Abstract
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.
Keywords: congenital cytomegalovirus, antiviral therapy, ganciclovir, valganciclovir, sirtuins, letermovir, cyclopropovir, maribavir, monoclonal antibodies
Advances in the prevention and treatment of cytomegalovirus (CMV) infections are limited but well documented. Realized successes indicate a need for improved therapies, if not combinations. Only 5 CMV therapeutics are approved by the US Food and Drug Administration (FDA): foscarnet (1991), ganciclovir (1994) and its prodrug valganciclovir (2001), cidofovir (1996), and letermovir (2017). Risk/benefit considerations of foscarnet and cidofovir preclude use in neonatal and infant populations; letermovir, while licensed for adult stem cell transplant recipients, is being evaluated in children undergoing transplantation. This document provides a glimpse of the limited achievements and offers hope for further improvements.
ADVANCES IN THE TREATMENT OF CONGENITAL CYTOMEGALOVIRUS INFECTIONS
CMV infection is the leading nongenetic cause of sensorineural hearing loss (SNHL) [1–4] and the most frequent known viral cause of mental retardation [5], affecting 0.5%–0.7% of live births in industrialized countries [6–8], including 19 000 to 26 600 congenital infections annually in the United States. Approximately 2300 (10%) have symptomatic disease at delivery, of whom 35% have SNHL, up to 66% have neurologic deficits, and 4% die in the newborn period [7–11]. SNHL occurs at a lower rate among the 90% of congenitally infected neonates who are asymptomatic at delivery, but accounts for the majority of cases of hearing loss overall [7, 12]. African American neonates disproportionately suffer from congenital CMV infection at triple the rate of white neonates and 9-fold higher than Asian American neonates [13]. As estimated in Table 1, each year 4000–5000 of these babies will develop CMV-associated disabilities. Congenital CMV accounts for 21% of hearing loss at birth and 24% of all cases of hearing loss by 4 years of age [1, 14]. The overall economic burden of congenital CMV infection exceeds $3 billion annually, adjusted for 2015 dollars [15–17].
Table 1.
Sequelae Following Congenital Cytomegalovirus Infection in the United States
| Parameter | Estimated Annual Value |
|---|---|
| No. of live births per year | 3 800 000 |
| Rate of congenital CMV infection | 0.6% |
| No. of infected infants | 22 800 |
| No. of infants symptomatic at birth (12.8%) | 2918 |
| Symptomatic at birth, who have or develop disability (50%) | 1459 |
| No. of infants asymptomatic at birth (87.2%) | 19 882 |
| Asymptomatic at birth, who have or develop disability (13.5%) | 2684 |
| Total with congenital CMV-related disabilities | 4143 |
Adapted from Cannon et al [18].
Abbreviation: CMV, cytomegalovirus.
Since the 1980s, the National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group (CASG) has conducted a series of clinical trials with parenteral ganciclovir and oral valganciclovir in infants <1 month of age born with symptomatic congenital CMV disease [19–25].
Study Design and Results of the Ganciclovir Study
After establishing the appropriate dose of intravenous (IV) ganciclovir for use in neonates in the 1980s [19, 24, 25], the CASG conducted a phase 3 randomized controlled trial of 6 weeks of IV ganciclovir (6 mg/kg/dose IV) vs no therapy for neonates with symptomatic neonatal disease [23]. One hundred patients with symptomatic congenital CMV disease involving the central nervous system (CNS) were randomized. The primary study endpoint was improved brain stem evoked response (BSER) audiometry by 1 gradation between baseline and the 6-month follow-up (or, for those patients with normal hearing at baseline, normal BSER at both time points). Clinical and laboratory improvement constituted secondary endpoints. Audiological analyses were performed on the best evaluable ear (“functional” assessment) and on total evaluable ears (“biologic” assessment).
For infants available at follow-up, 21 of 25 (84%) ganciclovir recipients either had improvement in hearing in their best ear between baseline and 6 months or had normal hearing at both time points, compared with 10 of 17 (59%) subjects in the no treatment group (odds ratio [OR], 5.03 [95% confidence interval {CI}, .84–45.94]; adjusted P = .06). None of the 25 ganciclovir recipients had hearing deterioration in their best ear between baseline and 6 months, compared with 7 of 17 (41%) subjects in the no treatment group (OR, 21.11 [95% CI, 2.84–∞]; adjusted P < .001). Five of 24 (21%) ganciclovir recipients had worsening in hearing in their best ear between baseline and ≥1 year, compared with 13 of 19 (68%) subjects in the no treatment group (OR, 10.26 [95% CI, 1.79–81.92]; adjusted P = .002). Ganciclovir-treated subjects also normalized alanine aminotransferase (ALT) more rapidly compared with the no treatment group (19 vs 66 days, respectively; P = .03).
Twenty-nine of 46 (63%) ganciclovir-treated subjects developed grade 3 or 4 neutropenia (Division of AIDS Toxicity Tables) compared with 9 of 43 (21%) subjects in the no treatment group (P < .01). Fourteen of the 29 (48%) required dosage adjustments, although only 4 patients discontinued drug permanently. Neutropenia in ganciclovir-treated subjects resolved in 12.8 (±13.6) days, and in the no treatment group in 14.2 (±13.5) days. All subjects resolved their neutropenia. The incidence of grade 3–4 thrombocytopenia was comparable across both study arms, as was the incidence of grade 3–4 increases in ALT, and total bilirubin levels.
Developmental Analyses
Denver Developmental Evaluations were performed at 6 weeks, 6 months, and 12 months [26]. At the 6-week assessment, the average number of developmental delays per subject was 1.5 for ganciclovir and 2.05 for the no treatment subjects (P = .13); at 6 months, delays were 4.46 and 7.51, respectively (P = .06) and at 12 months were 9.78 vs 17.14, respectively (P = .007). Multivariate analysis of variance tested independent factors related to poor development, indicating that the ganciclovir benefit remained statistically significant at 12 months (P = .007), and approached statistical significance at 6 weeks (P = .08) and 6 months (P = .08).
Study Design and Results of the Valganciclovir Therapy Study
Ganciclovir results suggested that therapeutic benefit waned over the first 2 years of life [23], implying that a new strategy of extending the duration of therapy may be beneficial. Thus, the CASG developed a pharmacokinetic and pharmacodynamic study to determine the dose of oral valganciclovir that achieved systemic ganciclovir exposure equivalent to IV ganciclovir [21], generally an area under the curve (AUC12) in the range of 27–32 µg × hour/mL, with a coefficient of variation in the range of 30%–40%.
Twenty-four subjects <1 month of age with symptomatic congenital CMV disease were assessed. A dose of 15.62 mg/kg provided an AUC12 of 27.4 µg × hour/mL. The oral bioavailability of valganciclovir increased in early infancy from 48% at approximately 4 weeks of life to 64% at approximately at 7 weeks of life, an increase proportionate to improved ganciclovir renal clearance.
Using the oral dose of valganciclovir of 16 mg/kg/dose twice daily, the CASG study assessed whether longer-term therapy provided prolonged beneficial effects. Ninety-six subjects were randomized to blinded study medication (47 drug, 49 placebo) after receiving 6 weeks of valganciclovir. Total ears from subjects receiving 6 months of valganciclovir were more likely to have improved hearing or to maintain normal hearing between baseline and 12 months (compared to 6 weeks) after a priori adjustment for CNS involvement at baseline (adjusted OR [aOR], 3.04 [95% CI, 1.26–7.35]; P = .01). Similar results were evident when prematurity and age at treatment initiation were added to the model (P = .01). The relative risk for improved or protected total ear hearing between baseline and 12 months for the 53 subjects with baseline CNS involvement who received 6 months of treatment and 6 weeks of treatment was 1.66 (95% CI, .92–2.4), and the risk difference was 0.27 (95% CI, .09–.45). The benefit of longer-term therapy in the total ear analysis was maintained at 24 months, with improved outcomes after adjusting for CNS involvement at baseline (aOR, 2.61 [95% CI, 1.05–6.43]; P = .04). Similar results were evident when prematurity at treatment initiation was modeled (P = .04). The relative risk for improved or protected total ear hearing between baseline and 24 months was 1.46 (95% CI, .87–2.05); the risk difference was 0.23 (95% CI, .05–.41), following 6 months of therapy.
Adjusting a priori for CNS involvement, valganciclovir therapy led to higher Bayley III Language Composite (P = .0046) and Receptive Communication Scale (P = .0031) developmental scores at 24 months compared with subjects randomized to 6 weeks of treatment. These differences were maintained when age at treatment initiation and prematurity were added to the model (P = .0037 and P = .0027, respectively).
Collectively, these trials led the FDA to modify the valganciclovir package insert to cite the dose used in CASG studies [22, 27], and the American Academy of Pediatrics now recommends 6 months of oral valganciclovir therapy for infants with symptomatic congenital CMV disease [28, 29].
