Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.048
Filter
1.
Drug Des Devel Ther ; 18: 3987-4001, 2024.
Article in English | MEDLINE | ID: mdl-39258274

ABSTRACT

Cytomegalovirus (CMV) infection is arguably the most important infectious complication that negatively affects the outcome of solid organ transplantation. For decades, CMV management after transplantation has relied on antiviral drugs that inhibit viral DNA polymerase (ganciclovir, foscarnet, and cidofovir). However, their use has been complicated by myelosuppression, nephrotoxicity, and selection of drug-resistant viruses. During the past few years, the therapeutic armamentarium for the management of CMV in solid organ transplant recipients has expanded with the approval of letermovir for CMV prophylaxis in high-risk CMV D+/R- kidney recipients, and maribavir for the treatment of refractory and resistant CMV infection. Both drugs offer significant improvement when compared to standard anti-CMV therapies; letermovir was as efficacious for CMV prevention, whereas maribavir was more effective in treating refractory and resistant CMV infections. Both letermovir and maribavir have favorable safety profiles compared to CMV DNA polymerase inhibitors, without the risk of neutropenia and leukopenia associated with ganciclovir and renal toxicities associated with foscarnet and cidofovir. Moreover, letermovir and maribavir are orally bioavailable, which allows convenient outpatient treatment. However, letermovir and maribavir have a significant drug interaction potential in solid organ transplant recipients, resulting in higher levels of calcineurin inhibitors (cyclosporine and tacrolimus) and mTOR inhibitors (sirolimus and everolimus). Both letermovir and maribavir are CMV-specific and do not have clinical efficacy against other herpes viruses. Thus, there is a need for additional antiviral drugs to prevent herpes simplex and other herpes viruses when clinically indicated. This article provides a comprehensive review of the clinical data supporting the use of letermovir and maribavir in clinical practice. The author provides perspectives on the role of these newly approved drugs in the current management landscape of CMV infection in solid organ transplantation.


Subject(s)
Acetates , Antiviral Agents , Cytomegalovirus Infections , Organ Transplantation , Quinazolines , Ribonucleosides , Humans , Cytomegalovirus Infections/drug therapy , Antiviral Agents/therapeutic use , Antiviral Agents/adverse effects , Antiviral Agents/pharmacology , Ribonucleosides/therapeutic use , Ribonucleosides/adverse effects , Ribonucleosides/pharmacology , Organ Transplantation/adverse effects , Acetates/therapeutic use , Acetates/adverse effects , Acetates/pharmacology , Quinazolines/therapeutic use , Quinazolines/pharmacology , Benzimidazoles/therapeutic use , Benzimidazoles/adverse effects , Benzimidazoles/pharmacology , Cytomegalovirus/drug effects , Dichlororibofuranosylbenzimidazole/analogs & derivatives
2.
Clin Transplant ; 38(9): e15453, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39229690

ABSTRACT

PURPOSE: Evaluate cytomegalovirus (CMV) post-prophylaxis surveillance in high-risk (D+/R-) kidney and liver transplant recipients. METHODS: Adult D+/R- patients were included if transplanted between 6/1/15 and 11/30/22 and divided into a pre-CMV-stewardship-era (6/1/15-5/31/18), CMV-stewardship-era (6/1/18-6/30/20), and a surveillance-era (7/1/2020-11/30/2022) then followed through 12 months. The primary objective was to evaluate CMV-related outcomes. The secondary objective was to assess graft and patient survival by era. RESULTS: There were 328 patients in the study period; 133 in the pre-stewardship-era, 103 in the stewardship-era, and 92 in the surveillance-era. Replication rates in the surveillance-era were significantly higher, as anticipated due to increased sampling (pre 38.4%, stewardship 33.0%, surveillance 52.2%, p = 0.02). Time from transplant to first replication was similar (pre 214.0 ± 79.0 days, stewardship 231.1 ± 65.5, surveillance 234.9 ± 61.4, p = 0.29). CMV viral load (VL) at first detection, maximum-VL, and incidence of VL > 100 000 IU/mL were numerically lower in the surveillance era, although not statistically significant. CMV end-organ disease (p < 0.0001) and ganciclovir-resistance (p = 0.002) were significantly lower in the surveillance era than in both previous eras. Rejection was not different between eras (p = 0.4). Graft (p = 0.0007) and patient survival (p = 0.008) were significantly improved in the surveillance era. CONCLUSIONS: Post-prophylaxis surveillance significantly reduced CMV end-organ disease and resistance. Despite observing increased replication rates in the surveillance era, rejection was not significantly different and there was no graft loss or patient mortality at 12 months.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Cytomegalovirus , Drug Resistance, Viral , Ganciclovir , Graft Rejection , Graft Survival , Kidney Transplantation , Liver Transplantation , Humans , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/epidemiology , Male , Female , Middle Aged , Cytomegalovirus/isolation & purification , Cytomegalovirus/drug effects , Antiviral Agents/therapeutic use , Ganciclovir/therapeutic use , Follow-Up Studies , Liver Transplantation/adverse effects , Risk Factors , Kidney Transplantation/adverse effects , Prognosis , Graft Rejection/prevention & control , Graft Rejection/etiology , Graft Rejection/virology , Postoperative Complications/prevention & control , Adult , Survival Rate , Retrospective Studies , Transplant Recipients/statistics & numerical data
3.
Cells ; 13(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39195228

