Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 62
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Infect Dis ; 229(2): 413-421, 2024 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-37506264

RESUMEN

BACKGROUND: This drug resistance analysis of a randomized trial includes 234 patients receiving maribavir and 116 receiving investigator-assigned standard therapy (IAT), where 56% and 24%, respectively, cleared cytomegalovirus DNA at week 8 (treatment responders). METHODS: Baseline and posttreatment plasma samples were tested for mutations conferring drug resistance in viral genes UL97, UL54, and UL27. RESULTS: At baseline, genotypic testing revealed resistance to ganciclovir, foscarnet, or cidofovir in 56% of patients receiving maribavir and 68% receiving IAT, including 9 newly phenotyped mutations. Among them, 63% (maribavir) and 21% (IAT) were treatment responders. Detected baseline maribavir resistance mutations were UL27 L193F (n = 1) and UL97 F342Y (n = 3). Posttreatment, emergent maribavir resistance mutations were detected in 60 (26%) of those randomized to maribavir, including 49 (48%) of 103 nonresponders and 25 (86%) of the 29 nonresponders where viral DNA initially cleared then rebounded while on maribavir. The most common maribavir resistance mutations were UL97 T409M (n = 34), H411Y (n = 26), and C480F (n = 21), first detected 26 to 130 (median 56) days after starting maribavir. CONCLUSIONS: Baseline maribavir resistance was rare. Drug resistance to standard cytomegalovirus antivirals did not preclude treatment response to maribavir. Rebound in plasma cytomegalovirus DNA while on maribavir strongly suggests emerging drug resistance. CLINICAL TRIALS REGISTRATION: NCT02931539.


Asunto(s)
Infecciones por Citomegalovirus , Diclororribofuranosil Benzoimidazol , Ribonucleósidos , Humanos , Antivirales/uso terapéutico , Antivirales/farmacología , Bencimidazoles/uso terapéutico , Citomegalovirus/genética , Infecciones por Citomegalovirus/tratamiento farmacológico , Diclororribofuranosil Benzoimidazol/análogos & derivados , ADN , Farmacorresistencia Viral/genética , Ganciclovir/uso terapéutico , Mutación , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Ribonucleósidos/uso terapéutico , Receptores de Trasplantes
2.
Clin Infect Dis ; 78(3): 562-572, 2024 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-38036487

RESUMEN

BACKGROUND: Neutropenia may limit the use of valganciclovir treatment for cytomegalovirus (CMV) infection following hematopoietic cell transplant (HCT). A phase 2 study indicated efficacy of maribavir with fewer treatment-limiting toxicities than valganciclovir. METHODS: In this multicenter, double-blind, phase 3 study, patients with first asymptomatic CMV infection post-HCT were stratified and randomized 1:1 to maribavir 400 mg twice daily or valganciclovir (dose-adjusted for renal clearance) for 8 weeks with 12 weeks of follow-up. The primary endpoint was confirmed CMV viremia clearance at week 8 (primary hypothesis of noninferiority margin of 7.0%). The key secondary endpoint was a composite of the primary endpoint with no findings of CMV tissue-invasive disease at week 8 through week 16. Treatment-emergent adverse events (TEAEs) were assessed. RESULTS: Among patients treated (273 maribavir; 274 valganciclovir), the primary endpoint of noninferiority of maribavir was not met (maribavir, 69.6%; valganciclovir, 77.4%; adjusted difference: -7.7%; 95% confidence interval [CI]: -14.98, -.36; lower limit of 95% CI of treatment difference exceeded -7.0%). At week 16, 52.7% and 48.5% of patients treated (maribavir and valganciclovir, respectively) maintained CMV viremia clearance without tissue-invasive disease (adjusted difference: 4.4%; 95% CI: -3.91, 12.76). With maribavir (vs valganciclovir), fewer patients experienced neutropenia (16.1% and 52.9%) or discontinued due to TEAEs (27.8% and 41.2%). Discontinuations were mostly due to neutropenia (maribavir, 4.0%; valganciclovir, 17.5%). CONCLUSIONS: Although noninferiority of maribavir to valganciclovir for the primary endpoint was not achieved based on the prespecified noninferiority margin, maribavir demonstrated comparable CMV viremia clearance during post-treatment follow-up, with fewer discontinuations due to neutropenia. Clinical Trials Registration. NCT02927067 [AURORA].


