Page last updated: 2024-11-13

letermovir

Description Research Excerpts Clinical Trials Roles Classes Pathways Study Profile Bioassays Related Drugs Related Conditions Protein Interactions Research Growth Market Indicators

Description

letermovir: has antiviral activity; structure in first source [Medical Subject Headings (MeSH), National Library of Medicine, extracted Dec-2023]

Cross-References

ID SourceID
PubMed CID45138674
CHEMBL ID1241951
SCHEMBL ID379403
MeSH IDM000594403

Synonyms (53)

Synonym
letermovir
2-[(4s)-8-fluoro-2-[4-(3-methoxyphenyl)piperazin-1-yl]-3-[2-methoxy-5-(trifluoromethyl)phenyl]-4h-quinazolin-4-yl]acetic acid
aic246
aic-246
CHEMBL1241951
aic 246
mk-8228
letermovir [usan:inn]
1h09y5wo1f ,
4-quinazolineacetic acid, 8-fluoro-3,4-dihydro-2-(4-(3-methoxyphenyl)-1-piperazinyl)-3-(2-methoxy-5-(trifluoromethyl)phenyl)-, (4s)-
2-((4s)-8-fluoro-2-(4-(3-methoxyphenyl)piperazin-1-yl)-3-(2-methoxy-5-(trifluoromethyl)phenyl)-4h-quinazolin-4-yl)acetic acid
917389-32-3
prevymis
unii-1h09y5wo1f
S8873
letermovir [who-dd]
letermovir [inn]
(4s)-2-{8-fluoro-2-[4-(3-methoxyphenyl)piperazin-1-yl]-3-[2-methoxy-5-(trifluoromethyl)phenyl]-3,4-dihydroquinazolin-4-yl}acetic acid
letermovir [mi]
letermovir [jan]
letermovir [orange book]
letermovir [usan]
CS-1571
HY-15233
(s)-[8-fluoro-2-[4-(3-methoxyphenyl)piperazine-1-yl]-3-(2-methoxy-5-trifluoromethylphenyl)-3,4-dihydroquinazoline-4-yl]acetic acid
(s)-{8-fluoro-2-[4-(3-methoxyphenyl)piperazin-1-yl]-3-(2-methoxy-5-trifluoromethylphenyl)-3,4-dihydro-quinazolin-4-yl}acetic acid
(s)-{8-fluoro-2-[4-(3-methoxyphenyl)piperazine-1-yl]-3-(2-methoxy-5-trifluoromethylphenyl)-3,4-dihydroquinazoline-4-yl}acetic acid
(s)-{8-fluoro-2-[4-(3-methoxyphenyl)piperazin-1-yl]-3-(2-methoxy-5-trifluoromethylphenyl)-3,4-dihydroquinazolin-4-yl}acetic acid
SCHEMBL379403
D10801
prevymis (tn)
letermovir (jan/usan/inn)
DTXSID40238683 ,
AKOS030526995
D71052
NCGC00378936-01
DB12070
(s)-2-(8-fluoro-3-(2-methoxy-5-(trifluoromethyl)phenyl)-2-(4-(3-methoxyphenyl)piperazin-1-yl)-3,4-dihydroquinazolin-4-yl)acetic acid
Q15409407
mk-8828
NCGC00378936-02
EX-A1871
AS-56206
letermovir(aic246)
(4s)-8-fluoro-3,4-dihydro-2-[4-(3-methoxyphenyl)-1-piperazinyl]-3-[2-methoxy-5-(trifluoromethyl)phenyl]-4-quinazolineacetic acid
A902281
EN300-18167086
2-[(4s)-8-fluoro-3-[2-methoxy-5-(trifluoromethyl)phenyl]-2-[4-(3-methoxyphenyl)piperazin-1-yl]-3,4-dihydroquinazolin-4-yl]acetic acid
j05ax18
letermovirum
(4s)-2-(8-fluoro-2-(4-(3-methoxyphenyl)piperazin-1-yl)-3-(2-methoxy-5-(trifluoromethyl)phenyl)-3,4-dihydroquinazolin-4-yl)acetic acid
dtxcid90161174
AC-35698

Research Excerpts

Overview

Letermovir is a human cytomegalovirus (CMV) terminase inhibitor for the prophylaxis of CMV infection and disease in allogeneic hematopoietic stem-cell transplant recipients. It is a substrate of UGT1A1/3, P-gp, and OATP1B, with its clearance primarily mediated by OATp1B.