ABSTRACT

Herpesviral protein kinases, such as the therapy-relevant pUL97 of human cytomegalovirus (HCMV), are important for viral replication efficiency as well as pathogenesis, and represent key antiviral drug targets. HCMV pUL97 is a viral cyclin-dependent kinase (CDK) ortholog, as it shares functional and structural properties with human CDKs. Recently, the formation of vCDK/pUL97-cyclin complexes and the phosphorylation of a variety of viral and cellular substrate proteins has been demonstrated. Genetic mapping and structural modeling approaches helped to define two pUL97 interfaces, IF1 and IF2, responsible for cyclin binding. In particular, the regulatory importance of interactions between vCDK/pUL97 and host cyclins as well as CDKs has been highlighted, both as determinants of virus replication and as a novel drug-targeting option. This aspect was substantiated by the finding that virus replication was impaired upon cyclin type H knock-down, and that such host-directed interference also affected viruses resistant to existing therapies. Beyond the formation of binary interactive complexes, a ternary pUL97-cyclin H-CDK7 complex has also been described, and in light of this, an experimental trans-stimulation of CDK7 activity by pUL97 appeared crucial for virus-host coregulation. In accordance with this understanding, several novel antiviral targeting options have emerged. These include kinase inhibitors directed to pUL97, to host CDKs, and to the pUL97-cyclin H interactive complexes. Importantly, a statistically significant drug synergy has recently been reported for antiviral treatment schemes using combinations of pharmacologically relevant CDK7 and vCDK/pUL97 inhibitors, including maribavir. Combined, such findings provide increased options for anti-HCMV control. This review focuses on regulatory interactions of vCDK/pUL97 with the host cyclin-CDK apparatus, and it addresses the functional relevance of these key effector complexes for viral replication and pathogenesis. On this basis, novel strategies of antiviral drug targeting are defined.


Subject(s)
Antiviral Agents , Cyclin-Dependent Kinases , Cytomegalovirus , Viral Proteins , Humans , Antiviral Agents/pharmacology , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Viral Proteins/metabolism , Viral Proteins/chemistry , Cyclin-Dependent Kinases/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , Virus Replication/drug effects , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Animals , Cyclins/metabolism , Phosphotransferases (Alcohol Group Acceptor)
4.
Viruses ; 16(8)2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39205242

ABSTRACT

This systematic review discusses the use of prophylaxis to prevent cytomegalovirus (CMV) infection in recipients who have undergone hematopoietic cell transplantation. It highlights the need for new approaches to control and prevent CMV infection. The approval of the anti-CMV drug letermovir has made antiviral prophylaxis more popular. CMV-specific T cell-mediated immunity tests are effective in identifying patients who have undergone immune reconstitution and predicting disease progression. Maribavir (MBV) has been approved for the treatment of post-transplant CMV infection/disease in adolescents. Adoptive T-cell therapy and the PepVax CMV vaccine show promise in tackling refractory and resistant CMV. However, the effectiveness of PepVax in reducing CMV viremia/disease was not demonstrated in a phase II trial. Cell-mediated immunity assays are valuable for personalized management plans, but more interventional studies are needed. MBV and adoptive T-cell therapy are promising treatments, and trials for CMV vaccines are ongoing.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Cytomegalovirus , Hematopoietic Stem Cell Transplantation , Humans , Antiviral Agents/administration & dosage , Cytomegalovirus/drug effects , Cytomegalovirus/immunology , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Cytomegalovirus Vaccines/administration & dosage , Cytomegalovirus Vaccines/immunology , Dichlororibofuranosylbenzimidazole/administration & dosage , Dichlororibofuranosylbenzimidazole/analogs & derivatives , Hematopoietic Stem Cell Transplantation/adverse effects , T-Lymphocytes/immunology
5.
J Antimicrob Chemother ; 79(10): 2500-2508, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39087737