Asunto(s)
Infecciones por Citomegalovirus , Diclororribofuranosil Benzoimidazol , Trasplante de Células Madre Hematopoyéticas , Neutropenia , Humanos , Antivirales/efectos adversos , Diclororribofuranosil Benzoimidazol/análogos & derivados , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Neutropenia/inducido químicamente , Valganciclovir/efectos adversos , Viremia/tratamiento farmacológico
3.
J Med Virol ; 96(4): e29609, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38647051

RESUMEN

This study evaluated the cost-effectiveness of maribavir versus investigator-assigned therapy (IAT; valganciclovir/ganciclovir, foscarnet, or cidofovir) for post-transplant refractory cytomegalovirus (CMV) infection with or without resistance. A two-stage Markov model was designed using data from the SOLSTICE trial (NCT02931539), real-world multinational observational studies, and published literature. Stage 1 (0-78 weeks) comprised clinically significant CMV (csCMV), non-clinically significant CMV (n-csCMV), and dead states; stage 2 (78 weeks-lifetime) comprised alive and dead states. Total costs (2022 USD) and quality-adjusted life years (QALYs) were estimated for the maribavir and IAT cohorts. An incremental cost-effectiveness ratio was calculated to determine cost-effectiveness against a willingness-to-pay threshold of $100 000/QALY. Compared with IAT, maribavir had lower costs ($139 751 vs $147 949) and greater QALYs (6.04 vs 5.83), making it cost-saving and more cost-effective. Maribavir had higher acquisition costs compared with IAT ($80 531 vs $65 285), but lower costs associated with administration/monitoring ($16 493 vs $27 563), adverse events (AEs) ($11 055 vs $16 114), hospitalization ($27 157 vs $33 905), and graft loss ($4516 vs $5081), thus making treatment with maribavir cost-saving. Maribavir-treated patients spent more time without CMV compared with IAT-treated patients (0.85 years vs 0.68 years), leading to lower retreatment costs for maribavir (cost savings: -$42 970.80). Compared with IAT, maribavir was more cost-effective for transplant recipients with refractory CMV, owing to better clinical efficacy and avoidance of high costs associated with administration, monitoring, AEs, and hospitalizations. These results can inform healthcare decision-makers on the most effective use of their resources for post-transplant refractory CMV treatment.


Asunto(s)
Antivirales , Bencimidazoles , Análisis Costo-Beneficio , Infecciones por Citomegalovirus , Diclororribofuranosil Benzoimidazol/análogos & derivados , Años de Vida Ajustados por Calidad de Vida , Ribonucleósidos , Humanos , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/economía , Antivirales/uso terapéutico , Antivirales/economía , Ribonucleósidos/uso terapéutico , Ribonucleósidos/economía , Bencimidazoles/uso terapéutico , Bencimidazoles/economía , Estados Unidos , Citomegalovirus/efectos de los fármacos , Citomegalovirus/genética , Farmacorresistencia Viral , Masculino , Femenino , Persona de Mediana Edad , Adulto , Genotipo , Receptores de Trasplantes
4.
Transpl Infect Dis ; 26(2): e14216, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38221739

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) infections among hematopoietic stem cell transplant (HSCT) and solid organ transplant (SOT) recipients impose a significant health care resource utilization (HCRU)-related economic burden. Maribavir (MBV), a novel anti-viral therapy (AVT), approved by the United States Food and Drug Administration for post-transplant CMV infections refractory (with/without resistance) to conventional AVTs has demonstrated lower hospital length of stay (LOS) versus investigator-assigned therapy (IAT; valgancilovir, ganciclovir, foscarnet, or cidofovir) in a phase 3 trial (SOLSTICE). This study estimated the HCRU costs of MBV versus IAT. METHODS: An economic model was developed to estimate HCRU costs for patients treated with MBV or IAT. Mean per-patient-per-year (PPPY) HCRU costs were calculated using (i) annualized mean hospital LOS in SOLSTICE, and (ii) CMV-related direct costs from published literature. Probabilistic sensitivity analysis with Monte-Carlo simulations assessed model robustness. RESULTS: Of 352 randomized patients receiving MBV (n = 235) or IAT (n = 117) for 8 weeks in SOLSTICE, 40% had HSCT and 60% had SOT. Mean overall PPPY HCRU costs of overall hospital-LOS were $67,205 (95% confidence interval [CI]: $33,767, $231,275) versus $145,501 (95% CI: $62,064, $589,505) for MBV and IAT groups, respectively. Mean PPPY ICU and non-ICU stay costs were: $32,231 (95% CI: $5,248, $184,524) versus $45,307 (95% CI: $3,957, $481,740) for MBV and IAT groups, and $82,237 (95% CI: $40,397, $156,945) MBV versus $228,329 (95% CI: $94,442, $517,476) for MBV and IAT groups, respectively. MBV demonstrated cost savings in over 99.99% of simulations. CONCLUSIONS: This analysis suggests that Mean PPPY HCRU costs were 29%-64% lower with MBV versus other-AVTs.