ExcerptReferenceRelevance
"Letermovir is a human cytomegalovirus (CMV) terminase inhibitor for the prophylaxis of CMV infection and disease in allogeneic hematopoietic stem-cell transplant recipients. "( Pharmacokinetics and Safety of Letermovir and Midazolam Coadministration in Healthy Subjects.
Erb-Zohar, K; Kropeit, D; McCormick, D; Rübsamen-Schaeff, H; Stobernack, HP; Zimmermann, H, 2022
)
2.45
"Letermovir is a human cytomegalovirus (CMV) terminase inhibitor approved in the United States, Canada, Japan, and the European Union for prophylaxis of CMV infection and disease in CMV-seropositive, allogeneic, hematopoietic stem-cell transplant recipients. "( The Effect of Oral Letermovir Administration on the Pharmacokinetics of a Single Oral Dose of P-Glycoprotein Substrate Digoxin in Healthy Volunteers.
Erb-Zohar, K; Kropeit, D; McCormick, D; Rübsamen-Schaeff, H; Scheuenpflug, J; Stobernack, HP; Theis, JGW; Zimmermann, H, 2022
)
2.49
"Letermovir is a substrate of UGT1A1/3, P-gp, and OATP1B, with its clearance primarily mediated by OATP1B."( Acute and Chronic Effects of Rifampin on Letermovir Suggest Transporter Inhibition and Induction Contribute to Letermovir Pharmacokinetics.
Chu, X; Fancourt, C; Iwamoto, M; Lassman, M; Marceau West, R; McCrea, JB; Menzel, K; Mostoller, K; Robbins, JA; Stoch, SA; Witter, R; Zhao, T, 2022
)
1.71
"Letermovir is a novel antiviral agent used off-label in this population for CMV prevention."( Cytomegalovirus prevention in thoracic organ transplantation: A single-center evaluation of letermovir prophylaxis.
Bacchus, M; Baker, AW; Berry, H; Eichenberger, EM; Ferrari, A; Kakoullis, SA; Maziarz, EK; Reynolds, JM; Saullo, JL; Snyder, LD; Steinbrink, JM; Wolfe, CR; Zaffiri, L, 2022
)
1.66
"Letermovir is a human cytomegalovirus terminase inhibitor for the prophylaxis of cytomegalovirus infection and disease in allogeneic hematopoietic stem cell transplant recipients. "( Pharmacokinetics, Safety, and Tolerability of Letermovir Following Single- and Multiple-Dose Administration in Healthy Japanese Subjects.
Asari, K; Fancourt, C; Furihata, K; Ishii, M; Iwamoto, M; McCrea, JB; Stoch, SA; Wakana, A; Yoon, E; Yoshitsugu, H, 2022
)
2.42
"Letermovir is a promising medication for use in cytomegalovirus prophylaxis in children."( Letermovir prophylaxis for cytomegalovirus reactivation in children who underwent hematopoietic stem cell transplantation: A single-institute experience in Taiwan.
Chen, JS; Cheng, CN; Li, SS; Shen, CF; Yeh, YH, 2022
)
2.89
"Letermovir (LMV) is a human cytomegalovirus (HCMV) terminase inhibitor indicated as prophylaxis for HCMV-positive stem-cell recipients. "( First clinical description of letermovir resistance mutation in cytomegalovirus UL51 gene and potential impact on the terminase complex structure.
Alain, S; Feghoul, L; Frobert, E; Garrigue, I; Hantz, S; Lepiller, Q; Mirand, A; Muller, C; Sidorov, E; Tilloy, V, 2022
)
2.45
"Letermovir (LET) is a novel antiviral drug used to prevent CMV infection."( Real-world efficacy of letermovir prophylaxis for cytomegalovirus infection after allogeneic hematopoietic stem cell transplantation: A single-center retrospective analysis.
Fujita, S; Hotta, M; Ichikawa, J; Ishii, K; Ishii, Y; Ito, T; Konishi, A; Nakanishi, T; Nomura, S; Saito, R; Satake, A; Yoshimura, H, 2022
)
1.75
"Letermovir is a highly effective, well-tolerated prophylaxis that mitigates CMV infection, CMV-related mortality, and antiviral therapy toxicities in CBT recipients."( Extended-duration letermovir prophylaxis for cytomegalovirus infection after cord blood transplantation in adults.
Barker, JN; Devlin, SM; Lau, C; Lin, A; Papanicolaou, GA; Perales, MA; Politikos, I; Quach, S; Seo, SK; Shah, GL, 2022
)
1.78
"Letermovir is a novel antiviral agent approved for CMV prophylaxis in hematopoietic stem cell transplant, but not for SOT (neither for prophylaxis nor for treatment)."( A Case Report of Successful Use of Twice-Daily Letermovir in the Treatment of Resistant Cytomegalovirus in a Small Bowel Transplant Recipient.
Al-Jedai, A; Alaidaros, F; Almaghrabi, RS; Broering, D; Hamad, A; Joharji, H; Koujan, H; Zidan, A,
)
1.11
"Letermovir is a novel agent for the prevention of cytomegalovirus (CMV) infection and disease that, unlike traditional CMV DNA polymerase inhibitors, does not carry the risk of myelosuppression. "( Letermovir for cytomegalovirus prophylaxis in high-risk heart transplant recipients.
Batra, J; Carey, M; Choe, J; Clerkin, K; Colombo, PC; DeFilippis, EM; Fried, J; Gaine, M; Golob, S; Hi Lee, S; Jennings, D; Latif, F; Mabasa, A; Majure, D; Pereira, MR; Raikelkar, J; Restaino, S; Sayer, G; Uriel, M; Uriel, N; Yuzefpolskaya, M, 2022
)
3.61
"Letermovir is an attractive cytomegalovirus (CMV) prophylactic agent, but published data in children are scarce. "( Letermovir as Cytomegalovirus Prophylaxis in a Pediatric Cohort: A Retrospective Analysis.
Dinnes, L; Ferdjallah, A; Khan, S; Kohorst, M; Kuhn, A; Madigan, T; Puttkammer, J, 2023
)
3.8
"Letermovir is a newly available and recently approved drug for CMV prophylaxis."( Letermovir Prophylaxis for CMV Reactivation in Allogeneic Stem Cell Recipients: A Retrospective Single Center Analysis.
Bassermann, F; Braitsch, K; Götze, K; Herhaus, P; Koch, K; Miller, I; Osswald, L; Verbeek, M, 2022
)
2.89
"Letermovir is a novel anti-CMV drug that is only approved for CMV prophylaxis in hematopoietic stem cell transplant recipients, with fewer side effects."( Successful Treatment with Letermovir in a Heart Transplant Recipient with UL97 Mutation Ganciclovir-Resistant Cytomegalovirus Colitis and Viremia.
Amiya, E; Ando, M; Bujo, C; Fukushi, S; Hatano, M; Ishida, J; Kinoshita, O; Komuro, I; Okamoto, K; Ono, M; Shimada, S; Tsuji, M; Yamada, S; Yanase, T, 2023
)
1.93
"Letermovir is a relatively new antiviral for prophylaxis against cytomegalovirus (CMV) after allogeneic hematopoietic cell transplantation (HCT). "( Cytomegalovirus breakthrough and resistance during letermovir prophylaxis.
Biernacki, MA; Boeckh, M; Castor, J; Greninger, AL; Jerome, KR; Joncas-Schronce, L; Kim, Y; Martin, PJ; Perchetti, GA; Sandmaier, BM; Ueda Oshima, M; Xie, H; Zamora, D, 2023
)
2.6
"Letermovir is an effective antiviral agent for CMV prevention and has demonstrated enhanced safety, which may allow for extended durations of primary prophylaxis among transplant recipients along with other improved clinical outcomes by mitigating the indirect effects of CMV."( A new direction for cytomegalovirus prophylaxis among transplant recipients: Benefits and nonviral outcomes of letermovir use as primary CMV prophylaxis.
Boutin, CA; Moore, WJ; Tanna, S, 2023
)
2.56
"Letermovir (LMV) is a new antiviral drug used to prevent cytomegalovirus infection in hematopoietic stem cell transplantation (HSCT) recipients. "( Tacrolimus Concentration after Letermovir Initiation in Hematopoietic Stem Cell Transplantation Recipients Receiving Voriconazole: A Retrospective, Observational Study.
Fukunaga, K; Higami, T; Hikasa, S; Ikegame, K; Kaida, K; Kimura, T; Osugi, Y; Shimabukuro, S; Tada, M; Tanaka, K; Yanai, M, 2020
)
2.29
"Letermovir is a prophylactic agent for cytomegalovirus infection and disease in adult cytomegalovirus-seropositive recipients of allogeneic hematopoietic stem cell transplant. "( Assessment of Pharmacokinetic Interaction Between Letermovir and Fluconazole in Healthy Participants.
Adedoyin, A; Fancourt, C; Iwamoto, M; McCrea, JB; Menzel, K; Panebianco, D; Stoch, SA; Tomek, C; Zhao, T, 2021
)
2.32
"Letermovir is a recently licensed antiviral agent for primary CMV prophylaxis in allogenic hematopoietic stem cell transplant (HSCT) recipients."( Emergence of letermovir resistance in solid organ transplant recipients with ganciclovir resistant cytomegalovirus infection: A case series and review of the literature.
Bittel, P; Dahdal, S; Hirzel, C; Hofmann, E; Manuel, O; Sidler, D; Suter-Riniker, F; Walti, LN, 2021
)
1.71
"Letermovir is a new agent approved for CMV prophylaxis in hematopoietic stem cell transplantation and is associated with less toxicity."( Letermovir prophylaxis in solid organ transplant-Assessing CMV breakthrough and tacrolimus drug interaction.
Brown, A; Gupta, G; Kumar, D; Song, C; Thacker, L; Winstead, RJ; Yakubu, I, 2021
)
2.79
"Letermovir (LTV) is a novel antiviral agent approved for CMV prophylaxis after allogeneic transplantation."( Prophylactic letermovir decreases cytomegalovirus reactivation after stem cell transplantation: a single-center real-world evidence study.
D'Addona, M; Ferrara, I; Fontana, R; Giudice, V; Guariglia, R; Langella, M; Martorelli, MC; Mettivier, L; Pezzullo, L; Selleri, C; Serio, B; Vaccaro, E, 2021
)
1.71
"Letermovir is a cytochrome P450 (CYP) 2C19 inducer."( Drug interaction between letermovir and voriconazole after allogeneic hematopoietic cell transplantation.
Doke, Y; Fukuda, T; Fukushi, Y; Hashimoto, H; Inamoto, Y; Nakashima, T; Yamaguchi, M, 2021
)
1.65
"Letermovir is an antiviral agent indicated for primary prophylaxis of cytomegalovirus (CMV) infection and disease in adult allogeneic hematopoietic stem cell transplant recipients. "( Successful Treatment of UL97 Mutation Ganciclovir-Resistant Cytomegalovirus Viremia in a Renal Transplant Recipient With Letermovir and Adjunct Hyperimmune Cytomegalovirus Immunoglobulin: A Case Report.
Gruber, J; Ingemi, AI; Khardori, N; McMahon, M; Pearston, AP; Ripley, K; Sutton, S; Wilson, TJ, 2021
)
2.27
"Letermovir is a new antiviral drug approved for the prophylaxis of CMV infection in allogeneic stem cell transplants. "( Letermovir in lung transplant recipients with cytomegalovirus infection: A retrospective observational study.
Barton, J; Behr, J; Kauke, T; Keppler, OT; Kneidinger, N; Meiser, B; Michel, S; Milger, K; Munker, D; Nitschko, H; Veit, T; Zoller, M, 2021
)
3.51
"Letermovir is a novel antiviral in clinical development for prophylaxis against human cytomegalovirus in immunocompromised transplant recipients. "( Intravenous Hydroxypropyl β-Cyclodextrin Formulation of Letermovir: A Phase I, Randomized, Single-Ascending, and Multiple-Dose Trial.
Erb-Zohar, K; Hulskotte, E; Kropeit, D; Rübsamen-Schaeff, H; Scheuenpflug, J; Stobernack, HP; van Schanke, A; Zimmermann, H, 2017
)
2.14
"Letermovir is a novel viral-terminase inhibitor that has demonstrated prophylactic/pre-emptive activity against human cytomegalovirus in Phase 2 and 3 transplant trials."( Pharmacokinetics and safety of the anti-human cytomegalovirus drug letermovir in subjects with hepatic impairment.
Erb-Zohar, K; Kobalava, ZD; Kropeit, D; McCormick, D; Moiseev, VS; Rübsamen-Schaeff, H; Stobernack, HP; Zimmermann, H, 2017
)
1.41
"Letermovir is a human cytomegalovirus (CMV) terminase inhibitor that was clinically effective in a Phase III prevention trial. "( A third component of the human cytomegalovirus terminase complex is involved in letermovir resistance.
Chou, S, 2017
)
2.12
"Letermovir is an antiviral drug that inhibits the CMV-terminase complex."( Letermovir Prophylaxis for Cytomegalovirus in Hematopoietic-Cell Transplantation.
Badshah, C; Blumberg, EA; Boeckh, M; Brown, J; Chemaly, RF; Dadwal, SS; DiNubile, MJ; Duarte, RF; Einsele, H; Haider, S; Kanda, Y; Kartsonis, NA; Katayama, Y; Leavitt, RY; Ljungman, P; Maertens, J; Marty, FM; Mullane, KM; Murata, Y; Snydman, DR; Teal, VL; Ullmann, AJ; Wan, H, 2017
)
2.62
"Letermovir (Prevymis™) is an orally or intravenously administered cytomegalovirus (CMV) DNA terminase complex inhibitor being developed by Merck & Co., Inc., under a global license from AiCuris Anti-infective Cures GmbH. "( Letermovir: First Global Approval.
Kim, ES, 2018
)
3.37
"Letermovir is a human cytomegalovirus terminase inhibitor for cytomegalovirus infection prophylaxis in hematopoietic stem cell transplant recipients. "( Pharmacokinetics and Tolerability of Letermovir Coadministered With Azole Antifungals (Posaconazole or Voriconazole) in Healthy Subjects.
Butterton, JR; Cho, CR; de Haes, JIU; Drexel, M; Hulskotte, EGJ; Hussaini, A; Iwamoto, M; Jordan, HR; Kantesaria, BS; Liu, F; Macha, S; Marshall, WL; McCrea, JB; Menzel, K; Tsai, C; van Schanke, A, 2018
)
2.2
"Letermovir is an important addition to the current strategies for CMV prevention after allogeneic HSCT. "( Role of letermovir for prevention of cytomegalovirus infection after allogeneic haematopoietic stem cell transplantation.
Razonable, RR, 2018
)
2.36
"Letermovir is a human cytomegalovirus (CMV) terminase inhibitor for the prophylaxis of CMV infection in allogeneic hematopoietic stem-cell transplant (HSCT) recipients. "( PBPK Modeling Strategy for Predicting Complex Drug Interactions of Letermovir as a Perpetrator in Support of Product Labeling.
Chen, D; Cho, CR; Hartmann, G; Menzel, K; Wang, YH, 2019
)
2.19
"Letermovir is a human cytomegalovirus (CMV) terminase inhibitor recently approved as prophylaxis in stem cell transplant recipients. "( New Locus of Drug Resistance in the Human Cytomegalovirus UL56 Gene Revealed by
Chou, S; Ercolani, RJ; Satterwhite, LE, 2018
)
1.92
"Letermovir is an antiviral agent recently approved by the Food and Drug Administration for prophylaxis of cytomegalovirus infection in adult patients that are cytomegalovirus-seropositive recipients of an allogeneic hematopoietic stem cell transplant. "( Letermovir associated hepatic transaminitis: A case report.
Geswein, L, 2019
)
3.4
"Letermovir is a new antiviral agent with activity against human cytomegalovirus (CMV). "( Letermovir for prophylaxis of cytomegalovirus in allogeneic hematopoietic stem cell recipients.
Cho, JC; Le, AD; Locke, SC, 2018
)
3.37
"Letermovir is a new antiviral approved to prevent cytomegalovirus infection in hematopoietic stem cell transplant recipients. "( Pharmacokinetic drug evaluation of letermovir prophylaxis for cytomegalovirus in hematopoietic stem cell transplantation.
Deleenheer, B; Maertens, J; Spriet, I, 2018
)
2.2
"Letermovir is a novel antiviral recently approved for CMV prophylaxis following hematopoietic cell transplantation, but its efficacy in other settings is unknown."( Use of Letermovir as Salvage Therapy for Drug-Resistant Cytomegalovirus Retinitis.
Arif, S; Baker, AW; Cox, G; Grewal, DS; Maziarz, EK; Saullo, JH; Strand, A; Turner, N; Wolfe, CR, 2019
)
1.69
"Letermovir is an anti-human cytomegalovirus (HCMV) drug with a novel mechanism of action. "( [Pharmacological and clinical effects of letermovir (Prevymis
Eto, T; Ogawa, M, 2019
)
2.22
"Letermovir is a human cytomegalovirus (HCMV) terminase inhibitor recently approved in the United States for prophylaxis of HCMV infection or disease in adult HCMV-seropositive recipients [R+] of an allogeneic hematopoietic stem cell transplant. "( In-depth genomic analyses identified novel letermovir resistance-associated substitutions in the cytomegalovirus UL56 and UL89 gene products.
Colberg-Poley, AM; Donaldson, EF; Hodowanec, AC; Komatsu, TE; O'Rear, JJ; Pikis, A; Singer, ME, 2019
)
2.22
"Letermovir (AIC246), is a novel anti-HCMV drug in development, acting via a novel mechanism of action."( Preemptive treatment of Cytomegalovirus infection in kidney transplant recipients with letermovir: results of a Phase 2a study.
Arns, W; Beelen, DW; Budde, K; Dürr, M; Fischereder, M; Gwinner, W; Hummel, J; Lischka, P; Michel, D; Mühlfeld, A; Renders, L; Rübsamen-Schaeff, H; Stangl, M; Stoelben, S; Suwelack, B; Witzke, O; Zimmermann, H, 2014
)
1.35
"Letermovir is a novel antiviral compound currently in clinical development for the prevention of human cytomegalovirus (HCMV) infections. "( Geno- and phenotypic characterization of human cytomegalovirus mutants selected in vitro after letermovir (AIC246) exposure.
Goldner, T; Hempel, C; Lischka, P; Ruebsamen-Schaeff, H; Zimmermann, H, 2014
)
2.06
"Letermovir is a new drug in Phase 3 clinical development for the prevention of human Cytomegalovirus (HCMV) infections in hematopoietic-stem-cell transplant recipients (HSCT). "( Phenotypic characterization of two naturally occurring human Cytomegalovirus sequence polymorphisms located in a distinct region of ORF UL56 known to be involved in in vitro resistance to letermovir.
Goldner, T; Lischka, P; Zimmermann, H, 2015
)
2.05
"Letermovir (LMV) is an experimental cytomegalovirus terminase inhibitor undergoing phase 3 clinical trials. "( Rapid In Vitro Evolution of Human Cytomegalovirus UL56 Mutations That Confer Letermovir Resistance.
Chou, S, 2015
)
2.09
"Letermovir is a novel anti-HCMV drug in Phase III development that targets the UL56 subunit of the viral terminase complex. "( Impact of glycoprotein B genotype and naturally occurring ORF UL56 polymorphisms upon susceptibility of clinical human cytomegalovirus isolates to letermovir.
Holder, D; Lischka, P; Zhang, D; Zimmermann, H, 2016
)
2.08
"Letermovir is a novel antiviral in the late stages of drug development for the treatment and prevention of CMV."( Letermovir for the management of cytomegalovirus infection.
Bowman, LJ; Brennan, DC; Melaragno, JI, 2017
)
2.62