ABSTRACT

BACKGROUND: The treatment of congenital cytomegalovirus (CMV) infection is usually administered to neonates after birth; however, it can be anticipated during the prenatal period by treating pregnant women in order to reduce the severity of the congenital disease. The most commonly used treatment for CMV during pregnancy is valaciclovir; however, valganciclovir has a higher potency against CMV and is the first choice for neonates with congenital CMV disease. OBJECTIVES: We investigated neonatal and maternal safety of tertiary prevention in infected fetuses showing ultrasound features of infection using valganciclovir. METHODS: Retrospective cohort study of pregnant women and their symptomatic infected fetuses taking valganciclovir, 3 × 450 mg per day. All fetuses presented at least one prenatal feature on ultrasound. We assessed fetal/neonatal and maternal safety, as well as neonatal efficacy of treatment. The main outcome was neutropenia. Secondary outcomes included other haematological side effects, symptoms at birth and neonatal CMV-PCR was positive. RESULTS: Seventeen women with singleton pregnancies received valganciclovir from a median (IQR) of 27.1 (26.0-30.3) to 11.6 (6.5-12.9) weeks of gestation. No neonatal neutropenia was reported. One pregnancy was terminated for severe features. Three newborns (18%) were asymptomatic at birth, including one with negative CMV-PCR from blood and saliva. CMV-PCR was positive for 12/13 symptomatic newborns, with a median (IQR) log10 viral load of 3.36 (3.30-4.20), 4.03 (1.75-4.27) and 3.04 (0.00-3.40) log10 copies/mL in blood, urine and saliva, respectively. CONCLUSIONS: Tertiary prevention by valganciclovir appears to be well tolerated for both fetus and mother. However, more extensive trials accompanied by long-term follow-up are needed.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Pregnancy Complications, Infectious , Valganciclovir , Humans , Valganciclovir/therapeutic use , Valganciclovir/administration & dosage , Valganciclovir/adverse effects , Female , Pregnancy , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/congenital , Antiviral Agents/therapeutic use , Antiviral Agents/adverse effects , Antiviral Agents/administration & dosage , Retrospective Studies , Infant, Newborn , Adult , Pregnancy Complications, Infectious/drug therapy , Pregnancy Complications, Infectious/virology , Treatment Outcome , Cytomegalovirus/drug effects , Ganciclovir/analogs & derivatives , Ganciclovir/therapeutic use , Ganciclovir/adverse effects , Infectious Disease Transmission, Vertical/prevention & control
6.
J Med Virol ; 96(8): e29842, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39115036

ABSTRACT

To explore the impacts of cytomegalovirus (CMV) infection and antiviral treatment (AVT) on native liver survival (NLS) in biliary atresia (BA) infants. This retrospective cohort study included infants diagnosed as BA between January 2015 and December 2021 at Hunan Children's Hospital. CMV infection was defined by DNA polymerase chain reaction alone (DNA data set) and combination of DNA and immunoglobulin M (CMV data set). In the DNA data set of 330 patients, 234 patients (70.9%) survived with their native liver in 2 years, with 113 (73.9%) in the DNA- cohort, 70 (65.4%) in the DNA+ and AVT- cohort and 51 (72.9%) in the DNA+ and AVT+ cohort, without significant differences by log-rank tests. In patients administrated between 2015 and March 2019, there were 206 evaluable patients in the DNA data set, with rates of 5-year NLS of 68.3% in the DNA- cohort, similar to that in the DNA+ and AVT+ cohort (62.2%, p = 0.546), but significantly higher than that in the DNA+ and AVT- cohort (51.4%, p = 0.031). Similar trends were also observed in the CMV data set, although statistically insignificant. CMV infection before or on the day of HPE can reduce the rate of 5-year NLS and AVT was recommended for CMV-infected BA infants.


Subject(s)
Antiviral Agents , Biliary Atresia , Cytomegalovirus Infections , Cytomegalovirus , Humans , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Retrospective Studies , Biliary Atresia/drug therapy , Antiviral Agents/therapeutic use , Female , Male , Infant , Cytomegalovirus/genetics , Cytomegalovirus/drug effects , Prognosis , DNA, Viral , Infant, Newborn
7.
Viruses ; 16(7)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39066247

ABSTRACT

Despite the significant progress made, CMV infection is one of the most frequent infectious complications in transplant recipients. CMV infections that become refractory or resistant (R/R) to the available antiviral drugs constitute a clinical challenge and are associated with increased morbidity and mortality. Novel anti-CMV therapies have been recently developed and introduced in clinical practice, which may improve the treatment of these infections. In this review, we summarize the treatment options for R/R CMV infections in adult hematopoietic cell transplant and solid organ transplant recipients, with a special focus on newly available antiviral agents with anti-CMV activity, including maribavir and letermovir.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Cytomegalovirus , Drug Resistance, Viral , Transplant Recipients , Humans , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/etiology , Antiviral Agents/therapeutic use , Cytomegalovirus/drug effects , Cytomegalovirus/physiology , Hematopoietic Stem Cell Transplantation/adverse effects , Organ Transplantation/adverse effects , Acetates , Dichlororibofuranosylbenzimidazole/analogs & derivatives , Quinazolines
8.
Clin Infect Dis ; 79(3): 787-794, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39041385