Asunto(s)
Infecciones por Citomegalovirus , Diclororribofuranosil Benzoimidazol/análogos & derivados , Trasplante de Órganos , Ribonucleósidos , Humanos , Citomegalovirus , Antivirales , Ganciclovir/uso terapéutico , Hospitalización , Receptores de Trasplantes , Bencimidazoles/uso terapéutico , Ribonucleósidos/uso terapéutico , Ribonucleósidos/efectos adversos , Trasplante de Órganos/efectos adversos , Células Madre Hematopoyéticas
5.
J Biol Chem ; 298(11): 102513, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36150501

RESUMEN

The human cytomegalovirus (HCMV) UL97 protein is a conserved herpesvirus protein kinase (CHPK) and a viral cyclin-dependent kinase (v-CDK). However, mechanisms regulating its activity in the context of infection are unknown. Here, we identified several cellular regulatory 14-3-3 proteins as UL97-interacting partners that promote UL97 stability. Humans are known to encode seven isoforms of 14-3-3 proteins (ß, ε, η, γ, σ, θ, and ζ) that bind phosphoserines or phosphothreonines to impact protein structure, stability, activity, and localization. Our proteomic analysis of UL97 identified 49 interacting partners, including 14-3-3 isoforms ß, η, and γ. Furthermore, coimmunoprecipitation with Western blotting assays demonstrated that UL97 interaction with 14-3-3 isoforms ß, ε, η, γ, and θ occurs in a kinase activity-dependent manner. Using mutational analysis, we determined the serine residue at amino acid 13 of UL97 is crucial for 14-3-3 interaction. We demonstrate UL97 S13A (serine to alanine substitution at residue 13) retains kinase activity but the mutant protein accumulated at lower levels than WT UL97. Finally, we show both laboratory (AD169) and clinical (TB40/E) strains of HCMV encoding UL97 S13A replicated with WT kinetics in fibroblasts but showed decreased UL97 accumulation. Taken together, we conclude that 14-3-3 proteins interact with and stabilize UL97 during HCMV infection.


Asunto(s)
Proteínas 14-3-3 , Citomegalovirus , Humanos , Citomegalovirus/fisiología , Proteínas 14-3-3/genética , Proteínas 14-3-3/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , Serina/metabolismo , Proteómica , Fosfotransferasas (Aceptor de Grupo Alcohol)/metabolismo
6.
Transpl Int ; 36: 11785, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37901297

RESUMEN

Despite advances in monitoring and treatment, cytomegalovirus (CMV) infections remain one of the most common complications after solid organ transplantation (SOT). CMV infection may fail to respond to standard first- and second-line antiviral therapies with or without the presence of antiviral resistance to these therapies. This failure to respond after 14 days of appropriate treatment is referred to as "resistant/refractory CMV." Limited data on refractory CMV without antiviral resistance are available. Reported rates of resistant CMV are up to 18% in SOT recipients treated for CMV. Therapeutic options for treating these infections are limited due to the toxicity of the agent used or transplant-related complications. This is often the challenge with conventional agents such as ganciclovir, foscarnet and cidofovir. Recent introduction of new CMV agents including maribavir and letermovir as well as the use of adoptive T cell therapy may improve the outcome of these difficult-to-treat infections in SOT recipients. In this expert review, we focus on new treatment options for resistant/refractory CMV infection and disease in SOT recipients, with an emphasis on maribavir, letermovir, and adoptive T cell therapy.


Asunto(s)
Antivirales , Infecciones por Citomegalovirus , Humanos , Antivirales/uso terapéutico , Citomegalovirus , Infecciones por Citomegalovirus/tratamiento farmacológico , Acetatos/uso terapéutico , Ganciclovir/uso terapéutico
7.
Ann Pharmacother ; 57(5): 597-608, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36003036

RESUMEN

OBJECTIVE: To review the efficacy and safety of maribavir for management of cytomegalovirus (CMV) in solid organ transplant recipients. DATA SOURCES: A literature search of PubMed and the Cochrane Controlled Trials Register (1960 to early July 2022) was performed using the following search terms: maribavir, 1263W94, and cytomegalovirus. STUDY SELECTION AND DATA EXTRACTION: All relevant English-language studies were reviewed and considered, with a focus on phase 3 trials. DATA SYNTHESIS: Maribavir, an orally available benzimidazole riboside with minimal adverse effects, was originally studied for universal prophylaxis in phase 3 trials but failed to demonstrate noninferiority over placebo and oral ganciclovir. It was effective for preemptive treatment in a dose-finding Phase 2 study. Maribavir is FDA approved for treatment of refractory/resistant CMV infection based on improved response rate at 8 weeks compared with investigator-assigned therapy (IAT) when initiated at median viral loads less than approximately 10 000 IU/mL (55.7% vs 23.9%, P < 0.001). Recurrence after 8-week treatment for refractory/resistant CMV was high (maribavir 50% vs IAT 39%). Significant drug interactions exist and must be managed by a pharmacotherapy expert to prevent harm. RELEVANCE TO PATIENT CARE AND CLINICAL PRACTICE: The addition of maribavir to the antiviral armamentarium should improve the management of refractory/resistant CMV, allowing early transition from toxic, high-cost, intravenous agents such as foscarnet and outpatient management. Optimal timing of initiation, duration, and potential alternative uses are unclear. CONCLUSION: Future studies are needed to fully elucidate the role of maribavir in the management of CMV after transplant.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Adulto , Humanos , Receptores de Trasplantes , Antivirales , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/prevención & control , Ganciclovir/uso terapéutico , Bencimidazoles/efectos adversos
8.
J Infect Dis ; 226(4): 576-584, 2022 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-32726419

RESUMEN

BACKGROUND: In separate phase 2 trials, 120 patients received maribavir for cytomegalovirus (CMV) infection failing conventional therapy (trial 202) and 119 received maribavir for asymptomatic infection (trial 203). Overall, 172 cleared their CMV infection (CMV DNA <200 copies/mL) within 6 weeks. METHODS: Baseline and posttreatment plasma samples were tested for mutations in viral genes UL97, UL54, and/or UL27. Selected viral mutants were phenotyped for drug susceptibility. RESULTS: Baseline samples revealed UL54 mutations newly phenotyped as conferring resistance to standard DNA polymerase inhibitor(s), including K493N, P497S, K513T, L565V, V823A, A987V, and E989D. Of 29 patients (including 25 from trial 202) who cleared but later experienced recurrent CMV infection while on maribavir, 23 had available UL97 genotyping data; 17 had known resistance mutations (T409M or H411Y) and 5 additional had UL97 C480F alone. The newly phenotyped mutation C480F conferred high-grade maribavir resistance and low-grade ganciclovir resistance. Among 25 who did not respond to >14 days of therapy, 9 showed T409M or H411Y and 4 others showed C480F alone. CONCLUSIONS: After maribavir therapy (400-1200 mg twice daily), UL97 mutations T409M, H411Y, or C480F emerge to confer maribavir resistance in patients with recurrent CMV infection while on therapy or no response to therapy. CLINICAL TRIALS REGISTRATION: NCT01611974 and EudraCT 2010-024247-32.


Asunto(s)
Infecciones por Citomegalovirus , Citomegalovirus , Humanos , Citomegalovirus/genética , Farmacorresistencia Viral/genética , Antivirales/farmacología , Antivirales/uso terapéutico , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Ganciclovir/farmacología , Ganciclovir/uso terapéutico , Mutación , Fenotipo
9.
Clin Infect Dis ; 75(4): 690-701, 2022 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34864943

RESUMEN

BACKGROUND: Therapies for refractory cytomegalovirus infections (with or without resistance [R/R]) in transplant recipients are limited by toxicities. Maribavir has multimodal anti-cytomegalovirus activity through the inhibition of UL97 protein kinase. METHODS: In this phase 3, open-label study, hematopoietic-cell and solid-organ transplant recipients with R/R cytomegalovirus were randomized 2:1 to maribavir 400 mg twice daily or investigator-assigned therapy (IAT; valganciclovir/ganciclovir, foscarnet, or cidofovir) for 8 weeks, with 12 weeks of follow-up. The primary endpoint was confirmed cytomegalovirus clearance at end of week 8. The key secondary endpoint was achievement of cytomegalovirus clearance and symptom control at end of week 8, maintained through week 16. RESULTS: 352 patients were randomized (235 maribavir; 117 IAT). Significantly more patients in the maribavir versus IAT group achieved the primary endpoint (55.7% vs 23.9%; adjusted difference [95% confidence interval (CI)]: 32.8% [22.80-42.74]; P < .001) and key secondary endpoint (18.7% vs 10.3%; adjusted difference [95% CI]: 9.5% [2.02-16.88]; P = .01). Rates of treatment-emergent adverse events (TEAEs) were similar between groups (maribavir, 97.4%; IAT, 91.4%). Maribavir was associated with less acute kidney injury versus foscarnet (8.5% vs 21.3%) and neutropenia versus valganciclovir/ganciclovir (9.4% vs 33.9%). Fewer patients discontinued treatment due to TEAEs with maribavir (13.2%) than IAT (31.9%). One patient per group had fatal treatment-related TEAEs. CONCLUSIONS: Maribavir was superior to IAT for cytomegalovirus viremia clearance and viremia clearance plus symptom control maintained post-therapy in transplant recipients with R/R cytomegalovirus. Maribavir had fewer treatment discontinuations due to TEAEs than IAT. Clinical Trials Registration. NCT02931539 (SOLSTICE).


Asunto(s)
Infecciones por Citomegalovirus , Viremia , Antivirales/efectos adversos , Citomegalovirus , Diclororribofuranosil Benzoimidazol/análogos & derivados , Farmacorresistencia Viral , Foscarnet/uso terapéutico , Ganciclovir/uso terapéutico , Humanos , Valganciclovir/uso terapéutico , Viremia/tratamiento farmacológico
10.
Antimicrob Agents Chemother ; 66(9): e0240521, 2022 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-35916518

RESUMEN

Maribavir was approved by the U.S. Food and Drug Administration in November 2021 for the treatment of adult and pediatric patients with post-transplant cytomegalovirus (CMV) infection/disease that is refractory to treatment (with or without genotypic resistance) with ganciclovir, valganciclovir, cidofovir, or foscarnet. Maribavir is an oral benzimidazole riboside with potent and selective multimodal anti-CMV activity. It utilizes a novel mechanism of action which confers activity against CMV strains that are resistant to traditional anti-CMV agents, and also offers a more favorable safety profile relative to the dose-limiting side effects of previously available therapies. Maribavir was initially studied as an agent for CMV prophylaxis in solid organ and hematopoietic stem cell recipients, but initial phase III trials failed to meet clinical efficacy endpoints. It has been more recently studied as a therapeutic agent at higher doses for refractory-resistant (R-R) CMV infections with favorable outcomes. After an overview of maribavir's chemistry and clinical pharmacology, this review will summarize clinical efficacy, safety, tolerability, and resistance data associated with maribavir therapy.


Asunto(s)
Antiinfecciosos , Infecciones por Citomegalovirus , Adulto , Antiinfecciosos/farmacología , Antivirales/farmacología , Antivirales/uso terapéutico , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Niño , Cidofovir , Infecciones por Citomegalovirus/prevención & control , Diclororribofuranosil Benzoimidazol/análogos & derivados , Farmacorresistencia Viral , Foscarnet , Ganciclovir/uso terapéutico , Humanos , Valganciclovir/uso terapéutico
11.
J Clin Pharm Ther ; 47(5): 699-702, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35023177

RESUMEN

WHAT IS KNOWN AND OBJECTIVE: Management of pan-resistant cytomegalovirus infection (CMVi) requires a multifaceted approach, including host defence optimization by reducing immunosuppression, and standard or experimental antiviral therapy. CASE DESCRIPTION: A 36-year-old man with anaplastic lymphoma kinase (ALK)-negative anaplastic large cell lymphoma, who underwent allogeneic haematopoietic stem cell transplant (alloHCT) with resultant graft-versus-host disease treated with immunosuppressive therapy, developed pan-resistant CMVi. He was successfully treated with combination therapy of maribavir and letermovir. WHAT IS NEW AND CONCLUSION: Combination therapy, used for other infections to prevent cross-resistant, may apply for CMVi.


