Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
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.
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
3.
Clin Cancer Res ; 30(15): 3167-3178, 2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-38836759

RESUMEN

PURPOSE: Patients with glioblastoma (GBM) have a dismal prognosis. Although the DNA alkylating agent temozolomide (TMZ) is the mainstay of chemotherapy, therapeutic resistance rapidly develops in patients. Base excision repair inhibitor TRC102 (methoxyamine) reverses TMZ resistance in preclinical glioma models. We aimed to investigate the efficacy and safety of oral TRC102+TMZ in recurrent GBM (rGBM). PATIENTS AND METHODS: A preregistered (NCT02395692), nonrandomized, multicenter, phase 2 clinical trial (BERT) was planned and conducted through the Adult Brain Tumor Consortium (ABTC-1402). Arm 1 included patients with bevacizumab-naïve GBM at the first recurrence, with the primary endpoint of response rates. If sufficient activity was identified, a second arm was planned for the bevacizumab-refractory patients. The secondary endpoints were overall survival (OS), progression-free survival (PFS), PFS at 6 months (PFS6), and toxicity. RESULTS: Arm 1 enrolled 19 patients with a median of two treatment cycles. Objective responses were not observed; hence, arm 2 did not open. The median OS was 11.1 months [95% confidence interval (CI), 8.2-17.9]. The median PFS was 1.9 months (95% CI, 1.8-3.7). The PFS6 was 10.5% (95% CI, 1.3%-33.1%). Most toxicities were grades 1 and 2, with two grade 3 lymphopenias and one grade 4 thrombocytopenia. Two patients with PFS ≥ 17 months and OS > 32 months were deemed "extended survivors." RNA sequencing of tumor tissue, obtained at diagnosis, demonstrated significantly enriched signatures of DNA damage response (DDR), chromosomal instability (CIN70, CIN25), and cellular proliferation (PCNA25) in "extended survivors." CONCLUSIONS: These findings confirm the safety and feasibility of TRC102+TMZ in patients with rGBM. They also warrant further evaluation of combination therapy in biomarker-enriched trials enrolling GBM patients with baseline hyperactivated DDR pathways.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Recurrencia Local de Neoplasia , Temozolomida , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Reparación por Escisión/efectos de los fármacos , Glioblastoma/tratamiento farmacológico , Glioblastoma/patología , Glioblastoma/genética , Glioblastoma/mortalidad , Hidroxilaminas/uso terapéutico , Hidroxilaminas/administración & dosificación , Recurrencia Local de Neoplasia/tratamiento farmacológico , Recurrencia Local de Neoplasia/patología , Pronóstico , Temozolomida/uso terapéutico , Temozolomida/administración & dosificación
4.
J Infect ; 89(2): 106190, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38834107

RESUMEN

OBJECTIVES: This study aimed to determine the association of early use of oral antiviral drugs (including nirmatrelvir-ritonavir and molnupiravir) with the risk of post COVID-19 condition (PCC) and compare the possible efficacy of nirmatrelvir-ritonavir and molnupiravir. METHODS: PubMed, Web of Science, Embase, Cochrane, MedRxiv, and Psycinfo were searched from inception until November 1, 2023. We included studies that assessed the effect of oral antiviral drugs on the incidence of PCC. Pairwise and network meta-analyses were conducted using a random-effects model. Risk ratios (RRs) for oral antiviral drugs were calculated with a confidence interval (CI). RESULTS: Nine observational studies containing 866,066 patients were included. Nirmatrelvir-ritonavir and molnupiravir were evaluated in eight and two studies respectively, with both drugs evaluated in one study. Pair-wise meta-analysis showed that early oral antiviral drugs reduced PCC risk (RR 0.77, 95% CI 0.68-0.88). Network meta-analysis showed that nirmatrelvir-ritonavir may perform better than molnupiravir (surface under the cumulative ranking curve: 95.5% vs. 31.6%) at reducing PCC risk. CONCLUSIONS: Early use of oral antiviral drugs may potentially protect against developing PCC in non-hospitalized patients with COVID-19. These findings support the standardized administration of oral antiviral drugs in patients during the acute phase of COVID-19 according to the guidelines.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , COVID-19 , Metaanálisis en Red , Ritonavir , SARS-CoV-2 , Humanos , Antivirales/uso terapéutico , Antivirales/administración & dosificación , Ritonavir/uso terapéutico , Ritonavir/administración & dosificación , Administración Oral , COVID-19/epidemiología , Combinación de Medicamentos , Hidroxilaminas/uso terapéutico , Hidroxilaminas/administración & dosificación , Síndrome Post Agudo de COVID-19 , Lactamas , Citidina/análogos & derivados , Nitrilos , Prolina , Leucina
5.
J Infect Public Health ; 17(8): 102465, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38878678