Implications for Potential Biomarkers on New Treatment Approaches
Overall clinical benefit is modest, raising the possibility that more effective drugs or treatment regimens could provide additional benefit. Subjects with symptomatic congenital CMV disease who achieve complete viral suppression (defined as ≤2.5 log) by day 14 of valganciclovir therapy and maintain it over the next 4 months are statistically more likely to have improved hearing across the first 2 years of life (Table 2) [30]. However, the ability of ganciclovir/valganciclovir to cause such a rapid decline is limited to a small number of patients; typically, viral load decreases by only 1 log over the first week and then <0.5 log over subsequent weeks (Figure 1) [22]. The rapidity of clearance of virus and cerebrospinal fluid findings require further evaluation. Combination therapy has the potential to significantly advance management options in the treatment of symptomatic congenital CMV disease, as has been the case with combination therapy in the treatment of human immunodeficiency virus (HIV) and hepatitis C virus infections. These data foreshadow the potential for linking CMV biomarker(s) to drug exposure. These relationships would serve to isolate the most important pharmacokinetic parameters linked to treatment response, and more precisely identify the optimum dosing strategy.
Table 2.
Improvement and Protection in Best-Ear and Total-Ear Hearing Between Baseline and Follow-up in Subjects With Complete Viral Suppression by Day 14 of Therapy Through Month 4, and Subjects Without Complete Viral Suppression
| Analysis | No. of Subjects or Ears | Hearing Between Baseline and Follow-up | P Value | |
|---|---|---|---|---|
| Improved/Protected | Others | |||
| 6-mo total-ear hearing | ||||
| With complete viral suppression | 19 | 17 (89) | 2 (11) | .0098 |
| Without complete viral suppression | 48 | 27 (56) | 21 (44) | |
| 12-mo total-ear hearing | ||||
| With complete viral suppression | 20 | 20 (100) | 0 | .0007 |
| Without complete viral suppression | 48 | 30 (63) | 18 (38) | |
| 24-mo total-ear hearing | ||||
| With complete viral suppression | 17 | 16 (94) | 1 (6) | .0458 |
| Without complete viral suppression | 41 | 28 (68) | 13 (32) |
Adapted from Marsico et al [30].
Figure 1.
Whole blood cytomegalovirus DNA viral load. Reprinted from Kimberlin et al [31] with permission. Clearance of virus in congenitally infected infants who received either 6 weeks or 6 months of therapy. A rebound in virus titer occurred upon drug discontinuation.
Current CASG Studies Assessing Additional Populations and Treatment Approaches
The CASG is currently evaluating oral valganciclovir therapy when started beyond the first month of life for treatment of infants and toddlers with CMV-associated hearing loss in a placebo-controlled trial (ClinicalTrials.gov identifier NCT01649869). Furthermore, screening of 50 000 babies has been undertaken to identify approximately 230 otherwise asymptomatic infected infants who will receive oral valganciclovir therapy for 4 months (ClinicalTrials.gov identifier NCT03301415) to determine if hearing loss can be prevented.
UTILIZATION OF LETERMOVIR IN THE PREVENTION OF CMV INFECTIONS IN STEM CELL TRANSPLANT RECIPIENTS
CMV is the most common clinically significant viral infection following stem cell or organ transplantation, causing morbidity due to direct effects (pneumonia, hepatitis, retinitis and encephalitis) and indirect effects (increased risk of opportunistic bacterial and invasive fungal infections, graft-vs-host disease, delayed engraftment, or graft failure/rejection), as well as increased overall mortality [32, 33].
Two approaches exist to preventing CMV disease in transplant recipients: (1) prophylaxis, whereby antiviral treatment is started prior to viremia; or (2) preemptive therapy, defined as active surveillance for viral replication, with treatment only initiated when CMV viremia is detected. Preemptive therapy has been the preferred approach for preventing CMV disease in hematopoietic stem cell transplant (HSCT) recipients to minimize the toxicities of the available nucleoside analog agents. Although the introduction of preemptive therapy for the management of CMV infection has reduced the incidence of CMV end-organ disease, this approach has remained suboptimal as (1) preemptive therapy is initiated only after patients develop CMV viremia, and any level of viremia is associated with an increased risk of overall mortality; and (2) nucleoside analog inhibitors have toxicities, including myelosuppression, delayed bone marrow engraftment, and nephrotoxicity. Prophylaxis would be a preferred strategy if a drug had reduced toxicity and/or improved potency compared with existing agents. PREVYMIS (letermovir) was developed to meet the unmet medical need for a prophylactic anti-CMV agent with potent activity and a favorable therapeutic index that could improve clinical outcomes in HSCT recipients [34].
PREVYMIS (letermovir) is an inhibitor of the CMV viral terminase that is responsible for the cleavage of newly synthesized CMV DNA into individual unit–length viral genomes. The first terminase inhibitors described for CMV were the halogenated benzimidazole analogs and were shown to eliminate the formation of monomer viral genomes in infected cells [35, 36]. This series is remarkably specific for CMV, as reviewed earlier [37]. While they are nucleoside analogs, they do not require phosphorylation for antiviral activity [38], but rather target components of the viral terminase, including UL56 and UL89 [39]. Letermovir demonstrated potent and selective inhibition of CMV activity in vitro and in preclinical models [40–45].
A series of phase 2 and 3 studies has defined antiviral activity and safety. First, proof of antiviral activity in humans was established in solid organ transplant recipients who had CMV viremia. Second, a phase 2b study demonstrated a dose response and excellent tolerability in the prevention of CMV viremia and/or disease in HSCT recipients. Last, these findings were confirmed in a pivotal randomized, placebo-controlled phase 3 study for the prophylaxis of CMV infection/disease in adult CMV-seropositive recipients of an allogeneic HSCT in the first 3 months posttransplant—a time frame for highest risk of CMV reactivation. Letermovir prophylaxis, started within 28 days and continued until 14 weeks posttransplant, effectively prevented clinically significant CMV infection/disease, with 37.5% of participants on letermovir developing CMV viremia/disease compared to 60.6% participants on placebo at 24 weeks posttransplant (P < .0001). These results demonstrated an approximate 40% relative reduction of CMV infection/disease compared to placebo with a number needed to treat to prevent 1 case of infection/disease of 5 patients. Furthermore, all-cause mortality was decreased by approximately 30% compared to placebo (10.2% letermovir group vs 15.9% placebo group) at week 24 post-HSCT, with a number needed to treat of 20. The mortality benefit of letermovir treatment was maintained through week 48 after transplantation. Letermovir was well tolerated with no evidence of myelotoxicity; participants who received letermovir had a similar time to engraftment compared with those who received placebo [46].
PREVYMIS was approved in the United States by the FDA on 9 November 2017 and by the European Union on 8 January 2018, after accelerated assessment. As the first CMV therapeutic approved since 2001, PREVYMIS marks the beginning of a new era for the prophylaxis of CMV infection and disease in HSCT recipients by enabling the adoption of a new prevention paradigm of CMV management.
DRUGS IN CLINICAL DEVELOPMENT
Maribavir, a benzimidazole L riboside structurally related to the halogenated benzimidazole terminase inhibitors, is a highly specific inhibitor of the CMV UL97 kinase, as reviewed [47, 48]. This molecule exhibits favorable pharmacokinetic properties, is well tolerated, and holds promise for the treatment of CMV infections [49–51]. In a phase 3 study, maribavir-treated patients failed to meet the clinical endpoints [52], possibly due to an inadequate dose. One phase 3 trial recently demonstrated equivalence of maribavir at higher doses compared with ganciclovir; another is ongoing (ClinicalTrials.gov identifiers NCT02931539 and NCT02927067) [53]. The inhibition of UL97 kinase activity by this drug would interfere with the activation of ganciclovir and filociclovir; thus, their concomitant administration would likely reduce the efficacy of the later dugs [54–56]. Clinical studies need to be designed with this issue in mind, but strategies to minimize this effect clearly exist.
The acyclic nucleoside phosphonates, such as cidofovir, have excellent antiviral activity against CMV, but its use is limited by toxicity [57, 58]. Brincidofovir, the new ether lipid ester prodrug of cidofovir [59], exhibits enhanced in vitro efficacy against all the human herpesviruses, including CMV [60], and retained activity against ganciclovir-resistant isolates of CMV having mutations in the UL97 gene. Brincidofovir also exhibits markedly enhanced activity in vitro against a spectrum of DNA viruses, including the herpesviruses, orthopoxviruses, adenoviruses, and polyomaviruses [60–63]. The drug was originally designed to improve the oral bioavailability of cidofovir and indeed proved to be readily absorbed by the oral route in pharmacokinetic studies [64]. More importantly, this modification essentially eliminated its uptake in the kidney by the organic anion transporter, thus reducing its nephrotoxicity and offering a potentially significant advantage over cidofovir [65, 66].