Asunto(s)
Infecciones por Citomegalovirus , Neoplasias Hematológicas , Trasplante de Células Madre Hematopoyéticas , Acetatos , Adulto , Antivirales/uso terapéutico , Bencimidazoles , Citomegalovirus , Infecciones por Citomegalovirus/tratamiento farmacológico , Neoplasias Hematológicas/complicaciones , Neoplasias Hematológicas/tratamiento farmacológico , Humanos , Masculino , Quinazolinas , Ribonucleósidos
12.
J Infect Dis ; 224(6): 1024-1028, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-33475730

RESUMEN

Two transplant recipients (1 kidney and 1 hematopoietic stem cell) received maribavir (MBV) after cytomegalovirus (CMV) infection clinically resistant to standard therapy. Both patients achieved CMV DNA clearance within 30 and 18 days; however, the UL97 C480F variant emerged, causing recurrent CMV infection after a cumulative 2 months of MBV and 15 or 4 weeks of ganciclovir treatment, respectively. C480F was not detected under ganciclovir before MBV treatment. Recombinant phenotyping showed that C480F conferred the highest level of MBV resistance and ganciclovir cross-resistance, with impaired viral growth. Clinical follow-up and genotypic and phenotypic studies are essential for the assessment and optimization of patients with suspected MBV resistance.


Asunto(s)
Bencimidazoles/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Farmacorresistencia Viral/genética , Ganciclovir/uso terapéutico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Riñón/efectos adversos , Ribonucleósidos/uso terapéutico , Receptores de Trasplantes , Adulto , Antivirales/farmacología , Antivirales/uso terapéutico , Bencimidazoles/farmacología , Citomegalovirus/genética , Farmacorresistencia Viral/efectos de los fármacos , Femenino , Ganciclovir/farmacología , Células Madre Hematopoyéticas , Humanos , Mutación/efectos de los fármacos , Trasplante de Órganos , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Fosfotransferasas (Aceptor de Grupo Alcohol)/uso terapéutico , Ribonucleósidos/farmacología , Resultado del Tratamiento
13.
Transpl Infect Dis ; 23(4): e13645, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34022099

RESUMEN

As some of those who were lucky enough to have been mentored by Dr Francisco Marty in transplant infectious diseases, we stand with the larger medical community in mourning his untimely death and in commemorating him as a uniquely exceptional and talented physician, investigator, teacher, mentor, friend, artist, and human being.


Asunto(s)
Médicos , Humanos , Masculino
14.
J Infect Dis ; 221(Suppl 1): S32-S44, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-32134483

RESUMEN

The development of therapeutics for cytomegalovirus (CMV) infections, while progressing, has not matched the pace of new treatments of human immunodeficiency virus (HIV) infections; nevertheless, recent developments in the treatment of CMV infections have resulted in improved human health and perhaps will encourage the development of new therapeutic approaches. First, the deployment of ganciclovir and valganciclovir for both the prevention and treatment of CMV infections and disease in transplant recipients has been further improved with the licensure of the efficacious and less toxic letermovir. Regardless, late-onset CMV disease, specifically pneumonia, remains problematic. Second, the treatment of congenital CMV infections with valganciclovir has beneficially improved both hearing and neurologic outcomes, both fundamental advances for these children. In these pediatric studies, viral load was decreased but not eliminated. Thus, an important lesson learned from studies in both populations is the need for new antiviral agents and the necessity for combination therapies as has been shown to be beneficial in the treatment of HIV infections, among others. The development of monoclonal antibodies, sirtuins, and cyclopropovir may provide new treatment options.


Asunto(s)
Antivirales/uso terapéutico , Infecciones por Citomegalovirus/tratamiento farmacológico , Citomegalovirus/efectos de los fármacos , Desarrollo de Medicamentos , Acetatos/administración & dosificación , Acetatos/efectos adversos , Acetatos/uso terapéutico , Antivirales/farmacología , Biomarcadores , Estudios Clínicos como Asunto , Infecciones por Citomegalovirus/prevención & control , Infecciones por Citomegalovirus/transmisión , Infecciones por Citomegalovirus/virología , Farmacorresistencia Viral , Humanos , Transmisión Vertical de Enfermedad Infecciosa , Quinazolinas/administración & dosificación , Quinazolinas/efectos adversos , Quinazolinas/uso terapéutico , Proyectos de Investigación , Sirtuinas/administración & dosificación , Sirtuinas/efectos adversos , Sirtuinas/uso terapéutico , Trasplante de Células Madre/efectos adversos , Trasplante de Células Madre/métodos , Resultado del Tratamiento , Carga Viral
15.
Artículo en Inglés | MEDLINE | ID: mdl-33077661