ABSTRACT

Cytomegalovirus (CMV) infection and disease are important causes of morbidity and mortality in transplant recipients. For the purpose of developing consistent reporting of CMV outcomes in clinical trials, definitions of CMV infection and disease were developed and most recently published in 2017. Since then, there have been major developments, including registration of new antiviral agents. Therefore, the Transplant Associated Virus Infections Forum, which consists of scientists, clinicians, regulators, and industry representatives, has produced an updated version of these definitions that incorporates recent knowledge with the aim of supporting clinical research and drug development. This also includes an update regarding the definition of resistant and refractory CMV infections previously published in 2019. As the field evolves, the need for updates of these definitions is clear, and collaborative efforts among clinicians, scientists, regulators, and industry representatives can provide a platform for this work.


Subject(s)
Antiviral Agents , Clinical Trials as Topic , Consensus , Cytomegalovirus Infections , Cytomegalovirus , Transplant Recipients , Humans , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/diagnosis , Antiviral Agents/therapeutic use , Cytomegalovirus/drug effects , Drug Resistance, Viral , Organ Transplantation/adverse effects
9.
mBio ; 15(8): e0315223, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38953361

ABSTRACT

There are no licensed vaccines for human cytomegalovirus (HCMV), and current antiviral drugs that target viral proteins are toxic and prone to resistance. Targeting host pathways essential for virus replication provides an alternate strategy that may reduce opportunities for drug resistance to occur. Oxidative stress is triggered by numerous viruses including HCMV. Peroxynitrite is a reactive nitrogen species that is formed during oxidative stress. Herein, we identified that HCMV rapidly induces the generation of intracellular peroxynitrite upon infection in a manner partially dependent upon xanthine oxidase generation. Peroxynitrite promoted HCMV infection in both cell-free and cell-associated infection systems in multiple cell types. Inhibiting peroxynitrite within the first 24 hours of infection prevented HCMV replication and peroxynitrite promoted cell entry and pp65 translocation into the host cell nuclei. Furthermore, using the murine cytomegalovirus model, we demonstrated that antagonizing peroxynitrite significantly reduces cytomegalovirus replication and pathogenesis in vivo. Overall, our study highlights a proviral role for peroxynitrite in CMV infection and implies that RNS and/or the mechanisms that induce their production could be targeted as a novel strategy to inhibit HCMV infection. IMPORTANCE: Human cytomegalovirus (HCMV) causes significant disease in individuals with impaired or immature immune systems, such as transplant patients and after congenital infection. Antiviral drugs that target the virus directly are toxic and are susceptible to antiviral drug resistance due to virus mutations. An alternate strategy is to target processes within host cells that are required by the virus for replication. Herein, we show that HCMV infection triggers a highly reactive molecule, peroxynitrite, during the initial stages of infection. Peroxynitrite was required for the initial entry of the virus into the cell and promotes virus replication in multiple cell types, suggesting a broad pro-viral function. Importantly, targeting peroxynitrite dramatically inhibited cytomegalovirus replication in cells in the laboratory and in mice, suggesting that therapeutic targeting of this molecule and/or the cellular functions it regulates could represent a novel strategy to inhibit HCMV infection.


Subject(s)
Cytomegalovirus Infections , Cytomegalovirus , Disease Models, Animal , Peroxynitrous Acid , Virus Internalization , Virus Replication , Peroxynitrous Acid/metabolism , Peroxynitrous Acid/pharmacology , Animals , Mice , Cytomegalovirus/physiology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Virus Internalization/drug effects , Humans , Cytomegalovirus Infections/virology , Virus Replication/drug effects , Muromegalovirus/physiology , Muromegalovirus/drug effects , Cell Line , Oxidative Stress
11.
Antiviral Res ; 228: 105935, 2024 08.
Article in English | MEDLINE | ID: mdl-38880196

ABSTRACT

Emergence of drug resistance is rare after use of letermovir (LMV) as prophylaxis for post-transplant cytomegalovirus (CMV) infection. In a recent study involving renal transplant recipients, no known LMV resistance mutations were detected in those receiving LMV prophylaxis. However, uncharacterized viral amino acid substitutions were detected in LMV recipients by deep sequencing in viral subpopulations of 5%-7%, at codons previously associated with drug resistance: UL56 S229Y (n = 1), UL56 M329I (n = 9) and UL89 D344Y (n = 5). Phenotypic analysis of these mutations in a cloned laboratory CMV strain showed that S229Y conferred a 2-fold increase in LMV EC50, M329I conferred no LMV resistance, and D344Y knocked out viral viability that was restored after the nonviable clone was reverted to wild type D344. As in previous CMV antiviral trials, the detection of nonviable mutations, even in multiple study subjects, raises strong suspicion of genotyping artifacts and encourages the use of replicate testing for authentication of atypical mutation readouts. The non-viability of UL89 D344Y also confirms the biologically important locus of the D344E substitution that confers resistance to benzimidazole CMV terminase complex inhibitors, but does not feature prominently in LMV resistance.