Effects

Letermovir has been approved in Canada and the USA for the prophylaxis of CMV infection and disease in adult CMV-seropositive recipients of an allogeneic haematopoietic stem cell transplant (HSCT) The drug has shown promising clinical efficacy and is generally well tolerated.

ExcerptReferenceRelevance
"Letermovir has been approved as a novel cytomegalovirus (CMV) prophylactic agent after allogeneic hematopoietic stem cell transplantation (HSCT). "( Risk factor analysis for cytomegalovirus reactivation under prophylaxis with letermovir after allogeneic hematopoietic stem cell transplantation.
Abe, R; Kataoka, K; Kato, J; Koda, Y; Mizuno, K; Mori, T; Sakurai, M; Yamaguchi, K, 2022
)
2.39
"Letermovir (LTV) has been recently introduced for HCMV prophylaxis in adult patients who received allogeneic HSCT."( Human Cytomegalovirus (HCMV) - specific T-cell response after letermovir prophylaxis is predictive for subsequent HCMV reactivation in haematopoietic stem cell transplant recipients.
Arena, F; Baldanti, F; Bergami, F; Bernasconi, P; Borsani, O; Caldera, D; Cassaniti, I; Colombo, AA; Lilleri, D; Vitello, D; Zavaglio, F, 2023
)
1.87
"Letermovir has been demonstrated to reduce the risk of CMV infection when used for prophylaxis in allogeneic hematopoietic cell transplantation."( Single-center experience with use of letermovir for CMV prophylaxis or treatment in thoracic organ transplant recipients.
Ahmad, K; Aryal, S; Brown, AW; Chun, J; Cochrane, A; Desai, S; Fregoso, M; Katugaha, SB; King, C; Marinak, L; Nathan, SD; Shlobin, OA, 2019
)
1.51
"Letermovir has been approved for the prevention of cytomegalovirus (CMV) infection after allogeneic hematopoietic stem cell transplantation (HCT). "( Drug interaction between letermovir and voriconazole after allogeneic hematopoietic cell transplantation.
Doke, Y; Fukuda, T; Fukushi, Y; Hashimoto, H; Inamoto, Y; Nakashima, T; Yamaguchi, M, 2021
)
2.37
"Letermovir has been approved in Canada and the USA for the prophylaxis of CMV infection and disease in adult CMV-seropositive recipients of an allogeneic haematopoietic stem cell transplant (HSCT)."( Letermovir: First Global Approval.
Kim, ES, 2018
)
2.64
"Letermovir has shown promising clinical efficacy and is generally well tolerated, thus providing a favorable new option in the prophylaxis of CMV infection and disease."( Letermovir for prophylaxis of cytomegalovirus in allogeneic hematopoietic stem cell recipients.
Cho, JC; Le, AD; Locke, SC, 2018
)
2.64

Treatment

Letermovir treatment led to a significantly reduced incidence of CMV reactivation after alloSCT (33.3% in letermovir group versus 76.2% in the control group, p<0.001) No patient developed CMV disease during the trial.

ExcerptReferenceRelevance
"Letermovir treatment led to a significantly reduced incidence of CMV reactivation after alloSCT (33.3% in the letermovir group versus 76.2% in the control group, p<0.001)."( Letermovir Prophylaxis for CMV Reactivation in Allogeneic Stem Cell Recipients: A Retrospective Single Center Analysis.
Bassermann, F; Braitsch, K; Götze, K; Herhaus, P; Koch, K; Miller, I; Osswald, L; Verbeek, M, 2022
)
2.89
"Letermovir treatment was generally well tolerated, no patient developed CMV disease during the trial."( Preemptive treatment of Cytomegalovirus infection in kidney transplant recipients with letermovir: results of a Phase 2a study.
Arns, W; Beelen, DW; Budde, K; Dürr, M; Fischereder, M; Gwinner, W; Hummel, J; Lischka, P; Michel, D; Mühlfeld, A; Renders, L; Rübsamen-Schaeff, H; Stangl, M; Stoelben, S; Suwelack, B; Witzke, O; Zimmermann, H, 2014
)
1.35

Toxicity

Letermovir is safe and effective compared with alternative prophylaxis approaches following CBT through day 100. Both letermovir and acyclovir showed significantly reduced risk for serious adverse events compared with ganciclovir, with RRs of .

ExcerptReferenceRelevance
" In terms of safety, letermovir was at least similar in comparison with placebo and most agents while both letermovir and acyclovir showed significantly reduced risk for serious adverse events compared with ganciclovir, with RRs of ."( Comparative Efficacy and Safety of Different Antiviral Agents for Cytomegalovirus Prophylaxis in Allogeneic Hematopoietic Cell Transplantation: A Systematic Review and Meta-Analysis.
Gagelmann, N; Kröger, N; Ljungman, P; Styczynski, J, 2018
)
0.8
" Letermovir is safe and effective compared with alternative prophylaxis approaches following CBT through day 100."( 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.
Abidi, MZ; Bajrovic, V; Benamu, E; Chase, S; Gakhar, N; Gutman, JA; Haverkos, BM; Kaiser, J; MacDonald, J; Miller, M; Purev, E; Sharma, P; Tobin, J; Weinberg, A, 2020
)
2.91
"9 % experienced adverse drug reactions, the most frequent of which were renal impairment (2."( Safety and Effectiveness of Letermovir in Allogenic Hematopoietic Stem Cell Transplantation Recipients: Interim Report of Post-marketing Surveillance in Japan.
Hiraishi, I; Maekawa, S; Ueno, R; Watanabe, A, 2021
)
0.92

Pharmacokinetics

The aim of this study was to identify factors affecting blood concentrations of voriconazole following letermovir coadministration in allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients.

ExcerptReferenceRelevance
"Phase 1, open-label, parallel-group pharmacokinetic and safety comparison of multiple once-daily oral letermovir in female subjects with hepatic impairment and healthy matched controls."( Pharmacokinetics and safety of the anti-human cytomegalovirus drug letermovir in subjects with hepatic impairment.
Erb-Zohar, K; Kobalava, ZD; Kropeit, D; McCormick, D; Moiseev, VS; Rübsamen-Schaeff, H; Stobernack, HP; Zimmermann, H, 2017
)
0.91
" In both trials, blood samples were collected for the assessment of the pharmacokinetic profiles of the antifungals, and safety was assessed."( Pharmacokinetics and Tolerability of Letermovir Coadministered With Azole Antifungals (Posaconazole or Voriconazole) in Healthy Subjects.
Butterton, JR; Cho, CR; de Haes, JIU; Drexel, M; Hulskotte, EGJ; Hussaini, A; Iwamoto, M; Jordan, HR; Kantesaria, BS; Liu, F; Macha, S; Marshall, WL; McCrea, JB; Menzel, K; Tsai, C; van Schanke, A, 2018
)
0.75
" Two-stage population pharmacokinetic (PK) modeling of letermovir was conducted to support dose rationale and evaluate the impact of intrinsic/extrinsic factors."( Population pharmacokinetics of letermovir following oral and intravenous administration in healthy participants and allogeneic hematopoietic cell transplantation recipients.
Cho, CR; Davis, C; de Alwis, D; Dykstra, K; Fancourt, C; Iwamoto, M; Macha, S; Prohn, M; Sabato, P; Viberg, A; Zhang, D, 2021
)
1.15
"The aim of this study was to identify factors affecting blood concentrations of voriconazole following letermovir coadministration using population pharmacokinetic (PPK) analysis in allogeneic hematopoietic stem cell transplant (allo-HSCT) recipients."( Effects of Letermovir and/or Methylprednisolone Coadministration on Voriconazole Pharmacokinetics in Hematopoietic Stem Cell Transplantation: A Population Pharmacokinetic Study.
Akashi, K; Egashira, N; Fukumoto, J; Hirota, T; Ieiri, I; Matsukane, R; Miyamoto, T; Mori, Y; Muraki, S; Suetsugu, K, 2021
)
1.23
" The presence of steady-state letermovir reduced digoxin area under the plasma concentration-time curve from administration until last quantifiable measurement by 12% and maximum plasma concentration by 22% compared with digoxin alone; digoxin half-life and elimination rate remained similar in both conditions."( The Effect of Oral Letermovir Administration on the Pharmacokinetics of a Single Oral Dose of P-Glycoprotein Substrate Digoxin in Healthy Volunteers.
Erb-Zohar, K; Kropeit, D; McCormick, D; Rübsamen-Schaeff, H; Scheuenpflug, J; Stobernack, HP; Theis, JGW; Zimmermann, H, 2022
)
1.34
" Following administration of oral single and multiple doses, letermovir was absorbed with a median time to maximum plasma concentration of 2 to 4 hours, and concentrations declined in a biphasic manner with a terminal half-life of ≈10 to 13 hours."( Pharmacokinetics, Safety, and Tolerability of Letermovir Following Single- and Multiple-Dose Administration in Healthy Japanese Subjects.
Asari, K; Fancourt, C; Furihata, K; Ishii, M; Iwamoto, M; McCrea, JB; Stoch, SA; Wakana, A; Yoon, E; Yoshitsugu, H, 2022
)
1.22
"Data describing the magnitude of the pharmacokinetic interaction between letermovir and tacrolimus in allogeneic hematopoietic cell transplantation (allo-HCT) recipients are limited, and varying outcomes have been reported."( Evaluation of the Pharmacokinetic Interaction Between Letermovir and Tacrolimus in Allogeneic Hematopoietic Cell Transplantation Recipients.
Cumpston, A; Dillaman, M; Marciano, KA; Ross, KG; Seago, K; Veltri, L, 2022
)
1.2

Compound-Compound Interactions

L Termovir may be a perpetrator of CYP2C9/19-mediated drug-drug interactions. For the future, drug combination therapies, including letermovir, might be indicated under special medical conditions.

ExcerptReferenceRelevance
" For the future, drug combination therapies, including letermovir, might be indicated under special medical conditions, such as the emergence of multidrug-resistant virus strains in transplant recipients or in HCMV-HIV-coinfected patients."( In vitro drug combination studies of Letermovir (AIC246, MK-8228) with approved anti-human cytomegalovirus (HCMV) and anti-HIV compounds in inhibition of HCMV and HIV replication.
Lischka, P; Wildum, S; Zimmermann, H, 2015
)
0.94
" These results suggest that letermovir may be a perpetrator of CYP2C9/19-mediated drug-drug interactions."( Pharmacokinetics and Tolerability of Letermovir Coadministered With Azole Antifungals (Posaconazole or Voriconazole) in Healthy Subjects.
Butterton, JR; Cho, CR; de Haes, JIU; Drexel, M; Hulskotte, EGJ; Hussaini, A; Iwamoto, M; Jordan, HR; Kantesaria, BS; Liu, F; Macha, S; Marshall, WL; McCrea, JB; Menzel, K; Tsai, C; van Schanke, A, 2018
)
1.05
" These results were used to inform the US prescribing information in the absence of clinical drug-drug interaction studies."( PBPK Modeling Strategy for Predicting Complex Drug Interactions of Letermovir as a Perpetrator in Support of Product Labeling.
Chen, D; Cho, CR; Hartmann, G; Menzel, K; Wang, YH, 2019
)
0.75
" Our results suggest that voriconazole trough concentration decreases when voriconazole is combined with letermovir in allogeneic HCT recipients."( Drug interaction between letermovir and voriconazole after allogeneic hematopoietic cell transplantation.
Doke, Y; Fukuda, T; Fukushi, Y; Hashimoto, H; Inamoto, Y; Nakashima, T; Yamaguchi, M, 2021
)
1.14
" A drug-drug interaction study was conducted in healthy participants (N = 16) to assess the effect of letermovir on acyclovir pharmacokinetics."( A drug-drug interaction study with letermovir and acyclovir in healthy participants.
Fancourt, C; Iwamoto, M; McCrea, JB; Menzel, K; Stoch, SA; Witter, R; Zhao, T, 2023
)
1.4

Bioavailability

Ltermovir may be considered a valuable off-label option for secondary prophylaxis of GCV-resistant CMV in SOT recipients. These staged analyses indicate that le Termovir PK in HSCT recipients and healthy participants differ only with respect to bioavailability and absorption rate.

ExcerptReferenceRelevance
"The ATP-binding cassette transporter P-glycoprotein (P-gp) is known to limit both brain penetration and oral bioavailability of many chemotherapy drugs."( A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
Ambudkar, SV; Brimacombe, KR; Chen, L; Gottesman, MM; Guha, R; Hall, MD; Klumpp-Thomas, C; Lee, OW; Lee, TD; Lusvarghi, S; Robey, RW; Shen, M; Tebase, BG, 2019
)
0.51
" Given the favorable safety profile and its oral bioavailability letermovir may be considered a valuable off-label option for secondary prophylaxis of GCV-resistant CMV in SOT recipients."( Emergence of letermovir resistance in solid organ transplant recipients with ganciclovir resistant cytomegalovirus infection: A case series and review of the literature.
Bittel, P; Dahdal, S; Hirzel, C; Hofmann, E; Manuel, O; Sidler, D; Suter-Riniker, F; Walti, LN, 2021
)
1.23
" These staged analyses indicate that letermovir PK in HSCT recipients and healthy participants differ only with respect to bioavailability and absorption rate."( Population pharmacokinetics of letermovir following oral and intravenous administration in healthy participants and allogeneic hematopoietic cell transplantation recipients.
Cho, CR; Davis, C; de Alwis, D; Dykstra, K; Fancourt, C; Iwamoto, M; Macha, S; Prohn, M; Sabato, P; Viberg, A; Zhang, D, 2021
)
1.18

Dosage Studied

Letermovir is available both orally and intravenously in 480-mg and 240-mg dosage forms. It is approved for use in the prophylaxis of CMV infection and disease in CMV-seropositive recipients of allogeneic hematopoietic stem cell transplant (HSCT)

ExcerptRelevanceReference
" Letermovir is available both orally and intravenously in 480-mg and 240-mg dosage forms, and is approved for use in the prophylaxis of CMV infection and disease in CMV-seropositive recipients of allogeneic hematopoietic stem cell transplant (HSCT) over the age of 18."( Letermovir for prophylaxis of cytomegalovirus in allogeneic hematopoietic stem cell recipients.
Cho, JC; Le, AD; Locke, SC, 2018
)
2.83
" These data support the reduction in clinical dosage of letermovir (to 240 mg) upon coadministration with cyclosporine."( Pharmacokinetic Drug-Drug Interactions Between Letermovir and the Immunosuppressants Cyclosporine, Tacrolimus, Sirolimus, and Mycophenolate Mofetil.
Adedoyin, A; Cho, CR; Iwamoto, M; Kraft, WK; Levine, V; Liu, F; Macha, S; Marshall, W; McCrea, JB; Menzel, K; Panebianco, D; Stoch, SA; Yoon, E; Zhao, T, 2019
)
1.02
" Detailed information on therapeutic-drug-monitoring measures and dosage adjustments for letermovir is provided."( Compassionate Use of Letermovir in a 2-Year-Old Immunocompromised Child With Resistant Cytomegalovirus Disease.
Andre, P; Asner, SA; Buclin, T; Decosterd, LA; Gengler, C; Jaton-Ogay, K; Mercier, T; Meylan, P; Murray, K; Natterer, J; Opota, O; Pérez Marín, M; Perez, MH; Rizzi, M, 2020
)
1.1
" Further study of the dosing and efficacy of letermovir for CMV prophylaxis or treatment in thoracic organ transplant recipients is warranted."( Single-center experience with use of letermovir for CMV prophylaxis or treatment in thoracic organ transplant recipients.
Ahmad, K; Aryal, S; Brown, AW; Chun, J; Cochrane, A; Desai, S; Fregoso, M; Katugaha, SB; King, C; Marinak, L; Nathan, SD; Shlobin, OA, 2019
)
1.05
"Data on the efficacy, dosing and safety of letermovir for the compassionate therapeutic use of CMV infections are limited."( Letermovir for the compassionate therapeutic use of cytomegalovirus infection.
Burgmann, H; Fisecker, L; Schubert, L; Steininger, C; Thalhammer, F, 2021
)
2.33
" Further studies on combination therapy or alternative LET dosing are needed."( Letermovir treatment of cytomegalovirus infection or disease in solid organ and hematopoietic cell transplant recipients.
Avery, R; Clark, NM; Gonzalez, AA; Hakki, M; Kaul, DR; Kotton, CN; Kovacs, C; La Hoz, RM; Limaye, AP; Linder, KA; Malinis, M; Mishkin, A; Mullane, KM; Ostrander, D; Prono, MD; Smith, J; Wolfe, C, 2021
)
2.06
" Due to the high toxicity of Foscarnet (FOS) and Cidofovir (CDV), Letermovir (LMV) monotherapy at the dosage of 480 mg per day was administered, with a gradual viral load reduction."( Emergence of Letermovir-resistant HCMV UL56 mutant during rescue treatment in a liver transplant recipient with ganciclovir-resistant infection HCMV: a case report.
Baldanti, F; Campanini, G; Cassaniti, I; Fratini, A; Girelli, F; Meini, A; Novazzi, F; Palumbo, L; Paolucci, S; Plebani, A; Tebaldi, A, 2021
)
1.23
" These results support current dosing recommendations in several countries and regions, including the United States and European Union."( Exposure-Response Analyses of Letermovir Following Oral and Intravenous Administration in Allogeneic Hematopoietic Cell Transplantation Recipients.
Badshah, C; Cho, CR; Davis, C; Dykstra, K; Fancourt, C; Leavitt, R; Macha, S; Murata, Y; Prohn, M; Sabato, P; Stone, J; Viberg, A, 2022
)
1.01
" Nonlinear pharmacokinetics (PKs) were observed in clinical studies after intravenous and oral dosing across a wide dose range, including the efficacious doses of 240 and 480 mg."( Developing a mechanistic understanding of the nonlinear pharmacokinetics of letermovir and prospective drug interaction with everolimus using physiological-based pharmacokinetic modeling.
Asari, K; Chen, D; Cho, CR; Hartmann, G; Kuo, Y; McCrea, JB; Menzel, K; Wang, YH, 2023
)
1.14
"CYP2C19-guided voriconazole dosing reduces pharmacokinetic variability, but many patients remain subtherapeutic."( Pharmacogenetic and clinical predictors of voriconazole concentration in hematopoietic stem cell transplant recipients receiving CYP2C19-guided dosing.
Avalos, B; Copelan, E; Druhan, LJ; Ghosh, N; Grunwald, MR; Hamilton, A; Jandrisevits, E; Lopes, KE; Morris, SA; Patel, JN; Robinson, M; Steuerwald, N, 2023
)
0.91
[information is derived through text-mining from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Protein Targets (6)

Potency Measurements

ProteinTaxonomyMeasurementAverage (µ)Min (ref.)Avg (ref.)Max (ref.)Bioassay(s)
cytochrome P450 family 3 subfamily A polypeptide 4Homo sapiens (human)Potency23.91850.01237.983543.2770AID1645841
GVesicular stomatitis virusPotency6.74120.01238.964839.8107AID1645842
Interferon betaHomo sapiens (human)Potency6.74120.00339.158239.8107AID1645842
HLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)Potency6.74120.01238.964839.8107AID1645842
Inositol hexakisphosphate kinase 1Homo sapiens (human)Potency6.74120.01238.964839.8107AID1645842
cytochrome P450 2C9, partialHomo sapiens (human)Potency6.74120.01238.964839.8107AID1645842
[prepared from compound, protein, and bioassay information from National Library of Medicine (NLM), extracted Dec-2023]

Biological Processes (45)

Processvia Protein(s)Taxonomy
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell activation involved in immune responseInterferon betaHomo sapiens (human)
cell surface receptor signaling pathwayInterferon betaHomo sapiens (human)
cell surface receptor signaling pathway via JAK-STATInterferon betaHomo sapiens (human)
response to virusInterferon betaHomo sapiens (human)
positive regulation of autophagyInterferon betaHomo sapiens (human)
cytokine-mediated signaling pathwayInterferon betaHomo sapiens (human)
natural killer cell activationInterferon betaHomo sapiens (human)
positive regulation of peptidyl-serine phosphorylation of STAT proteinInterferon betaHomo sapiens (human)
cellular response to interferon-betaInterferon betaHomo sapiens (human)
B cell proliferationInterferon betaHomo sapiens (human)
negative regulation of viral genome replicationInterferon betaHomo sapiens (human)
innate immune responseInterferon betaHomo sapiens (human)
positive regulation of innate immune responseInterferon betaHomo sapiens (human)
regulation of MHC class I biosynthetic processInterferon betaHomo sapiens (human)
negative regulation of T cell differentiationInterferon betaHomo sapiens (human)
positive regulation of transcription by RNA polymerase IIInterferon betaHomo sapiens (human)
defense response to virusInterferon betaHomo sapiens (human)
type I interferon-mediated signaling pathwayInterferon betaHomo sapiens (human)
neuron cellular homeostasisInterferon betaHomo sapiens (human)
cellular response to exogenous dsRNAInterferon betaHomo sapiens (human)
cellular response to virusInterferon betaHomo sapiens (human)
negative regulation of Lewy body formationInterferon betaHomo sapiens (human)
negative regulation of T-helper 2 cell cytokine productionInterferon betaHomo sapiens (human)
positive regulation of apoptotic signaling pathwayInterferon betaHomo sapiens (human)
response to exogenous dsRNAInterferon betaHomo sapiens (human)
B cell differentiationInterferon betaHomo sapiens (human)
natural killer cell activation involved in immune responseInterferon betaHomo sapiens (human)
adaptive immune responseInterferon betaHomo sapiens (human)
T cell activation involved in immune responseInterferon betaHomo sapiens (human)
humoral immune responseInterferon betaHomo sapiens (human)
positive regulation of T cell mediated cytotoxicityHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
adaptive immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
antigen processing and presentation of endogenous peptide antigen via MHC class I via ER pathway, TAP-independentHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of T cell anergyHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
defense responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
detection of bacteriumHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of interleukin-12 productionHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of interleukin-6 productionHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protection from natural killer cell mediated cytotoxicityHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
innate immune responseHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
regulation of dendritic cell differentiationHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
antigen processing and presentation of endogenous peptide antigen via MHC class IbHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
inositol phosphate metabolic processInositol hexakisphosphate kinase 1Homo sapiens (human)
phosphatidylinositol phosphate biosynthetic processInositol hexakisphosphate kinase 1Homo sapiens (human)
negative regulation of cold-induced thermogenesisInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol phosphate biosynthetic processInositol hexakisphosphate kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Molecular Functions (18)

Processvia Protein(s)Taxonomy
cytokine activityInterferon betaHomo sapiens (human)
cytokine receptor bindingInterferon betaHomo sapiens (human)
type I interferon receptor bindingInterferon betaHomo sapiens (human)
protein bindingInterferon betaHomo sapiens (human)
chloramphenicol O-acetyltransferase activityInterferon betaHomo sapiens (human)
TAP bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
signaling receptor bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protein bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
peptide antigen bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
TAP bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
protein-folding chaperone bindingHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
inositol-1,3,4,5,6-pentakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol heptakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 5-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
protein bindingInositol hexakisphosphate kinase 1Homo sapiens (human)
ATP bindingInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 1-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol hexakisphosphate 3-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol 5-diphosphate pentakisphosphate 5-kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
inositol diphosphate tetrakisphosphate kinase activityInositol hexakisphosphate kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Ceullar Components (22)

Processvia Protein(s)Taxonomy
extracellular spaceInterferon betaHomo sapiens (human)
extracellular regionInterferon betaHomo sapiens (human)
Golgi membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
endoplasmic reticulumHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
Golgi apparatusHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
plasma membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
cell surfaceHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
ER to Golgi transport vesicle membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
secretory granule membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
phagocytic vesicle membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
early endosome membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
recycling endosome membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular exosomeHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
lumenal side of endoplasmic reticulum membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
MHC class I protein complexHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
extracellular spaceHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
external side of plasma membraneHLA class I histocompatibility antigen, B alpha chain Homo sapiens (human)
fibrillar centerInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleoplasmInositol hexakisphosphate kinase 1Homo sapiens (human)
cytosolInositol hexakisphosphate kinase 1Homo sapiens (human)
nucleusInositol hexakisphosphate kinase 1Homo sapiens (human)
cytoplasmInositol hexakisphosphate kinase 1Homo sapiens (human)
[Information is prepared from geneontology information from the June-17-2024 release]

Bioassays (49)

Assay IDTitleYearJournalArticle
AID510774Antiviral activity against 0.3 MOI human cytomegalovirus AD169 infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510783Antiviral activity against ganciclovir-resistant human cytomegalovirus isolate 1947R infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510751Antiviral activity against human cytomegalovirus infected in human MRC5 cells after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510770Antiviral activity against 0.003 MOI human cytomegalovirus AD169 infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510755Antiviral activity against human cytomegalovirus RV-HG infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510775Antiviral activity against 1 MOI human cytomegalovirus AD169 infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510760Cytotoxicity against human HS27 cells infected with human cytomegalovirus after 7 days by alamar blue assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510781Antiviral activity against human cytomegalovirus isolate E16415S infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510764Cytotoxicity against HLF infected with human cytomegalovirus after 7 days by alamar blue assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510777Antiviral activity against human cytomegalovirus isolate Se infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510758Antiviral activity against human cytomegalovirus Davis infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID1695547Antiviral activity against human Cytomegalovirus UL56 infected in human MRC5 cells assessed as reduction in infection rate incubated for 6 days by Hoechst 33342 staining based microscopic analysis2020RSC medicinal chemistry, Jul-01, Volume: 11, Issue:7
A
AID510778Antiviral activity against human cytomegalovirus isolate Ba infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510792Antiviral activity against human cytomegalovirus Davis xenograft mouse model assessed as reduction of viral load administered via oral gavage QD for 9 days measured post last dose2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510762Cytotoxicity against HEL299 cells infected with human cytomegalovirus after 7 days by alamar blue assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510793Antiviral activity against human cytomegalovirus AD169 infected in NHDF assessed as inhibition of virus replication at 50 nM measured after 24 to 72 hrs of compound wash by fluorescence assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510782Antiviral activity against ganciclovir-resistant human cytomegalovirus isolate E17251S infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510787Antiviral activity against human cytomegalovirus AD169 infected in NHDF at 10 times EC50 treated after 168 hrs postinfection measured after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510759Antiviral activity against human cytomegalovirus infected in NHLF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510791Antiviral activity against human cytomegalovirus Davis xenograft mouse model assessed as reduction of viral load at 10 to 100 mg/kg, po QD for 9 days measured post last dose2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510756Antiviral activity against human cytomegalovirus AD169 infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510779Antiviral activity against human cytomegalovirus isolate La infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510772Antiviral activity against 0.03 MOI human cytomegalovirus AD169 infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510763Cytotoxicity against human MRC5 cells infected with human cytomegalovirus after 7 days by alamar blue assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510768Selectivity index, ratio of CC50 for NHLF to EC50 for human cytomegalovirus infected in NHLF2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510767Selectivity index, ratio of CC50 for HEL299 cells to EC50 for human cytomegalovirus infected in HEL299 cells2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510773Antiviral activity against 0.1 MOI human cytomegalovirus AD169 infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510780Antiviral activity against human cytomegalovirus isolate 472 infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510784Antiviral activity against human cytomegalovirus infected in human fibroblast assessed as decrease in infectious foci size at 50 nM after 5 days by DAPI staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510776Antiviral activity against ganciclovir-resistant human cytomegalovirus AD169 UL97 M460I infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510769Selectivity index, ratio of CC50 for human MRC5 cells to EC50 for human cytomegalovirus infected in human MRC5 cells2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510771Antiviral activity against 0.001 MOI human cytomegalovirus AD169 infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510754Antiviral activity against human cytomegalovirus infected in human HS27 cells after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID1230097Antiviral activity against Cytomegalovirus AD-169 infected in HEL cells assessed as reduction of virus-induced cytopathogenicity2015Bioorganic & medicinal chemistry, Jul-01, Volume: 23, Issue:13
Design, antiviral and cytostatic properties of isoxazolidine-containing amonafide analogues.
AID510786Antiviral activity against human cytomegalovirus AD169 infected in NHDF assessed as inhibition of virus replication at 50 nM after 96 hrs by fluorescence assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510765Selectivity index, ratio of CC50 for human HS27 cells to EC50 for human cytomegalovirus infected in human HS27 cells2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510789Antiviral activity against human cytomegalovirus AD169 infected in NHDF at 50 nM treated after 57 hrs postinfection measured after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510766Selectivity index, ratio of CC50 for NHDF to EC50 for human cytomegalovirus infected in NHDF2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510757Antiviral activity against human cytomegalovirus AD169 infected in NHDF assessed as inhibition of virus-induced cytopathic effect after 6 to 7 days by giemsa staining2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510761Cytotoxicity against NHDF infected with human cytomegalovirus after 7 days by alamar blue assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510753Antiviral activity against human cytomegalovirus infected in NHDF after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID510752Antiviral activity against human cytomegalovirus infected in HEL299 cells after 7 days by GFP-based fluorescent reduction assay2010Antimicrobial agents and chemotherapy, Mar, Volume: 54, Issue:3
In vitro and in vivo activities of the novel anticytomegalovirus compound AIC246.
AID1296008Cytotoxic Profiling of Annotated Libraries Using Quantitative High-Throughput Screening2020SLAS discovery : advancing life sciences R & D, 01, Volume: 25, Issue:1
Cytotoxic Profiling of Annotated and Diverse Chemical Libraries Using Quantitative High-Throughput Screening.
AID1347160Primary screen NINDS Rhodamine qHTS for Zika virus inhibitors2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1745855NCATS anti-infectives library activity on the primary C. elegans qHTS viability assay2023Disease models & mechanisms, 03-01, Volume: 16, Issue:3
In vivo quantitative high-throughput screening for drug discovery and comparative toxicology.
AID1346987P-glycoprotein substrates identified in KB-8-5-11 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
AID1745854NCATS anti-infectives library activity on HEK293 viability as a counter-qHTS vs the C. elegans viability qHTS2023Disease models & mechanisms, 03-01, Volume: 16, Issue:3
In vivo quantitative high-throughput screening for drug discovery and comparative toxicology.
AID1347159Primary screen GU Rhodamine qHTS for Zika virus inhibitors: Unlinked NS2B-NS3 protease assay2020Proceedings of the National Academy of Sciences of the United States of America, 12-08, Volume: 117, Issue:49
Therapeutic candidates for the Zika virus identified by a high-throughput screen for Zika protease inhibitors.
AID1346986P-glycoprotein substrates identified in KB-3-1 adenocarcinoma cell line, qHTS therapeutic library screen2019Molecular pharmacology, 11, Volume: 96, Issue:5
A High-Throughput Screen of a Library of Therapeutics Identifies Cytotoxic Substrates of P-glycoprotein.
[information is prepared from bioassay data collected from National Library of Medicine (NLM), extracted Dec-2023]

Research

Studies (204)

TimeframeStudies, This Drug (%)All Drugs %
pre-19900 (0.00)18.7374
1990's0 (0.00)18.2507
2000's0 (0.00)29.6817
2010's66 (32.35)24.3611
2020's138 (67.65)2.80
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Market Indicators

Research Demand Index: 68.84

According to the monthly volume, diversity, and competition of internet searches for this compound, as well the volume and growth of publications, there is estimated to be very strong demand-to-supply ratio for research on this compound.

MetricThis Compound (vs All)
Research Demand Index68.84 (24.57)
Research Supply Index5.43 (2.92)
Research Growth Index6.91 (4.65)
Search Engine Demand Index113.38 (26.88)
Search Engine Supply Index2.00 (0.95)

This Compound (68.84)

All Compounds (24.57)

Study Types

Publication TypeThis drug (%)All Drugs (%)
Trials20 (9.66%)5.53%
Reviews29 (14.01%)6.00%
Case Studies16 (7.73%)4.05%
Observational7 (3.38%)0.25%
Other135 (65.22%)84.16%
[information is prepared from research data collected from National Library of Medicine (NLM), extracted Dec-2023]

Clinical Trials (30)

Trial Overview

TrialPhaseEnrollmentStudy TypeStart DateStatus
An Open-label Pilot Protocol to Evaluate the Efficacy of Letermovir for the Prevention of Human Cytomegalovirus (CMV) Infection and Disease in Adult Lung Transplant Recipients With Idiopathic Pulmonary Fibrosis [NCT05041426]Phase 230 participants (Anticipated)Interventional2021-12-06Recruiting
A Phase 3 Randomized, Double-blind, Placebo-controlled Clinical Trial to Evaluate the Safety and Efficacy of Letermovir (LET) Prophylaxis When Extended From 100 Days to 200 Days Post-transplant in Cytomegalovirus (CMV) Seropositive Recipients (R+) of an A [NCT03930615]Phase 3220 participants (Actual)Interventional2019-06-21Completed
A Phase 2b Open-label, Single-arm Study to Evaluate Pharmacokinetics, Efficacy, Safety and Tolerability of Letermovir in Pediatric Participants From Birth to Less Than 18 Years of Age at Risk of Developing CMV Infection and/or Disease Following Allogeneic [NCT03940586]Phase 265 participants (Actual)Interventional2019-08-08Completed
A Pilot Trial of the Tolerability and Clinical Effectiveness of Letermovir When Used for Secondary Prophylaxis to Prevent Recurrent Cytomegalovirus Disease in Solid Organ Transplant Recipients [NCT05626530]Phase 425 participants (Anticipated)Interventional2023-02-02Recruiting
Influence of a 3-month Letermovir Treatment on Gut Inflammation in ART-treated HIV-infected Persons in an Open Labelled Controlled Randomized Study [NCT05362916]60 participants (Anticipated)Interventional2022-09-26Recruiting
Historical Controlled, Single Center Open Label Pilot Comparing the Effectiveness and Tolerability of De-novo Initiation of Letermovir Versus Valganciclovir for Cytomegalovirus Prophylaxis in AA Kidney Transplant Recipients [NCT06001320]Early Phase 1100 participants (Anticipated)Interventional2023-09-25Recruiting
Letermovir for Cytomegalovirus Prophylaxis in Patients With Hematological Malignancies Treated With Alemtuzumab [NCT04312841]Phase 225 participants (Anticipated)Interventional2020-09-15Recruiting
A Phase 2 Study of Letermovir Treatment for Patients Experiencing Refractory or Resistant Cytomegalovirus Infection or Disease With Concurrent Organ Dysfunction [NCT03728426]Phase 210 participants (Actual)Interventional2019-01-11Completed
Prenatal Treatment of Congenital Cytomegalovirus Infection With Letermovir Randomized Against Valaciclovir - STEP 1 [NCT04732260]7 participants (Actual)Interventional2021-06-11Completed
Letermovir for the Prevention of Cytomegalovirus Infection in Hematopoietic Cell Transplant Recipients Based on the Outcome of Metagenomic Next-Generation Sequencing: a Phase 2, Open Label, Single-Arm Clinical Trial. [NCT06021210]Phase 280 participants (Anticipated)Interventional2022-07-07Recruiting
Evaluation of the Tolerability and Clinical Effectiveness of Letermovir in Heart Transplantation [NCT04904614]Phase 427 participants (Anticipated)Interventional2021-10-01Recruiting
Letermovir Prophylaxis for CMV in Patients With Graft-versus-host Disease After Allogeneic Hematopoietic Cell Transplantation: a Proof of Concept Multicenter Study [NCT05969743]Phase 242 participants (Anticipated)Interventional2023-10-01Not yet recruiting
Randomized, Controlled Trial to Evaluate the Anti-inflammatory Efficacy of Letermovir (Prevymis) in Adults With Human Immunodeficiency Virus (HIV)-1 and Asymptomatic Cytomegalovirus (CMV) Who Are on Suppressive ART and Its Effect on Chronic Inflammation, [NCT04840199]Phase 2180 participants (Anticipated)Interventional2022-11-02Recruiting
An Open-label, Single-arm Study of Letermovir (LTV) for Prevention of Recurrent CMV Infection in High-risk Hematopoietic Cell Transplant (HCT) Recipients [NCT04017962]Phase 2106 participants (Anticipated)Interventional2019-07-19Recruiting
Letermovir Prophylaxis for Cytomegalovirus Infection in Haploidentical Allogeneic Hematopoietic Cell Transplant Recipients: Single-center Real-world Data in China [NCT05789615]50 participants (Anticipated)Observational2023-04-15Recruiting
A Phase III, Randomized, Double-Blind, Active Comparator-Controlled Study to Evaluate the Efficacy and Safety of MK-8228 (Letermovir) Versus Valganciclovir for the Prevention of Human Cytomegalovirus (CMV) Disease in Adult Kidney Transplant Recipients [NCT03443869]Phase 3601 participants (Actual)Interventional2018-05-03Completed
Prospective Study to Assess the Efficacy of Letermovir Prophylaxis in Preventing CMV Infection in Lung Transplant Recipients Compared to a Retrospective Cohort Treated With Standard Valganciclovir Prophylaxis for 12 Months (LETERCOR Study) [NCT06057194]Phase 290 participants (Anticipated)Interventional2023-10-31Not yet recruiting
Efficacy and Safety of Letermovir for Secondary Prophylaxis of Cytomegalovirus Infection After HLA-haploidentical Hematopoietic Stem Cell Transplantation [NCT05914701]Phase 431 participants (Anticipated)Interventional2023-07-01Not yet recruiting
A Randomized, Double-blind, Placebo Controlled Trial to Evaluate the Safety, Tolerability and Antiviral Activity of 12 Weeks' Treatment With a New Antiviral HCMV Drug [NCT01063829]Phase 2133 participants (Actual)Interventional2010-03-31Completed
A Phase 3, Open-Label, Single-Arm Clinical Study to Evaluate the Safety, Efficacy and Pharmacokinetics of MK-8228 (Letermovir) for the Prevention of Human Cytomegalovirus (CMV) Infection and Disease in Adult Japanese Kidney Transplant Recipients [NCT04129398]Phase 322 participants (Actual)Interventional2019-12-27Completed
Efficacy and Safety of Letermovir for Primary Prophylaxis of Cytomegalovirus Infection After HLA-haploidentical Hematopoietic Stem Cell Transplantation [NCT05914675]Phase 421 participants (Anticipated)Interventional2023-07-01Not yet recruiting
Letermovir Prophylaxis for Cytomegalovirus in Pediatric Hematopoietic Cell Transplantation [NCT05711667]Phase 3143 participants (Anticipated)Interventional2024-01-26Not yet recruiting
Immune Reconstitution to Cytomegalovirus After Allogeneic Hematopoietic Stem Cell Transplantation: Impact of Clinical Factors and Therapy Strategies [NCT05656599]120 participants (Anticipated)Observational2023-01-03Enrolling by invitation
Cytomegalovirus Prophylaxis With Letermovir in Heart Transplant Recipients: A Non-randomized Cohort Pilot Study [NCT05432778]Phase 290 participants (Anticipated)Interventional2023-05-01Recruiting
A Phase II Randomized, Placebo-Controlled, Multicenter Trial to Evaluate the Protective Function of CMV-MVA Triplex Vaccine in Adult Recipients of Haploidentical Hematopoietic Stem Cell Transplant [NCT04060277]Phase 212 participants (Actual)Interventional2019-11-27Active, not recruiting
Prenatal Treatment of Congenital Cytomegalovirus Infection With Letermovir Randomized Against Valaciclovir (Step 2) [NCT05446571]Phase 346 participants (Anticipated)Interventional2023-04-30Not yet recruiting
A Phase I Pharmacokinetic and Safety Assessment of Oral Letermovir in Infants With Symptomatic Congenital Cytomegalovirus [NCT06118515]Phase 112 participants (Anticipated)Interventional2023-11-16Not yet recruiting
A Phase 3, Open Label, Single-Arm Clinical Trial to Evaluate the Efficacy and Safety of MK-8228 (Letermovir) for the Prevention of Clinically Significant Cytomegalovirus (CMV) Infection in Chinese Adult, CMV-Seropositive Allogeneic Hematopoietic Stem Cell [NCT05763823]Phase 3120 participants (Anticipated)Interventional2023-03-24Active, not recruiting
Open Label Trial of Tolerability and Efficacy of Oral Letermovir for CMV Prophylaxis Among Heart and Lung Transplant Recipients [NCT06066957]Phase 280 participants (Anticipated)Interventional2024-01-01Not yet recruiting
A Phase III Randomized, Placebo-controlled Clinical Trial to Evaluate the Safety and Efficacy of MK-8228 (Letermovir) for the Prevention of Clinically Significant Human Cytomegalovirus (CMV) Infection in Adult, CMV-Seropositive Allogeneic Hematopoietic St [NCT02137772]Phase 3570 participants (Actual)Interventional2014-06-06Completed
[information is prepared from clinicaltrials.gov, extracted Sep-2024]