RESUMEN

Congenital cytomegalovirus (HCMV) infection may cause significant fetal malformation, lifelong disease, and, in severe cases, fetal or neonatal death. Placental infection with HCMV is the major mechanism of mother-to-child transmission (MTCT) and fetal injury. Thus, any pharmaceutical antiviral interference to reduce viral load may reduce placental damage, MTCT, and fetal disease. However, there is currently no licensed HCMV antiviral for use during pregnancy. In this study, aciclovir and the HCMV-specific antivirals letermovir, maribavir, and cidofovir were compared with ganciclovir for antiviral effects in model systems of pregnancy, including first-trimester TEV-1 trophoblast cell cultures and third-trimester ex vivo placental explant histocultures. HCMV-infected trophoblasts at 7 days postinfection (dpi) showed an EC50 of 21 µM for aciclovir, 0.0007 µM for letermovir, 0.11 µM for maribavir, and 0.29 µM for cidofovir, relative to 0.42 µM for ganciclovir. Antivirals added at 10 µM showed no cytotoxic effects and did not affect trophoblast cell proliferation (P > 0.9999). Multiple-round HCMV replication measured at 7 dpi showed letermovir, maribavir, and cidofovir treatment inhibited immediate early, early, and true late viral protein expression as assayed on Western blots. Antiviral treatment of HCMV-infected placental explants showed significant inhibition (P < 0.05) of viral replication with letermovir (83.3%), maribavir (83.6%), cidofovir (89.3%), and ganciclovir (82.4%), but not aciclovir (P > 0.9999). In ex vivo model systems, recently trialed HCMV antivirals letermovir and maribavir were effective at inhibiting HCMV replication. They partly fulfil requirements for use as safe and effective therapeutics during pregnancy to control congenital HCMV. Clinical trials of these newer agents would assist assessment of their utility in pregnancy.


Asunto(s)
Antivirales , Infecciones por Citomegalovirus , Antivirales/farmacología , Antivirales/uso terapéutico , Niño , Citomegalovirus , Infecciones por Citomegalovirus/tratamiento farmacológico , Femenino , Humanos , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Placenta , Embarazo , Replicación Viral
16.
J Virol ; 93(17)2019 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-31217241

RESUMEN

The herpesvirus human cytomegalovirus (HCMV) is a leading cause of congenital birth defects. Infection can result in infants born with a variety of symptoms, including hepatosplenomegaly, microcephaly, and developmental disabilities. Microcephaly is associated with disruptions in the neural progenitor cell (NPC) population. Here, we defined the impact of HCMV infection on neural tissue development and calcium regulation, a critical activity in neural development. Regulation of intracellular calcium involves purinergic receptors and voltage-gated calcium channels (VGCC). HCMV infection compromised the ability of both pathways in NPCs as well as fibroblasts to respond to stimulation. We observed significant drops in basal calcium levels in infected NPCs which were accompanied by loss in VGCC activity and purinergic receptor responses. However, uninfected cells in the population retained responsiveness. Addition of the HCMV inhibitor maribavir reduced viral spread but failed to restore activity in infected cells. To study neural development, we infected three-dimensional cortical organoids with HCMV. Infection spread to a subset of cells over time and disrupted organoid structure, with alterations in developmental and neural layering markers. Organoid-derived infected neurons and astrocytes were unable to respond to stimulation whereas uninfected cells retained nearly normal responses. Maribavir partially restored structural features, including neural rosette formation, and dampened the impact of infection on neural cellular function. Using a tissue model system, we have demonstrated that HCMV alters cortical neural layering and disrupts calcium regulation in infected cells.IMPORTANCE Human cytomegalovirus (HCMV) replicates in several cell types throughout the body, causing disease in the absence of an effective immune response. Studies on HCMV require cultured human cells and tissues due to species specificity. In these studies, we investigated the impact of infection on developing three-dimensional cortical organoid tissues, with specific emphasis on cell-type-dependent calcium signaling. Calcium signaling is an essential function during neural differentiation and cortical development. We observed that HCMV infects and spreads within these tissues, ultimately disrupting cortical structure. Infected cells exhibited depleted calcium stores and loss of ATP- and KCl-stimulated calcium signaling while uninfected cells in the population maintained nearly normal responses. Some protection was provided by the viral inhibitor maribavir. Overall, our studies provide new insights into the impact of HCMV on cortical tissue development and function.