In clinical studies, brincidofovir performed well in a phase 2 trial for CMV infections in stem cell transplant recipients and reduced the number of CMV events, the primary endpoint, at an oral dose of 100 mg twice weekly [67]. In a phase 3, double-blind, placebo-controlled, randomized trial, it did not meet its primary endpoint (above) (ClinicalTrials.gov identifier NCT00411645) [68]. An IV formulation of brincidofovir is in phase 2 development and was designed to reduce the incidence and severity of diarrhea and increase levels of the drug in other important compartments.
THERAPIES IN EARLY DEVELOPMENT
Overview of Small Molecules
Promising molecules in early stages of development have been reported. Distinct acyclic nucleoside phosphonate analogs, such as diaminopyrimidine derivatives, have broad-spectrum antiviral activity and may exhibit reduced toxicity [69]. Nonnucleoside inhibitors of the DNA polymerase also represent an important class of potent compounds, providing promise as broad-spectrum inhibitors of the herpesviruses without liabilities associated with nucleoside inhibitors [70]. The 4-oxo-dihydroquinoline derivatives provide an example of highly potent compounds with activity against all the human herpesviruses [71, 72] and have the advantage of remaining active against ganciclovir-resistant isolates of CMV [73].
The first terminase inhibitors described for CMV were the halogenated benzimidazole analogs and were shown to eliminate the formation of monomer viral genomes in infected cells [35, 36]. This series is remarkably specific for CMV, as reviewed previously [37]. Although nucleoside analogs, they do not require phosphorylation for antiviral activity [38], but rather target components of the viral terminase including UL56 and UL89 [39]. An advanced candidate is GW275175X, which inhibits the terminase complex [74] and retains good activity against CMV [75, 76].
Filociclovir, another acyclic nucleoside, is in clinical development for the treatment and/or prevention of CMV, human herpesvirus 6, and adenovirus. Filociclovir demonstrated good in vitro and in vivo potency against human CMV. Filociclovir has a dual mechanism of action, inhibiting both the CMV UL54 DNA polymerase and the CMV UL97 kinase while retaining activity against ganciclovir-resistant CMV, despite sharing a similar mechanism of action [55]. In addition, filociclovir is converted more efficiently in infected cells to the active triphosphate form than ganciclovir [77].
Oral bioavailability of filociclovir following single oral doses in rats and dogs was 22%–46% and 70%–91%, respectively. In safety pharmacology studies, filociclovir did not cause significant changes in respiratory, cardiovascular, or CNS parameters at doses up to 100 mg/kg. Filociclovir was not considered genotoxic. Toxicology studies established target organs similar to other drugs in the class: kidney, bone marrow, gastrointestinal tract, and testes [78].
The first-in-human, phase 1a clinical study (ClinicalTrials.gov identifier NCT01433835) [78] in healthy volunteers evaluated the safety and pharmacokinetics of filociclovir in single ascending oral doses. Forty-eight healthy volunteers (3 male, 45 female) were enrolled and completed the study. No serious adverse events were reported. The most common adverse events were dry mouth and headache. Maximum plasma concentrations (Cmax) and area under the curve values increased following filociclovir doses of 35–1000 mg and decreased at 1350 mg; exposure was less than dose proportional. Cmax values were 281.5–1175 ng/mL and area under the curve at last assessment (AUClast) values were 946.2–5696 ng × hour/mL following administration of 35, 100, 350, 700, 1000, and 1350 mg oral doses.
A phase 1b clinical study (ClinicalTrials.gov identifier NCT02454699; Rouphael et al, unpublished data) has been completed in healthy volunteers and evaluated the safety and pharmacokinetics following administration of 7 oral daily doses (100, 350, or 750 mg) of filociclovir, and the results are being analyzed.
Safety data support advancement of filociclovir following work to improve formulation. Phase 2 clinical trials will be undertaken [69–72, 77–81].
Application of Monoclonal Antibodies for Therapeutic Purposes
Administering antibodies as an alternative to vaccines has been used in pregnancy to treat blood group incompatibilities to and prevent rubella, hepatitis, varicella, respiratory syncytial virus, and measles [82]. Advantages include more reliable and rapid achievement of efficacious plasma concentrations. Passive human immunoglobulin (HIG) immunization, which had suggested value for the prevention of congenital CMV infection (2005–2012), became controversial when a 2014 study failed to prevent fetal infection [83]. Subsequent study of HIG pharmacokinetics established unexpectedly rapid clearance (11 days). Based on this observation, a phase 2 trial was conducted [84] in which 40 pregnant women with a primary CMV infection (9.6 weeks median gestational age) were treated with HIG biweekly rather than monthly as in previous trials. Transmission before amniocentesis at 20 weeks occurred in only 1 subject (2.5%), with 2 additional subjects infected after treatment. Including all 3 cases, the transmission rate was 7.5%, with no infected neonate symptomatic at birth. The results are significant (P < .0001) when compared to untreated historical controls matched for first trimester seroconversion with amniocentesis at 20 weeks (108 pregnancies), for whom the transmission rate was 35.2% and incidence of sequelae was 14%.
HIG is also effective in solid organ transplant. A meta-analysis of 11 randomized trials (N = 698; median follow-up of 12 months) concluded that HIG prophylaxis was associated with reduced CMV disease and improved survival [85]. For pediatric HSTC, CMV infection at 1 year was 13.4% for HIG-treated children vs 44.4% without HIG (P = .001) [86].
Replicating the activity of HIG with a monoclonal antibody (mAb) offers the prospect of improved consistency in manufacturing, lower infusion volume, improved pharmacokinetics, and reduced risk of off-target reactivity leading to toxicity. Moreover, HIG batches are not rigorously evaluated for titer to particular antigens, which may contribute to variability in efficacy. Antibodies to the CMV pentameric complex are abundant in HIG [87], although immunodominance is not necessarily correlated with neutralizing activity. The major clinical effort targeting the pentameric complex has been a mixture of 2 mAbs (RG7667), which showed promise in a phase 2 trial in renal transplant recipients by delaying onset of CMV excretion [88] but is no longer being developed, despite modest neutralizing activity.
A drawback to targeting the pentameric complex is that antibodies against it fail to neutralize infection of fibroblasts [89], whose ubiquitous distribution may be important for disease progression [90]. Antibodies to the gB fusion protein, which is essential for entry in all cell types, are also part of the natural immune response, and the adjuvanted gB/MF59 vaccine showed efficacy in a substantial number of subjects, both in transplant and congenital indications [91]. The first clinically tested mAb against gB (Theraclone: TCN-202) was well tolerated in a phase 1 trial, but development was discontinued. CSJ148 (Novartis) is a mixture of 2 mAbs (targets: gB [AD-4], an immunodominant region, and the pentameric complex) [92]. In a phase 1 trial, CSJ148 was well tolerated, but it too is no longer being advanced. Both AD-4 and the similarly immunodominant AD-1 are more variable than the AD-2 site I (which is also the least immunogenic site). TRL345 is a native human mAb that has 10-fold higher affinity than TCN-202 for this highly conserved epitope, and 6.5-fold higher potency in vitro than the anti-gB component of CSJ148 [93]. High affinity is important for the mAbs to prevent transmission by remaining bound to the virus following transcytosis across the placenta via the neonatal Fc receptor [94]. Furthermore, high affinity can prevent displacement of neutralizing antibodies to AD-2 site I by nonneutralizing antibodies to the adjacent site II present in approximately 25% of anti-CMV human serum samples [95]. Elevated titers to the TRL345 epitope is associated with reduced risk of congenital transmission [96].
TRL345 showed potent antiviral activity against 15 primary clinical isolates of diverse genotypes [97]. For several clinical strains, potency in a wide range of cell types was 10- to 50-fold better than HIG. The absence of a well-validated animal model has put a premium on studies using human placental tissue as a model. CMV replicates in trophoblast progenitor cells and impairs their capacity to self-renew and differentiate. TRL345 was approximately 10-fold more potent than HIG in these cells [98]. In an established ex vivo model in which explants of first-trimester human placenta are grown on Matrigel, TRL345 was highly effective at reducing infection [97].
TRL345 was also compared to a high-affinity antipentameric complex antibody cloned from published sequence data (mAb 1F11 binding to UL128-131A). The 1F11 mAb had more potent neutralizing activity on endothelial cells than TRL345, but it provided no protection against infection of smooth muscle cells, placental fibroblasts, or trophoblast progenitor cells [97]. Formation of multinucleated syncytia is a characteristic phenotype of CMV in vivo. In a study of syncytial spread in vitro, TRL345 was the most effective antibody in a comprehensive panel of 28 antibodies, both polyclonal and monoclonal, targeting all major CMV virion glycoproteins [99]. An additional observation from this study was that antibodies targeting epitopes in gH or gH/gL exhibited strain and cell type dependence. By contrast, the frequency of mutations in the TRL345 epitope is approximately 10-fold lower than for other envelope glycoprotein sites [100], despite continuous selective pressure (antibodies to the epitope represent approximately 1% of the anti-CMV activity in serum), suggesting that escape will be rare. TRL345 has been expressed in stably transformed Chinese hamster ovary (CHO) cells at 1.8 g/L. The no-observed-adverse-effect level in a 28-day toxicology study in Sprague-Dawley rats was 150 mg/kg/dose. At 15 mg/kg, the projected half-life in humans is 21 days. An investigational new drug application is expected to be filed in 2020.