RESUMEN

BACKGROUNDS: The effectiveness of oral antiviral therapy including nirmatrelvir plus ritonavir and molnupiravir in managing COVID-19 among individuals with pre-existing lung cancer was unclear. Therefore, this study was conducted to evaluate the usefulness of antiviral agents in the management of COVID-19 among patients with lung cancer. METHODS: Utilizing data from the TriNetX - a global health research network, a retrospective cohort study was conducted involving 2484 patients diagnosed with both lung cancer and COVID-19. Propensity score matching (PSM) was employed to create well-balanced cohorts. The study assessed the primary outcome of all-cause hospitalization or mortality within a 30-day follow-up. RESULTS: After PSM, the oral antiviral group exhibited a significantly lower risk of the primary composite outcome compared to the control group (6.1 % vs. 9.9 %; HR: 0.60; 95 % CI: 0.45-0.80). This association was consistent across various subgroups according to age, sex, vaccine status, type of oral antiviral agent, and lung cancer characteristics. Additionally, the oral antiviral group showed a lower risk of all-cause hospitalization (HR: 0.73; 95 % CI: 0.54-0.99) and a significantly lower risk of mortality (HR: 0.16; 95 % CI: 0.06-0.41). CONCLUSION: The study suggests a favorable impact of oral antiviral therapy on the outcomes of COVID-19 in individuals with lung cancer and support the potential utility of oral antiviral agents in improving outcomes in this vulnerable population.


Asunto(s)
Antivirales , Tratamiento Farmacológico de COVID-19 , Neoplasias Pulmonares , Ritonavir , SARS-CoV-2 , Humanos , Masculino , Femenino , Antivirales/uso terapéutico , Antivirales/administración & dosificación , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/mortalidad , Estudios Retrospectivos , Persona de Mediana Edad , Anciano , Ritonavir/uso terapéutico , Ritonavir/administración & dosificación , Administración Oral , Hospitalización/estadística & datos numéricos , COVID-19/mortalidad , Hidroxilaminas/uso terapéutico , Hidroxilaminas/administración & dosificación , Resultado del Tratamiento , Combinación de Medicamentos , Citidina/análogos & derivados
6.
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
7.
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
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
12.
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
13.
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
14.
Nature ; 592(7854): 340-343, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33854246
15.
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
16.
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
17.
J Cereb Blood Flow Metab ; 41(5): 958-974, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32703113

RESUMEN

Inhibition of histone deacetylases (HDACs) has been shown to reduce inflammation and white matter damage after various forms of brain injury via modulation of microglia/macrophage polarization. Previously we showed that the HDAC inhibitor scriptaid could attenuate white matter injury (WMI) after ICH. To access whether modulation of microglia/macrophage polarization might underlie this protection, we investigated the modulatory role of HDAC2 in microglia/macrophage polarization in response to WMI induced by intracerebral hemorrhage (ICH) and in primary microglia and oligodendrocyte co-cultures. HDAC2 activity was inhibited via conditional knockout of the Hdac2 gene in microglia or via administration of scriptaid. Conditional knockout of the Hdac2 gene in microglia and HDAC inhibition with scriptaid both improved neurological functional recovery and reduced WMI after ICH. Additionally, HDAC inhibition shifted microglia/macrophage polarization toward the M2 phenotype and reduced proinflammatory cytokine secretion after ICH in vivo. In vitro, a transwell co-culture model of microglia and oligodendrocytes also demonstrated that the HDAC inhibitor protected oligodendrocytes by modulating microglia polarization and mitigating neuroinflammation. Moreover, we found that scriptaid decreased the expression of pJAK2 and pSTAT1 in cultured microglia when stimulated with hemoglobin. Thus, HDAC inhibition ameliorated ICH-mediated neuroinflammation and WMI by modulating microglia/macrophage polarization.