Subject(s)
Acetates , Antiviral Agents , Cytomegalovirus Infections , Cytomegalovirus , Drug Resistance, Viral , Genotype , Phenotype , Quinazolines , Humans , Cytomegalovirus/genetics , Cytomegalovirus/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Drug Resistance, Viral/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/drug therapy , Quinazolines/pharmacology , Quinazolines/therapeutic use , Acetates/pharmacology , Acetates/therapeutic use , Amino Acid Substitution , Kidney Transplantation , Mutation , Genetic Variation , Genotyping Techniques/methods , Viral Proteins/genetics
12.
J Virol ; 98(7): e0021324, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38832789

ABSTRACT

Human cytomegalovirus (HCMV) is a ß-herpesvirus that poses severe disease risk for immunocompromised patients who experience primary infection or reactivation. Development and optimization of safe and effective anti-HCMV therapeutics is of urgent necessity for the prevention and treatment of HCMV-associated diseases in diverse populations. The use of neutralizing monoclonal antibodies (mAbs) to limit HCMV infection poses a promising therapeutic strategy, as anti-HCMV mAbs largely inhibit infection by targeting virion glycoprotein complexes. In contrast, the small-molecule compounds currently approved for patients (e.g., ganciclovir, letermovir, and maribavir) target later stages of the HCMV life cycle. Here, we present a broadly neutralizing human mAb, designated 1C10, elicited from a VelocImmune mouse immunized with infectious HCMV particles. Clone 1C10 neutralizes infection after virion binding to cells by targeting gH/gL envelope complexes and potently reduces infection of diverse HCMV strains in fibroblast, trophoblast, and epithelial cells. Antibody competition assays found that 1C10 recognizes a region of gH associated with broad neutralization and binds to soluble pentamer in the low nanomolar range. Importantly, 1C10 treatment significantly reduced virus proliferation in both fibroblast and epithelial cells. Further, the combination treatment of mAb 1C10 with ganciclovir reduced HCMV infection and proliferation in a synergistic manner. This work characterizes a neutralizing human mAb for potential use as a HCMV treatment, as well as a possible therapeutic strategy utilizing combination-based treatments targeting disparate steps of the viral life cycle. Collectively, the findings support an antibody-based therapy to effectively treat patients at risk for HCMV-associated diseases. IMPORTANCE: Human cytomegalovirus is a herpesvirus that infects a large proportion of the population and can cause significant disease in diverse patient populations whose immune systems are suppressed or compromised. The development and optimization of safe anti-HCMV therapeutics, especially those that have viral targets and inhibition mechanisms different from current HCMV treatments, are of urgent necessity to better public health. Human monoclonal antibodies (mAbs) that prevent HCMV entry of cells were identified by immunizing transgenic mice and screened for broad and effective neutralization capability. Here, we describe one such mAb, which was found to target gH/gL envelope complexes and effectively limit HCMV infection and dissemination. Further, administration of the antibody in combination with the antiviral drug ganciclovir inhibited HCMV in a synergistic manner, highlighting this approach and the use of anti-HCMV mAbs more broadly, as a potential therapeutic strategy for the treatment of diverse patient populations.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Cytomegalovirus Infections , Cytomegalovirus , Mice, Transgenic , Viral Envelope Proteins , Animals , Humans , Cytomegalovirus/immunology , Cytomegalovirus/drug effects , Mice , Antibodies, Viral/immunology , Cytomegalovirus Infections/immunology , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Viral Envelope Proteins/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Virion/immunology , Fibroblasts/virology , Virus Replication/drug effects , Broadly Neutralizing Antibodies/immunology , Antiviral Agents/pharmacology , Immunization
13.
Transplant Cell Ther ; 30(8): 792.e1-792.e12, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838781