ROLE OF SIRTUINS IN THE TREATMENT OF CMV INFECTIONS
Direct-Acting Antivirals Are Limited by Their Spectrum of Antiviral Effectiveness
Because direct-acting antivirals exert their therapeutic effect through direct interactions with a viral protein, they are also limited by drug resistance secondary to acquisition of viral mutations disrupting the molecular interaction of the drug with the targeted viral protein. Drug-resistant clinical isolates of CMV have been identified for all marketed CMV antivirals: ganciclovir/valganciclovir, foscarnet, cidofovir, and letermovir [101]. A single mutation at UL56 C325 confers absolute resistance to letermovir with minimal impact on viral fitness [102, 103].
Host-targeted antivirals (HTAs) are directed against the host-cell processes upon which viruses are dependent. Compared to DAAs, HTAs have the potential to reduce or eliminate viral resistance, demonstrate broad-spectrum effectiveness, and provide therapeutic utility in areas of unmet medical need. First-generation HTAs, such as interferons, broadly activate the host’s innate and adaptive immune responses (eg, hepatitis B and C) [104]. The clinical use of interferons is limited by toxicity. Next-generation HTAs are small molecules that target host sirtuin proteins. By modulating sirtuin-enzyme activity, these leads are predicted to reduce viral replication by restoring host-cell metabolism and intrinsic immunity within the infected cell.
The 7 human sirtuins (SIRT1–7) are nicotinamide adenine dinucleotide (NAD+)–dependent deacylases that regulate cellular metabolism and gene activity by posttranslationally removing acyl groups from target proteins [105]. In addition to the deacetylase activity of all 7 SIRTs, SIRT4, SIRT5, and SIRT6 can also remove lipoyl, methylglutaryl, hydroxymethylglutaryl, and 3-methylglutaconyl moieties [106, 107]; malonyl and succinyl moieties; and myristoyl moieties, respectively. The extent to which proteins in the cell are acylated by the distinct fatty-acyl chains is dependent on acyl–coenzyme A metabolism, which can be selectively regulated [108]. SIRTs localize to the nucleus (SIRT1, SIRT2, SIRT6), nucleolus (SIRT7), cytoplasm (SIRT2), and mitochondria (SIRT3, SIRT4, SIRT5). The NAD+ requirement for the deacylation reaction ties the activity of sirtuins to the metabolic capacity of the cell. As such, the emerging role of sirtuins is as sensors linking the metabolite profile of the cell to cellular signal transduction, as reviewed elsewhere [109]. Sirtuin-mediated deacylation of target proteins impacts numerous cell functions, including metabolism, cell cycle, apoptosis, stress response, DNA repair, and gene expression—all functions known to affect virus growth.
Sirtuins control cellular processes that impact the growth of many different viruses and function as elements of intrinsic immunity [110]. Modulation of sirtuins can restrict the growth of both RNA and DNA viruses. Viruses depend on host cell metabolism for energy, metabolic precursors for viral components, and cellular organization for replication, maturation, and dissemination. Not surprisingly, diverse, intracellular pathogens have been shown to directly interact with sirtuins in order to co-opt host-cell metabolism or epigenetic mechanisms into supporting the pathogen. These include Salmonella typhimurium [111], Listeria monocytogenes [112, 113], Leishmania infantum [114], Kaposi sarcoma–associated herpesvirus [115], HIV [116], and hepatitis B virus (HBV) [117, 118]. In particular, recent publications demonstrate the potential utility of SIRT2 inhibitors as antibacterial (Listeria) and antiviral (HBV) agents [112–117].
A small-molecule screen was carried out on a library of approximately 13 000 compounds to identify modulators of SIRT1, SIRT2, SIRT3, and/or SIRT6 deacetylase activity [119]. Belying the importance of metabolism on productive viral replication and the role of sirtuins as mediators of intrinsic immunity, among the 85 hits validated in the primary assay as significantly inhibiting or activating 1 or more of the 4 sirtuins tested, more than two-thirds proved to be antiviral with 50% inhibitory concentration <25 µM in a secondary CMV viral growth assay. A medicinal chemistry campaign was performed with >400 molecules synthesized to improve the antiviral activity of a SIRT2 inhibitor identified in the screen (designated FH-003, a 6-{1-[2-(1,2,3,4-tetrahydroisoquinolin-2-yl)-1,3-thiazol-5-yl]ethyl} quinoxaline). Table 3 shows approximately 100-fold improvement in anti-CMV activity with the current lead as well as broad-spectrum activity against multiple DNA and RNA viruses including BK virus, JC virus, influenza A and B, and respiratory syncytial virus. As would be expected for targeting an evolutionally conserved host protein, the broad-spectrum antiviral activity was measured in cells derived from multiple species, including human, monkey, and dog—all demonstrating a therapeutic index >200.
Table 3.
Broad-Spectrum Antiviral Effectiveness of Small-Molecule SIRT2 Inhibitors
| Compound | DNA Viruses | RNA Viruses | ||||||||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| HCMV TB40 | BK Virus | JC Virus | Influenza A A/PR/8/1934 H1N1 | Influenza A A/WSN/1933 H1N1 Oseltamivir | Influenza A A/CA/07/2000 H1N1 | Influenza A A/Perth/16/2009 H3N2 | Influenza B B/FL/4/2006 Yamagata | RSV Long Strain | ||||||||||
| Human MRC5 | Human HFF | Monkey Cos7 | Canine MDCK | Canine MDCK | Human HNBEc | Human HNBEc | Canine MDCK | Human MRC5 | ||||||||||
| IC50a | SIb | IC50 | SI | IC50 | SI | IC50 | SI | IC50 | SI | IC50 | SI | IC50 | SI | IC50 | SI | IC50 | SI | |
| FH-003 | 14 | … | … | … | … | … | … | … | … | … | … | … | … | … | … | … | … | … |
| cpd1 | 7.2 | … | … | … | … | … | 7.9 | … | … | … | … | … | … | … | … | … | … | … |
| cpd2 | 1.4 | >18 | 0.85 | >1592 | 0.05 | >382 | 9.1 | … | … | … | … | … | … | … | … | … | … | … |
| Lead cpd3 | 0.45 | >56 | … | … | … | … | 0.76 | >12 | 2.5 | >10 | 1.2 | >82 | 2.1 | >48 | 1.2 | >21 | 6.7 | >3.7 |
| Ganciclovir | 1.4 | … | … | … | … | … | … | … | … | … | … | … | … | … | … | … | … | … |
| Oseltamivir | … | … | … | … | … | … | 0.02 | >69 | 9.0 | … | … | … | … | … | >25 | … | … | … |
| Ribavirin | … | … | … | … | … | … | … | … | … | … | 0.71 | >141 | 0.73 | >138 | … | … | 16.1 | >3.1 |
| Cidofovir | … | … | 4.4 | … | 3.8 | … | … | … | … | … | … | … | … | … | … | … | … | … |
Host-targeted SIRT2 inhibitors FH-003, cpd1, cpd2, cpd3, and direct-acting antivirals tested on indicated virus in indicated cell line at a multiplicity of infection of 0.1.
Abbreviations: HCMV, human cytomegalovirus; HFF, human foreskin fibroblasts; HNBE, human normal bronchial epithelial cekks; IC50, 50% inhibitory concentration; MDCK, Madin-Darby canine kidney cells; RSV, respiratory syncytial virus; SI, selectivity index.
aIC50 of viral spread given in µM; values in bold indicate 90% inhibitory concentration instead of IC50 measurement.
bSI indicates the 50% cell cytotoxic concentration/IC50.
cAssays performed by the National Institute of Allergy and Infectious Diseases, Division of Microbiology and Infectious Diseases, Resources for Researchers, in vitro testing.
Host targeting predicts favorable antiviral properties, including a high barrier to development of resistance and synergy with known DAAs. Influenza A, a rapidly dividing RNA virus with a higher mutation rate and shorter doubling time than most DNA viruses, was used in these studies [120]. A complete absence of acquired drug resistance was observed after 10 influenza A passages in culture in the presence of the lead SIRT2 inhibitor; in contrast, resistance to the DAA oseltamivir was evident after 2 passages. In addition, HTA plus DAA synergy was demonstrated by testing the interaction between the lead SIRT2 inhibitor and oseltamivir across a wide spectrum of possible drug combinations. The degree of combined viral inhibition ranged from 50% to 90%. Using Chou-Talalay modeling [121], the combination index was 0.68 combination index, or overall combination index < 1, indicating synergy across all relative combinations used.
The mechanism of SIRT2 inhibition on microbe growth is pathogen specific. Infection by Listeria monocytogenes induces subcellular localization of SIRT2 from the cytoplasm to the nucleus where epigenetic effects manifest as repression of a significant program of host genes [116]. HBV replication upregulates protein expression of SIRT2 that acts in a feed-forward fashion to increase HBV transcription and replication [113, 114]. Conversely, parainfluenza virus type 3 (HPIV3) reportedly forms viral inclusion bodies dependent on acetylated tubulin for efficient fusion; HDAC6 and SIRT2-mediated deacetylation of α-tubulin can restrict HPIV3 inclusion body fusion [122]. The mechanism of SIRT2 inhibition on human CMV growth is under investigation. Pathways downstream of SIRT2 posttranslational modifications comprising potential interaction with CMV biology include c-MYC, p53, CDK9, and AKT. The lead SIRT2 inhibitor can potently down-regulate c-MYC protein known to accumulate during CMV infection, presumably to induce efficient glutamine utilization required for viral nucleic acids and lipids [123]. p53 is a direct target of SIRT2 deacetylation [124, 125]. SIRT2 inhibition leads to hyperacetylation and activation of p53, whose transcriptional regulatory activity is known to be substantially inhibited by CMV [126, 127]. SIRT2 also activates CDK9 [128] and AKT kinases [129, 130], known to block the replication [131] or persistence [132], respectively, of CMV.
The current lead in the FH-003 series demonstrates favorable pharmacokinetic properties in mice including 100% oral bioavailability, 1:1 plasma:tissue distribution, a half-life approaching 5 hours, development-amenable in vitro pharmaceutical properties, and manufacturing-scalable synthesis. Experiments are in progress to demonstrate effectiveness in animal models of infection with promising preliminary results. Successful development of HTAs, including sirtuin modulators, will provide an arsenal of agents to use as stand-alone or combination therapies with existing DAAs, foretelling significant patient benefits of improved control of viral load, minimized adverse events, and reduced emergence of viral resistance, as has proven to be the case with HIV combination therapies. Given the intimate relationship between viral infection and host metabolism, broadly effective sirtuin-targeted antivirals may also address the unmet need of infection by pandemic viruses yet to be identified.
In conclusion, human CMV infections remain a considerable cause of morbidity and mortality. As the number of immunosuppressed individuals increases, with organ transplantation becoming more common, together with the mandate of therapy for congenitally infected infants, improved outcome is essential [22]. Newer approaches should explore combination therapies.
Notes
Presented in part: Cytomegalovirus Infection: Advancing Strategies for Prevention and Treatment, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, 4–6 September 2018.
Financial support. This work was supported by the National Institute of Allergy and Infectious Diseases (NIAID) (contract numbers N0-AI-65306 and N0-AI-30025 to E. A., D. K., M. P., and R. W.); the National Institutes of Health (NIH) (contract number AI054135 to T. B., J. B., and I. H.); task orders under the NIAID (contract number HHSN2722001100016I (to T. B., J. B., and I. H.); the Vaccine and Treatment Evaluation Unit Program (contract number HHSN272201300018I to T. B., J. B., and I. H.); and the NIAID ((contract number 2SB1AI02396-06 to L. K.) and (contract numbers 1R43AI110048-01, 1R43AI114079-01, and 1R44AI122488-01 to L. C.)).
Supplement sponsorship. This supplement was sponsored by NIAID and NICHD.
Potential conflicts of interest. T. B., J. B., and I. H. have received grants and nonfinancial support from the NIH during the conduct of the study and are employees of Microbiotix, Inc, the company currently developing filociclovir. L. C.’s spouse serves on the board for or advises numerous nonprofit and for-profit organizations; ones with antiviral focus include the Hepatitis B Foundation (Board of Directors) and FORGE Life Science, LLC (Founder). L. K. has received grants from NIAID to Trellis Bioscience, LLC, and is an employee of Trellis, which has been awarded US patents for which he is an inventor (patent numbers 9017668, 9688744, and 10030069). D. K. has received grants from the NIH during the conduct of the study. R. L. is an employee of Merck. R. W. reports grants from the NIH during the conduct of the study. All other authors report no potential conflicts of interest.
All authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.
References
- 1. Morton CC, Nance WE. Newborn hearing screening—a silent revolution. N Engl J Med 2006; 354:2151–64. [DOI] [PubMed] [Google Scholar]
- 2. Fowler KB, McCollister FP, Dahle AJ, Boppana S, Britt WJ, Pass RF. Progressive and fluctuating sensorineural hearing loss in children with asymptomatic congenital cytomegalovirus infections. J Pediatr 1997; 130:624–30. [DOI] [PubMed] [Google Scholar]
- 3. Fowler KB, Dahle AJ, Boppana SB, Pass RF. Newborn hearing screening: will children with hearing loss caused by congenital cytomegalovirus infection be missed? J Pediatr 1999; 135:60–4. [DOI] [PubMed] [Google Scholar]
- 4. Fowler KB, Boppana SB. Congenital cytomegalovirus (CMV) infection and hearing deficit. J Clin Virol 2006; 35:226–31. [DOI] [PubMed] [Google Scholar]
- 5. Elek SD, Stern H. Development of a vaccine against mental retardation caused by cytomegalovirus infection in utero. Lancet 1974; 1:1–5. [DOI] [PubMed] [Google Scholar]
- 6. Kenneson A, Cannon MJ. Review and meta-analysis of the epidemiology of congenital cytomegalovirus (CMV) infection. Rev Med Virol 2007; 17:253–76. [DOI] [PubMed] [Google Scholar]
- 7. Dollard SC, Grosse SD, Ross DS. New estimates of the prevalence of neurological and sensory sequelae and mortality associated with congenital cytomegalovirus infection. Rev Med Virol 2007; 17:355–63. [DOI] [PubMed] [Google Scholar]
- 8. Manicklal S, Emery VC, Lazzarotto T, Boppana SB, Gupta RK. The “silent” global burden of congenital cytomegalovirus. Clin Microbiol Rev 2013; 26:86–102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Williamson WD, Desmond MM, LaFevers N, Taber LH, Catlin FI, Weaver TG. Symptomatic congenital cytomegalovirus. Disorders of language, learning, and hearing. Am J Dis Child 1982; 136:902–5. [DOI] [PubMed] [Google Scholar]
- 10. Dahle AJ, Fowler KB, Wright JD, Boppana SB, Britt WJ, Pass RF. Longitudinal investigation of hearing disorders in children with congenital cytomegalovirus. J Am Acad Audiol 2000; 11:283–90. [PubMed] [Google Scholar]
- 11. Boppana SB, Ross SA, Fowler KB. Congenital cytomegalovirus infection: clinical outcome. Clin Infect Dis 2013; 57(Suppl 4):S178–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Centers for Disease Control and Prevention. U.S. public health impact of congenital cytomegalovirus infection. MMWR Surveill Summ 1992; 41:35–9. [Google Scholar]
- 13. Fowler KB, Ross SA, Shimamura M, et al. Racial and ethnic differences in the prevalence of congenital cytomegalovirus infection. J Pediatr 2018; 200:196–201.e1. [DOI] [PubMed] [Google Scholar]
- 14. Grosse SD, Ross DS, Dollard SC. Congenital cytomegalovirus (CMV) infection as a cause of permanent bilateral hearing loss: a quantitative assessment. J Clin Virol 2008; 41:57–62. [DOI] [PubMed] [Google Scholar]
- 15. Yow MD, Demmler GJ. Congenital cytomegalovirus disease—20 years is long enough. N Engl J Med 1992; 326:702–3. [DOI] [PubMed] [Google Scholar]
- 16. Arvin AM, Fast P, Myers M, Plotkin S, Rabinovich R; National Vaccine Advisory Committee Vaccine development to prevent cytomegalovirus disease: report from the National Vaccine Advisory Committee. Clin Infect Dis 2004; 39:233–9. [DOI] [PubMed] [Google Scholar]
- 17. US Department of Labor. Bureau of Labor Statistics CPI inflation calculator. 2015. http://www.bls.gov/data/inflation_calculator.htm. Accessed 14 October 2019. [Google Scholar]
- 18. Cannon MJ, Griffiths PD, Aston V, Rawlinson WD. Universal newborn screening for congenital CMV infection: what is the evidence of potential benefit? Rev Med Virol 2014; 24:291–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Zhou XJ, Gruber W, Demmler G, et al. Population pharmacokinetics of ganciclovir in newborns with congenital cytomegalovirus infections. NIAID Collaborative Antiviral Study Group. Antimicrob Agents Chemother 1996; 40:2202–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Acosta EP, Brundage RC, King JR, et al. National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group Ganciclovir population pharmacokinetics in neonates following intravenous administration of ganciclovir and oral administration of a liquid valganciclovir formulation. Clin Pharmacol Ther 2007; 81:867–72. [DOI] [PubMed] [Google Scholar]
- 21. Kimberlin DW, Acosta EP, Sánchez PJ, et al. National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group Pharmacokinetic and pharmacodynamic assessment of oral valganciclovir in the treatment of symptomatic congenital cytomegalovirus disease. J Infect Dis 2008; 197:836–45. [DOI] [PubMed] [Google Scholar]
- 22. Kimberlin DW, Jester PM, Sánchez PJ, et al. National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group Valganciclovir for symptomatic congenital cytomegalovirus disease. N Engl J Med 2015; 372:933–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Kimberlin DW, Lin CY, Sánchez PJ, et al. National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group Effect of ganciclovir therapy on hearing in symptomatic congenital cytomegalovirus disease involving the central nervous system: a randomized, controlled trial. J Pediatr 2003; 143:16–25. [DOI] [PubMed] [Google Scholar]
- 24. Whitley RJ, Cloud G, Gruber W, et al. Ganciclovir treatment of symptomatic congenital cytomegalovirus infection: results of a phase II study. National Institute of Allergy and Infectious Diseases Collaborative Antiviral Study Group. J Infect Dis 1997; 175:1080–6. [DOI] [PubMed] [Google Scholar]
- 25. Trang JM, Kidd L, Gruber W, et al. Linear single-dose pharmacokinetics of ganciclovir in newborns with congenital cytomegalovirus infections. NIAID Collaborative Antiviral Study Group. Clin Pharmacol Ther 1993; 53:15–21. [DOI] [PubMed] [Google Scholar]
- 26. Oliver SE, Cloud GA, Sánchez PJ, et al. National Institute of Allergy, Infectious Diseases Collaborative Antiviral Study Group Neurodevelopmental outcomes following ganciclovir therapy in symptomatic congenital cytomegalovirus infections involving the central nervous system. J Clin Virol 2009; 46(Suppl 4):S22–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Genentech. Valganciclovir package insert. https://www.gene.com/download/pdf/valcyte_prescribing.pdf. Accessed 14 October 2019. [Google Scholar]
- 28. American Academy of Pediatrics. Cytomegalovirus infection. In: Kimberlin D, Brady M, Jackson M, Long S, eds. Red Book: 2015 Report of the Committee on Infectious Diseases. 30th ed Elk Grove Village, IL: American Academy of Pediatrics, 2015:317–22. [Google Scholar]
- 29. American Academy of Pediatrics. Cytomegalovirus infection. In: Kimberlin D, Brady M, Jackson M, Long S, eds. Red Book: 2018 Report of the Committee on Infectious Diseases. 31st ed Elk Grove Village, IL: American Academy of Pediatrics, 2018:310–7. [Google Scholar]
- 30. Marsico C, Aban I, Kuo H, et al. Blood viral load in symptomatic congenital cytomegalovirus infection. J Infect Dis 2019;219:1398– 1406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Kimberlin DW, Jester PM, Sanchez PJ, et al. Valganciclovir for symptomatic congenital cytomegalovirus disease. N Engl J Med 2015; 372:933–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Marty FM, Ljungman P, Chemaly RF, et al. Letermovir prophylaxis for cytomegalovirus in hematopoietic-cell transplantation. N Engl J Med 2017; 377:2433–44. [DOI] [PubMed] [Google Scholar]
- 33. Chemaly RF, Ullmann AJ, Stoelben S, et al. AIC246 Study Team Letermovir for cytomegalovirus prophylaxis in hematopoietic-cell transplantation. N Engl J Med 2014; 370:1781–9. [DOI] [PubMed] [Google Scholar]
- 34. Kim ES. Letermovir: first global approval. Drugs 2018; 78:147–52. [DOI] [PubMed] [Google Scholar]
- 35. Underwood MR, Harvey RJ, Stanat SC, et al. Inhibition of human cytomegalovirus DNA maturation by a benzimidazole ribonucleoside is mediated through the UL89 gene product. J Virol 1998; 72:717–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Townsend LB, Devivar RV, Turk SR, Nassiri MR, Drach JC. Design, synthesis, and antiviral activity of certain 2,5,6-trihalo-1-(beta-D-ribofuranosyl)benzimidazoles. J Med Chem 1995; 38:4098–105. [DOI] [PubMed] [Google Scholar]
- 37. Drach J, Townsend L, Bogner E.. Benzimidazoled-ribonucleosides as antiviral agents that target HCMV terminase. Dordrecht, Netherlands: Springer, 2006. [Google Scholar]
- 38. Krosky PM, Borysko KZ, Nassiri MR, et al. Phosphorylation of beta-D-ribosylbenzimidazoles is not required for activity against human cytomegalovirus. Antimicrob Agents Chemother 2002; 46:478–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Krosky PM, Underwood MR, Turk SR, et al. Resistance of human cytomegalovirus to benzimidazole ribonucleosides maps to two open reading frames: UL89 and UL56. J Virol 1998; 72:4721–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Lischka P, Hewlett G, Wunberg T, et al. In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246. Antimicrob Agents Chemother 2010; 54:1290–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Goldner T, Hewlett G, Ettischer N, Ruebsamen-Schaeff H, Zimmermann H, Lischka P. The novel anticytomegalovirus compound AIC246 (letermovir) inhibits human cytomegalovirus replication through a specific antiviral mechanism that involves the viral terminase. J Virol 2011; 85:10884–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Kaul DR, Stoelben S, Cober E, et al. First report of successful treatment of multidrug-resistant cytomegalovirus disease with the novel anti-CMV compound AIC246. Am J Transplant 2011; 11:1079–84. [DOI] [PubMed] [Google Scholar]
- 43. Stoelben S, Arns W, Renders L, et al. Preemptive treatment of cytomegalovirus infection in kidney transplant recipients with letermovir: results of a phase 2a study. Transpl Int 2014; 27:77–86. [DOI] [PubMed] [Google Scholar]
- 44. Marshall WL, McCrea JB, Macha S, et al. Pharmacokinetics and tolerability of letermovir coadministered with azole antifungals (posaconazole or voriconazole) in healthy subjects. J Clin Pharmacol 2018; 58:897–904. [DOI] [PubMed] [Google Scholar]
- 45. Kropeit D, von Richter O, Stobernack HP, Rübsamen-Schaeff H, Zimmermann H. Pharmacokinetics and safety of letermovir coadministered with cyclosporine A or tacrolimus in healthy subjects. Clin Pharmacol Drug Dev 2018; 7:9–21. [DOI] [PubMed] [Google Scholar]
- 46. Green ML, Leisenring W, Xie H, et al. Cytomegalovirus viral load and mortality after haemopoietic stem cell transplantation in the era of pre-emptive therapy: a retrospective cohort study. Lancet Haematol 2016; 3:e119–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Biron K. Maribavir: a promising new antiherpes therapeutic agent. Dordrecht, Netherlands: Springer, 2006. [Google Scholar]
- 48. Prichard MN. Function of human cytomegalovirus UL97 kinase in viral infection and its inhibition by maribavir. Rev Med Virol 2009; 19:215–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Koszalka GW, Johnson NW, Good SS, et al. Preclinical and toxicology studies of 1263W94, a potent and selective inhibitor of human cytomegalovirus replication. Antimicrob Agents Chemother 2002; 46:2373–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Lalezari JP, Aberg JA, Wang LH, et al. Phase I dose escalation trial evaluating the pharmacokinetics, anti-human cytomegalovirus (HCMV) activity, and safety of 1263W94 in human immunodeficiency virus-infected men with asymptomatic HCMV shedding. Antimicrob Agents Chemother 2002; 46:2969–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Ma JD, Nafziger AN, Villano SA, Gaedigk A, Bertino JS Jr. Maribavir pharmacokinetics and the effects of multiple-dose maribavir on cytochrome P450 (CYP) 1A2, CYP 2C9, CYP 2C19, CYP 2D6, CYP 3A, N-acetyltransferase-2, and xanthine oxidase activities in healthy adults. Antimicrob Agents Chemother 2006; 50:1130–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Marty FM, Ljungman P, Papanicolaou GA, et al. Maribavir prophylaxis for prevention of cytomegalovirus disease in recipients of allogeneic stem-cell transplants: a phase 3, double-blind, placebo-controlled, randomised trial. Lancet Infect Dis 2011; 11:284–92. [DOI] [PubMed] [Google Scholar]
- 53. Maertens J, Cordonnier C, Jaksch P, et al. Maribavir for preemptive treatment of cytomegalovirus reactivation. N Engl J Med 2019; 381:1136–47. [DOI] [PubMed] [Google Scholar]
- 54. Evers DL, Komazin G, Shin D, Hwang DD, Townsend LB, Drach JC. Interactions among antiviral drugs acting late in the replication cycle of human cytomegalovirus. Antiviral Res 2002; 56:61–72. [DOI] [PubMed] [Google Scholar]
- 55. James SH, Hartline CB, Harden EA, et al. Cyclopropavir inhibits the normal function of the human cytomegalovirus UL97 kinase. Antimicrob Agents Chemother 2011; 55:4682–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Chou S, Marousek GI. Maribavir antagonizes the antiviral action of ganciclovir on human cytomegalovirus. Antimicrob Agents Chemother 2006; 50:3470–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. De Clercq E, Holý A. Acyclic nucleoside phosphonates: a key class of antiviral drugs. Nat Rev Drug Discov 2005; 4:928–40. [DOI] [PubMed] [Google Scholar]
- 58. De Clercq E. Selective anti-herpesvirus agents. Antivir Chem Chemother 2013; 23:93–101. [DOI] [PubMed] [Google Scholar]
- 59. Beadle JR, Hartline C, Aldern KA, et al. Alkoxyalkyl esters of cidofovir and cyclic cidofovir exhibit multiple-log enhancement of antiviral activity against cytomegalovirus and herpesvirus replication in vitro. Antimicrob Agents Chemother 2002; 46:2381–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Williams-Aziz SL, Hartline CB, Harden EA, et al. Comparative activities of lipid esters of cidofovir and cyclic cidofovir against replication of herpesviruses in vitro. Antimicrob Agents Chemother 2005; 49:3724–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Keith KA, Wan WB, Ciesla SL, Beadle JR, Hostetler KY, Kern ER. Inhibitory activity of alkoxyalkyl and alkyl esters of cidofovir and cyclic cidofovir against orthopoxvirus replication in vitro. Antimicrob Agents Chemother 2004; 48:1869–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Hartline CB, Gustin KM, Wan WB, et al. Ether lipid-ester prodrugs of acyclic nucleoside phosphonates: activity against adenovirus replication in vitro. J Infect Dis 2005; 191:396–9. [DOI] [PubMed] [Google Scholar]
- 63. Randhawa P, Farasati NA, Shapiro R, Hostetler KY. Ether lipid ester derivatives of cidofovir inhibit polyomavirus BK replication in vitro. Antimicrob Agents Chemother 2006; 50:1564–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Ciesla SL, Trahan J, Wan WB, et al. Esterification of cidofovir with alkoxyalkanols increases oral bioavailability and diminishes drug accumulation in kidney. Antiviral Res 2003; 59:163–71. [DOI] [PubMed] [Google Scholar]
- 65. Tippin TK, Morrison ME, Brundage TM, Momméja-Marin H. Brincidofovir is not a substrate for the human organic anion transporter 1: a mechanistic explanation for the lack of nephrotoxicity observed in clinical studies. Ther Drug Monit 2016; 38:777–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66. Hostetler KY. Alkoxyalkyl prodrugs of acyclic nucleoside phosphonates enhance oral antiviral activity and reduce toxicity: current state of the art. Antiviral Res 2009; 82:A84–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67. Marty FM, Winston DJ, Rowley SD, et al. CMX001-201 Clinical Study Group CMX001 to prevent cytomegalovirus disease in hematopoietic-cell transplantation. N Engl J Med 2013; 369:1227–36. [DOI] [PubMed] [Google Scholar]
- 68.Marty FM, Winston DJ, Chemaly RF, et al. A randomized, double-blind, placebo-controlled phase 3 trial of oral brincidofovir for cytomegalovirus prophylaxis in allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2019; 25:369–381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. De Clercq E, Andrei G, Balzarini J, et al. Antiviral potential of a new generation of acyclic nucleoside phosphonates, the 6-[2-(phosphonomethoxy)alkoxy]-2,4-diaminopyrimidines. Nucleosides Nucleotides Nucleic Acids 2005; 24:331–41. [DOI] [PubMed] [Google Scholar]
- 70. Wathen MW. Non-nucleoside inhibitors of herpesviruses. Rev Med Virol 2002; 12:167–78. [DOI] [PubMed] [Google Scholar]
- 71. Brideau RJ, Knechtel ML, Huang A, et al. Broad-spectrum antiviral activity of PNU-183792, a 4-oxo-dihydroquinoline, against human and animal herpesviruses. Antiviral Res 2002; 54:19–28. [DOI] [PubMed] [Google Scholar]
- 72. Oien NL, Brideau RJ, Hopkins TA, et al. Broad-spectrum antiherpes activities of 4-hydroxyquinoline carboxamides, a novel class of herpesvirus polymerase inhibitors. Antimicrob Agents Chemother 2002; 46:724–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Hartline CB, Harden EA, Williams-Aziz SL, Kushner NL, Brideau RJ, Kern ER. Inhibition of herpesvirus replication by a series of 4-oxo-dihydroquinolines with viral polymerase activity. Antiviral Res 2005; 65:97–105. [DOI] [PubMed] [Google Scholar]
- 74. Underwood MR, Ferris RG, Selleseth DW, et al. Mechanism of action of the ribopyranoside benzimidazole GW275175X against human cytomegalovirus. Antimicrob Agents Chemother 2004; 48:1647–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Kern ER, Hartline CB, Rybak RJ, et al. Activities of benzimidazole D- and L-ribonucleosides in animal models of cytomegalovirus infections. Antimicrob Agents Chemother 2004; 48:1749–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Williams SL, Hartline CB, Kushner NL, et al. In vitro activities of benzimidazole D- and L-ribonucleosides against herpesviruses. Antimicrob Agents Chemother 2003; 47:2186–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Gentry BG, Drach JC. Metabolism of cyclopropavir and ganciclovir in human cytomegalovirus-infected cells. Antimicrob Agents Chemother 2014; 58:2329–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Rouphael NG, Hurwitz SJ,. Hart M, et al. Phase IB trial to evaluate the safety and pharmacokinetics of multiple ascending doses of filociclovir (MBX-400, Cyclopropavir) in healthy volunteers. Antimicrob Agents Chemother 2019; 63:717–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Wiltshire H, Paya CV, Pescovitz MD, et al. Valganciclovir Solid Organ Transplant Study Group Pharmacodynamics of oral ganciclovir and valganciclovir in solid organ transplant recipients. Transplantation 2005; 79:1477–83. [DOI] [PubMed] [Google Scholar]
- 80. Chou S, Bowlin TL. Cytomegalovirus UL97 mutations affecting cyclopropavir and ganciclovir susceptibility. Antimicrob Agents Chemother 2011; 55:382–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Gentry BG, Kamil JP, Coen DM, Zemlicka J, Drach JC. Stereoselective phosphorylation of cyclopropavir by pUL97 and competitive inhibition by maribavir. Antimicrob Agents Chemother 2010; 54:3093–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Clark AL, Gall SA. Clinical uses of intravenous immunoglobulin in pregnancy. Am J Obstet Gynecol 1997; 176:241–53. [DOI] [PubMed] [Google Scholar]
- 83. Adler SP. Primary maternal cytomegalovirus infection during pregnancy: do we have a treatment option? Clin Infect Dis 2012; 55:504–6. [DOI] [PubMed] [Google Scholar]
- 84. Kagan KO, Enders M, Schampera MS, et al. Prevention of maternal-fetal transmission of cytomegalovirus after primary maternal infection in the first trimester by biweekly hyperimmunoglobulin administration. Ultrasound Obstet Gynecol 2019; 53:383–9. [DOI] [PubMed] [Google Scholar]
- 85. Bonaros N, Mayer B, Schachner T, Laufer G, Kocher A. CMV-hyperimmune globulin for preventing cytomegalovirus infection and disease in solid organ transplant recipients: a meta-analysis. Clin Transplant 2008; 22:89–97. [DOI] [PubMed] [Google Scholar]
- 86. Goldstein G, Rutenberg TF, Mendelovich SL, et al. The role of immunoglobulin prophylaxis for prevention of cytomegalovirus infection in pediatric hematopoietic stem cell transplantation recipients. Pediatr Blood Cancer 2017; 64. doi:10.1002/pbc.26420. [DOI] [PubMed] [Google Scholar]
- 87. Fouts AE, Chan P, Stephan JP, Vandlen R, Feierbach B. Antibodies against the gH/gL/UL128/UL130/UL131 complex comprise the majority of the anti-cytomegalovirus (anti-CMV) neutralizing antibody response in CMV hyperimmune globulin. J Virol 2012; 86:7444–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Ishida JH, Patel A, Mehta AK, et al. Phase 2 randomized, double-blind, placebo-controlled trial of RG7667, a combination monoclonal antibody, for prevention of cytomegalovirus infection in high-risk kidney transplant recipients. Antimicrob Agents Chemother 2017; 61. doi:10.1128/AAC.01794-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Macagno A, Bernasconi NL, Vanzetta F, et al. Isolation of human monoclonal antibodies that potently neutralize human cytomegalovirus infection by targeting different epitopes on the gH/gL/UL128-131A complex. J Virol 2010; 84:1005–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Scrivano L, Sinzger C, Nitschko H, Koszinowski UH, Adler B. HCMV spread and cell tropism are determined by distinct virus populations. PLoS Pathog 2011; 7:e1001256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Pass RF, Zhang C, Evans A, et al. Vaccine prevention of maternal cytomegalovirus infection. N Engl J Med 2009; 360:1191–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Patel HD, Nikitin P, Gesner T, et al. In vitro characterization of human cytomegalovirus-targeting therapeutic monoclonal antibodies LJP538 and LJP539. Antimicrob Agents Chemother 2016; 60:4961–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. McVoy MM, Tenorio E, Kauvar LM. A native human monoclonal antibody targeting HCMV gB (AD-2 site I). Int J Mol Sci 2018; 19. doi:10.3390/ijms19123982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Maidji E, McDonagh S, Genbacev O, Tabata T, Pereira L. Maternal antibodies enhance or prevent cytomegalovirus infection in the placenta by neonatal Fc receptor-mediated transcytosis. Am J Pathol 2006; 168:1210–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Lantto J, Fletcher JM, Ohlin M. Binding characteristics determine the neutralizing potential of antibody fragments specific for antigenic domain 2 on glycoprotein B of human cytomegalovirus. Virology 2003; 305:201–9. [DOI] [PubMed] [Google Scholar]
- 96. Bialas KM, Westreich D, Cisneros de la Rosa E, et al. Maternal antibody responses and nonprimary congenital cytomegalovirus infection of HIV-1-exposed infants. J Infect Dis 2016; 214:1916–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97. Kauvar LM, Liu K, Park M, et al. A high-affinity native human antibody neutralizes human cytomegalovirus infection of diverse cell types. Antimicrob Agents Chemother 2015; 59:1558–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Pereira L, Tabata T, Petitt M, Fang-Hoover J. Congenital cytomegalovirus infection undermines early development and functions of the human placenta. Placenta 2017; 59(Suppl 1):S8–16. [DOI] [PubMed] [Google Scholar]
- 99. Cui X, Freed DC, Wang D, et al. Impact of antibodies and strain polymorphisms on cytomegalovirus entry and spread in fibroblasts and epithelial cells. J Virol 2017; 91. doi:10.1128/JVI.01650-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Renzette N, Bhattacharjee B, Jensen JD, Gibson L, Kowalik TF. Extensive genome-wide variability of human cytomegalovirus in congenitally infected infants. PLoS Pathog 2011; 7:e1001344. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101. Razonable RR. Drug-resistant cytomegalovirus: clinical implications of specific mutations. Curr Opin Organ Transplant 2018; 23:388–94. [DOI] [PubMed] [Google Scholar]
- 102. Chou S. Rapid in vitro evolution of human cytomegalovirus UL56 mutations that confer letermovir resistance. Antimicrob Agents Chemother 2015; 59:6588–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Goldner T, Hempel C, Ruebsamen-Schaeff H, Zimmermann H, Lischka P. Geno- and phenotypic characterization of human cytomegalovirus mutants selected in vitro after letermovir (AIC246) exposure. Antimicrob Agents Chemother 2014; 58:610–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Hayes CN, Chayama K. Interferon stimulated genes and innate immune activation following infection with hepatitis B and C viruses. J Med Virol 2017; 89:388–96. [DOI] [PubMed] [Google Scholar]
- 105. Bheda P, Jing H, Wolberger C, Lin H. The substrate specificity of sirtuins. Annu Rev Biochem 2016; 85:405–29. [DOI] [PubMed] [Google Scholar]
- 106. Mathias RA, Greco TM, Oberstein A, et al. Sirtuin 4 is a lipoamidase regulating pyruvate dehydrogenase complex activity. Cell 2014; 159:1615–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Anderson KA, Huynh FK, Fisher-Wellman K, et al. SIRT4 is a lysine deacylase that controls leucine metabolism and insulin secretion. Cell Metab 2017; 25:838–55.e15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Sabari BR, Zhang D, Allis CD, Zhao Y. Metabolic regulation of gene expression through histone acylations. Nat Rev Mol Cell Biol 2017; 18:90–101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Kauppinen A, Suuronen T, Ojala J, Kaarniranta K, Salminen A. Antagonistic crosstalk between NF-κB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell Signal 2013; 25:1939–48. [DOI] [PubMed] [Google Scholar]
- 110. Koyuncu E, Budayeva HG, Miteva YV, et al. Sirtuins are evolutionarily conserved viral restriction factors. mBio 2014; 5. doi:10.1128/mBio.02249-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Gogoi M, Chandra K, Sarikhani M, Ramani R, Sundaresan NR, Chakravortty D. Salmonella escapes adaptive immune response via SIRT2 mediated modulation of innate immune response in dendritic cells. PLoS Pathog 2018; 14:e1007437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Eskandarian HA, Impens F, Nahori MA, et al. A role for SIRT2-dependent histone H3K18 deacetylation in bacterial infection. Science 2013; 341:1238858. [DOI] [PubMed] [Google Scholar]
- 113. Pereira JM, Chevalier C, Chaze T, et al. Infection reveals a modification of SIRT2 critical for chromatin association. Cell Rep 2018; 23:1124–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Moreira D, Rodrigues V, Abengozar M, et al. Leishmania infantum modulates host macrophage mitochondrial metabolism by hijacking the SIRT1-AMPK axis. PLoS Pathog 2015; 11:e1004684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Li Q, He M, Zhou F, Ye F, Gao SJ. Activation of Kaposi’s sarcoma-associated herpesvirus (KSHV) by inhibitors of class III histone deacetylases: identification of sirtuin 1 as a regulator of the KSHV life cycle. J Virol 2014; 88:6355–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Pinzone MR, Cacopardo B, Condorelli F, Di Rosa M, Nunnari G. Sirtuin-1 and HIV-1: an overview. Curr Drug Targets 2013; 14:648–52. [DOI] [PubMed] [Google Scholar]
- 117. Cheng ST, Ren JH, Cai XF, Jiang H, Chen J. HBx-elevated SIRT2 promotes HBV replication and hepatocarcinogenesis. Biochem Biophys Res Commun 2018; 496:904–10. [DOI] [PubMed] [Google Scholar]
- 118. Piracha ZZ, Kwon H, Saeed U, et al. Sirtuin 2 isoform 1 enhances hepatitis B virus RNA transcription and DNA synthesis through the AKT/GSK-3beta/beta-catenin signaling pathway. J Virol 2018; 92. doi:10.1128/JVI.00955-18. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 119.Oberstein A, Perlman DH, Shenk T, Terry LJ. Human cytomegalovirus pUL97 kinase induces global changes in the infected cell phosphoproteome. Proteomics 2015; 15:2006–2022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Sanjuán R, Nebot MR, Chirico N, Mansky LM, Belshaw R. Viral mutation rates. J Virol 2010; 84:9733–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Chou TC. Drug combination studies and their synergy quantification using the Chou-Talalay method. Cancer Res 2010; 70:440–6. [DOI] [PubMed] [Google Scholar]
- 122. Zhang S, Jiang Y, Cheng Q, Zhong Y, Qin Y, Chen M. Inclusion body fusion of human parainfluenza virus type 3 regulated by acetylated alpha-tubulin enhances viral replication. J Virol 2017; 91. doi:10.1128/JVI.01802-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Shenk T, Alwine JC. Human cytomegalovirus: coordinating cellular stress, signaling, and metabolic pathways. Annu Rev Virol 2014; 1:355–74. [DOI] [PubMed] [Google Scholar]
- 124. Hoffmann G, Breitenbücher F, Schuler M, Ehrenhofer-Murray AE. A novel sirtuin 2 (SIRT2) inhibitor with p53-dependent pro-apoptotic activity in non-small cell lung cancer. J Biol Chem 2014; 289:5208–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Peck B, Chen CY, Ho KK, et al. SIRT inhibitors induce cell death and p53 acetylation through targeting both SIRT1 and SIRT2. Mol Cancer Ther 2010; 9:844–55. [DOI] [PubMed] [Google Scholar]
- 126. Bresnahan WA, Boldogh I, Thompson EA, Albrecht T. Human cytomegalovirus inhibits cellular DNA synthesis and arrests productively infected cells in late G1. Virology 1996; 224:150–60. [DOI] [PubMed] [Google Scholar]
- 127. Jault FM, Jault JM, Ruchti F, et al. Cytomegalovirus infection induces high levels of cyclins, phosphorylated Rb, and p53, leading to cell cycle arrest. J Virol 1995; 69:6697–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Zhang H, Park SH, Pantazides BG, et al. SIRT2 directs the replication stress response through CDK9 deacetylation. Proc Natl Acad Sci U S A 2013; 110:13546–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129. Ramakrishnan G, Davaakhuu G, Kaplun L, et al. Sirt2 deacetylase is a novel AKT binding partner critical for AKT activation by insulin. J Biol Chem 2014; 289:6054–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130. Dan L, Klimenkova O, Klimiankou M, et al. The role of sirtuin 2 activation by nicotinamide phosphoribosyltransferase in the aberrant proliferation and survival of myeloid leukemia cells. Haematologica 2012; 97:551–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131. Yamamoto M, Onogi H, Kii I, et al. CDK9 inhibitor FIT-039 prevents replication of multiple DNA viruses. J Clin Invest 2014; 124:3479–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132. Peppenelli MA, Miller MJ, Altman AM, Cojohari O, Chan GC. Aberrant regulation of the Akt signaling network by human cytomegalovirus allows for targeting of infected monocytes. Antiviral Res 2018; 158:13–24. [DOI] [PMC free article] [PubMed] [Google Scholar]

