 (COVID-19). The study used three-dimensional pharmacophores for ENT1 and ENT2 substrates and inhibitors and Bayesian machine learning models to identify potential interactions with these transporters. In vitro transport experiments demonstrated that remdesivir was the most potent inhibitor of ENT-mediated [3H]uridine uptake (ENT1 IC50: 39 µM; ENT2 IC50: 77 µM), followed by EIDD-1931 (ENT1 IC50: 259 µM; ENT2 IC50: 467 µM), whereas molnupiravir was a modest inhibitor (ENT1 IC50: 701 µM; ENT2 IC50: 851 µM). Other proposed antivirals failed to inhibit ENT-mediated [3H]uridine uptake below 1 mM. Remdesivir accumulation decreased in the presence of 6-S-[(4-nitrophenyl)methyl]-6-thioinosine (NBMPR) by 30% in ENT1 cells (P = 0.0248) and 27% in ENT2 cells (P = 0.0054). EIDD-1931 accumulation decreased in the presence of NBMPR by 77% in ENT1 cells (P = 0.0463) and by 64% in ENT2 cells (P = 0.0132), which supported computational predictions that both are ENT substrates that may be important for efficacy against COVID-19. NBMPR failed to decrease molnupiravir uptake, suggesting that ENT interaction is likely inhibitory. Our combined computational and in vitro data can be used to identify additional ENT-drug interactions to improve our understanding of drugs that can circumvent the BTB. SIGNIFICANCE STATEMENT: This study identified remdesivir and EIDD-1931 as substrates of equilibrative nucleoside transporters 1 and 2. This provides a potential mechanism for uptake of these drugs into cells and may be important for antiviral potential in the testes and other tissues expressing these transporters.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/metabolismo , Citidina/análogos & derivados , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Transportador Equilibrativo 2 de Nucleósido/metabolismo , SARS-CoV-2/metabolismo , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/metabolismo , Alanina/administración & dosificación , Alanina/metabolismo , Antivirales/administración & dosificación , COVID-19/metabolismo , Citidina/administración & dosificación , Citidina/metabolismo , Relación Dosis-Respuesta a Droga , Interacciones Farmacológicas/fisiología , Células HeLa , Humanos , Unión Proteica/efectos de los fármacos , Unión Proteica/fisiología , SARS-CoV-2/efectos de los fármacos , Tratamiento Farmacológico de COVID-19
12.
EBioMedicine ; 68: 103390, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-34127431

RESUMEN

BACKGROUND: Coronavirus Disease 2019 (Covid-19) continues to challenge the limits of our knowledge and our healthcare system. Here we sought to define the host immune response, a.k.a, the "cytokine storm" that has been implicated in fatal COVID-19 using an AI-based approach. METHOD: Over 45,000 transcriptomic datasets of viral pandemics were analyzed to extract a 166-gene signature using ACE2 as a 'seed' gene; ACE2 was rationalized because it encodes the receptor that facilitates the entry of SARS-CoV-2 (the virus that causes COVID-19) into host cells. An AI-based approach was used to explore the utility of the signature in navigating the uncharted territory of Covid-19, setting therapeutic goals, and finding therapeutic solutions. FINDINGS: The 166-gene signature was surprisingly conserved across all viral pandemics, including COVID-19, and a subset of 20-genes classified disease severity, inspiring the nomenclatures ViP and severe-ViP signatures, respectively. The ViP signatures pinpointed a paradoxical phenomenon wherein lung epithelial and myeloid cells mount an IL15 cytokine storm, and epithelial and NK cell senescence and apoptosis determine severity/fatality. Precise therapeutic goals could be formulated; these goals were met in high-dose SARS-CoV-2-challenged hamsters using either neutralizing antibodies that abrogate SARS-CoV-2•ACE2 engagement or a directly acting antiviral agent, EIDD-2801. IL15/IL15RA were elevated in the lungs of patients with fatal disease, and plasma levels of the cytokine prognosticated disease severity. INTERPRETATION: The ViP signatures provide a quantitative and qualitative framework for titrating the immune response in viral pandemics and may serve as a powerful unbiased tool to rapidly assess disease severity and vet candidate drugs. FUNDING: This work was supported by the National Institutes for Health (NIH) [grants CA151673 and GM138385 (to DS) and AI141630 (to P.G), DK107585-05S1 (SD) and AI155696 (to P.G, D.S and S.D), U19-AI142742 (to S. C, CCHI: Cooperative Centers for Human Immunology)]; Research Grants Program Office (RGPO) from the University of California Office of the President (UCOP) (R00RG2628 & R00RG2642 to P.G, D.S and S.D); the UC San Diego Sanford Stem Cell Clinical Center (to P.G, D.S and S.D); LJI Institutional Funds (to S.C); the VA San Diego Healthcare System Institutional funds (to L.C.A). GDK was supported through The American Association of Immunologists Intersect Fellowship Program for Computational Scientists and Immunologists. ONE SENTENCE SUMMARY: The host immune response in COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/genética , Antivirales/administración & dosificación , COVID-19/genética , Perfilación de la Expresión Génica/métodos , Interleucina-15/genética , Receptores de Interleucina-15/genética , Virosis/genética , Animales , Anticuerpos Neutralizantes/administración & dosificación , Anticuerpos Neutralizantes/farmacología , Antivirales/farmacología , Inteligencia Artificial , Autopsia , COVID-19/inmunología , Cricetinae , Citidina/administración & dosificación , Citidina/análogos & derivados , Citidina/farmacología , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Redes Reguladoras de Genes/efectos de los fármacos , Marcadores Genéticos/efectos de los fármacos , Humanos , Hidroxilaminas/administración & dosificación , Hidroxilaminas/farmacología , Interleucina-15/sangre , Pulmón/inmunología , Mesocricetus , Pandemias , Receptores de Interleucina-15/sangre , Virosis/inmunología , Tratamiento Farmacológico de COVID-19
13.
Nat Commun ; 12(1): 3309, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34083527

RESUMEN

The ongoing pandemic caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), necessitates strategies to identify prophylactic and therapeutic drug candidates for rapid clinical deployment. Here, we describe a screening pipeline for the discovery of efficacious SARS-CoV-2 inhibitors. We screen a best-in-class drug repurposing library, ReFRAME, against two high-throughput, high-content imaging infection assays: one using HeLa cells expressing SARS-CoV-2 receptor ACE2 and the other using lung epithelial Calu-3 cells. From nearly 12,000 compounds, we identify 49 (in HeLa-ACE2) and 41 (in Calu-3) compounds capable of selectively inhibiting SARS-CoV-2 replication. Notably, most screen hits are cell-line specific, likely due to different virus entry mechanisms or host cell-specific sensitivities to modulators. Among these promising hits, the antivirals nelfinavir and the parent of prodrug MK-4482 possess desirable in vitro activity, pharmacokinetic and human safety profiles, and both reduce SARS-CoV-2 replication in an orthogonal human differentiated primary cell model. Furthermore, MK-4482 effectively blocks SARS-CoV-2 infection in a hamster model. Overall, we identify direct-acting antivirals as the most promising compounds for drug repurposing, additional compounds that may have value in combination therapies, and tool compounds for identification of viral host cell targets.


Asunto(s)
Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Reposicionamiento de Medicamentos/métodos , Pandemias , SARS-CoV-2 , Animales , COVID-19/prevención & control , COVID-19/virología , Línea Celular , Citidina/administración & dosificación , Citidina/análogos & derivados , Citidina/farmacología , Bases de Datos Farmacéuticas , Descubrimiento de Drogas/métodos , Evaluación Preclínica de Medicamentos/métodos , Células HeLa , Ensayos Analíticos de Alto Rendimiento/métodos , Humanos , Hidroxilaminas/administración & dosificación , Hidroxilaminas/farmacología , Mesocricetus , Nelfinavir/farmacología , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Replicación Viral/efectos de los fármacos
14.
Nature ; 592(7854): 340-343, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33854246
15.
Philos Trans R Soc Lond B Biol Sci ; 376(1826): 20200125, 2021 06 07.
Artículo en Inglés | MEDLINE | ID: mdl-33866803

RESUMEN

In response to novel environments, invasive populations often evolve rapidly. Standing genetic variation is an important predictor of evolutionary response but epigenetic variation may also play a role. Here, we use an iconic invader, the cane toad (Rhinella marina), to investigate how manipulating epigenetic status affects phenotypic traits. We collected wild toads from across Australia, bred them, and experimentally manipulated DNA methylation of the subsequent two generations (G1, G2) through exposure to the DNA methylation inhibitor zebularine and/or conspecific tadpole alarm cues. Direct exposure to alarm cues (an indicator of predation risk) increased the potency of G2 tadpole chemical cues, but this was accompanied by reductions in survival. Exposure to alarm cues during G1 also increased the potency of G2 tadpole cues, indicating intergenerational plasticity in this inducible defence. In addition, the negative effects of alarm cues on tadpole viability (i.e. the costs of producing the inducible defence) were minimized in the second generation. Exposure to zebularine during G1 induced similar intergenerational effects, suggesting a role for alteration in DNA methylation. Accordingly, we identified intergenerational shifts in DNA methylation at some loci in response to alarm cue exposure. Substantial demethylation occurred within the sodium channel epithelial 1 subunit gamma gene (SCNN1G) in alarm cue exposed individuals and their offspring. This gene is a key to the regulation of sodium in epithelial cells and may help to maintain the protective epidermal barrier. These data suggest that early life experiences of tadpoles induce intergenerational effects through epigenetic mechanisms, which enhance larval fitness. This article is part of the theme issue 'How does epigenetics influence the course of evolution?'


Asunto(s)
Bufo marinus/fisiología , Metilación de ADN , Epigénesis Genética , Rasgos de la Historia de Vida , Animales , Australia , Bufo marinus/genética , Bufo marinus/crecimiento & desarrollo , Señales (Psicología) , Citidina/administración & dosificación , Citidina/análogos & derivados , Especies Introducidas , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Larva/fisiología
16.
Nat Commun ; 12(1): 2295, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863887

RESUMEN

The COVID-19 pandemic progresses unabated in many regions of the world. An effective antiviral against SARS-CoV-2 that could be administered orally for use following high-risk exposure would be of substantial benefit in controlling the COVID-19 pandemic. Herein, we show that MK-4482, an orally administered nucleoside analog, inhibits SARS-CoV-2 replication in the Syrian hamster model. The inhibitory effect of MK-4482 on SARS-CoV-2 replication is observed in animals when the drug is administered either beginning 12 h before or 12 h following infection in a high-risk exposure model. These data support the potential utility of MK-4482 to control SARS-CoV-2 infection in humans following high-risk exposure as well as for treatment of COVID-19 patients.


Asunto(s)
Antivirales/administración & dosificación , Tratamiento Farmacológico de COVID-19 , Citidina/análogos & derivados , Hidroxilaminas/administración & dosificación , SARS-CoV-2/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Administración Oral , Animales , COVID-19/virología , Chlorocebus aethiops , Citidina/administración & dosificación , Modelos Animales de Enfermedad , Humanos , Mesocricetus , SARS-CoV-2/aislamiento & purificación , SARS-CoV-2/fisiología , Células Vero
17.
Mol Ther ; 29(5): 1758-1771, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33571681

RESUMEN

DNA methylation abnormality is closely related to tumor occurrence and development. Chemical inhibitors targeting DNA methyltransferase (DNMTis) have been used in treating cancer. However, the impact of DNMTis on antitumor immunity has not been well elucidated. In this study, we show that zebularine (a demethylating agent) treatment of cancer cells led to increased levels of interferon response in a cyclic guanosine monophosphate-AMP (cGAMP) synthase (cGAS)- and stimulator of interferon genes (STING)-dependent manner. This treatment also specifically sensitized the cGAS-STING pathway in response to DNA stimulation. Incorporation of zebularine into genomic DNA caused demethylation and elevated expression of a group of genes, including STING. Without causing DNA damage, zebularine led to accumulation of DNA species in the cytoplasm of treated cells. In syngeneic tumor models, administration of zebularine alone reduced tumor burden and extended mice survival. This effect synergized with cGAMP and immune checkpoint blockade therapy. The efficacy of zebularine was abolished in nude mice and in cGAS-/- or STING-/- mice, indicating its dependency on host immunity. Analysis of tumor cells indicates upregulation of interferon-stimulated genes (ISGs) following zebularine administration. Zebularine promoted infiltration of CD8 T cells and natural killer (NK) cells into tumor and therefore suppressed tumor growth. This study unveils the role of zebularine in sensitizing the cGAS-STING pathway to promote anti-tumor immunity and provides the foundation for further therapeutic development.


Asunto(s)
Citidina/análogos & derivados , Melanoma Experimental/tratamiento farmacológico , Proteínas de la Membrana/genética , Nucleótidos Cíclicos/administración & dosificación , Nucleotidiltransferasas/genética , Administración Oral , Animales , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Citidina/administración & dosificación , Citidina/farmacología , Sinergismo Farmacológico , Humanos , Células Asesinas Naturales/metabolismo , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Ratones Desnudos , Nucleótidos Cíclicos/farmacología , Regiones Promotoras Genéticas , Células THP-1 , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nature ; 591(7850): 451-457, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33561864

RESUMEN

All coronaviruses known to have recently emerged as human pathogens probably originated in bats1. Here we use a single experimental platform based on immunodeficient mice implanted with human lung tissue (hereafter, human lung-only mice (LoM)) to demonstrate the efficient in vivo replication of severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), as well as two endogenous SARS-like bat coronaviruses that show potential for emergence as human pathogens. Virus replication in this model occurs in bona fide human lung tissue and does not require any type of adaptation of the virus or the host. Our results indicate that bats contain endogenous coronaviruses that are capable of direct transmission to humans. Our detailed analysis of in vivo infection with SARS-CoV-2 in human lung tissue from LoM showed a predominant infection of human lung epithelial cells, including type-2 pneumocytes that are present in alveoli and ciliated airway cells. Acute infection with SARS-CoV-2 was highly cytopathic and induced a robust and sustained type-I interferon and inflammatory cytokine and chemokine response. Finally, we evaluated a therapeutic and pre-exposure prophylaxis strategy for SARS-CoV-2 infection. Our results show that therapeutic and prophylactic administration of EIDD-2801-an oral broad-spectrum antiviral agent that is currently in phase II/III clinical trials-markedly inhibited SARS-CoV-2 replication in vivo, and thus has considerable potential for the prevention and treatment of COVID-19.


Asunto(s)
Tratamiento Farmacológico de COVID-19 , COVID-19/prevención & control , Citidina/análogos & derivados , Hidroxilaminas/administración & dosificación , Hidroxilaminas/uso terapéutico , Administración Oral , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/patología , Células Epiteliales Alveolares/virología , Animales , COVID-19/inmunología , Quimioprevención , Quirópteros/virología , Ensayos Clínicos Fase II como Asunto , Ensayos Clínicos Fase III como Asunto , Citidina/administración & dosificación , Citidina/uso terapéutico , Citocinas/inmunología , Células Epiteliales/virología , Femenino , Xenoinjertos , Humanos , Inmunidad Innata , Interferón Tipo I/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Trasplante de Pulmón , Masculino , Ratones , Profilaxis Posexposición , Profilaxis Pre-Exposición , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Replicación Viral
19.
Sci Transl Med ; 12(541)2020 04 29.
Artículo en Inglés | MEDLINE | ID: mdl-32253226

RESUMEN

Coronaviruses (CoVs) traffic frequently between species resulting in novel disease outbreaks, most recently exemplified by the newly emerged SARS-CoV-2, the causative agent of COVID-19. Here, we show that the ribonucleoside analog ß-d-N4-hydroxycytidine (NHC; EIDD-1931) has broad-spectrum antiviral activity against SARS-CoV-2, MERS-CoV, SARS-CoV, and related zoonotic group 2b or 2c bat-CoVs, as well as increased potency against a CoV bearing resistance mutations to the nucleoside analog inhibitor remdesivir. In mice infected with SARS-CoV or MERS-CoV, both prophylactic and therapeutic administration of EIDD-2801, an orally bioavailable NHC prodrug (ß-d-N4-hydroxycytidine-5'-isopropyl ester), improved pulmonary function and reduced virus titer and body weight loss. Decreased MERS-CoV yields in vitro and in vivo were associated with increased transition mutation frequency in viral, but not host cell RNA, supporting a mechanism of lethal mutagenesis in CoV. The potency of NHC/EIDD-2801 against multiple CoVs and oral bioavailability highlights its potential utility as an effective antiviral against SARS-CoV-2 and other future zoonotic CoVs.


Asunto(s)
Antivirales/administración & dosificación , Infecciones por Coronavirus/tratamiento farmacológico , Neumonía Viral/tratamiento farmacológico , Ribonucleósidos/administración & dosificación , Replicación Viral/efectos de los fármacos , Adenosina Monofosfato/administración & dosificación , Adenosina Monofosfato/análogos & derivados , Alanina/administración & dosificación , Alanina/análogos & derivados , Animales , Profilaxis Antibiótica , Betacoronavirus/fisiología , COVID-19 , Línea Celular , Infecciones por Coronavirus/patología , Citidina/administración & dosificación , Citidina/análogos & derivados , Modelos Animales de Enfermedad , Farmacorresistencia Viral , Humanos , Hidroxilaminas , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Modelos Moleculares , Mutación/efectos de los fármacos , Pandemias , Neumonía Viral/patología , Cultivo Primario de Células , ARN Viral , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Distribución Aleatoria , Sistema Respiratorio/citología , SARS-CoV-2
20.
Endocrinology ; 161(1)2020 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-31742329

RESUMEN

Many neural sex differences are differences in the number of neurons of a particular phenotype. For example, male rodents have more calbindin-expressing neurons in the medial preoptic area (mPOA) and bed nucleus of the stria terminalis (BNST), and females have more neurons expressing estrogen receptor alpha (ERα) and kisspeptin in the ventromedial nucleus of the hypothalamus (VMH) and the anteroventral periventricular nucleus (AVPV), respectively. These sex differences depend on neonatal exposure to testosterone, but the underlying molecular mechanisms are unknown. DNA methylation is important for cell phenotype differentiation throughout the developing organism. We hypothesized that testosterone causes sex differences in neurochemical phenotype via changes in DNA methylation, and tested this by inhibiting DNA methylation neonatally in male and female mice, and in females given a masculinizing dose of testosterone. Neonatal testosterone treatment masculinized calbindin, ERα and kisspeptin cell number of females at weaning. Inhibiting DNA methylation with zebularine increased calbindin cell number only in control females, thus eliminating sex differences in calbindin in the mPOA and BNST. Zebularine also reduced the sex difference in ERα cell number in the VMH, in this case by increasing ERα neuron number in males and testosterone-treated females. In contrast, the neonatal inhibition of DNA methylation had no effect on kisspeptin cell number. We conclude that testosterone normally increases the number of calbindin cells and reduces ERα cells in males through orchestrated changes in DNA methylation, contributing to, or causing, the sex differences in both cell types.


Asunto(s)
Encéfalo/efectos de los fármacos , Metilación de ADN/efectos de los fármacos , Diferenciación Sexual/efectos de los fármacos , Testosterona/farmacología , Animales , Animales Recién Nacidos , Encéfalo/citología , Encéfalo/metabolismo , Química Encefálica/efectos de los fármacos , Calbindinas/metabolismo , Citidina/administración & dosificación , Citidina/análogos & derivados , Citidina/farmacología , Receptor alfa de Estrógeno/metabolismo , Femenino , Kisspeptinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Diferenciación Sexual/fisiología , Factores Sexuales , Testosterona/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...