ABSTRACT

Preemptive therapy (PET) historically has been the primary strategy to reduce early-onset cytomegalovirus (CMV) reactivation after allogeneic hematopoietic cell transplantation (HCT) but is associated with antiviral-associated toxicities and increases in healthcare resource utilization and cost. Despite its high cost, letermovir (LTV) prophylaxis has largely supplanted PET due to its effectiveness and tolerability. Direct comparisons between LTV and PET approaches on economic and clinical outcomes after allogeneic HCT remain limited. Objective: To compare total cost of care (inpatient and outpatient) between LTV prophylaxis and PET through day+180 after allogeneic HCT. Adult allogeneic CMV seropositive (R+) HCT recipients who initiated LTV <30 days after HCT between 01/01/18 and 12/31/18 were matched 1:1 to allogeneic CMV R+ HCT recipients between 01/01/15 and 12/31/17 (PET cohort). Patients were grouped into high-risk (HR) or standard-risk (SR) for CMV to compare the LTV and PET cohorts. Direct costs for each patient's index HCT admission and all subsequent inpatient and outpatient care through day+180 after HCT were determined and converted into 2021 US dollars and then to Medicare proportional dollars (MPD). A secondary analysis using 2019 average wholesale price was conducted to specifically evaluate anti-CMV medication costs. There were a total of 176 patients with 54 HR CMV pairs and 34 SR CMV pairs. No differences in survival between LTV and PET for both HR and SR CMV groups were observed. The rate of clinically significant CMV infection decreased for both HR CMV (11/54, 20.4% versus 38/54, 70.4%, P < .001) and SR CMV (1/34, 2.9% versus 12/34, 35.3%, P < .001) patients who were given LTV prophylaxis with corresponding reductions in val(ganciclovir) and foscarnet (HR CMV only) use. Among HR CMV patients, LTV prophylaxis was associated with reductions in CMV-related readmissions (3/54, 5.6% versus 18/54, 33.3%, P < .001) and outpatient visits within the first 100 days after HCT (20 versus 25, P = .002), and a decreased median total cost of care ($36,018 versus $75,525, P < .001) in MPD was observed. For SR CMV patients on LTV, a significant reduction in the median inpatient cost ($15,668 versus $27,818, P < .001) was found, but this finding was offset by a higher median outpatient cost ($26,145 versus $20,307, P = .030) that was not CMV-driven. LTV prophylaxis is highly effective in reducing clinically significant CMV reactivations for both HR and SR HCT recipients. In this study, LTV prophylaxis was associated with a decreased total cost of care for HR CMV patients through day+180. Specifically, reductions in CMV-related readmissions, exposure to CMV-directed antiviral agents, and outpatient visits in the first 100 days after HCT were observed. SR CMV patients receiving LTV prophylaxis benefited by having a reduced inpatient cost of care due to lowered room and pharmacy costs.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Cytomegalovirus , Hematopoietic Stem Cell Transplantation , Quinazolines , Humans , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/economics , Antiviral Agents/economics , Antiviral Agents/therapeutic use , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/economics , Cytomegalovirus Infections/drug therapy , Male , Female , Quinazolines/therapeutic use , Quinazolines/economics , Middle Aged , Adult , Cytomegalovirus/drug effects , Acetates/therapeutic use , Acetates/economics , Acetates/administration & dosage , Aged , Treatment Outcome , Retrospective Studies , Health Care Costs/statistics & numerical data , Cost-Benefit Analysis
15.
Viruses ; 16(6)2024 May 30.
Article in English | MEDLINE | ID: mdl-38932169

ABSTRACT

Repression of human cytomegalovirus (HCMV) immediate-early (IE) gene expression is a key regulatory step in the establishment and maintenance of latent reservoirs. Viral IE transcription and protein accumulation can be elevated during latency by treatment with histone deacetylase inhibitors such as valproic acid (VPA), rendering infected cells visible to adaptive immune responses. However, the latency-associated viral protein UL138 inhibits the ability of VPA to enhance IE gene expression during infection of incompletely differentiated myeloid cells that support latency. UL138 also limits the accumulation of IFNß transcripts by inhibiting the cGAS-STING-TBK1 DNA-sensing pathway. Here, we show that, in the absence of UL138, the cGAS-STING-TBK1 pathway promotes both IFNß accumulation and VPA-responsive IE gene expression in incompletely differentiated myeloid cells. Inactivation of this pathway by either genetic or pharmacological inhibition phenocopied UL138 expression and reduced VPA-responsive IE transcript and protein accumulation. This work reveals a link between cytoplasmic pathogen sensing and epigenetic control of viral lytic phase transcription and suggests that manipulation of pattern recognition receptor signaling pathways could aid in the refinement of MIEP regulatory strategies to target latent viral reservoirs.


Subject(s)
Cytomegalovirus , Membrane Proteins , Myeloid Cells , Nucleotidyltransferases , Protein Serine-Threonine Kinases , Signal Transduction , Valproic Acid , Humans , Valproic Acid/pharmacology , Myeloid Cells/virology , Myeloid Cells/metabolism , Myeloid Cells/drug effects , Signal Transduction/drug effects , Membrane Proteins/metabolism , Membrane Proteins/genetics , Cytomegalovirus/physiology , Cytomegalovirus/drug effects , Cytomegalovirus/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Protein Serine-Threonine Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Cytomegalovirus Infections/virology , Cytomegalovirus Infections/metabolism , Cytomegalovirus Infections/genetics , Virus Latency/drug effects , Transcription, Genetic/drug effects , Cell Differentiation/drug effects , Gene Expression Regulation, Viral/drug effects , Genes, Immediate-Early , Interferon-beta/metabolism , Interferon-beta/genetics
16.
Transpl Infect Dis ; 26(3): e14282, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38824435

ABSTRACT

OBJECTIVES: Prophylaxis (P) or pre-emptive strategy (PS) in high-risk liver transplant recipients (LTRs) are either recommended. We compared the results of each strategy. METHODS: Two groups of LTR transplanted during two consecutive periods were compared. Only cytomegalovirus (CMV)-mismatched LTR (Donor +/ Recipient -) were included. The primary endpoints were: the onset of polymerase chain reaction-based DNAemia and the proportion of patients with CMV disease. A number of episodes of CMV infection, antiviral therapy, ganciclovir resistance, infectious or immunological complications, cost of both strategies, and survival (1, 5, and 10 years) were also compared. RESULTS: Forty-eight and 60 patients were respectively included in the P and PS groups. Eighteen (38%) in the P group and 56 (93%) in the PS group had CMV DNAemia (p <.0001) with a similar CMV disease rate (16.7% and 15%). Duration of curative therapy was longer in the PS group: 91 days versus 16 (p <.0001). Acute rejection was less frequent (p = .04) and more patients experienced a ganciclovir-resistant CMV infection in the PS group (10% vs. 0, p = .03). The drug-associated cost of PS was higher (10 004 vs. 4804€) and the median number of rehospitalization days tended to be higher (6 vs. 4, p = .06). Survival at any time was similar. CONCLUSION: We reported more CMV DNAemias and ganciclovir-resistant CMV events with PS. The cost of the PS strategy was higher.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Cytomegalovirus , Ganciclovir , Liver Transplantation , Humans , Cytomegalovirus Infections/prevention & control , Cytomegalovirus Infections/virology , Liver Transplantation/adverse effects , Male , Middle Aged , Antiviral Agents/therapeutic use , Antiviral Agents/administration & dosage , Female , Cytomegalovirus/drug effects , Ganciclovir/therapeutic use , Ganciclovir/administration & dosage , Adult , Aged , Transplant Recipients/statistics & numerical data , DNA, Viral/blood , Graft Rejection/prevention & control , Retrospective Studies , Drug Resistance, Viral
17.
Clin Lab ; 70(6)2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38868867

ABSTRACT

BACKGROUND: Both humoral and cell-mediated immunity of the patient affected by multiple myeloma (MM) are impaired; thus, infection is the main cause of the onset of symptoms and death caused by MM. Bortezomib is a first-line drug approved for patients with multiple myeloma (MM) and has significantly increased their overall survival. However, bortezomib-induced peripheral neuropathy (PN) remains a significant side effect that has led to its discontinuation in some patients. Guillain-Barre syndrome (GBS) is thought to be related to immune damage, and most patients have cytomegalovirus (CMV), Epstein-Barr virus (EBV), or mycoplasma infection before onset. Cases of GBS secondary to MM are rare. METHODS: We provide a case of GBS caused by cytomegalovirus infection after MM treatment, and briefly review the existing literature. RESULTS: Secondary GBS after MM. This patient received active treatment. The clinical symptoms are gradually improving. CONCLUSIONS: The use of bortezomib has the risk of reactivating the virus. It is more about the reactivation of hep-atitis B virus. Nonetheless, cytomegalovirus and Epstein-Barr virus shall have our attention. Patients with MM need to monitor CMV, regularly, especially during the treatment of bortezomib. At the same time, they also need to closely monitor the symptoms and signs of the nervous system to guard against the occurrence of GBS.


Subject(s)
Bortezomib , Cytomegalovirus Infections , Guillain-Barre Syndrome , Multiple Myeloma , Female , Humans , Middle Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Bortezomib/therapeutic use , Bortezomib/adverse effects , Cytomegalovirus/immunology , Cytomegalovirus/drug effects , Cytomegalovirus Infections/diagnosis , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/complications , Cytomegalovirus Infections/virology , Guillain-Barre Syndrome/diagnosis , Guillain-Barre Syndrome/drug therapy , Guillain-Barre Syndrome/etiology , Multiple Myeloma/drug therapy , Multiple Myeloma/complications
18.
Virology ; 595: 110096, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38710129

ABSTRACT

Herpes stromal keratitis is the leading cause of infectious blindness in the western world. Infection by HSV1 is most common, but VZV and hCMV also infect the cornea. Multiple models of HSV1 corneal infection exist, but none for VZV and hCMV because of their host specificity. Here, we used commercially available 3D human corneal epithelial equivalents (HCEE) to study infection by these herpesviruses. HCEE was infected by HSV-1 and hCMV without requiring scarification and resulted in spreading infections. Spread of HSV-1 infection was rapid, while that of hCMV was slow. In contrast, infections with VZV required damage to the HCEE and did not spread. Acyclovir dramatically reduced replication of HSV-1 in this model. We conclude that highly quality-controlled, readily available HCEE is a useful model to study human-restricted herpesvirus infection of the human corneal epithelium and for screening of antiviral drugs for treating HSK in an 3D model system.


Subject(s)
Antiviral Agents , Epithelium, Corneal , Herpesvirus 1, Human , Keratitis, Herpetic , Humans , Keratitis, Herpetic/virology , Keratitis, Herpetic/drug therapy , Epithelium, Corneal/virology , Epithelium, Corneal/pathology , Herpesvirus 1, Human/physiology , Herpesvirus 1, Human/drug effects , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Herpesvirus 3, Human/physiology , Herpesvirus 3, Human/drug effects , Cytomegalovirus/physiology , Cytomegalovirus/drug effects , Virus Replication , Acyclovir/pharmacology , Acyclovir/therapeutic use , Epithelial Cells/virology , Models, Biological
19.
Sci Rep ; 14(1): 10253, 2024 05 04.
Article in English | MEDLINE | ID: mdl-38704431

ABSTRACT

The tegument protein pp150 of Human Cytomegalovirus (HCMV) is known to be essential for the final stages of virus maturation and mediates its functions by interacting with capsid proteins. Our laboratory has previously identified the critical regions in pp150 important for pp150-capsid interactions and designed peptides similar in sequence to these regions, with a goal to competitively inhibit capsid maturation. Treatment with a specific peptide (PepCR2 or P10) targeted to pp150 conserved region 2 led to a significant reduction in murine CMV (MCMV) growth in cell culture, paving the way for in vivo testing in a mouse model of CMV infection. However, the general pharmacokinetic parameters of peptides, including rapid degradation and limited tissue and cell membrane permeability, pose a challenge to their successful use in vivo. Therefore, we designed a biopolymer-stabilized elastin-like polypeptide (ELP) fusion construct (ELP-P10) to enhance the bioavailability of P10. Antiviral efficacy and cytotoxic effects of ELP-P10 were studied in cell culture, and pharmacokinetics, biodistribution, and antiviral efficacy were studied in a mouse model of CMV infection. ELP-P10 maintained significant antiviral activity in cell culture, and this conjugation significantly enhanced P10 bioavailability in mouse tissues. The fluorescently labeled ELP-P10 accumulated to higher levels in mouse liver and kidneys as compared to the unconjugated P10. Moreover, viral titers from vital organs of MCMV-infected mice indicated a significant reduction of virus load upon ELP-P10 treatment. Therefore, ELP-P10 has the potential to be developed into an effective antiviral against CMV infection.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Elastin , Muromegalovirus , Peptides , Phosphoproteins , Viral Matrix Proteins , Animals , Mice , Antiviral Agents/pharmacology , Antiviral Agents/pharmacokinetics , Antiviral Agents/chemistry , Capsid/metabolism , Capsid/drug effects , Capsid Proteins/metabolism , Capsid Proteins/chemistry , Cytomegalovirus/drug effects , Cytomegalovirus Infections/drug therapy , Cytomegalovirus Infections/virology , Disease Models, Animal , Elastin/chemistry , Elastin/metabolism , Elastin-Like Polypeptides , Muromegalovirus/drug effects , Peptides/pharmacology , Peptides/chemistry , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/pharmacokinetics
20.
Ugeskr Laeger ; 186(15)2024 Apr 08.
Article in Danish | MEDLINE | ID: mdl-38708697

ABSTRACT

Cytomegalovirus infection (CMV) can be fatal for organ transplant recipients as shown in this case report. Maribavir is a recently approved drug, which can be used for therapy-refractory CMV infection or when other treatment options cannot be used. The patient in this case report was a CMV-infected liver transplant recipient, who developed a severe erythema and high CMV DNA during valganciclovir therapy. Toxic epidermal necrolysis was suspected. The patient was treated with maribavir, and both CMV DNA and the skin normalised. This case illustrates that maribavir is a useful alternative to other antiviral drugs for CMV infection.


Subject(s)
Antiviral Agents , Cytomegalovirus Infections , Dichlororibofuranosylbenzimidazole/analogs & derivatives , Liver Transplantation , Ribonucleosides , Humans , Cytomegalovirus Infections/drug therapy , Liver Transplantation/adverse effects , Antiviral Agents/therapeutic use , Ribonucleosides/therapeutic use , Ribonucleosides/administration & dosage , Benzimidazoles/therapeutic use , Benzimidazoles/adverse effects , Male , Middle Aged , Cytomegalovirus/isolation & purification , Cytomegalovirus/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL