Skip to main content
Top
Published in: Current Hematologic Malignancy Reports 2/2020

01-04-2020 | Stem Cell Transplantation (R Maziarz, Section Editor)

Moving Past Ganciclovir and Foscarnet: Advances in CMV Therapy

Author: Morgan Hakki

Published in: Current Hematologic Malignancy Reports | Issue 2/2020

Login to get access

Abstract

Purpose of Review

CMV DNA polymerase inhibitors such as ganciclovir and foscarnet have dramatically reduced the burden of CMV infection in the HCT recipient. However, their use is often limited by toxicities and resistance. Agents with novel mechanisms and favorable toxicity profiles are critically needed. We review recent developments in CMV antivirals and immune-based approaches to mitigating CMV infection.

Recent Findings

Letermovir, an inhibitor of the CMV terminase complex, was approved in 2017 for primary CMV prophylaxis in adult seropositive allogeneic HCT recipients. Maribavir, an inhibitor of the CMV UL97 kinase, is currently in two phase 3 treatment studies. Adoptive immunotherapy using third-party T cells has proven safe and effective in preliminary studies. Vaccine development continues, with several promising candidates currently under study.

Summary

No longer limited to DNA polymerase inhibitors, the prevention and treatment of CMV infections in the HCT recipient is a rapidly evolving field which should translate into improvements in CMV-related outcomes.
Literature
3.
go back to reference Bogner E. Human cytomegalovirus terminase as a target for antiviral chemotherapy. Rev Med Virol. 2002;12(2):115–27.PubMedCrossRef Bogner E. Human cytomegalovirus terminase as a target for antiviral chemotherapy. Rev Med Virol. 2002;12(2):115–27.PubMedCrossRef
4.
go back to reference Neuber S, Wagner K, Goldner T, Lischka P, Steinbrueck L, Messerle M, et al. Mutual interplay between the human cytomegalovirus terminase subunits pUL51, pUL56, and pUL89 promotes terminase complex formation. J Virol. 2017;91(12). Neuber S, Wagner K, Goldner T, Lischka P, Steinbrueck L, Messerle M, et al. Mutual interplay between the human cytomegalovirus terminase subunits pUL51, pUL56, and pUL89 promotes terminase complex formation. J Virol. 2017;91(12).
5.
go back to reference Borst EM, Kleine-Albers J, Gabaev I, Babic M, Wagner K, Binz A, et al. The human cytomegalovirus UL51 protein is essential for viral genome cleavage-packaging and interacts with the terminase subunits pUL56 and pUL89. J Virol. 2013;87(3):1720–32.PubMedPubMedCentralCrossRef Borst EM, Kleine-Albers J, Gabaev I, Babic M, Wagner K, Binz A, et al. The human cytomegalovirus UL51 protein is essential for viral genome cleavage-packaging and interacts with the terminase subunits pUL56 and pUL89. J Virol. 2013;87(3):1720–32.PubMedPubMedCentralCrossRef
6.
go back to reference Dunn W, Chou C, Li H, Hai R, Patterson D, Stolc V, et al. Functional profiling of a human cytomegalovirus genome. Proc Natl Acad Sci U S A. 2003;100(24):14223–8.PubMedPubMedCentralCrossRef Dunn W, Chou C, Li H, Hai R, Patterson D, Stolc V, et al. Functional profiling of a human cytomegalovirus genome. Proc Natl Acad Sci U S A. 2003;100(24):14223–8.PubMedPubMedCentralCrossRef
7.
go back to reference Underwood MR, Ferris RG, Selleseth DW, Davis MG, Drach JC, Townsend LB, et al. Mechanism of action of the ribopyranoside benzimidazole GW275175X against human cytomegalovirus. Antimicrob Agents Chemother. 2004;48(5):1647–51.PubMedPubMedCentralCrossRef Underwood MR, Ferris RG, Selleseth DW, Davis MG, Drach JC, Townsend LB, et al. Mechanism of action of the ribopyranoside benzimidazole GW275175X against human cytomegalovirus. Antimicrob Agents Chemother. 2004;48(5):1647–51.PubMedPubMedCentralCrossRef
8.
go back to reference Buerger I, Reefschlaeger J, Bender W, Eckenberg P, Popp A, Weber O, et al. A novel nonnucleoside inhibitor specifically targets cytomegalovirus DNA maturation via the UL89 and UL56 gene products. J Virol. 2001;75(19):9077–86.PubMedPubMedCentralCrossRef Buerger I, Reefschlaeger J, Bender W, Eckenberg P, Popp A, Weber O, et al. A novel nonnucleoside inhibitor specifically targets cytomegalovirus DNA maturation via the UL89 and UL56 gene products. J Virol. 2001;75(19):9077–86.PubMedPubMedCentralCrossRef
9.
go back to reference Lischka P, Hewlett G, Wunberg T, Baumeister J, Paulsen D, Goldner T, et al. In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246. Antimicrob Agents Chemother. 2010;54(3):1290–7.PubMedPubMedCentralCrossRef Lischka P, Hewlett G, Wunberg T, Baumeister J, Paulsen D, Goldner T, et al. In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246. Antimicrob Agents Chemother. 2010;54(3):1290–7.PubMedPubMedCentralCrossRef
10.
go back to reference 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(20):10884–93.PubMedPubMedCentralCrossRef 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(20):10884–93.PubMedPubMedCentralCrossRef
11.
go back to reference Marschall M, Stamminger T, Urban A, Wildum S, Ruebsamen-Schaeff H, Zimmermann H, et al. In vitro evaluation of the activities of the novel anticytomegalovirus compound AIC246 (letermovir) against herpesviruses and other human pathogenic viruses. Antimicrob Agents Chemother. 2012;56(2):1135–7.PubMedPubMedCentralCrossRef Marschall M, Stamminger T, Urban A, Wildum S, Ruebsamen-Schaeff H, Zimmermann H, et al. In vitro evaluation of the activities of the novel anticytomegalovirus compound AIC246 (letermovir) against herpesviruses and other human pathogenic viruses. Antimicrob Agents Chemother. 2012;56(2):1135–7.PubMedPubMedCentralCrossRef
12.
go back to reference Piret J, Boivin G. Clinical development of letermovir and maribavir: overview of human cytomegalovirus drug resistance. Antivir Res. 2019;163:91–105.PubMedCrossRef Piret J, Boivin G. Clinical development of letermovir and maribavir: overview of human cytomegalovirus drug resistance. Antivir Res. 2019;163:91–105.PubMedCrossRef
13.
go back to reference Kropeit D, von Richter O, Stobernack HP, Rubsamen-Schaeff H, Zimmermann H. Pharmacokinetics and safety of letermovir coadministered with cyclosporine A or tacrolimus in healthy subjects. Clin Pharmacol Drug Dev. 2018;7(1):9–21.PubMedCrossRef Kropeit D, von Richter O, Stobernack HP, Rubsamen-Schaeff H, Zimmermann H. Pharmacokinetics and safety of letermovir coadministered with cyclosporine A or tacrolimus in healthy subjects. Clin Pharmacol Drug Dev. 2018;7(1):9–21.PubMedCrossRef
14.
go back to reference McCrea JB, Macha S, Adedoyin A, Marshall W, Menzel K, Cho CR, et al. Pharmacokinetic drug-drug interactions between letermovir and the immunosuppressants cyclosporine, tacrolimus, sirolimus, and mycophenolate mofetil. J Clin Pharmacol. 2019;59(10):1331–9.PubMedCrossRef McCrea JB, Macha S, Adedoyin A, Marshall W, Menzel K, Cho CR, et al. Pharmacokinetic drug-drug interactions between letermovir and the immunosuppressants cyclosporine, tacrolimus, sirolimus, and mycophenolate mofetil. J Clin Pharmacol. 2019;59(10):1331–9.PubMedCrossRef
15.
go back to reference Duong A, Sweet A, Jain R, Hill JA, Pergam SA, Boeckh M, et al. Clinically significant drug interaction: letermovir and voriconazole. J Antimicrob Chemother. 2019. Duong A, Sweet A, Jain R, Hill JA, Pergam SA, Boeckh M, et al. Clinically significant drug interaction: letermovir and voriconazole. J Antimicrob Chemother. 2019.
16.
go back to reference Marshall WL, McCrea JB, Macha S, Menzel K, Liu F, van Schanke A, et al. Pharmacokinetics and tolerability of letermovir coadministered with azole antifungals (posaconazole or voriconazole) in healthy subjects. J Clin Pharmacol. 2018;58(7):897–904.PubMedCrossRef Marshall WL, McCrea JB, Macha S, Menzel K, Liu F, van Schanke A, et al. Pharmacokinetics and tolerability of letermovir coadministered with azole antifungals (posaconazole or voriconazole) in healthy subjects. J Clin Pharmacol. 2018;58(7):897–904.PubMedCrossRef
17.
go back to reference PREVYMIS: Package insert and label information [press release]. 2017. PREVYMIS: Package insert and label information [press release]. 2017.
18.
go back to reference Kropeit D, McCormick D, Erb-Zohar K, Moiseev VS, Kobalava ZD, Stobernack HP, et al. Pharmacokinetics and safety of the anti-human cytomegalovirus drug letermovir in subjects with hepatic impairment. Br J Clin Pharmacol. 2017;83(12):2678–86.PubMedPubMedCentralCrossRef Kropeit D, McCormick D, Erb-Zohar K, Moiseev VS, Kobalava ZD, Stobernack HP, et al. Pharmacokinetics and safety of the anti-human cytomegalovirus drug letermovir in subjects with hepatic impairment. Br J Clin Pharmacol. 2017;83(12):2678–86.PubMedPubMedCentralCrossRef
19.
go back to reference Kropeit D, Scheuenpflug J, Erb-Zohar K, Halabi A, Stobernack HP, Hulskotte EGJ, et al. Pharmacokinetics and safety of letermovir, a novel anti-human cytomegalovirus drug, in patients with renal impairment. Br J Clin Pharmacol. 2017;83(9):1944–53.PubMedPubMedCentralCrossRef Kropeit D, Scheuenpflug J, Erb-Zohar K, Halabi A, Stobernack HP, Hulskotte EGJ, et al. Pharmacokinetics and safety of letermovir, a novel anti-human cytomegalovirus drug, in patients with renal impairment. Br J Clin Pharmacol. 2017;83(9):1944–53.PubMedPubMedCentralCrossRef
20.
go back to reference Erb-Zohar K, Kropeit D, Scheuenpflug J, Stobernack HP, Hulskotte E, van Schanke A, et al. Intravenous hydroxypropyl beta-cyclodextrin formulation of letermovir: a phase I, randomized, single-ascending, and multiple-dose trial. Clin Transl Sci. 2017;10(6):487–95.PubMedPubMedCentralCrossRef Erb-Zohar K, Kropeit D, Scheuenpflug J, Stobernack HP, Hulskotte E, van Schanke A, et al. Intravenous hydroxypropyl beta-cyclodextrin formulation of letermovir: a phase I, randomized, single-ascending, and multiple-dose trial. Clin Transl Sci. 2017;10(6):487–95.PubMedPubMedCentralCrossRef
21.
go back to reference Chemaly RF, Ullmann AJ, Stoelben S, Richard MP, Bornhauser M, Groth C, et al. Letermovir for cytomegalovirus prophylaxis in hematopoietic-cell transplantation. N Engl J Med. 2014;370(19):1781–9.PubMedCrossRef Chemaly RF, Ullmann AJ, Stoelben S, Richard MP, Bornhauser M, Groth C, et al. Letermovir for cytomegalovirus prophylaxis in hematopoietic-cell transplantation. N Engl J Med. 2014;370(19):1781–9.PubMedCrossRef
22.
go back to reference •• Marty FM, Ljungman P, Chemaly RF, Maertens J, Dadwal SS, Duarte RF, et al. Letermovir prophylaxis for cytomegalovirus in hematopoietic-cell transplantation. N Engl J Med. 2017;377(25):2433–44 Phase 3 study of letermovir that led to FDA approval for use as prophylaxis in CMV seropositive allogeneic HCT recipients. PubMedCrossRef •• Marty FM, Ljungman P, Chemaly RF, Maertens J, Dadwal SS, Duarte RF, et al. Letermovir prophylaxis for cytomegalovirus in hematopoietic-cell transplantation. N Engl J Med. 2017;377(25):2433–44 Phase 3 study of letermovir that led to FDA approval for use as prophylaxis in CMV seropositive allogeneic HCT recipients. PubMedCrossRef
23.
go back to reference • Ljungman P, Schmitt M, Marty FM, Maertens J, Chemaly RF, Kartsonis NA, et al. A mortality analysis of letermovir prophylaxis for cytomegalovirus (CMV) in CMV-seropositive recipients of allogeneic hematopoietic-cell transplantation. Clin Infect Dis. 2019; Analysis of mortality benefit observed in HCT recipients receiving prophylaxis as part of the phase 3 study. • Ljungman P, Schmitt M, Marty FM, Maertens J, Chemaly RF, Kartsonis NA, et al. A mortality analysis of letermovir prophylaxis for cytomegalovirus (CMV) in CMV-seropositive recipients of allogeneic hematopoietic-cell transplantation. Clin Infect Dis. 2019; Analysis of mortality benefit observed in HCT recipients receiving prophylaxis as part of the phase 3 study.
24.
go back to reference Nichols WG, Corey L, Gooley T, Davis C, Boeckh M. High risk of death due to bacterial and fungal infection among cytomegalovirus (CMV)-seronegative recipients of stem cell transplants from seropositive donors: evidence for indirect effects of primary CMV infection. J Infect Dis. 2002;185(3):273–82.PubMedCrossRef Nichols WG, Corey L, Gooley T, Davis C, Boeckh M. High risk of death due to bacterial and fungal infection among cytomegalovirus (CMV)-seronegative recipients of stem cell transplants from seropositive donors: evidence for indirect effects of primary CMV infection. J Infect Dis. 2002;185(3):273–82.PubMedCrossRef
25.
go back to reference Schmidt-Hieber M, Labopin M, Beelen D, Volin L, Ehninger G, Finke J, et al. CMV serostatus still has an important prognostic impact in de novo acute leukemia patients after allogeneic stem cell transplantation: a report from the Acute Leukemia Working Party of EBMT. Blood. 2013;122(19):3359–64.PubMedCrossRef Schmidt-Hieber M, Labopin M, Beelen D, Volin L, Ehninger G, Finke J, et al. CMV serostatus still has an important prognostic impact in de novo acute leukemia patients after allogeneic stem cell transplantation: a report from the Acute Leukemia Working Party of EBMT. Blood. 2013;122(19):3359–64.PubMedCrossRef
26.
go back to reference Teira P, Battiwalla M, Ramanathan M, Barrett AJ, Ahn KW, Chen M, et al. Early cytomegalovirus reactivation remains associated with increased transplant-related mortality in the current era: a CIBMTR analysis. Blood. 2016;127(20):2427–38.PubMedPubMedCentralCrossRef Teira P, Battiwalla M, Ramanathan M, Barrett AJ, Ahn KW, Chen M, et al. Early cytomegalovirus reactivation remains associated with increased transplant-related mortality in the current era: a CIBMTR analysis. Blood. 2016;127(20):2427–38.PubMedPubMedCentralCrossRef
27.
go back to reference Chou S. Rapid in vitro evolution of human cytomegalovirus UL56 mutations that confer letermovir resistance. Antimicrob Agents Chemother. 2015;59(10):6588–93.PubMedPubMedCentralCrossRef Chou S. Rapid in vitro evolution of human cytomegalovirus UL56 mutations that confer letermovir resistance. Antimicrob Agents Chemother. 2015;59(10):6588–93.PubMedPubMedCentralCrossRef
28.
go back to reference 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(1):610–3.PubMedPubMedCentralCrossRef 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(1):610–3.PubMedPubMedCentralCrossRef
29.
go back to reference Chou S. A third component of the human cytomegalovirus terminase complex is involved in letermovir resistance. Antivir Res. 2017;148:1–4.PubMedCrossRef Chou S. A third component of the human cytomegalovirus terminase complex is involved in letermovir resistance. Antivir Res. 2017;148:1–4.PubMedCrossRef
30.
go back to reference Chou S. Comparison of cytomegalovirus terminase gene mutations selected after exposure to three distinct inhibitor compounds. Antimicrobial agents and chemotherapy. 2017;61(11). Chou S. Comparison of cytomegalovirus terminase gene mutations selected after exposure to three distinct inhibitor compounds. Antimicrobial agents and chemotherapy. 2017;61(11).
31.
go back to reference Lischka P, Michel D, Zimmermann H. Characterization of cytomegalovirus breakthrough events in a phase 2 prophylaxis trial of letermovir (AIC246, MK 8228). J Infect Dis. 2016;213(1):23–30.PubMedCrossRef Lischka P, Michel D, Zimmermann H. Characterization of cytomegalovirus breakthrough events in a phase 2 prophylaxis trial of letermovir (AIC246, MK 8228). J Infect Dis. 2016;213(1):23–30.PubMedCrossRef
32.
go back to reference • Douglas CM, Barnard R, Holder D, Leavitt R, Levitan D, Maguire M, et al. Letermovir resistance analysis in a clinical trial of cytomegalovirus prophylaxis for hematopoietic stem cell transplant recipients. J Infect Dis. 2019; Analysis of letermovir resistance from the phase 3 prophylaxis study in HCT recipients. • Douglas CM, Barnard R, Holder D, Leavitt R, Levitan D, Maguire M, et al. Letermovir resistance analysis in a clinical trial of cytomegalovirus prophylaxis for hematopoietic stem cell transplant recipients. J Infect Dis. 2019; Analysis of letermovir resistance from the phase 3 prophylaxis study in HCT recipients.
33.
go back to reference Kilgore JT, Becken B, Varga MG, Parikh S, Prasad V, Lugo D, et al. Use of letermovir for salvage therapy for resistant cytomegalovirus in a pediatric hematopoietic stem cell transplant recipient. J Pediatric Infect Dis Soc. 2019. Kilgore JT, Becken B, Varga MG, Parikh S, Prasad V, Lugo D, et al. Use of letermovir for salvage therapy for resistant cytomegalovirus in a pediatric hematopoietic stem cell transplant recipient. J Pediatric Infect Dis Soc. 2019.
34.
go back to reference Knoll BM, Seiter K, Phillips A, Soave R. Breakthrough cytomegalovirus pneumonia in hematopoietic stem cell transplant recipient on letermovir prophylaxis. Bone Marrow Transplant. 2019;54(6):911–2.PubMedCrossRef Knoll BM, Seiter K, Phillips A, Soave R. Breakthrough cytomegalovirus pneumonia in hematopoietic stem cell transplant recipient on letermovir prophylaxis. Bone Marrow Transplant. 2019;54(6):911–2.PubMedCrossRef
35.
go back to reference Popping S, Dalm V, Lubke N, Cristanziano VD, Kaiser R, Boucher CAB, et al. Emergence and persistence of letermovir-resistant cytomegalovirus in a patient with primary immunodeficiency. Open Forum Infect Dis. 2019;6(9):ofz375.PubMedPubMedCentralCrossRef Popping S, Dalm V, Lubke N, Cristanziano VD, Kaiser R, Boucher CAB, et al. Emergence and persistence of letermovir-resistant cytomegalovirus in a patient with primary immunodeficiency. Open Forum Infect Dis. 2019;6(9):ofz375.PubMedPubMedCentralCrossRef
36.
go back to reference Humar A, Lebranchu Y, Vincenti F, Blumberg EA, Punch JD, Limaye AP, et al. The efficacy and safety of 200 days valganciclovir cytomegalovirus prophylaxis in high-risk kidney transplant recipients. Am J Transplant. 2010;10(5):1228–37.PubMedCrossRef Humar A, Lebranchu Y, Vincenti F, Blumberg EA, Punch JD, Limaye AP, et al. The efficacy and safety of 200 days valganciclovir cytomegalovirus prophylaxis in high-risk kidney transplant recipients. Am J Transplant. 2010;10(5):1228–37.PubMedCrossRef
37.
go back to reference Sharma P, Gakhar N, MacDonald J, Abidi MZ, Benamu E, Bajrovic V, et al. Letermovir prophylaxis through day 100 post transplant is safe and effective compared with alternative CMV prophylaxis strategies following adult cord blood and haploidentical cord blood transplantation. Bone Marrow Transplant. 2019. Sharma P, Gakhar N, MacDonald J, Abidi MZ, Benamu E, Bajrovic V, et al. Letermovir prophylaxis through day 100 post transplant is safe and effective compared with alternative CMV prophylaxis strategies following adult cord blood and haploidentical cord blood transplantation. Bone Marrow Transplant. 2019.
38.
go back to reference Lin A, Maloy M, Su Y, Bhatt V, DeRespiris L, Griffin M, et al. Letermovir for primary and secondary cytomegalovirus prevention in allogeneic hematopoietic cell transplant recipients: real-world experience. Transpl Infect Dis. 2019:e13187. Lin A, Maloy M, Su Y, Bhatt V, DeRespiris L, Griffin M, et al. Letermovir for primary and secondary cytomegalovirus prevention in allogeneic hematopoietic cell transplant recipients: real-world experience. Transpl Infect Dis. 2019:e13187.
39.
go back to reference Marty FM, Ljungman PT, Chemaly RF, Wan H, Teal VL, Butterton JR, et al. Outcomes of patients with detectable CMV DNA at randomization in the phase III trial of letermovir for the prevention of CMV infection in allogeneic hematopoietic cell transplantation. Am J Transplant. 2019. Marty FM, Ljungman PT, Chemaly RF, Wan H, Teal VL, Butterton JR, et al. Outcomes of patients with detectable CMV DNA at randomization in the phase III trial of letermovir for the prevention of CMV infection in allogeneic hematopoietic cell transplantation. Am J Transplant. 2019.
40.
go back to reference Frietsch JJ, Michel D, Stamminger T, Hunstig F, Birndt S, Schnetzke U, et al. In vivo emergence of UL56 C325Y cytomegalovirus resistance to letermovir in a patient with acute myeloid leukemia after hematopoietic cell transplantation. Mediterr J Hematol Infect Dis. 2019;11(1):e2019001.PubMedPubMedCentralCrossRef Frietsch JJ, Michel D, Stamminger T, Hunstig F, Birndt S, Schnetzke U, et al. In vivo emergence of UL56 C325Y cytomegalovirus resistance to letermovir in a patient with acute myeloid leukemia after hematopoietic cell transplantation. Mediterr J Hematol Infect Dis. 2019;11(1):e2019001.PubMedPubMedCentralCrossRef
41.
go back to reference Cherrier L, Nasar A, Goodlet KJ, Nailor MD, Tokman S, Chou S. Emergence of letermovir resistance in a lung transplant recipient with ganciclovir-resistant cytomegalovirus infection. Am J Transplant. 2018;18(12):3060–4.PubMedPubMedCentralCrossRef Cherrier L, Nasar A, Goodlet KJ, Nailor MD, Tokman S, Chou S. Emergence of letermovir resistance in a lung transplant recipient with ganciclovir-resistant cytomegalovirus infection. Am J Transplant. 2018;18(12):3060–4.PubMedPubMedCentralCrossRef
42.
go back to reference Jung S, Michel M, Stamminger T, Michel D. Fast breakthrough of resistant cytomegalovirus during secondary letermovir prophylaxis in a hematopoietic stem cell transplant recipient. BMC Infect Dis. 2019;19(1):388.PubMedPubMedCentralCrossRef Jung S, Michel M, Stamminger T, Michel D. Fast breakthrough of resistant cytomegalovirus during secondary letermovir prophylaxis in a hematopoietic stem cell transplant recipient. BMC Infect Dis. 2019;19(1):388.PubMedPubMedCentralCrossRef
43.
go back to reference Turner N, Strand A, Grewal DS, Cox G, Arif S, Baker AW, et al. Use of letermovir as salvage therapy for drug-resistant cytomegalovirus retinitis. Antimicrobial agents and chemotherapy. 2019;63(3). Turner N, Strand A, Grewal DS, Cox G, Arif S, Baker AW, et al. Use of letermovir as salvage therapy for drug-resistant cytomegalovirus retinitis. Antimicrobial agents and chemotherapy. 2019;63(3).
44.
go back to reference Wildum S, Zimmermann H, Lischka P. In vitro drug combination studies of letermovir (AIC246, MK-8228) with approved anti human cytomegalovirus (HCMV) and HIV compounds in inhibition of HCMV and HIV replication. Antimicrob Agents Chemother. 2015. Wildum S, Zimmermann H, Lischka P. In vitro drug combination studies of letermovir (AIC246, MK-8228) with approved anti human cytomegalovirus (HCMV) and HIV compounds in inhibition of HCMV and HIV replication. Antimicrob Agents Chemother. 2015.
45.
go back to reference O'Brien MS, Markovich KC, Selleseth D, DeVita AV, Sethna P, Gentry BG. In vitro evaluation of current and novel antivirals in combination against human cytomegalovirus. Antivir Res. 2018;158:255–63.PubMedCrossRef O'Brien MS, Markovich KC, Selleseth D, DeVita AV, Sethna P, Gentry BG. In vitro evaluation of current and novel antivirals in combination against human cytomegalovirus. Antivir Res. 2018;158:255–63.PubMedCrossRef
46.
go back to reference Biron KK, Harvey RJ, Chamberlain SC, Good SS, Smith AA 3rd, Davis MG, et al. Potent and selective inhibition of human cytomegalovirus replication by 1263W94, a benzimidazole L-riboside with a unique mode of action. Antimicrob Agents Chemother. 2002;46(8):2365–72.PubMedPubMedCentralCrossRef Biron KK, Harvey RJ, Chamberlain SC, Good SS, Smith AA 3rd, Davis MG, et al. Potent and selective inhibition of human cytomegalovirus replication by 1263W94, a benzimidazole L-riboside with a unique mode of action. Antimicrob Agents Chemother. 2002;46(8):2365–72.PubMedPubMedCentralCrossRef
47.
48.
go back to reference Azzeh M, Honigman A, Taraboulos A, Rouvinski A, Wolf DG. Structural changes in human cytomegalovirus cytoplasmic assembly sites in the absence of UL97 kinase activity. Virology. 2006;354(1):69–79.PubMedCrossRef Azzeh M, Honigman A, Taraboulos A, Rouvinski A, Wolf DG. Structural changes in human cytomegalovirus cytoplasmic assembly sites in the absence of UL97 kinase activity. Virology. 2006;354(1):69–79.PubMedCrossRef
49.
go back to reference Prichard MN, Gao N, Jairath S, Mulamba G, Krosky P, Coen DM, et al. A recombinant human cytomegalovirus with a large deletion in UL97 has a severe replication deficiency. J Virol. 1999;73(7):5663–70.PubMedPubMedCentralCrossRef Prichard MN, Gao N, Jairath S, Mulamba G, Krosky P, Coen DM, et al. A recombinant human cytomegalovirus with a large deletion in UL97 has a severe replication deficiency. J Virol. 1999;73(7):5663–70.PubMedPubMedCentralCrossRef
50.
go back to reference Webel R, Hakki M, Prichard MN, Rawlinson WD, Marschall M, Chou S. Differential properties of cytomegalovirus pUL97 kinase isoforms affect viral replication and maribavir susceptibility. J Virol. 2014;88(9):4776–85.PubMedPubMedCentralCrossRef Webel R, Hakki M, Prichard MN, Rawlinson WD, Marschall M, Chou S. Differential properties of cytomegalovirus pUL97 kinase isoforms affect viral replication and maribavir susceptibility. J Virol. 2014;88(9):4776–85.PubMedPubMedCentralCrossRef
51.
go back to reference Chou S, Ercolani RJ, Marousek G, Bowlin TL. Cytomegalovirus UL97 kinase catalytic domain mutations that confer multidrug resistance. Antimicrob Agents Chemother. 2013;57(7):3375–9.PubMedPubMedCentralCrossRef Chou S, Ercolani RJ, Marousek G, Bowlin TL. Cytomegalovirus UL97 kinase catalytic domain mutations that confer multidrug resistance. Antimicrob Agents Chemother. 2013;57(7):3375–9.PubMedPubMedCentralCrossRef
52.
go back to reference Hamirally S, Kamil JP, Ndassa-Colday YM, Lin AJ, Jahng WJ, Baek MC, et al. Viral mimicry of Cdc2/cyclin-dependent kinase 1 mediates disruption of nuclear lamina during human cytomegalovirus nuclear egress. PLoS Pathog. 2009;5(1):e1000275.PubMedPubMedCentralCrossRef Hamirally S, Kamil JP, Ndassa-Colday YM, Lin AJ, Jahng WJ, Baek MC, et al. Viral mimicry of Cdc2/cyclin-dependent kinase 1 mediates disruption of nuclear lamina during human cytomegalovirus nuclear egress. PLoS Pathog. 2009;5(1):e1000275.PubMedPubMedCentralCrossRef
53.
go back to reference Chou S, Van Wechel LC, Marousek GI. Effect of cell culture conditions on the anticytomegalovirus activity of maribavir. Antimicrob Agents Chemother. 2006;50(7):2557–9.PubMedPubMedCentralCrossRef Chou S, Van Wechel LC, Marousek GI. Effect of cell culture conditions on the anticytomegalovirus activity of maribavir. Antimicrob Agents Chemother. 2006;50(7):2557–9.PubMedPubMedCentralCrossRef
54.
go back to reference Drew WL, Miner RC, Marousek GI, Chou S. Maribavir sensitivity of cytomegalovirus isolates resistant to ganciclovir, cidofovir or foscarnet. J Clin Virol. 2006;37(2):124–7.PubMedCrossRef Drew WL, Miner RC, Marousek GI, Chou S. Maribavir sensitivity of cytomegalovirus isolates resistant to ganciclovir, cidofovir or foscarnet. J Clin Virol. 2006;37(2):124–7.PubMedCrossRef
55.
go back to reference Williams SL, Hartline CB, Kushner NL, Harden EA, Bidanset DJ, Drach JC, et al. In vitro activities of benzimidazole D- and L-ribonucleosides against herpesviruses. Antimicrob Agents Chemother. 2003;47(7):2186–92.PubMedPubMedCentralCrossRef Williams SL, Hartline CB, Kushner NL, Harden EA, Bidanset DJ, Drach JC, et al. In vitro activities of benzimidazole D- and L-ribonucleosides against herpesviruses. Antimicrob Agents Chemother. 2003;47(7):2186–92.PubMedPubMedCentralCrossRef
56.
go back to reference Wang LH, Peck RW, Yin Y, Allanson J, Wiggs R, Wire MB. Phase I safety and pharmacokinetic trials of 1263W94, a novel oral anti-human cytomegalovirus agent, in healthy and human immunodeficiency virus-infected subjects. Antimicrob Agents Chemother. 2003;47(4):1334–42.PubMedPubMedCentralCrossRef Wang LH, Peck RW, Yin Y, Allanson J, Wiggs R, Wire MB. Phase I safety and pharmacokinetic trials of 1263W94, a novel oral anti-human cytomegalovirus agent, in healthy and human immunodeficiency virus-infected subjects. Antimicrob Agents Chemother. 2003;47(4):1334–42.PubMedPubMedCentralCrossRef
57.
go back to reference Koszalka GW, Johnson NW, Good SS, Boyd L, Chamberlain SC, Townsend LB, et al. Preclinical and toxicology studies of 1263W94, a potent and selective inhibitor of human cytomegalovirus replication. Antimicrob Agents Chemother. 2002;46(8):2373–80.PubMedPubMedCentralCrossRef Koszalka GW, Johnson NW, Good SS, Boyd L, Chamberlain SC, Townsend LB, et al. Preclinical and toxicology studies of 1263W94, a potent and selective inhibitor of human cytomegalovirus replication. Antimicrob Agents Chemother. 2002;46(8):2373–80.PubMedPubMedCentralCrossRef
58.
go back to reference Swan SK, Smith WB, Marbury TC, Schumacher M, Dougherty C, Mico BA, et al. Pharmacokinetics of maribavir, a novel oral anticytomegalovirus agent, in subjects with varying degrees of renal impairment. J Clin Pharmacol. 2007;47(2):209–17.PubMedCrossRef Swan SK, Smith WB, Marbury TC, Schumacher M, Dougherty C, Mico BA, et al. Pharmacokinetics of maribavir, a novel oral anticytomegalovirus agent, in subjects with varying degrees of renal impairment. J Clin Pharmacol. 2007;47(2):209–17.PubMedCrossRef
59.
go back to reference Song IH, Ilic K, Murphy J, Lasseter K, Martin P. Effects of maribavir on P-glycoprotein and CYP2D6 in healthy volunteers. J Clin Pharmacol. 2019. Song IH, Ilic K, Murphy J, Lasseter K, Martin P. Effects of maribavir on P-glycoprotein and CYP2D6 in healthy volunteers. J Clin Pharmacol. 2019.
60.
go back to reference Pescovitz MD, Bloom R, Pirsch J, Johnson J, Gelone S, Villano SA. A randomized, double-blind, pharmacokinetic study of oral maribavir with tacrolimus in stable renal transplant recipients. Am J Transplant. 2009;9(10):2324–30.PubMedCrossRef Pescovitz MD, Bloom R, Pirsch J, Johnson J, Gelone S, Villano SA. A randomized, double-blind, pharmacokinetic study of oral maribavir with tacrolimus in stable renal transplant recipients. Am J Transplant. 2009;9(10):2324–30.PubMedCrossRef
61.
go back to reference • Papanicolaou GA, Silveira FP, Langston AA, Pereira MR, Avery RK, Uknis M, et al. Maribavir for refractory or resistant cytomegalovirus infections in hematopoietic-cell or solid-organ transplant recipients: a randomized, dose-ranging, double-blind, phase 2 study. Clin Infect Dis. 2019;68(8):1255–64 This study demonstrated that maribavir may be a viable option for the treatment of resistant/refractory CMV infection in HCT and SOT recipients. Results from the phase 3 study will be most informative. PubMedCrossRef • Papanicolaou GA, Silveira FP, Langston AA, Pereira MR, Avery RK, Uknis M, et al. Maribavir for refractory or resistant cytomegalovirus infections in hematopoietic-cell or solid-organ transplant recipients: a randomized, dose-ranging, double-blind, phase 2 study. Clin Infect Dis. 2019;68(8):1255–64 This study demonstrated that maribavir may be a viable option for the treatment of resistant/refractory CMV infection in HCT and SOT recipients. Results from the phase 3 study will be most informative. PubMedCrossRef
62.
go back to reference Lalezari JP, Aberg JA, Wang LH, Wire MB, Miner R, Snowden W, 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(9):2969–76.PubMedPubMedCentralCrossRef Lalezari JP, Aberg JA, Wang LH, Wire MB, Miner R, Snowden W, 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(9):2969–76.PubMedPubMedCentralCrossRef
63.
go back to reference Winston DJ, Young JA, Pullarkat V, Papanicolaou GA, Vij R, Vance E, et al. Maribavir prophylaxis for prevention of cytomegalovirus infection in allogeneic stem-cell transplant recipients: a multicenter, randomized, double-blind, placebo-controlled, dose-ranging study. Blood. 2008;111:5403–10.PubMedPubMedCentralCrossRef Winston DJ, Young JA, Pullarkat V, Papanicolaou GA, Vij R, Vance E, et al. Maribavir prophylaxis for prevention of cytomegalovirus infection in allogeneic stem-cell transplant recipients: a multicenter, randomized, double-blind, placebo-controlled, dose-ranging study. Blood. 2008;111:5403–10.PubMedPubMedCentralCrossRef
64.
go back to reference Marty FM, Ljungman P, Papanicolaou GA, Winston DJ, Chemaly RF, Strasfeld L, 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(4):284–92.PubMedCrossRef Marty FM, Ljungman P, Papanicolaou GA, Winston DJ, Chemaly RF, Strasfeld L, 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(4):284–92.PubMedCrossRef
65.
go back to reference Winston DJ, Saliba F, Blumberg E, Abouljoud M, Garcia-Diaz JB, Goss JA, et al. Efficacy and safety of maribavir dosed at 100 mg orally twice daily for the prevention of cytomegalovirus disease in liver transplant recipients: a randomized, double-blind, multicenter controlled trial. Am J Transplant. 2012;12(11):3021–30.PubMedCrossRef Winston DJ, Saliba F, Blumberg E, Abouljoud M, Garcia-Diaz JB, Goss JA, et al. Efficacy and safety of maribavir dosed at 100 mg orally twice daily for the prevention of cytomegalovirus disease in liver transplant recipients: a randomized, double-blind, multicenter controlled trial. Am J Transplant. 2012;12(11):3021–30.PubMedCrossRef
66.
go back to reference Marty FM, Boeckh M. Maribavir and human cytomegalovirus-what happened in the clinical trials and why might the drug have failed? Current opinion in virology. 2011;1(6):555–62.PubMedCrossRef Marty FM, Boeckh M. Maribavir and human cytomegalovirus-what happened in the clinical trials and why might the drug have failed? Current opinion in virology. 2011;1(6):555–62.PubMedCrossRef
67.
go back to reference Snydman DR. Why did maribavir fail in stem-cell transplants? Lancet Infect Dis. 2011;11(4):255–7.PubMedCrossRef Snydman DR. Why did maribavir fail in stem-cell transplants? Lancet Infect Dis. 2011;11(4):255–7.PubMedCrossRef
68.
go back to reference Avery RK, Marty FM, Strasfeld L, Lee I, Arrieta A, Chou S, et al. Oral maribavir for treatment of refractory or resistant cytomegalovirus infections in transplant recipients. Transpl Infect Dis. 2010;12(6):489–96.PubMedCrossRef Avery RK, Marty FM, Strasfeld L, Lee I, Arrieta A, Chou S, et al. Oral maribavir for treatment of refractory or resistant cytomegalovirus infections in transplant recipients. Transpl Infect Dis. 2010;12(6):489–96.PubMedCrossRef
69.
go back to reference • Maertens J, Cordonnier C, Jaksch P, Poire X, Uknis M, Wu J, et al. Maribavir for preemptive treatment of cytomegalovirus reactivation. N Engl J Med. 2019;381(12):1136–47 This study demonstrated the potential utility of maribavir as preemptive therapy in HCT and SOT recipients. A phase 3 study in HCT recipients is underway. PubMedCrossRef • Maertens J, Cordonnier C, Jaksch P, Poire X, Uknis M, Wu J, et al. Maribavir for preemptive treatment of cytomegalovirus reactivation. N Engl J Med. 2019;381(12):1136–47 This study demonstrated the potential utility of maribavir as preemptive therapy in HCT and SOT recipients. A phase 3 study in HCT recipients is underway. PubMedCrossRef
70.
go back to reference Chou S, Marousek GI. Accelerated evolution of maribavir resistance in a cytomegalovirus exonuclease domain II mutant. J Virol. 2008;82(1):246–53.PubMedCrossRef Chou S, Marousek GI. Accelerated evolution of maribavir resistance in a cytomegalovirus exonuclease domain II mutant. J Virol. 2008;82(1):246–53.PubMedCrossRef
71.
go back to reference Chou S, Van Wechel LC, Marousek GI. Cytomegalovirus UL97 kinase mutations that confer maribavir resistance. J Infect Dis. 2007;196(1):91–4.PubMedCrossRef Chou S, Van Wechel LC, Marousek GI. Cytomegalovirus UL97 kinase mutations that confer maribavir resistance. J Infect Dis. 2007;196(1):91–4.PubMedCrossRef
72.
go back to reference Strasfeld L, Lee I, Villano S, Chou S. Virologic characterization of multi-drug-resistant cytomegalovirus infection in two transplant recipients treated with maribavir. J Infect Dis. 2010;202(1):104–8.PubMedCrossRef Strasfeld L, Lee I, Villano S, Chou S. Virologic characterization of multi-drug-resistant cytomegalovirus infection in two transplant recipients treated with maribavir. J Infect Dis. 2010;202(1):104–8.PubMedCrossRef
73.
go back to reference Chou S. Cytomegalovirus UL97 mutations in the era of ganciclovir and maribavir. Rev Med Virol. 2008;18(4):233–46.PubMedCrossRef Chou S. Cytomegalovirus UL97 mutations in the era of ganciclovir and maribavir. Rev Med Virol. 2008;18(4):233–46.PubMedCrossRef
74.
go back to reference Chou S, Wu J, Song K, Bo T. Novel UL97 drug resistance mutations identified at baseline in a clinical trial of maribavir for resistant or refractory cytomegalovirus infection. Antivir Res. 2019;172:104616.PubMedCrossRef Chou S, Wu J, Song K, Bo T. Novel UL97 drug resistance mutations identified at baseline in a clinical trial of maribavir for resistant or refractory cytomegalovirus infection. Antivir Res. 2019;172:104616.PubMedCrossRef
75.
go back to reference Chou S. Diverse cytomegalovirus UL27 mutations adapt to loss of viral UL97 kinase activity under maribavir. Antimicrob Agents Chemother. 2009;53(1):81–5.PubMedCrossRef Chou S. Diverse cytomegalovirus UL27 mutations adapt to loss of viral UL97 kinase activity under maribavir. Antimicrob Agents Chemother. 2009;53(1):81–5.PubMedCrossRef
76.
go back to reference Chou S, Marousek GI, Senters AE, Davis MG, Biron KK. Mutations in the human cytomegalovirus UL27 gene that confer resistance to maribavir. J Virol. 2004;78(13):7124–30.PubMedPubMedCentralCrossRef Chou S, Marousek GI, Senters AE, Davis MG, Biron KK. Mutations in the human cytomegalovirus UL27 gene that confer resistance to maribavir. J Virol. 2004;78(13):7124–30.PubMedPubMedCentralCrossRef
77.
go back to reference Komazin G, Ptak RG, Emmer BT, Townsend LB, Drach JC. Resistance of human cytomegalovirus to the benzimidazole L-ribonucleoside maribavir maps to UL27. J Virol. 2003;77(21):11499–506.PubMedPubMedCentralCrossRef Komazin G, Ptak RG, Emmer BT, Townsend LB, Drach JC. Resistance of human cytomegalovirus to the benzimidazole L-ribonucleoside maribavir maps to UL27. J Virol. 2003;77(21):11499–506.PubMedPubMedCentralCrossRef
78.
go back to reference Zhou S, Breitenbach JM, Borysko KZ, Drach JC, Kern ER, Gullen E, et al. Synthesis and antiviral activity of (Z)- and (E)-2,2-[bis(hydroxymethyl)cyclopropylidene]methylpurines and -pyrimidines: second-generation methylenecyclopropane analogues of nucleosides. J Med Chem. 2004;47(3):566–75.PubMedCrossRef Zhou S, Breitenbach JM, Borysko KZ, Drach JC, Kern ER, Gullen E, et al. Synthesis and antiviral activity of (Z)- and (E)-2,2-[bis(hydroxymethyl)cyclopropylidene]methylpurines and -pyrimidines: second-generation methylenecyclopropane analogues of nucleosides. J Med Chem. 2004;47(3):566–75.PubMedCrossRef
79.
go back to reference Gentry BG, Gentry SN, Jackson TL, Zemlicka J, Drach JC. Phosphorylation of antiviral and endogenous nucleotides to di- and triphosphates by guanosine monophosphate kinase. Biochem Pharmacol. 2011;81(1):43–9.PubMedCrossRef Gentry BG, Gentry SN, Jackson TL, Zemlicka J, Drach JC. Phosphorylation of antiviral and endogenous nucleotides to di- and triphosphates by guanosine monophosphate kinase. Biochem Pharmacol. 2011;81(1):43–9.PubMedCrossRef
80.
go back to reference 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(8):3093–8.PubMedPubMedCentralCrossRef 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(8):3093–8.PubMedPubMedCentralCrossRef
81.
go back to reference Kern ER, Kushner NL, Hartline CB, Williams-Aziz SL, Harden EA, Zhou S, et al. In vitro activity and mechanism of action of methylenecyclopropane analogs of nucleosides against herpesvirus replication. Antimicrob Agents Chemother. 2005;49(3):1039–45.PubMedPubMedCentralCrossRef Kern ER, Kushner NL, Hartline CB, Williams-Aziz SL, Harden EA, Zhou S, et al. In vitro activity and mechanism of action of methylenecyclopropane analogs of nucleosides against herpesvirus replication. Antimicrob Agents Chemother. 2005;49(3):1039–45.PubMedPubMedCentralCrossRef
82.
go back to reference Chen H, Li C, Zemlicka J, Gentry BG, Bowlin TL, Coen DM. Potency and stereoselectivity of cyclopropavir triphosphate action on human cytomegalovirus DNA polymerase. Antimicrob Agents Chemother. 2016;60(7):4176–82.PubMedPubMedCentralCrossRef Chen H, Li C, Zemlicka J, Gentry BG, Bowlin TL, Coen DM. Potency and stereoselectivity of cyclopropavir triphosphate action on human cytomegalovirus DNA polymerase. Antimicrob Agents Chemother. 2016;60(7):4176–82.PubMedPubMedCentralCrossRef
83.
go back to reference Chou S, Bowlin TL. Cytomegalovirus UL97 mutations affecting cyclopropavir and ganciclovir susceptibility. Antimicrob Agents Chemother. 2011;55(1):382–4.PubMedCrossRef Chou S, Bowlin TL. Cytomegalovirus UL97 mutations affecting cyclopropavir and ganciclovir susceptibility. Antimicrob Agents Chemother. 2011;55(1):382–4.PubMedCrossRef
84.
go back to reference Chou S. Recombinant phenotyping of cytomegalovirus UL97 kinase sequence variants for ganciclovir resistance. Antimicrob Agents Chemother. 2010;54(6):2371–8.PubMedPubMedCentralCrossRef Chou S. Recombinant phenotyping of cytomegalovirus UL97 kinase sequence variants for ganciclovir resistance. Antimicrob Agents Chemother. 2010;54(6):2371–8.PubMedPubMedCentralCrossRef
85.
go back to reference Chou S, Marousek G, Bowlin TL. Cyclopropavir susceptibility of cytomegalovirus DNA polymerase mutants selected after antiviral drug exposure. Antimicrob Agents Chemother. 2012;56(1):197–201.PubMedPubMedCentralCrossRef Chou S, Marousek G, Bowlin TL. Cyclopropavir susceptibility of cytomegalovirus DNA polymerase mutants selected after antiviral drug exposure. Antimicrob Agents Chemother. 2012;56(1):197–201.PubMedPubMedCentralCrossRef
86.
go back to reference Gentry BG, Drach JC. Metabolism of cyclopropavir and ganciclovir in human cytomegalovirus-infected cells. Antimicrob Agents Chemother. 2014;58(4):2329–33.PubMedPubMedCentralCrossRef Gentry BG, Drach JC. Metabolism of cyclopropavir and ganciclovir in human cytomegalovirus-infected cells. Antimicrob Agents Chemother. 2014;58(4):2329–33.PubMedPubMedCentralCrossRef
87.
go back to reference Kern ER, Bidanset DJ, Hartline CB, Yan Z, Zemlicka J, Quenelle DC. Oral activity of a methylenecyclopropane analog, cyclopropavir, in animal models for cytomegalovirus infections. Antimicrob Agents Chemother. 2004;48(12):4745–53.PubMedPubMedCentralCrossRef Kern ER, Bidanset DJ, Hartline CB, Yan Z, Zemlicka J, Quenelle DC. Oral activity of a methylenecyclopropane analog, cyclopropavir, in animal models for cytomegalovirus infections. Antimicrob Agents Chemother. 2004;48(12):4745–53.PubMedPubMedCentralCrossRef
88.
go back to reference Bowlin TL, Brooks J, Zemlicka J. Preclinical pharmacokinetic, Toxicokinetic and toxicology results for cyclopropavir, a promising new agent for the treatment of beta- and gamma-herpesviruses. Antivir Res. 2009;82(2):A46–A7.CrossRef Bowlin TL, Brooks J, Zemlicka J. Preclinical pharmacokinetic, Toxicokinetic and toxicology results for cyclopropavir, a promising new agent for the treatment of beta- and gamma-herpesviruses. Antivir Res. 2009;82(2):A46–A7.CrossRef
89.
go back to reference Rouphael NG, Hurwitz SJ, Hart M, Beck A, Anderson EJ, Deye G, et al. Phase Ib trial to evaluate the safety and pharmacokinetics of multiple ascending doses of filociclovir (MBX-400, cyclopropavir) in healthy volunteers. Antimicrobial agents and chemotherapy. 2019;63(9). Rouphael NG, Hurwitz SJ, Hart M, Beck A, Anderson EJ, Deye G, et al. Phase Ib trial to evaluate the safety and pharmacokinetics of multiple ascending doses of filociclovir (MBX-400, cyclopropavir) in healthy volunteers. Antimicrobial agents and chemotherapy. 2019;63(9).
90.
go back to reference Wu Z, Drach JC, Prichard MN, Yanachkova M, Yanachkov I, Bowlin TL, et al. L-valine ester of cyclopropavir: a new antiviral prodrug. Antivir Chem Chemother. 2009;20(1):37–46.PubMedPubMedCentralCrossRef Wu Z, Drach JC, Prichard MN, Yanachkova M, Yanachkov I, Bowlin TL, et al. L-valine ester of cyclopropavir: a new antiviral prodrug. Antivir Chem Chemother. 2009;20(1):37–46.PubMedPubMedCentralCrossRef
91.
go back to reference James SH, Hartline CB, Harden EA, Driebe EM, Schupp JM, Engelthaler DM, et al. Cyclopropavir inhibits the normal function of the human cytomegalovirus UL97 kinase. Antimicrob Agents Chemother. 2011;55(10):4682–91.PubMedPubMedCentralCrossRef James SH, Hartline CB, Harden EA, Driebe EM, Schupp JM, Engelthaler DM, et al. Cyclopropavir inhibits the normal function of the human cytomegalovirus UL97 kinase. Antimicrob Agents Chemother. 2011;55(10):4682–91.PubMedPubMedCentralCrossRef
92.
go back to reference Beadle JR, Hartline C, Aldern KA, Rodriguez N, Harden E, Kern ER, 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(8):2381–6.PubMedPubMedCentralCrossRef Beadle JR, Hartline C, Aldern KA, Rodriguez N, Harden E, Kern ER, 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(8):2381–6.PubMedPubMedCentralCrossRef
93.
go back to reference • Marty FM, Winston DJ, Chemaly RF, Mullane KM, Shore TB, Papanicolaou GA, 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(2):369–81 This study reported the disappointing results using brincidofovir prophylaxis in HCT recipients. PubMedCrossRef • Marty FM, Winston DJ, Chemaly RF, Mullane KM, Shore TB, Papanicolaou GA, 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(2):369–81 This study reported the disappointing results using brincidofovir prophylaxis in HCT recipients. PubMedCrossRef
94.
go back to reference Marty FM, Winston DJ, Rowley SD, Vance E, Papanicolaou GA, Mullane KM, et al. CMX001 to prevent cytomegalovirus disease in hematopoietic-cell transplantation. N Engl J Med. 2013;369(13):1227–36.PubMedCrossRef Marty FM, Winston DJ, Rowley SD, Vance E, Papanicolaou GA, Mullane KM, et al. CMX001 to prevent cytomegalovirus disease in hematopoietic-cell transplantation. N Engl J Med. 2013;369(13):1227–36.PubMedCrossRef
95.
go back to reference Arvin AM, Fast P, Myers M, Plotkin S, Rabinovich R. Vaccine development to prevent cytomegalovirus disease: report from the National Vaccine Advisory Committee. Clin Infect Dis. 2004;39(2):233–9.PubMedCrossRef Arvin AM, Fast P, Myers M, Plotkin S, Rabinovich R. Vaccine development to prevent cytomegalovirus disease: report from the National Vaccine Advisory Committee. Clin Infect Dis. 2004;39(2):233–9.PubMedCrossRef
96.
go back to reference Diamond DJ, La Rosa C, Chiuppesi F, Contreras H, Dadwal S, Wussow F, et al. A fifty-year odyssey: prospects for a cytomegalovirus vaccine in transplant and congenital infection. Expert review of vaccines. 2018;17(10):889–911.PubMedPubMedCentralCrossRef Diamond DJ, La Rosa C, Chiuppesi F, Contreras H, Dadwal S, Wussow F, et al. A fifty-year odyssey: prospects for a cytomegalovirus vaccine in transplant and congenital infection. Expert review of vaccines. 2018;17(10):889–911.PubMedPubMedCentralCrossRef
97.
go back to reference Sylwester AW, Mitchell BL, Edgar JB, Taormina C, Pelte C, Ruchti F, et al. Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med. 2005;202(5):673–85.PubMedPubMedCentralCrossRef Sylwester AW, Mitchell BL, Edgar JB, Taormina C, Pelte C, Ruchti F, et al. Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med. 2005;202(5):673–85.PubMedPubMedCentralCrossRef
98.
go back to reference Vincenti F, Budde K, Merville P, Shihab F, Ram Peddi V, Shah M, et al. A randomized, phase 2 study of ASP0113, a DNA-based vaccine, for the prevention of CMV in CMV-seronegative kidney transplant recipients receiving a kidney from a CMV-seropositive donor. Am J Transplant. 2018;18(12):2945–54.PubMedCrossRef Vincenti F, Budde K, Merville P, Shihab F, Ram Peddi V, Shah M, et al. A randomized, phase 2 study of ASP0113, a DNA-based vaccine, for the prevention of CMV in CMV-seronegative kidney transplant recipients receiving a kidney from a CMV-seropositive donor. Am J Transplant. 2018;18(12):2945–54.PubMedCrossRef
100.
go back to reference Wang D, Freed DC, He X, Li F, Tang A, Cox KS, et al. A replication-defective human cytomegalovirus vaccine for prevention of congenital infection. Science translational medicine. 2016;8(362):362ra145.PubMedCrossRef Wang D, Freed DC, He X, Li F, Tang A, Cox KS, et al. A replication-defective human cytomegalovirus vaccine for prevention of congenital infection. Science translational medicine. 2016;8(362):362ra145.PubMedCrossRef
101.
go back to reference Adler SP, Lewis N, Conlon A, Christiansen MP, Al-Ibrahim M, Rupp R, et al. Phase 1 clinical trial of a conditionally replication-defective human cytomegalovirus (CMV) vaccine in CMV-seronegative subjects. J Infect Dis. 2019;220(3):411–9.PubMedCrossRef Adler SP, Lewis N, Conlon A, Christiansen MP, Al-Ibrahim M, Rupp R, et al. Phase 1 clinical trial of a conditionally replication-defective human cytomegalovirus (CMV) vaccine in CMV-seronegative subjects. J Infect Dis. 2019;220(3):411–9.PubMedCrossRef
102.
go back to reference Liu Y, Freed DC, Li L, Tang A, Li F, Murray EM, et al. A replication-defective human cytomegalovirus vaccine elicits humoral immune responses analogous to those with natural infection. J Virol. 2019;93(23). Liu Y, Freed DC, Li L, Tang A, Li F, Murray EM, et al. A replication-defective human cytomegalovirus vaccine elicits humoral immune responses analogous to those with natural infection. J Virol. 2019;93(23).
103.
go back to reference Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science (New York), NY. 1992;257(5067):238–241. Riddell SR, Watanabe KS, Goodrich JM, Li CR, Agha ME, Greenberg PD. Restoration of viral immunity in immunodeficient humans by the adoptive transfer of T cell clones. Science (New York), NY. 1992;257(5067):238–241.
104.
go back to reference Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED, et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med. 1995;333(16):1038–44.PubMedCrossRef Walter EA, Greenberg PD, Gilbert MJ, Finch RJ, Watanabe KS, Thomas ED, et al. Reconstitution of cellular immunity against cytomegalovirus in recipients of allogeneic bone marrow by transfer of T-cell clones from the donor. N Engl J Med. 1995;333(16):1038–44.PubMedCrossRef
105.
go back to reference Feuchtinger T, Opherk K, Bethge WA, Topp MS, Schuster FR, Weissinger EM, et al. Adoptive transfer of pp65-specific T cells for the treatment of chemorefractory cytomegalovirus disease or reactivation after haploidentical and matched unrelated stem cell transplantation. Blood. 2010;116(20):4360–7.PubMedCrossRef Feuchtinger T, Opherk K, Bethge WA, Topp MS, Schuster FR, Weissinger EM, et al. Adoptive transfer of pp65-specific T cells for the treatment of chemorefractory cytomegalovirus disease or reactivation after haploidentical and matched unrelated stem cell transplantation. Blood. 2010;116(20):4360–7.PubMedCrossRef
106.
go back to reference Peggs KS, Thomson K, Samuel E, Dyer G, Armoogum J, Chakraverty R, et al. Directly selected cytomegalovirus-reactive donor T cells confer rapid and safe systemic reconstitution of virus-specific immunity following stem cell transplantation. Clin Infect Dis. 2011;52(1):49–57.PubMedCrossRef Peggs KS, Thomson K, Samuel E, Dyer G, Armoogum J, Chakraverty R, et al. Directly selected cytomegalovirus-reactive donor T cells confer rapid and safe systemic reconstitution of virus-specific immunity following stem cell transplantation. Clin Infect Dis. 2011;52(1):49–57.PubMedCrossRef
107.
go back to reference Einsele H, Roosnek E, Rufer N, Sinzger C, Riegler S, Loffler J, et al. Infusion of cytomegalovirus (CMV)-specific T cells for the treatment of CMV infection not responding to antiviral chemotherapy. Blood. 2002;99(11):3916–22.PubMedCrossRef Einsele H, Roosnek E, Rufer N, Sinzger C, Riegler S, Loffler J, et al. Infusion of cytomegalovirus (CMV)-specific T cells for the treatment of CMV infection not responding to antiviral chemotherapy. Blood. 2002;99(11):3916–22.PubMedCrossRef
108.
go back to reference Pei XY, Zhao XY, Chang YJ, Liu J, Xu LP, Wang Y, et al. Cytomegalovirus-specific T-cell transfer for refractory cytomegalovirus infection after Haploidentical stem cell transplantation: the quantitative and qualitative immune recovery for cytomegalovirus. J Infect Dis. 2017;216(8):945–56.PubMedCrossRef Pei XY, Zhao XY, Chang YJ, Liu J, Xu LP, Wang Y, et al. Cytomegalovirus-specific T-cell transfer for refractory cytomegalovirus infection after Haploidentical stem cell transplantation: the quantitative and qualitative immune recovery for cytomegalovirus. J Infect Dis. 2017;216(8):945–56.PubMedCrossRef
109.
go back to reference Blyth E, Clancy L, Simms R, Ma CK, Burgess J, Deo S, et al. Donor-derived CMV-specific T cells reduce the requirement for CMV-directed pharmacotherapy after allogeneic stem cell transplantation. Blood. 2013;121(18):3745–58.PubMedCrossRef Blyth E, Clancy L, Simms R, Ma CK, Burgess J, Deo S, et al. Donor-derived CMV-specific T cells reduce the requirement for CMV-directed pharmacotherapy after allogeneic stem cell transplantation. Blood. 2013;121(18):3745–58.PubMedCrossRef
110.
go back to reference 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(4):1130–5.PubMedPubMedCentralCrossRef 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(4):1130–5.PubMedPubMedCentralCrossRef
111.
go back to reference Mackinnon S, Thomson K, Verfuerth S, Peggs K, Lowdell M. Adoptive cellular therapy for cytomegalovirus infection following allogeneic stem cell transplantation using virus-specific T cells. Blood Cells Mol Dis. 2008;40(1):63–7.PubMedCrossRef Mackinnon S, Thomson K, Verfuerth S, Peggs K, Lowdell M. Adoptive cellular therapy for cytomegalovirus infection following allogeneic stem cell transplantation using virus-specific T cells. Blood Cells Mol Dis. 2008;40(1):63–7.PubMedCrossRef
112.
go back to reference Micklethwaite K, Hansen A, Foster A, Snape E, Antonenas V, Sartor M, et al. Ex vivo expansion and prophylactic infusion of CMV-pp65 peptide-specific cytotoxic T-lymphocytes following allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2007;13(6):707–14.PubMedCrossRef Micklethwaite K, Hansen A, Foster A, Snape E, Antonenas V, Sartor M, et al. Ex vivo expansion and prophylactic infusion of CMV-pp65 peptide-specific cytotoxic T-lymphocytes following allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2007;13(6):707–14.PubMedCrossRef
113.
go back to reference Micklethwaite KP, Clancy L, Sandher U, Hansen AM, Blyth E, Antonenas V, et al. Prophylactic infusion of cytomegalovirus-specific cytotoxic T lymphocytes stimulated with Ad5f35pp65 gene-modified dendritic cells after allogeneic hemopoietic stem cell transplantation. Blood. 2008;112(10):3974–81.PubMedCrossRef Micklethwaite KP, Clancy L, Sandher U, Hansen AM, Blyth E, Antonenas V, et al. Prophylactic infusion of cytomegalovirus-specific cytotoxic T lymphocytes stimulated with Ad5f35pp65 gene-modified dendritic cells after allogeneic hemopoietic stem cell transplantation. Blood. 2008;112(10):3974–81.PubMedCrossRef
114.
go back to reference Peggs KS, Verfuerth S, Pizzey A, Khan N, Guiver M, Moss PA, et al. Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines. Lancet. 2003;362(9393):1375–7.PubMedCrossRef Peggs KS, Verfuerth S, Pizzey A, Khan N, Guiver M, Moss PA, et al. Adoptive cellular therapy for early cytomegalovirus infection after allogeneic stem-cell transplantation with virus-specific T-cell lines. Lancet. 2003;362(9393):1375–7.PubMedCrossRef
115.
go back to reference Leen AM, Bollard CM, Mendizabal AM, Shpall EJ, Szabolcs P, Antin JH, et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood. 2013;121(26):5113–23.PubMedPubMedCentralCrossRef Leen AM, Bollard CM, Mendizabal AM, Shpall EJ, Szabolcs P, Antin JH, et al. Multicenter study of banked third-party virus-specific T cells to treat severe viral infections after hematopoietic stem cell transplantation. Blood. 2013;121(26):5113–23.PubMedPubMedCentralCrossRef
116.
go back to reference • Tzannou I, Papadopoulou A, Naik S, Leung K, Martinez CA, Ramos CA, et al. Off-the-shelf virus-specific T cells to treat BK virus, human herpesvirus 6, cytomegalovirus, Epstein-Barr virus, and adenovirus infections after allogeneic hematopoietic stem-cell transplantation. J Clin Oncol. 2017;35(31):3547–57 Provides additional support for the safety of adoptive immunotherapy using third-party donor T cells. PubMedPubMedCentralCrossRef • Tzannou I, Papadopoulou A, Naik S, Leung K, Martinez CA, Ramos CA, et al. Off-the-shelf virus-specific T cells to treat BK virus, human herpesvirus 6, cytomegalovirus, Epstein-Barr virus, and adenovirus infections after allogeneic hematopoietic stem-cell transplantation. J Clin Oncol. 2017;35(31):3547–57 Provides additional support for the safety of adoptive immunotherapy using third-party donor T cells. PubMedPubMedCentralCrossRef
117.
go back to reference • Withers B, Blyth E, Clancy LE, Yong A, Fraser C, Burgess J, et al. Long-term control of recurrent or refractory viral infections after allogeneic HSCT with third-party virus-specific T cells. Blood Adv. 2017;1(24):2193–205 Provides additional support for the safety of adoptive immunotherapy using third-party donor T cells. PubMedPubMedCentralCrossRef • Withers B, Blyth E, Clancy LE, Yong A, Fraser C, Burgess J, et al. Long-term control of recurrent or refractory viral infections after allogeneic HSCT with third-party virus-specific T cells. Blood Adv. 2017;1(24):2193–205 Provides additional support for the safety of adoptive immunotherapy using third-party donor T cells. PubMedPubMedCentralCrossRef
118.
go back to reference Boeckh M, Corey L. Adoptive immunotherapy of viral infections: should infectious disease embrace cellular immunotherapy? J Infect Dis. 2017;216(8):926–8.PubMedCrossRef Boeckh M, Corey L. Adoptive immunotherapy of viral infections: should infectious disease embrace cellular immunotherapy? J Infect Dis. 2017;216(8):926–8.PubMedCrossRef
119.
go back to reference Ljungman P, Cordonnier C, Einsele H, Bender-Gotze C, Bosi A, Dekker A, et al. Use of intravenous immune globulin in addition to antiviral therapy in the treatment of CMV gastrointestinal disease in allogeneic bone marrow transplant patients: a report from the European Group for Blood and Marrow Transplantation (EBMT). Infectious Diseases Working Party of the EBMT. Bone Marrow Transplant. 1998;21(5):473–6.PubMedCrossRef Ljungman P, Cordonnier C, Einsele H, Bender-Gotze C, Bosi A, Dekker A, et al. Use of intravenous immune globulin in addition to antiviral therapy in the treatment of CMV gastrointestinal disease in allogeneic bone marrow transplant patients: a report from the European Group for Blood and Marrow Transplantation (EBMT). Infectious Diseases Working Party of the EBMT. Bone Marrow Transplant. 1998;21(5):473–6.PubMedCrossRef
120.
go back to reference Alexander BT, Hladnik LM, Augustin KM, Casabar E, McKinnon PS, Reichley RM, et al. Use of cytomegalovirus intravenous immune globulin for the adjunctive treatment of cytomegalovirus in hematopoietic stem cell transplant recipients. Pharmacotherapy. 2010;30(6):554–61.PubMedPubMedCentralCrossRef Alexander BT, Hladnik LM, Augustin KM, Casabar E, McKinnon PS, Reichley RM, et al. Use of cytomegalovirus intravenous immune globulin for the adjunctive treatment of cytomegalovirus in hematopoietic stem cell transplant recipients. Pharmacotherapy. 2010;30(6):554–61.PubMedPubMedCentralCrossRef
121.
go back to reference Emanuel D, Cunningham I, Jules-Elysee K, Brochstein JA, Kernan NA, Laver J, et al. Cytomegalovirus pneumonia after bone marrow transplantation successfully treated with the combination of ganciclovir and high-dose intravenous immune globulin. Ann Intern Med. 1988;109(10):777–82.PubMedCrossRef Emanuel D, Cunningham I, Jules-Elysee K, Brochstein JA, Kernan NA, Laver J, et al. Cytomegalovirus pneumonia after bone marrow transplantation successfully treated with the combination of ganciclovir and high-dose intravenous immune globulin. Ann Intern Med. 1988;109(10):777–82.PubMedCrossRef
122.
go back to reference Reed EC, Bowden RA, Dandliker PS, Lilleby KE, Meyers JD. Treatment of cytomegalovirus pneumonia with ganciclovir and intravenous cytomegalovirus immunoglobulin in patients with bone marrow transplants. Ann Intern Med. 1988;109(10):783–8.PubMedCrossRef Reed EC, Bowden RA, Dandliker PS, Lilleby KE, Meyers JD. Treatment of cytomegalovirus pneumonia with ganciclovir and intravenous cytomegalovirus immunoglobulin in patients with bone marrow transplants. Ann Intern Med. 1988;109(10):783–8.PubMedCrossRef
123.
go back to reference Erard V, Guthrie KA, Seo S, Smith J, Huang M, Chien J, et al. Reduced mortality of cytomegalovirus pneumonia after hematopoietic cell transplantation due to antiviral therapy and changes in transplantation practices. Clin Infect Dis. 2015;61(1):31–9.PubMedPubMedCentralCrossRef Erard V, Guthrie KA, Seo S, Smith J, Huang M, Chien J, et al. Reduced mortality of cytomegalovirus pneumonia after hematopoietic cell transplantation due to antiviral therapy and changes in transplantation practices. Clin Infect Dis. 2015;61(1):31–9.PubMedPubMedCentralCrossRef
124.
go back to reference Bowden RA, Fisher LD, Rogers K, Cays M, Meyers JD. Cytomegalovirus (CMV)-specific intravenous immunoglobulin for the prevention of primary CMV infection and disease after marrow transplant [see comments]. J Infect Dis. 1991;164(3):483–7.PubMedCrossRef Bowden RA, Fisher LD, Rogers K, Cays M, Meyers JD. Cytomegalovirus (CMV)-specific intravenous immunoglobulin for the prevention of primary CMV infection and disease after marrow transplant [see comments]. J Infect Dis. 1991;164(3):483–7.PubMedCrossRef
125.
go back to reference Bowden RA, Sayers M, Flournoy N, Newton B, Banaji M, Thomas ED, et al. Cytomegalovirus immune globulin and seronegative blood products to prevent primary cytomegalovirus infection after marrow transplantation. N Engl J Med. 1986;314(16):1006–10.PubMedCrossRef Bowden RA, Sayers M, Flournoy N, Newton B, Banaji M, Thomas ED, et al. Cytomegalovirus immune globulin and seronegative blood products to prevent primary cytomegalovirus infection after marrow transplantation. N Engl J Med. 1986;314(16):1006–10.PubMedCrossRef
126.
go back to reference Ruutu T, Ljungman P, Brinch L, Lenhoff S, Lonnqvist B, Ringden O, et al. No prevention of cytomegalovirus infection by anti-cytomegalovirus hyperimmune globulin in seronegative bone marrow transplant recipients. The Nordic BMT Group. Bone Marrow Transplant. 1997;19(3):233–6.PubMedCrossRef Ruutu T, Ljungman P, Brinch L, Lenhoff S, Lonnqvist B, Ringden O, et al. No prevention of cytomegalovirus infection by anti-cytomegalovirus hyperimmune globulin in seronegative bone marrow transplant recipients. The Nordic BMT Group. Bone Marrow Transplant. 1997;19(3):233–6.PubMedCrossRef
127.
go back to reference Bass E, Powe N, Goodman S, Graziano S, Griffiths R, Kickler T, et al. Efficacy of immune globulin in preventing complications of bone marrow transplantation: a meta-analysis. Bone Marrow Transplant. 1993;12:179–83. Bass E, Powe N, Goodman S, Graziano S, Griffiths R, Kickler T, et al. Efficacy of immune globulin in preventing complications of bone marrow transplantation: a meta-analysis. Bone Marrow Transplant. 1993;12:179–83.
128.
go back to reference Messori A, Rampazzo R, Scroccaro G, Martini N. Efficacy of hyperimmune anti-cytomegalovirus immunoglobulins for the prevention of cytomegalovirus infection in recipients of allogeneic bone marrow transplantation: a meta analysis. Bone Marrow Transplant. 1994;13:163–8.PubMed Messori A, Rampazzo R, Scroccaro G, Martini N. Efficacy of hyperimmune anti-cytomegalovirus immunoglobulins for the prevention of cytomegalovirus infection in recipients of allogeneic bone marrow transplantation: a meta analysis. Bone Marrow Transplant. 1994;13:163–8.PubMed
129.
go back to reference Raanani P, Gafter-Gvili A, Paul M, Ben-Bassat I, Leibovici L, Shpilberg O. Immunoglobulin prophylaxis in patients undergoing haematopoietic stem cell transplantation: systematic review and meta-analysis. Bone Marrow Transplantation. 2008:S46 (abstract O267). Raanani P, Gafter-Gvili A, Paul M, Ben-Bassat I, Leibovici L, Shpilberg O. Immunoglobulin prophylaxis in patients undergoing haematopoietic stem cell transplantation: systematic review and meta-analysis. Bone Marrow Transplantation. 2008:S46 (abstract O267).
130.
go back to reference Sullivan KM, Kopecky KJ, Jocom J, Fisher L, Buckner CD, Meyers JD, et al. Immunomodulatory and antimicrobial efficacy of intravenous immunoglobulin in bone marrow transplantation. N Engl J Med. 1990;323(11):705–12.PubMedCrossRef Sullivan KM, Kopecky KJ, Jocom J, Fisher L, Buckner CD, Meyers JD, et al. Immunomodulatory and antimicrobial efficacy of intravenous immunoglobulin in bone marrow transplantation. N Engl J Med. 1990;323(11):705–12.PubMedCrossRef
131.
go back to reference Winston DJ, Ho WG, Lin CH, Bartoni K, Budinger MD, Gale RP, et al. Intravenous immune globulin for prevention of cytomegalovirus infection and interstitial pneumonia after bone marrow transplantation. Ann Intern Med. 1987;106(1):12–8.PubMedCrossRef Winston DJ, Ho WG, Lin CH, Bartoni K, Budinger MD, Gale RP, et al. Intravenous immune globulin for prevention of cytomegalovirus infection and interstitial pneumonia after bone marrow transplantation. Ann Intern Med. 1987;106(1):12–8.PubMedCrossRef
132.
go back to reference Zikos P, Van Lint MT, Lamparelli T, Gualandi F, Occhini D, Mordini N, et al. A randomized trial of high dose polyvalent intravenous immunoglobulin (HDIgG) vs. cytomegalovirus (CMV) hyperimmune IgG in allogeneic hemopoietic stem cell transplants (HSCT). Haematologica. 1998;83(2):132–7.PubMed Zikos P, Van Lint MT, Lamparelli T, Gualandi F, Occhini D, Mordini N, et al. A randomized trial of high dose polyvalent intravenous immunoglobulin (HDIgG) vs. cytomegalovirus (CMV) hyperimmune IgG in allogeneic hemopoietic stem cell transplants (HSCT). Haematologica. 1998;83(2):132–7.PubMed
133.
go back to reference Dole K, Segal FP, Feire A, Magnusson B, Rondon JC, Vemula J, et al. A first-in-human study to assess the safety and pharmacokinetics of monoclonal antibodies against human cytomegalovirus in healthy volunteers. Antimicrob Agents Chemother. 2016;60(5):2881–7.PubMedPubMedCentralCrossRef Dole K, Segal FP, Feire A, Magnusson B, Rondon JC, Vemula J, et al. A first-in-human study to assess the safety and pharmacokinetics of monoclonal antibodies against human cytomegalovirus in healthy volunteers. Antimicrob Agents Chemother. 2016;60(5):2881–7.PubMedPubMedCentralCrossRef
134.
go back to reference Boeckh M, Bowden R, Storer B, Chao N, Spielberger R, Tierney D, et al. Randomized, placebo-controlled, double-blind study of a cytomegalovirus-specific monoclonal antibody (MSL-109) for prevention of cytomegalovirus infection after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2001;7(6):343–51.PubMedCrossRef Boeckh M, Bowden R, Storer B, Chao N, Spielberger R, Tierney D, et al. Randomized, placebo-controlled, double-blind study of a cytomegalovirus-specific monoclonal antibody (MSL-109) for prevention of cytomegalovirus infection after allogeneic hematopoietic stem cell transplantation. Biol Blood Marrow Transplant. 2001;7(6):343–51.PubMedCrossRef
135.
go back to reference Gentry BG, Bogner E, Drach JC. Targeting the terminase: an important step forward in the treatment and prophylaxis of human cytomegalovirus infections. Antivir Res. 2019;161:116–24.PubMedCrossRef Gentry BG, Bogner E, Drach JC. Targeting the terminase: an important step forward in the treatment and prophylaxis of human cytomegalovirus infections. Antivir Res. 2019;161:116–24.PubMedCrossRef
136.
go back to reference Saag MS, Benson CA, Gandhi RT, Hoy JF, Landovitz RJ, Mugavero MJ, et al. Antiretroviral drugs for treatment and prevention of HIV infection in adults: 2018 recommendations of the International Antiviral Society-USA Panel. JAMA. 2018;320(4):379–96.PubMedPubMedCentralCrossRef Saag MS, Benson CA, Gandhi RT, Hoy JF, Landovitz RJ, Mugavero MJ, et al. Antiretroviral drugs for treatment and prevention of HIV infection in adults: 2018 recommendations of the International Antiviral Society-USA Panel. JAMA. 2018;320(4):379–96.PubMedPubMedCentralCrossRef
137.
go back to reference Panel A-IHG. Hepatitis C guidance 2018 update: AASLD-IDSA recommendations for testing, managing, and treating hepatitis C virus infection. Clin Infect Dis. 2018;67(10):1477–92.CrossRef Panel A-IHG. Hepatitis C guidance 2018 update: AASLD-IDSA recommendations for testing, managing, and treating hepatitis C virus infection. Clin Infect Dis. 2018;67(10):1477–92.CrossRef
138.
go back to reference Kornblit B, Maloney DG, Storer BE, Maris MB, Vindelov L, Hari P, et al. A randomized phase II trial of tacrolimus, mycophenolate mofetil and sirolimus after non-myeloablative unrelated donor transplantation. Haematologica. 2014;99(10):1624–31.PubMedPubMedCentralCrossRef Kornblit B, Maloney DG, Storer BE, Maris MB, Vindelov L, Hari P, et al. A randomized phase II trial of tacrolimus, mycophenolate mofetil and sirolimus after non-myeloablative unrelated donor transplantation. Haematologica. 2014;99(10):1624–31.PubMedPubMedCentralCrossRef
139.
go back to reference Kudchodkar SB, Yu Y, Maguire TG, Alwine JC. Human cytomegalovirus infection induces rapamycin-insensitive phosphorylation of downstream effectors of mTOR kinase. J Virol. 2004;78(20):11030–9.PubMedPubMedCentralCrossRef Kudchodkar SB, Yu Y, Maguire TG, Alwine JC. Human cytomegalovirus infection induces rapamycin-insensitive phosphorylation of downstream effectors of mTOR kinase. J Virol. 2004;78(20):11030–9.PubMedPubMedCentralCrossRef
140.
go back to reference Kudchodkar SB, Yu Y, Maguire TG, Alwine JC. Human cytomegalovirus infection alters the substrate specificities and rapamycin sensitivities of raptor- and rictor-containing complexes. Proc Natl Acad Sci U S A. 2006;103(38):14182–7.PubMedPubMedCentralCrossRef Kudchodkar SB, Yu Y, Maguire TG, Alwine JC. Human cytomegalovirus infection alters the substrate specificities and rapamycin sensitivities of raptor- and rictor-containing complexes. Proc Natl Acad Sci U S A. 2006;103(38):14182–7.PubMedPubMedCentralCrossRef
141.
go back to reference Marty FM, Bryar J, Browne SK, Schwarzberg T, Ho VT, Bassett IV, et al. Sirolimus-based graft-versus-host disease prophylaxis protects against cytomegalovirus reactivation after allogeneic hematopoietic stem cell transplantation: a cohort analysis. Blood. 2007;110(2):490–500.PubMedPubMedCentralCrossRef Marty FM, Bryar J, Browne SK, Schwarzberg T, Ho VT, Bassett IV, et al. Sirolimus-based graft-versus-host disease prophylaxis protects against cytomegalovirus reactivation after allogeneic hematopoietic stem cell transplantation: a cohort analysis. Blood. 2007;110(2):490–500.PubMedPubMedCentralCrossRef
142.
go back to reference Sandmaier BM, Kornblit B, Storer BE, Olesen G, Maris MB, Langston AA, et al. Addition of sirolimus to standard cyclosporine plus mycophenolate mofetil-based graft-versus-host disease prophylaxis for patients after unrelated non-myeloablative haemopoietic stem cell transplantation: a multicentre, randomised, phase 3 trial. Lancet Haematol. 2019;6(8):e409–e18.PubMedCrossRef Sandmaier BM, Kornblit B, Storer BE, Olesen G, Maris MB, Langston AA, et al. Addition of sirolimus to standard cyclosporine plus mycophenolate mofetil-based graft-versus-host disease prophylaxis for patients after unrelated non-myeloablative haemopoietic stem cell transplantation: a multicentre, randomised, phase 3 trial. Lancet Haematol. 2019;6(8):e409–e18.PubMedCrossRef
143.
go back to reference Efferth T, Romero MR, Wolf DG, Stamminger T, Marin JJ, Marschall M. The antiviral activities of artemisinin and artesunate. Clin Infect Dis. 2008;47(6):804–11.PubMedCrossRef Efferth T, Romero MR, Wolf DG, Stamminger T, Marin JJ, Marschall M. The antiviral activities of artemisinin and artesunate. Clin Infect Dis. 2008;47(6):804–11.PubMedCrossRef
144.
go back to reference Shapira MY, Resnick IB, Chou S, Neumann AU, Lurain NS, Stamminger T, et al. Artesunate as a potent antiviral agent in a patient with late drug-resistant cytomegalovirus infection after hematopoietic stem cell transplantation. Clin Infect Dis. 2008;46(9):1455–7.PubMedCrossRef Shapira MY, Resnick IB, Chou S, Neumann AU, Lurain NS, Stamminger T, et al. Artesunate as a potent antiviral agent in a patient with late drug-resistant cytomegalovirus infection after hematopoietic stem cell transplantation. Clin Infect Dis. 2008;46(9):1455–7.PubMedCrossRef
145.
go back to reference Wolf DG, Shimoni A, Resnick IB, Stamminger T, Neumann AU, Chou S, et al. Human cytomegalovirus kinetics following institution of artesunate after hematopoietic stem cell transplantation. Antivir Res. 2011;90(3):183–6.PubMedCrossRef Wolf DG, Shimoni A, Resnick IB, Stamminger T, Neumann AU, Chou S, et al. Human cytomegalovirus kinetics following institution of artesunate after hematopoietic stem cell transplantation. Antivir Res. 2011;90(3):183–6.PubMedCrossRef
146.
go back to reference Chong AS, Zeng H, Knight DA, Shen J, Meister GT, Williams JW, et al. Concurrent antiviral and immunosuppressive activities of leflunomide in vivo. Am J Transplant. 2006;6(1):69–75.PubMedCrossRef Chong AS, Zeng H, Knight DA, Shen J, Meister GT, Williams JW, et al. Concurrent antiviral and immunosuppressive activities of leflunomide in vivo. Am J Transplant. 2006;6(1):69–75.PubMedCrossRef
147.
go back to reference Waldman WJ, Knight DA, Blinder L, Shen J, Lurain NS, Miller DM, et al. Inhibition of cytomegalovirus in vitro and in vivo by the experimental immunosuppressive agent leflunomide. Intervirology. 1999;42(5–6):412–8.PubMedCrossRef Waldman WJ, Knight DA, Blinder L, Shen J, Lurain NS, Miller DM, et al. Inhibition of cytomegalovirus in vitro and in vivo by the experimental immunosuppressive agent leflunomide. Intervirology. 1999;42(5–6):412–8.PubMedCrossRef
148.
go back to reference Waldman WJ, Knight DA, Lurain NS, Miller DM, Sedmak DD, Williams JW, et al. Novel mechanism of inhibition of cytomegalovirus by the experimental immunosuppressive agent leflunomide. Transplantation. 1999;68(6):814–25.PubMedCrossRef Waldman WJ, Knight DA, Lurain NS, Miller DM, Sedmak DD, Williams JW, et al. Novel mechanism of inhibition of cytomegalovirus by the experimental immunosuppressive agent leflunomide. Transplantation. 1999;68(6):814–25.PubMedCrossRef
149.
go back to reference Zhang C, Chu M. Leflunomide: a promising drug with good antitumor potential. Biochem Biophys Res Commun. 2018;496(2):726–30.PubMedCrossRef Zhang C, Chu M. Leflunomide: a promising drug with good antitumor potential. Biochem Biophys Res Commun. 2018;496(2):726–30.PubMedCrossRef
150.
go back to reference Herget T, Freitag M, Morbitzer M, Kupfer R, Stamminger T, Marschall M. Novel chemical class of pUL97 protein kinase-specific inhibitors with strong anticytomegaloviral activity. Antimicrob Agents Chemother. 2004;48(11):4154–62.PubMedPubMedCentralCrossRef Herget T, Freitag M, Morbitzer M, Kupfer R, Stamminger T, Marschall M. Novel chemical class of pUL97 protein kinase-specific inhibitors with strong anticytomegaloviral activity. Antimicrob Agents Chemother. 2004;48(11):4154–62.PubMedPubMedCentralCrossRef
151.
go back to reference Hutterer C, Hamilton S, Steingruber M, Zeittrager I, Bahsi H, Thuma N, et al. The chemical class of quinazoline compounds provides a core structure for the design of anticytomegaloviral kinase inhibitors. Antivir Res. 2016;134:130–43.PubMedCrossRef Hutterer C, Hamilton S, Steingruber M, Zeittrager I, Bahsi H, Thuma N, et al. The chemical class of quinazoline compounds provides a core structure for the design of anticytomegaloviral kinase inhibitors. Antivir Res. 2016;134:130–43.PubMedCrossRef
152.
go back to reference Schleiss M, Eickhoff J, Auerochs S, Leis M, Abele S, Rechter S, et al. Protein kinase inhibitors of the quinazoline class exert anti-cytomegaloviral activity in vitro and in vivo. Antivir Res. 2008;79(1):49–61.PubMedCrossRef Schleiss M, Eickhoff J, Auerochs S, Leis M, Abele S, Rechter S, et al. Protein kinase inhibitors of the quinazoline class exert anti-cytomegaloviral activity in vitro and in vivo. Antivir Res. 2008;79(1):49–61.PubMedCrossRef
153.
go back to reference Avery RK, Mossad SB, Poggio E, Lard M, Budev M, Bolwell B, et al. Utility of leflunomide in the treatment of complex cytomegalovirus syndromes. Transplantation. 2010;90(4):419–26.PubMedCrossRef Avery RK, Mossad SB, Poggio E, Lard M, Budev M, Bolwell B, et al. Utility of leflunomide in the treatment of complex cytomegalovirus syndromes. Transplantation. 2010;90(4):419–26.PubMedCrossRef
Metadata
Title
Moving Past Ganciclovir and Foscarnet: Advances in CMV Therapy
Author
Morgan Hakki
Publication date
01-04-2020
Publisher
Springer US
Published in
Current Hematologic Malignancy Reports / Issue 2/2020
Print ISSN: 1558-8211
Electronic ISSN: 1558-822X
DOI
https://doi.org/10.1007/s11899-020-00557-6

Other articles of this Issue 2/2020

Current Hematologic Malignancy Reports 2/2020 Go to the issue

Multiple Myeloma (P Kapoor, Section Editor)

Supportive Care in Multiple Myeloma

T-Cell and Other Lymphoproliferative Malignancies (J Zain, Section Editor)

Advances and Perspectives in the Treatment of T-PLL

T-Cell and Other Lymphoproliferative Malignancies (J Zain, Section Editor)

Advances in the Diagnosis and Treatment of Large Granular Lymphocytic Leukemia

Multiple Myelomas (P Kapoor, Section Editor)

Extramedullary Disease in Multiple Myeloma

T-Cell and Other Lymphoproliferative Malignancies (J Zain, Section Editor)

Polatuzumab Vedotin: a New Target for B Cell Malignancies

T-Cell and Other Lymphoproliferative Malignancies (J Zain, Section Editor)

The Cardiovascular Complications of Chimeric Antigen Receptor T Cell Therapy