Trial Outcomes

TrialOutcome
NCT01063829 (3) [back to overview]Number of Patients With Systemic Detectable HCMV Replication.
NCT01063829 (3) [back to overview]"Time to Onset of HCMV Prophylaxis Failure"
NCT01063829 (3) [back to overview]"Number of Participants With HCMV Prophylaxis Failure"
NCT02137772 (10) [back to overview]Time to Onset of Clinically-significant CMV Infection (Kaplan-Meier Estimate of Percentage of Participants With a Qualifying Event at Week 24 Post-transplant)
NCT02137772 (10) [back to overview]Percentage of Participants With CMV End-organ Disease up to Week 14 Post-transplant
NCT02137772 (10) [back to overview]Percentage of Participants With Clinically-significant CMV Infection up to Week 24 Post-transplant
NCT02137772 (10) [back to overview]Percentage of Participants With Clinically-significant CMV Infection up to Week 14 Post-transplant
NCT02137772 (10) [back to overview]Percentage of Participants Discontinued From Study Medication Due to an Adverse Event
NCT02137772 (10) [back to overview]Percentage of Participants With CMV End-organ Disease up to Week 24 Post-transplant
NCT02137772 (10) [back to overview]Percentage of Participants With One or More Adverse Events up to Week 48 Post-transplant
NCT02137772 (10) [back to overview]Percentage of Participants With Pre-emptive Therapy for CMV Viremia up to Week 14 Post-transplant
NCT02137772 (10) [back to overview]Percentage of Participants With Pre-emptive Therapy for CMV Viremia up to Week 24 Post-transplant
NCT02137772 (10) [back to overview]Time to Initiation of Pre-emptive Therapy for CMV Viremia (Kaplan-Meier Estimate of Percentage of Participants With a Qualifying Event at Week 24 Post-transplant)
NCT03443869 (5) [back to overview]Time to Onset of Adjudicated CMV Disease Through 52 Weeks Post-transplant
NCT03443869 (5) [back to overview]Percentage of Participants With Any Drug-related Serious Adverse Event (SAE)
NCT03443869 (5) [back to overview]Percentage of Participants With Any AE
NCT03443869 (5) [back to overview]Percentage of Participants With Adjudicated Cytomegalovirus (CMV) Disease Through 52 Weeks Post-transplant
NCT03443869 (5) [back to overview]Percentage of Participants With Adjudicated CMV Disease Through 28 Weeks Post-transplant
NCT03728426 (6) [back to overview]Virological Response on Treatment Week 6
NCT03728426 (6) [back to overview]CMV Progression-free Survival
NCT03728426 (6) [back to overview]Emergence of Letermovir-resistant CMV Virus in Patients Treated in This Setting
NCT03728426 (6) [back to overview]Kinetics of Viral Clearance
NCT03728426 (6) [back to overview]Overall Survival
NCT03728426 (6) [back to overview]Percent of Patients With a Clinically Meaningful Treatment Response to Letermovir Treatment
NCT03930615 (13) [back to overview]Percentage of Participants With CMV Viremia Who Started PET From Week 14 Post-transplant to Week 48 Post-transplant
NCT03930615 (13) [back to overview]Percentage of Participants With CMV Viremia Who Started PET From Week 14 Post-transplant to Week 28 Post-transplant
NCT03930615 (13) [back to overview]Percentage of Participants With Clinically Significant CMV Infection From Week 14 Post-transplant Through Week 48 Post-transplant
NCT03930615 (13) [back to overview]Percentage of Participants With Clinically Significant CMV Infection From Week 14 Post-transplant Through Week 38 Post-transplant
NCT03930615 (13) [back to overview]Percentage of Participants With Clinically Significant CMV Infection From Week 14 (~100 Days) Post-transplant Through Week 28 (~200 Days) Post-transplant
NCT03930615 (13) [back to overview]Percentage of Participants With All-cause Mortality From Week 14 Post-transplant to Week 48 Post-transplant
NCT03930615 (13) [back to overview]Percentage of Participants With All-cause Mortality From Week 14 Post-transplant to Week 28 Post-transplant
NCT03930615 (13) [back to overview]Time to Onset of Clinically Significant CMV Infection From Week 14 Post-transplant to Week 48 Post-transplant
NCT03930615 (13) [back to overview]Time to Onset of Clinically Significant CMV Infection From Week 14 Post-transplant to Week 28 Post-transplant
NCT03930615 (13) [back to overview]Time to All-cause Mortality From Week 14 Post-transplant to Week 48 Post-transplant
NCT03930615 (13) [back to overview]Time to All-cause Mortality From Week 14 Post-transplant to Week 28 Post-transplant
NCT03930615 (13) [back to overview]Percentage of Participants Withdrawing From Study Drug Due to an AE From Week 14 (~100 Days) Post-transplant Through Week 28 (~200 Days) Post-transplant
NCT03930615 (13) [back to overview]Percentage of Participants Experiencing ≥1 Adverse Events (AEs) From Week 14 (~100 Days) Post-transplant Through Week 28 (~200 Days) Post-transplant

Number of Patients With Systemic Detectable HCMV Replication.

(NCT01063829)
Timeframe: 84 days

InterventionParticipants (Count of Participants)
AIC246 (60 mg)7
AIC246 (120 mg)5
AIC246 (240 mg)2
Placebo12

[back to top]

"Time to Onset of HCMV Prophylaxis Failure"

(NCT01063829)
Timeframe: 84 days

Interventiondays (Number)
AIC246 (60 mg)7
AIC246 (120 mg)6
AIC246 (240 mg)2
Placebo12

[back to top]

"Number of Participants With HCMV Prophylaxis Failure"

(NCT01063829)
Timeframe: 84 days

InterventionParticipants (Count of Participants)
AIC246 (60 mg)16
AIC246 (120 mg)10
AIC246 (240 mg)10
Placebo21

[back to top]

Time to Onset of Clinically-significant CMV Infection (Kaplan-Meier Estimate of Percentage of Participants With a Qualifying Event at Week 24 Post-transplant)

Clinically-significant CMV infection was defined as either one of the following: 1) onset of CMV end-organ disease, or 2) initiation of anti-CMV pre-emptive therapy based on documented CMV viremia and the clinical condition of the participant. Time to onset of clinically-significant CMV infection was defined from the day of transplantation to the day the participant developed clinically-significant CMV infection, and was analyzed by the Kaplan-Meier method. Participants were censored at the last assessment for participants who discontinued or did not develop clinically-significant CMV infection. (NCT02137772)
Timeframe: Up to Week 24 post-transplant

InterventionPercentage of participants (Number)
Letermovir18.9
Placebo44.3

[back to top]

Percentage of Participants With CMV End-organ Disease up to Week 14 Post-transplant

CMV end-organ disease met per-protocol diagnostic criteria for CMV-pneumonia, gastrointestinal disease, hepatitis, central nervous system disease, retinitis, nephritis, cystitis, myocarditis, pancreatitis, or other disease categories. Only Clinical Adjudication Committee-confirmed CMV end-organ disease was included in this analysis. The percentage of participants with CMV end-organ disease was assessed. (NCT02137772)
Timeframe: Up to Week 14 post-transplant

InterventionPercentage of participants (Number)
Letermovir0.4
Placebo1.4

[back to top]

Percentage of Participants With Clinically-significant CMV Infection up to Week 24 Post-transplant

Clinically-significant CMV infection was defined as either one of the following: 1) onset of CMV end-organ disease, or 2) initiation of anti-CMV pre-emptive therapy based on documented CMV viremia and the clinical condition of the participant. The percentage of participants with clinically-significant CMV infection was assessed. (NCT02137772)
Timeframe: Up to Week 24 post-transplant

InterventionPercentage of participants (Number)
Letermovir37.5
Placebo60.6

[back to top]

Percentage of Participants With Clinically-significant CMV Infection up to Week 14 Post-transplant

Clinically-significant CMV infection was defined as either one of the following: 1) onset of CMV end-organ disease, or 2) initiation of anti-CMV pre-emptive therapy based on documented CMV viremia and the clinical condition of the participant. The percentage of participants with clinically-significant CMV infection was assessed. (NCT02137772)
Timeframe: Up to Week 14 post-transplant

InterventionPercentage of participants (Number)
Letermovir19.1
Placebo50.0

[back to top]

Percentage of Participants Discontinued From Study Medication Due to an Adverse Event

An adverse event is defined as any untoward medical occurrence in a patient or clinical investigation participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. An adverse event can therefore be any unfavourable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening of a preexisting condition that is temporally associated with the use of the Sponsor's product, is also an adverse event. (NCT02137772)
Timeframe: Up to Week 14 post-transplant

InterventionPercentage of participants (Number)
Letermovir19.6
Placebo51.6

[back to top]

Percentage of Participants With CMV End-organ Disease up to Week 24 Post-transplant

CMV end-organ disease met per-protocol diagnostic criteria for CMV-pneumonia, gastrointestinal disease, hepatitis, central nervous system disease, retinitis, nephritis, cystitis, myocarditis, pancreatitis, or other disease categories. Only Clinical Adjudication Committee-confirmed CMV end-organ disease was included in this analysis. The percentage of participants with CMV end-organ disease was assessed. (NCT02137772)
Timeframe: Up to Week 24 post-transplant

InterventionPercentage of participants (Number)
Letermovir2.0
Placebo2.4

[back to top]

Percentage of Participants With One or More Adverse Events up to Week 48 Post-transplant

An adverse event is defined as any untoward medical occurrence in a patient or clinical investigation participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. An adverse event can therefore be any unfavourable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening of a preexisting condition that is temporally associated with the use of the Sponsor's product, is also an adverse event. (NCT02137772)
Timeframe: Up to Week 48 post-transplant

InterventionPercentage of participants (Number)
Letermovir98.4
Placebo100.0

[back to top]

Percentage of Participants With Pre-emptive Therapy for CMV Viremia up to Week 14 Post-transplant

Initiation of anti-CMV pre-emptive therapy was based on documented CMV viremia and the clinical condition of the participant. The percentage of participants with initiation of anti-CMV pre-emptive anti-CMV therapy was assessed. (NCT02137772)
Timeframe: Up to Week 14 post-transplant

InterventionPercentage of participants (Number)
Letermovir18.8
Placebo49.4

[back to top]

Percentage of Participants With Pre-emptive Therapy for CMV Viremia up to Week 24 Post-transplant

Initiation of anti-CMV pre-emptive therapy was based on documented CMV viremia and the clinical condition of the participant. The percentage of participants with initiation of anti-CMV pre-emptive anti-CMV therapy was assessed. (NCT02137772)
Timeframe: Up to Week 24 post-transplant

InterventionPercentage of participants (Number)
Letermovir36.6
Placebo59.4

[back to top]

Time to Initiation of Pre-emptive Therapy for CMV Viremia (Kaplan-Meier Estimate of Percentage of Participants With a Qualifying Event at Week 24 Post-transplant)

The need for anti-CMV pre-emptive therapy was based on documented CMV viremia and the clinical condition of the participant. The outcome was calculated from the day of transplantation to the start of anti-CMV pre-emptive therapy, and was analyzed by the Kaplan-Meier method. Participants were censored at the last assessment for participants who discontinued or did not initiate pre-emptive therapy. (NCT02137772)
Timeframe: Up to Week 24 post-transplant

InterventionPercentage of participants (Number)
Letermovir17.2
Placebo42.4

[back to top]

Time to Onset of Adjudicated CMV Disease Through 52 Weeks Post-transplant

The time to onset of adjudicated CMV disease was calculated in days, from the day of randomization to the day of onset of CMV disease as determined by the CAC. (NCT03443869)
Timeframe: Up to 52 weeks

InterventionDays (Median)
LetermovirNA
ValganciclovirNA

[back to top] [back to top]

Percentage of Participants With Any AE

An AE was defined as any untoward medical occurrence in a participant or clinical investigation participant administered a pharmaceutical product and which does not necessarily have to have a causal relationship with this treatment. An AE could therefore be any unfavourable and unintended sign, symptom, or disease temporally associated with the use of a medicinal product or protocol-specified procedure, whether or not considered related to the medicinal product or protocol-specified procedure. Any worsening of a preexisting condition that is temporally associated with the use of the Sponsor's product, was also an AE. (NCT03443869)
Timeframe: Up to 52 weeks

InterventionPercentage of Participants (Number)
Letermovir92.8
Valganciclovir92.9

[back to top]

Percentage of Participants With Adjudicated Cytomegalovirus (CMV) Disease Through 52 Weeks Post-transplant

"CMV disease was defined as the presence of either CMV end-organ disease or CMV syndrome and was confirmed by an independent, blinded Clinical Adjudication Committee (CAC). Only CAC-confirmed (adjudicated) cases were included in percentage of participants who met the endpoint. Investigator-assessed cases which were not confirmed by the CAC were not included." (NCT03443869)
Timeframe: Up to 52 weeks

InterventionPercentage of Participants (Number)
Letermovir10.4
Valganciclovir11.8

[back to top]

Percentage of Participants With Adjudicated CMV Disease Through 28 Weeks Post-transplant

"CMV disease was defined as the presence of either CMV end-organ disease or CMV syndrome and was confirmed by an independent, blinded CAC. Only CAC-confirmed (adjudicated) cases were included in percentage of participants who met the endpoint. Investigator-assessed cases which were not confirmed by the CAC were not included." (NCT03443869)
Timeframe: Up to 28 weeks

InterventionPercentage of Participants (Number)
Letermovir0.0
Valganciclovir1.7

[back to top]

Virological Response on Treatment Week 6

"The virologic response milestones are defined as: 1) Any decrease in CMV DNA from baseline (Week 0), measured on week 3 and 2) A ≥2 log decrease in CMV DNA from baseline, or an undetectable CMV DNA, measured on week 6.~For patients with clinical CMV disease, the clinical response milestones are defined as: 1) Stabilization of clinical disease by week 3 (i.e. no worsening signs or symptoms compared to week 1) as assessed by the site investigator and 2) Improvement or resolution of clinical disease by week 6 (i.e., improvement in signs and symptoms of affected organs [resolution of diarrhea, pneumonia, hepatitis, retinitis, etc.]) 3) Patients with clinical CMV disease should also meet virological response milestones outlined above to support the continuation of letermovir therapy. Patients who enter the study based solely on documented CMV disease by histopathology in the absence of quantifiable CMV virus load should remain nonquantifiable (less than 137 IU/mL)." (NCT03728426)
Timeframe: 6 weeks

InterventionParticipants (Count of Participants)
Letermovir5

[back to top]

CMV Progression-free Survival

Time from study enrollment to CMV progression or death whichever occurs first (NCT03728426)
Timeframe: 6 months

Interventiondays (Median)
Letermovir13

[back to top]

Emergence of Letermovir-resistant CMV Virus in Patients Treated in This Setting

Number of participants with breakthrough letermovir-resistant CMV infection in patients receiving letermovir treatment who experienced an initial virological response. (NCT03728426)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Letermovir0

[back to top]

Kinetics of Viral Clearance

time to undetectable plasma CMV DNA (NCT03728426)
Timeframe: 6 months

Interventiondays (Median)
Letermovir166

[back to top]

Overall Survival

number of patients alive (NCT03728426)
Timeframe: 6 months

InterventionParticipants (Count of Participants)
Letermovir7

[back to top]

Percent of Patients With a Clinically Meaningful Treatment Response to Letermovir Treatment

"Virological response and a concomitant clinical response in patients with CMV disease by Week 6 of treatment.~For patients with clinical CMV disease, the clinical response milestones are defined as:~Stabilization of clinical disease by week 3 (i.e. no worsening signs or symptoms compared to week 1) as assessed by the site investigator and~Improvement or resolution of clinical disease by week 6 (i.e., improvement in signs and symptoms of affected organs [resolution of diarrhea, pneumonia, hepatitis, retinitis, etc.])~Patients with clinical CMV disease should also meet virological response milestones outlined above to support the continuation of letermovir therapy. Patients who enter the study based solely on documented CMV disease by histopathology in the absence of quantifiable CMV virus load should remain nonquantifiable (less than 137 IU/mL)." (NCT03728426)
Timeframe: 6 Weeks

InterventionParticipants (Count of Participants)
Letermovir0

[back to top]

Percentage of Participants With CMV Viremia Who Started PET From Week 14 Post-transplant to Week 48 Post-transplant

The percentage of participants with CMV viremia who initiated PET of anti-CMV agents (ganciclovir, valganciclovir, foscarnet, and/or cidofovir) was determined. Missing values were handled with the OF approach, where failure was defined as all participants who develop clinically significant CMV infection or discontinue prematurely from the study with CMV viremia from week 14 through week 48 post-transplant. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 48 post-transplant (approximately 34 weeks)

InterventionPercentage of participants (Number)
Letermovir13.2
Placebo18.9

[back to top]

Percentage of Participants With CMV Viremia Who Started PET From Week 14 Post-transplant to Week 28 Post-transplant

The percentage of participants with CMV viremia who initiated PET of anti-CMV agents (ganciclovir, valganciclovir, foscarnet, and/or cidofovir) was determined. Missing values were handled with the OF approach, where failure was defined as all participants who develop clinically significant CMV infection or discontinue prematurely from the study with CMV viremia from week 14 through week 28 post-transplant. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 28 post-transplant (approximately 14 weeks)

InterventionPercentage of participants (Number)
Letermovir2.1
Placebo16.2

[back to top]

Percentage of Participants With Clinically Significant CMV Infection From Week 14 Post-transplant Through Week 48 Post-transplant

Clinically significant CMV infection is either the onset of probable or proven CMV end-organ disease or initiation of anti-CMV PET with approved anti-CMV agents (ganciclovir, valganciclovir, foscarnet, and/or cidofovir) based on documented CMV viremia and the clinical condition of the participant. Missing values were handled by the OF approach where failure was defined as all participants who develop clinically significant CMV infection or discontinue prematurely from the study with CMV viremia from week 14 (~100 days) through week 48 post-transplant. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 48 post-transplant (approximately 34 weeks)

InterventionPercentage of participants (Number)
Letermovir14.6
Placebo20.3

[back to top]

Percentage of Participants With Clinically Significant CMV Infection From Week 14 Post-transplant Through Week 38 Post-transplant

Clinically significant CMV infection is either the onset of probable or proven CMV end-organ disease or initiation of anti-CMV PET with approved anti-CMV agents (ganciclovir, valganciclovir, foscarnet, and/or cidofovir) based on documented CMV viremia and the clinical condition of the participant. Missing values were handled by the OF approach where failure was defined as all participants who develop clinically significant CMV infection or discontinue prematurely from the study with CMV viremia from week 14 (~100 days) through week 38 post-transplant. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 38 post-transplant (approximately 24 weeks)

InterventionPercentage of participants (Number)
Letermovir14.6
Placebo20.3

[back to top]

Percentage of Participants With Clinically Significant CMV Infection From Week 14 (~100 Days) Post-transplant Through Week 28 (~200 Days) Post-transplant

Clinically significant CMV infection is either the onset of probable or proven CMV end-organ disease or initiation of anti-CMV preemptive therapy (PET) with approved anti-CMV agents (ganciclovir, valganciclovir, foscarnet, and/or cidofovir) based on documented CMV viremia and the clinical condition of the participant. Missing values were handled by the observed failure (OF) approach where failure was defined as all participants who develop clinically significant CMV infection or discontinue prematurely from the study with CMV viremia from week 14 (~100 days) through week 28 post-transplant. It was hypothesized that LET is superior to placebo in the prevention of clinically significant CMV infection when LET prophylaxis is extended from 100 to 200 days. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 28 post-transplant (approximately 14 weeks)

InterventionPercentage of participants (Number)
Letermovir2.8
Placebo18.9

[back to top]

Percentage of Participants With All-cause Mortality From Week 14 Post-transplant to Week 48 Post-transplant

The percentage of participants who died due to any cause (all-cause mortality) from Week 14 to Week 48 was determined. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 48 post-transplant (approximately 34 weeks)

InterventionPercentage of participants (Number)
Letermovir8.3
Placebo8.1

[back to top]

Percentage of Participants With All-cause Mortality From Week 14 Post-transplant to Week 28 Post-transplant

The percentage of participants who died due to any cause (all-cause mortality) from Week 14 to Week 28 was determined. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 28 post-transplant (approximately 14 weeks)

InterventionPercentage of participants (Number)
Letermovir2.1
Placebo1.4

[back to top]

Time to Onset of Clinically Significant CMV Infection From Week 14 Post-transplant to Week 48 Post-transplant

Clinically significant CMV infection is either the onset of probable or proven CMV end-organ disease or initiation of anti-CMV PET with approved anti-CMV agents (ganciclovir, valganciclovir, foscarnet, and/or cidofovir) based on documented CMV viremia and the clinical condition of the participant. Time to onset of clinically significant CMV infection is the elapsed time from transplant to the onset of CMV end-organ disease or to the initiation of anti-CMV PET. Time to onset was determined from the Kaplan-Meier method for censored data. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 48 post-transplant (approximately 34 weeks)

InterventionDays (Median)
LetermovirNA
PlaceboNA

[back to top]

Time to Onset of Clinically Significant CMV Infection From Week 14 Post-transplant to Week 28 Post-transplant

Clinically significant CMV infection is either the onset of probable or proven CMV end-organ disease or initiation of anti-CMV PET with approved anti-CMV agents (ganciclovir, valganciclovir, foscarnet, and/or cidofovir) based on documented CMV viremia and the clinical condition of the participant. Time to onset of clinically significant CMV infection is the elapsed time from transplant to the onset of CMV end-organ disease or to the initiation of anti-CMV PET. Time to onset was determined from the Kaplan-Meier method for censored data. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 28 post-transplant (approximately 14 weeks)

InterventionDays (Median)
LetermovirNA
PlaceboNA

[back to top]

Time to All-cause Mortality From Week 14 Post-transplant to Week 48 Post-transplant

Time to all-cause mortality is the time elapsed after Week 14 post-transplant and death due to any cause, and was determined from the Kaplan-Meier method for censored data. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 48 post-transplant (approximately 34 weeks)

InterventionDays (Median)
LetermovirNA
PlaceboNA

[back to top]

Time to All-cause Mortality From Week 14 Post-transplant to Week 28 Post-transplant

Time to all-cause mortality is the time elapsed after Week 14 post-transplant and death due to any cause, and was determined from the Kaplan-Meier method for censored data. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 28 post-transplant (approximately 14 weeks)

InterventionDays (Median)
LetermovirNA
PlaceboNA

[back to top]

Percentage of Participants Withdrawing From Study Drug Due to an AE From Week 14 (~100 Days) Post-transplant Through Week 28 (~200 Days) Post-transplant

An AE is any untoward medical occurrence in a clinical study participant, temporally associated with the use of study intervention, whether or not considered related to the study intervention. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 28 post-transplant (approximately 14 weeks)

InterventionPercentage of participants (Number)
Letermovir4.9
Placebo1.4

[back to top]

Percentage of Participants Experiencing ≥1 Adverse Events (AEs) From Week 14 (~100 Days) Post-transplant Through Week 28 (~200 Days) Post-transplant

An adverse event (AE) is any untoward medical occurrence in a clinical study participant, temporally associated with the use of study intervention, whether or not considered related to the study intervention. (NCT03930615)
Timeframe: From Week 14 post-transplant to Week 28 post-transplant (approximately 14 weeks)

InterventionPercentage of participants (Number)
Letermovir88.9
Placebo93.2

[back to top]