Asunto(s)
Hemorragia Cerebral/complicaciones , Inhibidores de Histona Desacetilasas/farmacología , Hidroxilaminas/farmacología , Quinolinas/farmacología , Sustancia Blanca/lesiones , Animales , Factores de Coagulación Sanguínea/efectos de los fármacos , Factores de Coagulación Sanguínea/metabolismo , Estudios de Casos y Controles , Técnicas de Cocultivo , Citocinas/efectos de los fármacos , Modelos Animales de Enfermedad , Histona Desacetilasa 2/efectos de los fármacos , Histona Desacetilasa 2/metabolismo , Inhibidores de Histona Desacetilasas/uso terapéutico , Hidroxilaminas/administración & dosificación , Hidroxilaminas/uso terapéutico , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Evaluación de Resultado en la Atención de Salud , Quinolinas/administración & dosificación , Quinolinas/uso terapéutico , Recuperación de la Función/fisiología , Sustancia Blanca/efectos de los fármacos , Sustancia Blanca/metabolismo , Sustancia Blanca/ultraestructura
18.
Invest New Drugs ; 39(1): 142-151, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32556884

RESUMEN

Temozolomide (TMZ) generates DNA adducts that are repaired by direct DNA and base excision repair mechanisms. Methoxyamine (MX, TRC-102) potentiates TMZ activity by binding to apurinic and apyrimidinic (AP) sites after removal of N3-methyladenine and N7-methylguanine, inhibiting site recognition of AP endonuclease. We conducted a phase I trial to determine the maximum tolerated dose and dose-limiting toxicities (DLTs) of intravenous MX when given with oral TMZ. Patients with advanced solid tumors and progression on standard treatment were enrolled to a standard 3 + 3 dose escalation trial assessing escalating doses of TMZ and MX. Tumor response was assessed per RECIST and adverse events (AEs) by CTCAEv3. Pharmacokinetics (PK) of MX and COMET assays on peripheral blood mononuclear cells were performed. 38 patients were enrolled-median age 59.5 years (38-76), mean number of cycles 2.9 [1-13]. No DLTs were observed. Cycle 1 grade 3 AEs included fatigue, lymphopenia, anemia, INR, leukopenia, neutropenia, allergic reaction, constipation, psychosis and paranoia. Cycle 2-13 grade 4 AEs included thrombocytopenia and confusion. A partial response was seen in 1 patient with a pancreatic neuroendocrine tumor (PNET) and six additional patients, each with different tumor types, demonstrated prolonged stable disease. MX PK was linear with dose and was not affected by concomitant TMZ. TMZ 200 mg/m2 daily × 5 may be safely administered with MX 150 mg/m2 intravenously once on day 1 with minimal toxicity. Further studies assessing this drug combination in select tumor types where temozolomide has activity may be warranted.


Asunto(s)
Antineoplásicos Alquilantes/uso terapéutico , Hidroxilaminas/uso terapéutico , Neoplasias/tratamiento farmacológico , Temozolomida/uso terapéutico , Adulto , Anciano , Antineoplásicos Alquilantes/administración & dosificación , Antineoplásicos Alquilantes/efectos adversos , Antineoplásicos Alquilantes/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Área Bajo la Curva , Reparación del ADN/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Femenino , Semivida , Humanos , Hidroxilaminas/administración & dosificación , Hidroxilaminas/efectos adversos , Hidroxilaminas/farmacocinética , Masculino , Dosis Máxima Tolerada , Tasa de Depuración Metabólica , Persona de Mediana Edad , Temozolomida/efectos adversos , Temozolomida/farmacocinética
19.
Br J Cancer ; 118(10): 1359-1368, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29695769

RESUMEN

BACKGROUND: Cancer-associated fibroblasts (CAFs) support tumour progression and invasion, and they secrete abundant extracellular matrix (ECM) that may shield tumour cells from immune checkpoint or kinase inhibitors. Targeting CAFs using drugs that revert their differentiation, or inhibit their tumour-supportive functions, has been considered as an anti-cancer strategy. METHODS: We have used human and murine cell culture models, atomic force microscopy (AFM), microarray analyses, CAF/tumour cell spheroid co-cultures and transgenic fibroblast reporter mice to study how targeting HDACs using small molecule inhibitors or siRNAs re-directs CAF differentiation and function in vitro and in vivo. RESULTS: From a small molecule screen, we identified Scriptaid, a selective inhibitor of HDACs 1/3/8, as a repressor of TGFß-mediated CAF differentiation. Scriptaid inhibits ECM secretion, reduces cellular contraction and stiffness, and impairs collective cell invasion in CAF/tumour cell spheroid co-cultures. Scriptaid also reduces CAF abundance and delays tumour growth in vivo. CONCLUSIONS: Scriptaid is a well-tolerated and effective HDACi that reverses many of the functional and phenotypic properties of CAFs. Impeding or reversing CAF activation/function by altering the cellular epigenetic regulatory machinery could control tumour growth and invasion, and be beneficial in combination with additional therapies that target cancer cells or immune cells directly.


Asunto(s)
Fibroblastos Asociados al Cáncer/efectos de los fármacos , Inhibidores de Histona Desacetilasas/administración & dosificación , Hidroxilaminas/administración & dosificación , Neoplasias/tratamiento farmacológico , Quinolinas/administración & dosificación , Factor de Crecimiento Transformador beta/genética , Animales , Fibroblastos Asociados al Cáncer/metabolismo , Fibroblastos Asociados al Cáncer/ultraestructura , Diferenciación Celular/efectos de los fármacos , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/genética , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/ultraestructura , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/ultraestructura , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Ratones , Análisis por Micromatrices , Microscopía de Fuerza Atómica , Neoplasias/patología , Neoplasias/ultraestructura , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Ensayos Antitumor por Modelo de Xenoinjerto
20.
Oncotarget ; 7(44): 71841-71855, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27708247

RESUMEN

Non-small cell lung cancer (NSCLC), comprising 85% of lung cancer cases, has been associated with resistance to chemo/radiotherapy. The hypoxic tumor micro-environment, where insufficient vasculature results in poor drug penetrance and sub-optimal chemotherapy in the tumor interiors contributes heavily to this resistance. Additionally, epigenetic changes in tumorigenic cells also change their response to different forms of therapy. In our study, we have investigated the effectiveness of a combination of cisplatin with scriptaid [a pan-Histone Deacetylase inhibitor (HDACi)] in a model that mimics the tumor microenvironment of hypoxia and sub-lethal chemotherapy. Scriptaid synergistically increases the efficacy of cisplatin in normoxia as well as hypoxia, accompanied with reduced metastasis and enhanced DNA damage. Addition of scriptaid also overcomes the cisplatin resistance exhibited in lung cancer cells with stabilized hypoxia inducible factor 1 (HIF1)-α (mutant) and mutant p53. Molecular studies showed that the combination treatment increased apoptotic cell death in both normoxia and hypoxia with a dual role of p38MAPK. Together, our results suggest that the combination of low dose cisplatin and scriptaid is cytotoxic to NSCLC lines, can overcome hypoxia induced resistance and mutant p53- induced instability often associated with this cancer, and has the potential to be an effective therapeutic modality.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Recuento de Células , Cisplatino/farmacología , Genes p53 , Inhibidores de Histona Desacetilasas/inmunología , Hidroxilaminas/farmacología , Neoplasias Pulmonares/tratamiento farmacológico , Mutación , Quinolinas/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoptosis/efectos de los fármacos , Carcinoma de Pulmón de Células no Pequeñas/genética , Línea Celular Tumoral , Cisplatino/administración & dosificación , Resistencia a Antineoplásicos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Hidroxilaminas/administración & dosificación , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Neoplasias Pulmonares/genética , Quinolinas/administración & dosificación , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...