Asunto(s)
Señalización del Calcio , Infecciones por Citomegalovirus/metabolismo , Citomegalovirus/patogenicidad , Células-Madre Neurales/virología , Organoides/virología , Bencimidazoles/farmacología , Diferenciación Celular , Línea Celular , Citomegalovirus/efectos de los fármacos , Citomegalovirus/fisiología , Humanos , Células Madre Pluripotentes Inducidas/citología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Técnicas de Cultivo de Órganos , Organoides/citología , Organoides/metabolismo , Receptores Purinérgicos/metabolismo , Ribonucleósidos/farmacología , Replicación Viral/efectos de los fármacos , Canales de Sodio Activados por Voltaje/metabolismo
17.
Intern Med J ; 50(3): 277-284, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31403736

RESUMEN

Cytomegalovirus (CMV) viraemia continues to be a frequent complication in the post-haemopoietic stem cell transplantation period despite a low incidence of CMV end-organ disease. Several significant advances in the understanding and management of CMV infection have occurred in the last few years including improved diagnostics, monitoring of CMV immunity, availability of novel anti-CMV drugs, and emerging use of immunotherapies including CMV-specific T-cell infusions. In addition to reviewing these advances we also explore some of the more practical prescribing issues of the older and newer CMV drugs including cost, toxicity and drug interactions to help clinicians navigate this new era of CMV management.


Asunto(s)
Infecciones por Citomegalovirus , Trasplante de Células Madre Hematopoyéticas , Antivirales/uso terapéutico , Citomegalovirus , Infecciones por Citomegalovirus/tratamiento farmacológico , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Linfocitos T
18.
Clin Infect Dis ; 68(8): 1255-1264, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30329038

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) infections that are refractory or resistant (RR) to available antivirals ([val]ganciclovir, foscarnet, cidofovir) are associated with higher mortality in transplant patients. Maribavir is active against RR CMV strains. METHODS: Hematopoietic-cell or solid-organ transplant recipients ≥12 years old with RR CMV infections and plasma CMV deoxyribonucleic acid (DNA) ≥1000 copies/mL were randomized (1:1:1) to twice-daily dose-blinded maribavir 400, 800, or 1200 mg for up to 24 weeks. The primary efficacy endpoint was the proportion of patients with confirmed undetectable plasma CMV DNA within 6 weeks of treatment. Safety analyses included the frequency and severity of treatment-emergent adverse events (TEAEs). RESULTS: From July 2012 to December 2014, 120 patients were randomized and treated (40 per dose group): 80/120 (67%) patients achieved undetectable CMV DNA within 6 weeks of treatment (95% confidence interval, 57-75%), with rates of 70%, 63%, and 68%, respectively, for maribavir 400, 800, and 1200 mg twice daily. Recurrent on-treatment CMV infections occurred in 25 patients; 13 developed mutations conferring maribavir resistance. Maribavir was discontinued due to adverse events in 41/120 (34%) patients, and 17/41 discontinued due to CMV infections. During the study, 32 (27%) patients died, 4 due to CMV disease. Dysgeusia was the most common TEAE (78/120; 65%) and led to maribavir discontinuation in 1 patient. Absolute neutrophil counts <1000/µL were noted in 12/106 (11%) evaluable patients, with rates similar across doses. CONCLUSIONS: Maribavir ≥400 mg twice daily was active against RR CMV infections in transplant recipients; no new safety signals were identified. CLINICAL TRIALS REGISTRATION: NCT01611974.


Asunto(s)
Antivirales/administración & dosificación , Bencimidazoles/administración & dosificación , Infecciones por Citomegalovirus/tratamiento farmacológico , Huésped Inmunocomprometido , Ribonucleósidos/administración & dosificación , Adolescente , Adulto , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Farmacorresistencia Viral , Femenino , Trasplante de Células Madre Hematopoyéticas , Humanos , Masculino , Receptores de Trasplantes , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA