1
|
Gourin C, Alain S, Hantz S. Anti-CMV therapy, what next? A systematic review. Front Microbiol 2023; 14:1321116. [PMID: 38053548 PMCID: PMC10694278 DOI: 10.3389/fmicb.2023.1321116] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 11/06/2023] [Indexed: 12/07/2023] Open
Abstract
Human cytomegalovirus (HCMV) is one of the main causes of serious complications in immunocompromised patients and after congenital infection. There are currently drugs available to treat HCMV infection, targeting viral polymerase, whose use is complicated by toxicity and the emergence of resistance. Maribavir and letermovir are the latest antivirals to have been developed with other targets. The approval of letermovir represents an important innovation for CMV prevention in hematopoietic stem cell transplant recipients, whereas maribavir allowed improving the management of refractory or resistant infections in transplant recipients. However, in case of multidrug resistance or for the prevention and treatment of congenital CMV infection, finding new antivirals or molecules able to inhibit CMV replication with the lowest toxicity remains a critical need. This review presents a range of molecules known to be effective against HCMV. Molecules with a direct action against HCMV include brincidofovir, cyclopropavir and anti-terminase benzimidazole analogs. Artemisinin derivatives, quercetin and baicalein, and anti-cyclooxygenase-2 are derived from natural molecules and are generally used for different indications. Although they have demonstrated indirect anti-CMV activity, few clinical studies were performed with these compounds. Immunomodulating molecules such as leflunomide and everolimus have also demonstrated indirect antiviral activity against HCMV and could be an interesting complement to antiviral therapy. The efficacy of anti-CMV immunoglobulins are discussed in CMV congenital infection and in association with direct antiviral therapy in heart transplanted patients. All molecules are described, with their mode of action against HCMV, preclinical tests, clinical studies and possible resistance. All these molecules have shown anti-HCMV potential as monotherapy or in combination with others. These new approaches could be interesting to validate in clinical trials.
Collapse
Affiliation(s)
- Claire Gourin
- INSERM, CHU Limoges, University of Limoges, RESINFIT, Limoges, France
| | - Sophie Alain
- INSERM, CHU Limoges, University of Limoges, RESINFIT, Limoges, France
- CHU Limoges, Laboratoire de Bactériologie-Virologie-Hygiène, National Reference Center for Herpesviruses, Limoges, France
| | - Sébastien Hantz
- INSERM, CHU Limoges, University of Limoges, RESINFIT, Limoges, France
- CHU Limoges, Laboratoire de Bactériologie-Virologie-Hygiène, National Reference Center for Herpesviruses, Limoges, France
| |
Collapse
|
2
|
Bottino P, Pastrone L, Curtoni A, Bondi A, Sidoti F, Zanotto E, Cavallo R, Solidoro P, Costa C. Antiviral Approach to Cytomegalovirus Infection: An Overview of Conventional and Novel Strategies. Microorganisms 2023; 11:2372. [PMID: 37894030 PMCID: PMC10608897 DOI: 10.3390/microorganisms11102372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 09/16/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Human cytomegalovirus (HCMV) is a herpesvirus capable of establishing a lifelong persistence in the host through a chronic state of infection and remains an essential global concern due to its distinct life cycle, mutations, and latency. It represents a life-threatening pathogen for immunocompromised patients, such as solid organ transplanted patients, HIV-positive individuals, and hematopoietic stem cell recipients. Multiple antiviral approaches are currently available and administered in order to prevent or manage viral infections in the early stages. However, limitations due to side effects and the onset of antidrug resistance are a hurdle to their efficacy, especially for long-term therapies. Novel antiviral molecules, together with innovative approaches (e.g., genetic editing and RNA interference) are currently in study, with promising results performed in vitro and in vivo. Since HCMV is a virus able to establish latent infection, with a consequential risk of reactivation, infection management could benefit from preventive treatment for critical patients, such as immunocompromised individuals and seronegative pregnant women. This review will provide an overview of conventional antiviral clinical approaches and their mechanisms of action. Additionally, an overview of proposed and developing new molecules is provided, including nucleic-acid-based therapies and immune-mediated approaches.
Collapse
Affiliation(s)
- Paolo Bottino
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Lisa Pastrone
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Antonio Curtoni
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Alessandro Bondi
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Francesca Sidoti
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Elisa Zanotto
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Rossana Cavallo
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| | - Paolo Solidoro
- Pneumology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy;
| | - Cristina Costa
- Microbiology and Virology Unit, A.O.U. “Città della Salute e della Scienza di Torino”, 10126 Turin, Italy; (L.P.); (A.C.); (A.B.); (F.S.); (E.Z.); (R.C.)
| |
Collapse
|
3
|
Martino M, Pitino A, Gori M, Bruno B, Crescimanno A, Federico V, Picardi A, Tringali S, Ingrosso C, Carluccio P, Pastore D, Musuraca G, Paviglianiti A, Vacca A, Serio B, Storti G, Mordini N, Leotta S, Cimminiello M, Prezioso L, Loteta B, Ferreri A, Colasante F, Merla E, Giaccone L, Busca A, Musso M, Scalone R, Di Renzo N, Marotta S, Mazza P, Musto P, Attolico I, Selleri C, Canale FA, Pugliese M, Tripepi G, Porto G, Martinelli G, Carella AM, Cerchione C. Letermovir Prophylaxis for Cytomegalovirus Infection in Allogeneic Stem Cell Transplantation: A Real-World Experience. Front Oncol 2021; 11:740079. [PMID: 34616684 PMCID: PMC8489185 DOI: 10.3389/fonc.2021.740079] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/13/2021] [Indexed: 12/15/2022] Open
Abstract
Despite effective treatments, cytomegalovirus (CMV) continues to have a significant impact on morbidity and mortality in allogeneic stem cell transplant (allo-SCT) recipients. This multicenter, retrospective, cohort study aimed to evaluate the reproducibility of the safety and efficacy of commercially available letermovir for CMV prophylaxis in a real-world setting. Endpoints were rates of clinically significant CMV infection (CSCI), defined as CMV disease or CMV viremia reactivation within day +100-+168. 204 adult CMV-seropositive allo-SCT recipients from 17 Italian centres (median age 52 years) were treated with LET 240 mg/day between day 0 and day +28. Overall, 28.9% of patients underwent a haploidentical, 32.4% a matched related, and 27.5% a matched unrelated donor (MUD) transplant. 65.7% were considered at high risk of CSCI and 65.2% had a CMV seropositive donor. Low to mild severe adverse events were observed in 40.7% of patients during treatment [gastrointestinal toxicity (36.3%) and skin rash (10.3%)]. Cumulative incidence of CSCI at day +100 and day +168 was 5.4% and 18.1%, respectively, whereas the Kaplan-Meier event rate was 5.8% (95% CI: 2.4-9.1) and 23.3% (95% CI: 16.3-29.7), respectively. Overall mortality was 6.4% at day +100 and 7.3% at day +168. This real-world experience confirms the efficacy and safety of CMV.
Collapse
Affiliation(s)
- Massimo Martino
- Centro Unico Regionale Trapianti Cellule Staminali e Terapie Cellulari (CTMO), Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Annalisa Pitino
- Istituto di Fisiologia Clinica del Consiglio Nazionale delle Ricerche (CNR), Roma, Italy
| | - Mercedes Gori
- Istituto di Fisiologia Clinica del Consiglio Nazionale delle Ricerche (CNR), Roma, Italy
| | - Benedetto Bruno
- Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, AOU Città della Salute e della Scienza di Torino, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Divisione di Ematologia, Università di Torino, Torino, Italy
| | | | - Vincenzo Federico
- Ematologia e Trapianto di Cellule Staminali, Polo Ospedaliero "Vito Fazzi", Lecce, Italy
| | - Alessandra Picardi
- UOC Ematologia con Trapianto CSE, AORN "Antonio Cardarelli", Napoli, Italy.,Dipartimento di Biomedicina e Prevenzione, Università di Roma Tor Vergata, Roma, Italy
| | | | - Claudia Ingrosso
- Ematologia e Trapianto di Midollo Osseo, Ospedale "San Giuseppe Moscati", Taranto, Italy
| | - Paola Carluccio
- UOC di Ematologia con Trapianto, Dipartimento di Emergenza e Trapianti d'Organo, Università degli Studi "Aldo Moro" e AOUC Policlinico di Bari, Bari, Italy
| | - Domenico Pastore
- Divisione di Ematologia, Ospedale "Antonio Perrino", Brindisi, Italy
| | - Gerardo Musuraca
- Unità di Ematologia, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Annalisa Paviglianiti
- Centro Unico Regionale Trapianti Cellule Staminali e Terapie Cellulari (CTMO), Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Adriana Vacca
- UO Ematologia - CTMO, Polo Ospedaliero "Armando Businco", Cagliari, Italy
| | - Bianca Serio
- Dipartimento di Medicina, Chirurgia e Odontoiatria, Università di Salerno, Salerno, Italy
| | - Gabriella Storti
- Unità di Ematologia, Azienda Ospedaliera "San Giuseppe Moscati", Avellino, Italy
| | - Nicola Mordini
- SC Ematologia, Azienda Ospedaliera "S. Croce e Carle", Cuneo, Italy
| | - Salvatore Leotta
- Programma di Trapianto Emopoietico, Azienda Policlinico "Vittorio Emanuele", Catania, Italy
| | | | - Lucia Prezioso
- Ematologia e Centro Trapianti Midollo Osseo (CTMO), Dipartimento ad Attività Integrata Medicina Generale e Specialistica, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Barbara Loteta
- Centro Unico Regionale Trapianti Cellule Staminali e Terapie Cellulari (CTMO), Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Anna Ferreri
- Centro Unico Regionale Trapianti Cellule Staminali e Terapie Cellulari (CTMO), Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Fabrizia Colasante
- Ospedale I.R.C.C.S. Casa Sollievo della Sofferenza - Centro Trapianti di Cellule Staminali, San Giovanni Rotondo, Italy
| | - Emanuela Merla
- Ospedale I.R.C.C.S. Casa Sollievo della Sofferenza - Centro Trapianti di Cellule Staminali, San Giovanni Rotondo, Italy
| | - Luisa Giaccone
- Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, AOU Città della Salute e della Scienza di Torino, Torino, Italy.,Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Divisione di Ematologia, Università di Torino, Torino, Italy
| | - Alessandro Busca
- Dipartimento di Oncologia, SSD Trapianto Allogenico di Cellule Staminali, AOU Città della Salute e della Scienza di Torino, Torino, Italy
| | - Maurizio Musso
- Unità Operativa di Oncoematologia e TMO, Istituto "La Maddalena", Palermo, Italy
| | - Renato Scalone
- Unità Operativa di Oncoematologia e TMO, Istituto "La Maddalena", Palermo, Italy
| | - Nicola Di Renzo
- Ematologia e Trapianto di Cellule Staminali, Polo Ospedaliero "Vito Fazzi", Lecce, Italy
| | - Serena Marotta
- UOC Ematologia con Trapianto CSE, AORN "Antonio Cardarelli", Napoli, Italy
| | - Patrizio Mazza
- Ematologia e Trapianto di Midollo Osseo, Ospedale "San Giuseppe Moscati", Taranto, Italy
| | - Pellegrino Musto
- UOC di Ematologia con Trapianto, Dipartimento di Emergenza e Trapianti d'Organo, Università degli Studi "Aldo Moro" e AOUC Policlinico di Bari, Bari, Italy
| | - Immacolata Attolico
- UOC di Ematologia con Trapianto, Dipartimento di Emergenza e Trapianti d'Organo, Università degli Studi "Aldo Moro" e AOUC Policlinico di Bari, Bari, Italy
| | - Carmine Selleri
- Dipartimento di Medicina, Chirurgia e Odontoiatria, Università di Salerno, Salerno, Italy
| | - Filippo Antonio Canale
- Centro Unico Regionale Trapianti Cellule Staminali e Terapie Cellulari (CTMO), Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Marta Pugliese
- Centro Unico Regionale Trapianti Cellule Staminali e Terapie Cellulari (CTMO), Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Giovanni Tripepi
- Istituto di Fisiologia Clinica del Consiglio Nazionale delle Ricerche (CNR), Reggio Calabria, Italy
| | - Gaetana Porto
- Centro Unico Regionale Trapianti Cellule Staminali e Terapie Cellulari (CTMO), Grande Ospedale Metropolitano "Bianchi-Melacrino-Morelli", Reggio Calabria, Italy
| | - Giovanni Martinelli
- Unità di Ematologia, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| | - Angelo Michele Carella
- Ospedale I.R.C.C.S. Casa Sollievo della Sofferenza - Centro Trapianti di Cellule Staminali, San Giovanni Rotondo, Italy
| | - Claudio Cerchione
- Unità di Ematologia, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy
| |
Collapse
|
4
|
Chen K, Arbona-Haddad E, Cheng MP, McDonnell AM, Gooptu M, Orejas JL, Timblin K, Silverman E, Al-Hamed R, Soiffer RJ, Hammond SP, Marty FM. Cytomegalovirus events in high-risk allogeneic hematopoietic-cell transplantation patients who received letermovir prophylaxis. Transpl Infect Dis 2021; 23:e13619. [PMID: 33866648 DOI: 10.1111/tid.13619] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/01/2021] [Accepted: 04/05/2021] [Indexed: 11/30/2022]
Abstract
Letermovir is approved for the prevention of cytomegalovirus (CMV) reactivation and clinical disease in patients undergoing allogeneic hematopoietic-cell transplantation (HCT). However, there is uncertainty about letermovir's ability to prevent clinical events during the period of prophylaxis as well as after the discontinuation of prophylaxis in the post-transplant setting. We performed a retrospective cohort study in CMV-seropositive allogeneic HCT recipients at high risk of CMV events, who received letermovir for primary prophylaxis from November 2017 through December 2019. We analyzed CMV outcomes for these patients during and after prophylaxis was discontinued. Patient outcomes were followed through June 2020. Sixty patients received letermovir for a median of 13 weeks (range, 1-72 weeks). Thirteen (22%) patients had quantifiable CMV DNAemia (reactivation) during letermovir prophylaxis a median of 9 days (range, 1-59 days) after starting letermovir. Five (8%) of these patients discontinued prophylaxis and received preemptive therapy (PET) with valganciclovir; eight (13%) continued letermovir as prophylaxis and CMV DNAemia resolved without PET. Thirteen patients (22%) had post-prophylaxis CMV reactivation a median of 33 days (range, 14-109 days) after letermovir discontinuation. In four (7%) of these patients, CMV DNAemia resolved without PET, and nine (15%) received PET. No patient developed CMV disease. Patients who developed CMV reactivation during prophylaxis did so shortly after initiation of letermovir, and most patients who developed CMV reactivation post-prophylaxis did so within 60 days after discontinuation of letermovir. Letermovir prophylaxis has changed the presentation of CMV infection in high-risk HCT patients.
Collapse
Affiliation(s)
- Kaiwen Chen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Esther Arbona-Haddad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Matthew P Cheng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Anne M McDonnell
- Department of Pharmacy, Brigham and Women's Hospital, Boston, MA, USA
| | - Mahasweta Gooptu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jose L Orejas
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Kaitlyn Timblin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Emily Silverman
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rama Al-Hamed
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Robert J Soiffer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sarah P Hammond
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Francisco M Marty
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Derigs P, Radujkovic A, Schubert ML, Schnitzler P, Schöning T, Müller-Tidow C, Hegenbart U, Schönland SO, Luft T, Dreger P, Schmitt M. Letermovir prophylaxis is effective in preventing cytomegalovirus reactivation after allogeneic hematopoietic cell transplantation: single-center real-world data. Ann Hematol 2020; 100:2087-2093. [PMID: 33270162 PMCID: PMC8285358 DOI: 10.1007/s00277-020-04362-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 11/23/2020] [Indexed: 11/28/2022]
Abstract
Morbidity and mortality after allogeneic hematopoietic cell transplantation (alloHCT) are still essentially affected by reactivation of cytomegalovirus (CMV). We evaluated 80 seropositive patients transplanted consecutively between March 2018 and March 2019 who received letermovir (LET) prophylaxis from engraftment until day +100 and retrospectively compared them with 80 patients without LET allografted between January 2017 and March 2018. The primary endpoint of this study was the cumulative incidence (CI) of clinically significant CMV infection (CS-CMVi) defined as CMV reactivation demanding preemptive treatment or CMV disease. With 14% CI of CS-CMVi at day +100 (11 events) was significantly lower in the LET cohort when compared to the control group (33 events, 41%; HR 0.29; p < 0.001). Whereas therapy with foscarnet could be completely avoided in the LET group, 7 out of 80 patients in the control cohort received foscarnet, resulting in 151 extra in-patient days for foscarnet administration (p = 0.002). One-year overall survival was 72% in the control arm vs 84% in the LET arm (HR 0.75 [95%CI 0.43–1.30]; p < 0.306). This study confirms efficacy and safety of LET for prophylaxis of CS-CMVi after alloHCT in a real-world setting, resulting in a significant patient benefit by reducing hospitalization needs and exposure to potentially toxic antiviral drugs for treatment of CMV reactivation.
Collapse
Affiliation(s)
- Patrick Derigs
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| | - Aleksandar Radujkovic
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Maria-Luisa Schubert
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Paul Schnitzler
- Center for Infectious Diseases, Virology, Heidelberg University Hospital, Heidelberg, Germany
| | - Tilman Schöning
- Department of Pharmacy, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Ute Hegenbart
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Stefan O Schönland
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Thomas Luft
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V (Hematology/Oncology/Rheumatology), Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| |
Collapse
|
6
|
Rübsamen-Schaeff H. From academic research to founding a company: the story of AiCuris. PURE APPL CHEM 2019. [DOI: 10.1515/pac-2018-1105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractThis contribution describes the experiences with three careers: leading and building an academic research institute, heading a research area in a multinational pharma company and founding and leading a biotech company, which saw its first drug successfully enter the market in its 11thyear of existence. The three positions had very different challenges, the common denominator for success was good and innovative science. However, research in a commercial environment, in addition to scientific excellence, also means to demonstrate the likely commercial success of the particular research. The most challenging, but at the same time the most interesting mission was the foundation of a new company, securing the financial means and developing the drugs, which had been discovered, in the clinics.
Collapse
Affiliation(s)
- Helga Rübsamen-Schaeff
- Founding CEO of AiCuris Anti-Infective Cures GmbH, Chair of the Scientific Advisory Board, AiCuris , Friedrich-Ebert-Strasse 475 , Wuppertal 42117 , Germany
| |
Collapse
|
7
|
Antiviral prophylaxis for cytomegalovirus infection in allogeneic hematopoietic cell transplantation. Blood Adv 2019; 2:2159-2175. [PMID: 30154125 DOI: 10.1182/bloodadvances.2018016493] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 07/13/2018] [Indexed: 01/07/2023] Open
Abstract
Patients treated with allogeneic hematopoietic cell transplantation (HCT) are at risk of cytomegalovirus (CMV) reactivation and disease, which results in increased morbidity and mortality. Although universal antiviral prophylaxis against CMV improves outcomes in solid organ transplant recipients, data have been conflicting regarding such prophylaxis in patients undergoing allogeneic HCT. We conducted a systematic review of randomized trials of prophylactic antivirals against CMV after allogeneic HCT to summarize the evolution of the field over the last 35 years and evaluate the prophylactic potential of antiviral agents against CMV after allogeneic HCT. Electronic databases were queried from database inception through 31 December 2017. For included studies, incidence of CMV infection and all-cause mortality were collected as primary outcomes; CMV disease incidence, use of preemptive therapy, and drug toxicities were collected as secondary outcomes. Nineteen clinical trials conducted between 1981 and 2017 involving a total of 4173 patients were included for review. Prophylactic strategies included use of acyclovir, valacyclovir, ganciclovir, maribavir, brincidofovir, and letermovir compared with placebo or a comparator antiviral. Fourteen trials that compared antiviral prophylaxis with placebo demonstrated overall effectiveness in reducing incidence of CMV infection (odds ratio [OR], 0.49; 95% confidence interval [CI], 0.42-0.58), CMV disease (OR, 0.56; 95% CI, 0.40-0.80), and use of preemptive therapy (OR, 0.51; 95% CI, 0.42-0.62; 6 trials); however, none demonstrated reduction in all-cause mortality (OR, 0.96; 95% CI, 0.78-1.18) except the phase 3 trial of letermovir (week-24 OR, 0.59; 95% CI, 0.38-0.98). Additional research is warranted to determine patient groups most likely to benefit from antiviral prophylaxis and its optimal deployment after allogeneic HCT.
Collapse
|
8
|
Gentry BG, Bogner E, Drach JC. Targeting the terminase: An important step forward in the treatment and prophylaxis of human cytomegalovirus infections. Antiviral Res 2018; 161:116-124. [PMID: 30472161 DOI: 10.1016/j.antiviral.2018.11.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/07/2018] [Accepted: 11/13/2018] [Indexed: 10/27/2022]
Abstract
A key step in the replication of human cytomegalovirus (HCMV) in the host cell is the generation and packaging of unit-length genomes into preformed capsids. Enzymes required for this process are so-called terminases, first described for double-stranded DNA bacteriophages. The HCMV terminase consists of the two subunits, the ATPase pUL56 and the nuclease pUL89, and a potential third component pUL51. The terminase subunits are essential for virus replication and are highly conserved throughout the Herpesviridae family. Together with the portal protein pUL104 they form a powerful biological nanomotor. It has been shown for tailed dsDNA bacteriophages that DNA translocation into preformed capsid needs an extraordinary amount of energy. The HCMV terminase subunit pUL56 provides the required ATP hydrolyzing activity. The necessary nuclease activity to cleave the concatemers into unit-length genomes is mediated by the terminase subunit pUL89. Whether this cleavage is mediated by site-specific duplex nicking has not been demonstrated, however, it is required for packaging. Binding to the portal is a prerequisite for DNA translocation. To date, it is a common view that during translocation the terminase moves along some domains of the DNA by a binding and release mechanism. These critical structures have proven to be outstanding targets for drugs to treat HCMV infections because corresponding structures do not exist in mammalian cells. Herein we examine the HCMV terminase as a target for drugs and review several inhibitors discovered by both lead-directed medicinal chemistry and by target-specific design. In addition to producing clinically active compounds the research also has furthered the understanding of the role and function of the terminase itself.
Collapse
Affiliation(s)
- Brian G Gentry
- Drake University College of Pharmacy and Health Sciences, 2507 University Ave., Des Moines, 50311, IA, USA.
| | - Elke Bogner
- Institute of Virology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - John C Drach
- University of Michigan School of Dentistry, 1101 N. University Ave., Ann Arbor, 48109, MI, USA.
| |
Collapse
|
9
|
Marty FM, Ljungman P, Chemaly RF, Maertens J, Dadwal SS, Duarte RF, Haider S, Ullmann AJ, Katayama Y, Brown J, Mullane KM, Boeckh M, Blumberg EA, Einsele H, Snydman DR, Kanda Y, DiNubile MJ, Teal VL, Wan H, Murata Y, Kartsonis NA, Leavitt RY, Badshah C. Letermovir Prophylaxis for Cytomegalovirus in Hematopoietic-Cell Transplantation. N Engl J Med 2017; 377:2433-2444. [PMID: 29211658 DOI: 10.1056/nejmoa1706640] [Citation(s) in RCA: 771] [Impact Index Per Article: 96.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND Cytomegalovirus (CMV) infection remains a common complication after allogeneic hematopoietic-cell transplantation. Letermovir is an antiviral drug that inhibits the CMV-terminase complex. METHODS In this phase 3, double-blind trial, we randomly assigned CMV-seropositive transplant recipients, 18 years of age or older, in a 2:1 ratio to receive letermovir or placebo, administered orally or intravenously, through week 14 after transplantation; randomization was stratified according to trial site and CMV disease risk. Letermovir was administered at a dose of 480 mg per day (or 240 mg per day in patients taking cyclosporine). Patients in whom clinically significant CMV infection (CMV disease or CMV viremia leading to preemptive treatment) developed discontinued the trial regimen and received anti-CMV treatment. The primary end point was the proportion of patients, among patients without detectable CMV DNA at randomization, who had clinically significant CMV infection through week 24 after transplantation. Patients who discontinued the trial or had missing end-point data at week 24 were imputed as having a primary end-point event. Patients were followed through week 48 after transplantation. RESULTS From June 2014 to March 2016, a total of 565 patients underwent randomization and received letermovir or placebo beginning a median of 9 days after transplantation. Among 495 patients with undetectable CMV DNA at randomization, fewer patients in the letermovir group than in the placebo group had clinically significant CMV infection or were imputed as having a primary end-point event by week 24 after transplantation (122 of 325 patients [37.5%] vs. 103 of 170 [60.6%], P<0.001). The frequency and severity of adverse events were similar in the two groups overall. Vomiting was reported in 18.5% of the patients who received letermovir and in 13.5% of those who received placebo; edema in 14.5% and 9.4%, respectively; and atrial fibrillation or flutter in 4.6% and 1.0%, respectively. The rates of myelotoxic and nephrotoxic events were similar in the letermovir group and the placebo group. All-cause mortality at week 48 after transplantation was 20.9% among letermovir recipients and 25.5% among placebo recipients. CONCLUSIONS Letermovir prophylaxis resulted in a significantly lower risk of clinically significant CMV infection than placebo. Adverse events with letermovir were mainly of low grade. (Funded by Merck; ClinicalTrials.gov number, NCT02137772 ; EudraCT number, 2013-003831-31 .).
Collapse
Affiliation(s)
- Francisco M Marty
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Per Ljungman
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Roy F Chemaly
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Johan Maertens
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Sanjeet S Dadwal
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Rafael F Duarte
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Shariq Haider
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Andrew J Ullmann
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Yuta Katayama
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Janice Brown
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Kathleen M Mullane
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Michael Boeckh
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Emily A Blumberg
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Hermann Einsele
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - David R Snydman
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Yoshinobu Kanda
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Mark J DiNubile
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Valerie L Teal
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Hong Wan
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Yoshihiko Murata
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Nicholas A Kartsonis
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Randi Y Leavitt
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| | - Cyrus Badshah
- From the Dana-Farber Cancer Institute and Brigham and Women's Hospital (F.M.M.) and Tufts Medical Center and Tufts University School of Medicine (D.R.S.), Boston; Karolinska University Hospital and Karolinska Institutet, Stockholm (P.L.); University of Texas M.D. Anderson Cancer Center, Houston (R.F.C.); Universitaire Ziekenhuizen Leuven, Leuven, Belgium (J.M.); City of Hope National Medical Center, Duarte (S.S.D.), and Stanford University School of Medicine, Palo Alto (J.B.) - both in California; Hospital Universitario Puerta de Hierro-Majadahonda, Madrid (R.F.D.); Juravinski Hospital and Cancer Center, McMaster University, Hamilton, ON, Canada (S.H.); Universitätsklinikum Würzburg, Würzburg, Germany (A.J.U., H.E.); Hiroshima Red Cross Hospital and Atomic-Bomb Survivors Hospital, Hiroshima (Y. Katayama), and Saitama Medical Center, Jichi Medical University, Saitama (Y. Kanda) - both in Japan; University of Chicago, Chicago (K.M.M.); Fred Hutchinson Cancer Research Center, Seattle (M.B.); Perelman School of Medicine at the University of Pennsylvania, Philadelphia (E.A.B.); and Merck, Kenilworth, NJ (M.J.D., V.L.T., H.W., Y.M., N.A.K., R.Y.L., C.B.)
| |
Collapse
|
10
|
Kropeit D, von Richter O, Stobernack HP, Rübsamen-Schaeff H, Zimmermann H. Pharmacokinetics and Safety of Letermovir Coadministered With Cyclosporine A or Tacrolimus in Healthy Subjects. Clin Pharmacol Drug Dev 2017; 7:9-21. [PMID: 28967706 DOI: 10.1002/cpdd.388] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 07/27/2017] [Indexed: 11/10/2022]
Abstract
Letermovir is being developed for human cytomegalovirus infection treatment and prophylaxis. In patients receiving transplants, antivirals are coadministered with cyclosporine A (CsA) or tacrolimus (TAC) immunosuppressants. Therefore, we investigated the potential for letermovir-immunosuppressant interactions. In 2 phase 1 clinical trials either CsA 50 mg or TAC 5 mg was administered to healthy males. Following washout, letermovir 80 mg was dosed twice daily for 7 and 11 days in the CsA and TAC trials, respectively, with a second dose of immunosuppressant coadministered with letermovir at steady state. In addition, letermovir 40 mg twice daily was administered for 14 days, and either CsA 50 or 200 mg administered on days 7 and 14. Pharmacokinetics and tolerability were assessed. Letermovir increased CsA and TAC Cmax by 37% and 70%, respectively, and exposure by 70% and 78%, respectively, compared with immunosuppressant alone; t½ was also increased from 10.7 to 17.9 hours for CsA. CsA (50/200 mg) increased letermovir Cmax,ss (109%/167%) and AUCss,τ (126%/237%) and decreased t½ (4.33 to 3.68/3.04 hours) versus letermovir alone. TAC did not significantly affect letermovir pharmacokinetics. All treatments were well tolerated. Concomitant letermovir increased TAC and CsA exposure. CsA altered letermovir pharmacokinetics, whereas TAC did not.
Collapse
Affiliation(s)
- Dirk Kropeit
- AiCuris Anti-infective Cures GmbH, Wuppertal, Germany
| | | | | | | | | |
Collapse
|
11
|
Kropeit D, Scheuenpflug J, Erb-Zohar K, Halabi A, Stobernack HP, Hulskotte EGJ, van Schanke A, Zimmermann H, Rübsamen-Schaeff H. Pharmacokinetics and safety of letermovir, a novel anti-human cytomegalovirus drug, in patients with renal impairment. Br J Clin Pharmacol 2017; 83:1944-1953. [PMID: 28345163 DOI: 10.1111/bcp.13292] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 03/08/2017] [Accepted: 03/14/2017] [Indexed: 12/18/2022] Open
Abstract
AIMS Human cytomegalovirus remains a significant issue for immunocompromised patients and existing viral polymerase targeting therapies are associated with significant toxicity. Accordingly, the viral terminase complex inhibitor, letermovir, is in development. We assessed letermovir pharmacokinetics in renal impairment. METHODS This was a Phase 1, open-label, nonrandomised trial. Estimated glomerular filtration rate based on the Modification of Diet Renal Disease equation was used to create three groups of eight subjects: healthy function (estimated glomerular filtration rate ≥ 90 ml min-1 1.73m-2 ), moderate (30-59 ml min-1 1.73m-2 ) and severe (<30 ml min-1 1.73m-2 ) impairment. Oral letermovir 120 mg was dosed once-daily for 8 days and blood collected for pharmacokinetic analyses. RESULTS All 24 subjects enrolled completed the trial. Moderate and severe renal impairment increased mean unbound letermovir fractions by 11% and 26%, respectively, vs. healthy subjects. Exposure (AUCτ,ss and Css,max ) was increased with renal impairment [least square mean ratios (90% confidence intervals) total letermovir vs. healthy subjects, AUCτ,ss 192% (143-258%) and 142% (83-243%) for moderate and severe impairment, respectively; Css,max 125% (87-182%) and 106% (75-151%), respectively]. Clearance was decreased vs. healthy subjects. Correlation analyses indicated a correlation between decreasing renal function and increased unbound letermovir concentration (R2 = 0.5076, P < 0.0001). Correlations were identified between decreased clearance with both decreased renal function (R2 = 0.0662, P = 0.2249 and R2 = 0.1861, P = 0.0353 total and unbound clearance, respectively) and increased age (R2 = 0.3548, P = 0.0021 and R2 = 0.3166, P = 0.0042 total and unbound clearance, respectively). Multiple-dose letermovir 120 mg was well tolerated across groups. CONCLUSIONS Renal impairment increased exposure to letermovir, although age was a confounding factor.
Collapse
Affiliation(s)
- Dirk Kropeit
- AiCuris Anti-infective Cures GmbH, Wuppertal, Germany
| | | | | | - Atef Halabi
- CRS Clinical Research Services Kiel GmbH, Kiel, Germany
| | | | | | | | | | | |
Collapse
|
12
|
Vahedi F, Giles EC, Ashkar AA. The Application of Humanized Mouse Models for the Study of Human Exclusive Viruses. Methods Mol Biol 2017; 1656:1-56. [PMID: 28808960 DOI: 10.1007/978-1-4939-7237-1_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
The symbiosis between humans and viruses has allowed human tropic pathogens to evolve intricate means of modulating the human immune response to ensure its survival among the human population. In doing so, these viruses have developed profound mechanisms that mesh closely with our human biology. The establishment of this intimate relationship has created a species-specific barrier to infection, restricting the virus-associated pathologies to humans. This specificity diminishes the utility of traditional animal models. Humanized mice offer a model unique to all other means of study, providing an in vivo platform for the careful examination of human tropic viruses and their interaction with human cells and tissues. These types of animal models have provided a reliable medium for the study of human-virus interactions, a relationship that could otherwise not be investigated without questionable relevance to humans.
Collapse
Affiliation(s)
- Fatemeh Vahedi
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada, L8N 3Z5
- MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON, Canada, L8N 3Z5
| | - Elizabeth C Giles
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada, L8N 3Z5
- MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON, Canada, L8N 3Z5
| | - Ali A Ashkar
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, Hamilton, ON, Canada, L8N 3Z5.
- MG DeGroote Institute for Infectious Disease Research, McMaster Immunology Research Centre, Hamilton, ON, Canada, L8N 3Z5.
| |
Collapse
|
13
|
Crawford LB, Streblow DN, Hakki M, Nelson JA, Caposio P. Humanized mouse models of human cytomegalovirus infection. Curr Opin Virol 2015; 13:86-92. [PMID: 26118890 DOI: 10.1016/j.coviro.2015.06.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/10/2015] [Indexed: 02/07/2023]
Abstract
The generation of humanized mouse models in which immune deficient mice are engrafted with human tissues allows for the direct in vivo investigation of human-restricted viruses. These humanized mouse models have been developed and improved over the past 30 years. It is now possible to achieve high levels of human cell engraftment producing human myeloid and lymphoid lineage cells. Humanized mouse models have been increasingly utilized in the study of human cytomegalovirus (HCMV), a human-specific beta-herpesvirus that infects myeloprogenitor cells and establishes a life-long latency in the infected host. This review focuses on the strengths and limitations of the current humanized mouse models used to study HCMV replication, pathogenesis and treatment.
Collapse
Affiliation(s)
| | - Daniel N Streblow
- VGTI, OHSU West Campus, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - Morgan Hakki
- Division of Infectious Diseases, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Jay A Nelson
- VGTI, OHSU West Campus, 505 NW 185th Avenue, Beaverton, OR 97006, USA
| | - Patrizia Caposio
- VGTI, OHSU West Campus, 505 NW 185th Avenue, Beaverton, OR 97006, USA.
| |
Collapse
|
14
|
Schleiss MR, McVoy MA. Overview of congenitally and perinatally acquired cytomegalovirus infections: recent advances in antiviral therapy. Expert Rev Anti Infect Ther 2014; 2:389-403. [PMID: 15482204 DOI: 10.1586/14787210.2.3.389] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Congenital and perinatal infection with human cytomegalovirus (CMV) are commonly encountered in newborns. In recent years there has been increased awareness of the disabilities that result from congenital CMV infection, which in turn has prompted interest in examining the potential efficacy of antiviral agents to prevent or ameliorate neurodevelopmental injury. Currently, there are three licensed systemic antivirals for the treatment of CMV: ganciclovir (Cytovene, Roche] and its prodrug valganciclovir [Valcyte, Roche); foscarnet (Foscavir, AstraZeneca); and cidofovir (Vistide, Pharmacia). A CMV-specific immunoglobulin is also available. Experience with these agents in the setting of congenital and perinatal CMV infection is very limited, but there are encouraging data from a controlled clinical trial indicating that ganciclovir therapy may be of value in limiting one form of neurodevelopmental injury caused by congenital infection, that of sensorineural hearing loss. Licensed antivirals for the treatment of CMV all share the common mechanism of targeting the viral DNA polymerase, but novel therapies that employ alternative modes of action are in development. Ultimately, the problem of perinatal CMV infection may be best controlled by the development of CMV vaccines, which could be administered to young women of childbearing age to help control this important public health problem.
Collapse
Affiliation(s)
- Mark R Schleiss
- Pediatrics and Molecular and Developmental Biology, Children's Hospital Research Foundation, Cincinnati, Ohio 45229, USA.
| | | |
Collapse
|
15
|
Vadlapudi AD, Vadlapatla RK, Mitra AK. Current and emerging antivirals for the treatment of cytomegalovirus (CMV) retinitis: an update on recent patents. ACTA ACUST UNITED AC 2013; 7:8-18. [PMID: 22044356 DOI: 10.2174/157489112799829765] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2011] [Revised: 09/30/2011] [Accepted: 10/04/2011] [Indexed: 12/17/2022]
Abstract
Cytomegalovirus (CMV) retinitis is the most common ocular opportunistic complication and a serious cause of vision loss in immunocompromised patients. Even though, a rise in human immunodeficiency virus (HIV) infected individuals seems to be a major factor responsible for the prevalence of CMV retinitis, the introduction of highly active antiretroviral therapy (HAART) significantly reduced the incidence and severity of CMV retinitis. Thorough evaluation of the patient's immune status and an exact classification of the retinal lesions may provide better understanding of the disease etiology, which would be necessary for optimizing the treatment conditions. Current drugs such as ganciclovir, valganciclovir, cidofovir and foscarnet have been highly active against CMV, but prolonged therapy with these approved drugs is associated with dose-limiting toxicities thus limiting their utility. Moreover development of drug-resistant mutants has been observed particularly in patients with acquired immunodeficiency syndrome (AIDS). Continuous efforts by researchers in the industry and academia have led to the development of newer candidates with enhanced antiviral efficacy and apparently minimal side effects. These novel compounds can suppress viral replication and prevent reactivation in the target population. Though some of the novel therapeutics possess potent viral inhibitory activity, these compounds are still in stages of clinical development and yet to be approved. This review provides an overview of disease etiology, existing anti-CMV drugs, advances in emerging therapeutics in clinical development and related recent patents for the treatment of CMV retinitis.
Collapse
Affiliation(s)
- Aswani D Vadlapudi
- Division of Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, 2464 Charlotte Street, Kansas City, MO 64108-2718, USA
| | | | | |
Collapse
|
16
|
Abstract
INTRODUCTION Cytomegalovirus (CMV) is a ubiquitous pathogen that establishes a lifelong asymptomatic infection in healthy individuals. Infection of immunesuppressed individuals causes serious illness. Transplant and AIDS patients are highly susceptible to CMV leading to life-threatening end-organ disease. Another vulnerable population is the developing fetus in utero, where congenital infection can result in surviving newborns with long-term developmental problems. There is no vaccine licensed for CMV and current antivirals suffer from complications associated with prolonged treatment. These include drug toxicity and emergence of resistant strains. There is an obvious need for new antivirals. Candidate intervention strategies are tested in controlled preclinical animal models but species specificity of human CMV precludes the direct study of the virus in an animal model. AREAS COVERED This review explores the current status of CMV antivirals and development of new drugs. This includes the use of animal models and the development of new improved models such as humanized animal CMV and bioluminescent imaging of virus in animals in real time. EXPERT OPINION Various new CMV antivirals are in development, some with greater spectrum of activity against other viruses. Although the greatest need is in the setting of transplant patients, there remains an unmet need for a safe antiviral strategy against congenital CMV. This is especially important as an effective CMV vaccine remains an elusive goal. In this regard, greater emphasis should be placed on suitable preclinical animal models and greater collaboration between industry and academia.
Collapse
Affiliation(s)
- Alistair McGregor
- University of Minnesota Medical School, Center for Infectious Diseases and Microbiology Translational Research, 2001 6th Street SE, MN 55455, USA.
| | | |
Collapse
|
17
|
In vivo imaging assay for the convenient evaluation of antiviral compounds against cytomegalovirus in mice. Antiviral Res 2010; 88:45-52. [DOI: 10.1016/j.antiviral.2010.07.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2009] [Revised: 06/29/2010] [Accepted: 07/06/2010] [Indexed: 11/22/2022]
|
18
|
Abstract
Most of the antiviral agents that have been approved, and are currently used in the treatment of virus infections, are targeted at HIV, HBV, herpes simplex virus (HSV), varicella-zoster virus (VZV), cytomegalovirus (CMV) and HCV or influenza virus. Additional compounds for HIV, HBV, HSV, VZV, CMV, HCV, influenza virus and several other viral infections, for example poxvirus (e.g., variola, vaccinia and monkeypox), respiratory syncytial virus, hemorrhagic fever virus (e.g., Lassa, Rift Valley and Ebola) and enterovirus (e.g., polio, Coxsackie and echo), are still in the experimental stage, that is, under clinical or preclinical development.
Collapse
Affiliation(s)
- Erik De Clercq
- Rega Institute for Medical Research, KU Leuven, Minderbroedersstraat 10, B-3000 Leuven, Belgium
| |
Collapse
|
19
|
Visalli RJ, Nicolosi DM, Irven KL, Goshorn B, Khan T, Visalli MA. The Varicella-zoster virus DNA encapsidation genes: Identification and characterization of the putative terminase subunits. Virus Res 2007; 129:200-11. [PMID: 17868947 PMCID: PMC2669082 DOI: 10.1016/j.virusres.2007.07.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2007] [Revised: 07/24/2007] [Accepted: 07/24/2007] [Indexed: 11/23/2022]
Abstract
The putative DNA encapsidation genes encoded by open reading frames (ORFs) 25, 26, 30, 34, 43, 45/42 and 54 were cloned from Varicella-zoster virus (VZV) strain Ellen. Sequencing revealed that the Ellen ORFs were highly conserved at the amino acid level when compared to those of 19 previously published VZV isolates. Additionally, RT-PCR provided the first evidence that ORF45/42 was expressed as a spliced transcript in VZV-infected cells. All seven ORFs were expressed in vitro and full length products were identified using a C-terminal V5 epitope tag. The in vitro products of the putative VZV terminase subunits encoded by ORFs 30 and 45/42 proved useful in protein-protein interaction assays. Previous studies have reported the formation of a heterodimeric terminase complex involved in DNA encapsidation for both herpes simplex virus-type 1 (HSV-1) and human cytomegalovirus (HCMV). Here we report that the C-terminal portion of exon II of ORF45/42 (ORF42-C269) interacted in GST-pull down experiments with in vitro synthesized ORF30 and ORF45/42. The interactions were maintained in the presence of anionic detergents and in buffers of increasing ionic strength. Cells transiently transfected with epitope tagged ORF45/42 or ORF30 showed primarily cytoplasmic staining. In contrast, an antiserum directed to the N-terminal portion of ORF45 showed nearly exclusive nuclear localization of the ORF45/42 gene product in infected cells. An ORF30 specific antiserum detected an 87 kDa protein in both the cytoplasmic and nuclear fractions of VZV infected cells. The results were consistent with the localization and function of herpesviral terminase subunits. This is the first study aimed at the identification and characterization of the VZV DNA encapsidation gene products.
Collapse
Affiliation(s)
- Robert J Visalli
- Department of Biology, Indiana University Purdue University Fort Wayne, 2101 E. Coliseum Blvd., Fort Wayne, IN 46805-1499, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Bravo FJ, Cardin RD, Bernstein DI. A model of human cytomegalovirus infection in severe combined immunodeficient mice. Antiviral Res 2007; 76:104-10. [PMID: 17658624 PMCID: PMC2099693 DOI: 10.1016/j.antiviral.2007.06.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 06/07/2007] [Accepted: 06/10/2007] [Indexed: 10/23/2022]
Abstract
Animal models for the evaluation of therapies against human cytomegalovirus (HCMV) are limited due to the species-specific replication of CMV. Models utilizing human fetal tissues implanted into SCID mice have been used but tend to be labor intensive and require human tissues. We therefore developed a model using HCMV-infected human foreskin fibroblasts (HFF) seeded onto a biodegradable gelatin matrix (Gelfoam). Infected HFFs are then implanted subcutaneously into SCID mice. We next evaluated two antivirals in this model. Treatment from days 0 to 5 with ganciclovir (GCV) produced a marginally significant reduction in viral titer while treatment from days 0 to 14 resulted in a more significant reduction in viral titers of 1.47 log(10)pfu/ml (P<0.0001). Viral titers were similarly reduced in a group receiving GCV treatment from days 7 to 14 post-implantation (1.50 log(10)pfu/ml, P<0.0001). Cidofovir therapy from days 7 to 14 reduced viral titers by almost 2 log(10)pfu/ml (from 3.51+/-0.31 log(10)pfu/ml in untreated animals to 1.56+/-0.40 log(10)pfu/ml in treated animals, P<0.0001). These results indicate that the Gelfoam-HCMV SCID mouse model is suitable for the in vivo evaluation of new antivirals against HCMV and is simpler and more convenient than previous models.
Collapse
Affiliation(s)
- Fernando J Bravo
- Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, ML 6014, Cincinnati, OH 45229-3039, USA
| | | | | |
Collapse
|
21
|
Biron KK. Antiviral drugs for cytomegalovirus diseases. Antiviral Res 2006; 71:154-63. [PMID: 16765457 DOI: 10.1016/j.antiviral.2006.05.002] [Citation(s) in RCA: 324] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2006] [Revised: 05/01/2006] [Accepted: 05/04/2006] [Indexed: 11/25/2022]
Abstract
Cytomegalovirus infections are associated with severe morbidity and mortality is patients at risk for disease because of immune system disabilities; in particular, recipients of stem cell (HSCT) or solid organ (SOT) transplants. There are three systemic drugs approved for CMV treatment: ganciclovir, or its prodrug valganciclovir, foscarnet, and cidofovir. An anti-sense therapeutic, ISIS 2922, is also approved specifically as in intravitreal treatment for CMV retinitis. Ganciclovir, and more recently, valganciclovir, have been useful in proactive approaches of CMV disease management; in both prophylactic and preemptive regimens in HSCT and SOT populations. The major anti-herpes agent valacyclovir has also been approved for prophylaxis of renal transplant recipients, or SOTs outside of the US. These drugs have provided major advances in CMV disease management, although they are limited by intolerable toxicities, oral bioavailability and efficacy, and risk of drug resistance with extended use. Several drugs are in early clinical development which may address these limitations; this review will provide an overview of our current arsenal of available drugs, and of those in the early clinical development pipeline.
Collapse
Affiliation(s)
- Karen K Biron
- Department of Clinical Virology, Division of Virology, GlaxoSmithKline Inc., RTP, NC, United States.
| |
Collapse
|
22
|
Abstract
The long-term treatment of herpesvirus infections with current antivirals leads to the development of drug-resistant viruses. Because currently available antivirals finally target the viral DNA polymerase, mutant resistant to one drug often shows cross-resistance to other drugs. This evidence highlights the need for the development of new antivirals that have the different viral targets. Recently, high-through-put screening of large compound collections for inhibiting specific viral enzymes, or in vitro cell culture assay, has identified several new antivirals. These include the inhibitors of helicase/primase complex, terminase complex, portal protein and UL97 protein kinase. This review will focus on these new compounds that directly inhibit viral replication.
Collapse
Affiliation(s)
- Yoshito Eizuru
- Center for Chronic Viral Diseases, Graduate School of Medical and Dental Sciences, Kagoshima University, Japan.
| |
Collapse
|
23
|
Schleiss MR, Bernstein DI, McVoy MA, Stroup G, Bravo F, Creasy B, McGregor A, Henninger K, Hallenberger S. The non-nucleoside antiviral, BAY 38-4766, protects against cytomegalovirus (CMV) disease and mortality in immunocompromised guinea pigs. Antiviral Res 2005; 65:35-43. [PMID: 15652969 PMCID: PMC2768478 DOI: 10.1016/j.antiviral.2004.09.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2004] [Accepted: 09/21/2004] [Indexed: 11/27/2022]
Abstract
New antiviral drugs are needed for the treatment of cytomegalovirus (CMV) infections, particularly in immunocompromised patients. These studies evaluated the in vitro and in vivo activity of the non-nucleosidic CMV inhibitor, BAY 38-4766, against guinea pig cytomegalovirus (GPCMV). Plaque reduction assays indicated that BAY 38-4766 was active against GPCMV, with an IC(50) of 0.5muM. Yield reduction assays demonstrated an ED(90) and ED(99) of 0.4 and 0.6muM, respectively, of BAY 38-4766 against GPCMV. Guinea pigs tolerated oral administration of 50mg/kg/day of BAY 38-4766 without evidence of biochemical or hematologic toxicity. Plasma concentrations of BAY 38-4766 were high following oral dosing, with a mean peak level at 1-h post-dose of 26.7mg/ml (n=6; range, 17.8-35.4). Treatment with BAY 38-4766 reduced both viremia and DNAemia, as determined by a real-time PCR assay, following GPCMV infection of cyclophosphamide-immunosuppressed strain 2 guinea pigs (p<0.05, Mann-Whitney test). BAY 38-4766 also reduced mortality following lethal GPCMV challenge in immunosuppressed Hartley guinea pigs, from 83% (20/24) in placebo-treated guinea pigs, to 17% (4/24) in BAY 38-4766-treated animals (p<0.0001, Fisher's exact test). Mortality differences were accompanied by reduction in DNAemia in Hartley guinea pigs. Based upon its favorable safety, pharmacokinetic, and therapeutic profiles, BAY 38-4766 warrants further investigation in the GPCMV model.
Collapse
Affiliation(s)
- Mark R Schleiss
- Division of Infectious Diseases, Children's Hospital Research Foundation, 3333 Burnet Avenue, Cincinnati, OH 45229, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
The long-term treatment of herpesvirus infections with current antivirals in immunocompromised hosts leads to the development of drug-resistant viruses. Because nearly all currently available antivirals finally target viral DNA polymerase, virus resistant to one drug often shows cross-resistance to other drugs. In addition, nearly all the antivirals show various kinds of side effects or poor bioavailability. This evidence highlights the need for developing new antivirals for herpesviruses that have the different viral targets. Recently, high-throughput screening of large compound collections for inhibiting specific viral enzymes, or in vitro cell culture assay, has identified several new antivirals that target different viral proteins. These include the inhibitors of helicase/primase complex, terminase complex, portal protein and UL97 protein kinase. In addition, non-nucleoside inhibitors for viral DNA polymerase have been also developed. This review will focus on these new compounds that directly inhibit viral replication.
Collapse
Affiliation(s)
- Yoshito Eizuru
- Division of Persistent & Oncogenic Viruses, Centre for Chronic Viral Diseases, Faculty of Medicine, Kagoshima University, Kagoshima, Japan.
| |
Collapse
|
25
|
Evers DL, Komazin G, Shin D, Hwang DD, Townsend LB, Drach JC. Interactions among antiviral drugs acting late in the replication cycle of human cytomegalovirus. Antiviral Res 2002; 56:61-72. [PMID: 12323400 DOI: 10.1016/s0166-3542(02)00094-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
This study describes the extent of cross-resistance and interactions for selected inhibitors of human cytomegalovirus (HCMV) DNA synthesis and DNA processing. HCMV isolates resistant to the benzimidazole D-ribonucleoside viral DNA processing inhibitors TCRB and BDCRB were sensitive to BAY 38-4766, a non-nucleoside inhibitor of viral DNA processing. This indicates that these two drug types have distinct interactions with the products of HCMV genes UL56 and UL89 required for viral DNA cleavage and packaging. These virus isolates also were sensitive to ganciclovir (GCV) but slightly resistant to the L-benzimidazole ribonucleoside viral DNA synthesis inhibitor 1263W94. Virus resistant to 1263W94 remained sensitive to BDCRB, GCV, and BAY 38-4766. Examination of drug-drug interactions in cell culture assays measuring inhibition of HCMV replication revealed strong synergism for the combination of BDCRB with 1263W94, and for combinations of 1263W94 with cidofovir (CDV) and foscarnet (PFA), but not with GCV. Combinations of GCV with CDV and PFA were synergistic as well. The combination of GCV with 1263W94 showed additive antiviral interactions, whereas, a combination of BAY 38-4766 with GCV showed antagonism. Interaction of BDCRB with BAY 38-4766 showed a mixed pattern of synergy and antagonism. The antiviral synergy observed between GCV and PFA or CDV serves to validate clinical combination therapies for these drugs. Antagonism seen for BAY 38-4766 with GCV indicates that these two drugs are unlikely to be useful for combination therapies. Notably, 1263W94 demonstrated greater synergy in combination with PFA or CDV than did GCV, suggesting some promise for this benzimidazole L-riboside in such combination therapies.
Collapse
Affiliation(s)
- David L Evers
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | | | | | | | | | | |
Collapse
|
26
|
Ulgar V, Maya I, Fuentes J, Fernández-Bolaños JG. New N-alkylsulfonamides and alkyl sulfonates derived from 6-C-sulfosugars. Tetrahedron 2002. [DOI: 10.1016/s0040-4020(02)00874-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
27
|
Abstract
While the treatment of herpes simplex virus with acyclovir and similar nucleoside analogues was one of the first success stories in antiviral chemotherapy, substantial unmet medical needs remain for herpesvirus diseases. In particular, the increasing numbers of immunosuppressed people due to AIDS, transplantation, cancer and aging has driven the need for improved antivirals to treat diseases caused by human cytomegalovirus (HCMV). Currently available drugs for the treatment of HCMV diseases are less than ideal agents due to issues of toxicity, modest efficacy and poor oral bioavailability. High throughput screening of large compound collections for inhibitors of specific viral enzymes or inhibition of viral growth in cell culture have identified a number of new HCMV inhibitors at several pharmaceutical companies. These compounds act by inhibition of novel molecular targets such as the viral protein kinase, viral protease and viral proteins involved in DNA cleavage/packaging. In addition, novel non-nucleoside inhibitors of the herpesvirus DNA polymerase have recently been described. This review will summarise some of these research efforts and will focus on non-nucleoside compounds that directly inhibit a viral process.
Collapse
Affiliation(s)
- Michael W Wathen
- Infectious Diseases Research, Pharmacia Corp, Kalamazoo, MI 49001, USA.
| |
Collapse
|
28
|
McSharry JJ, McDonough A, Olson B, Hallenberger S, Reefschlaeger J, Bender W, Drusano GL. Susceptibilities of human cytomegalovirus clinical isolates to BAY38-4766, BAY43-9695, and ganciclovir. Antimicrob Agents Chemother 2001; 45:2925-7. [PMID: 11557492 PMCID: PMC90754 DOI: 10.1128/aac.45.10.2925-2927.2001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
BAY38-4766 and BAY43-9695 are nonnucleosidic compounds with activities against human cytomegalovirus (HCMV). Two phenotypic assays were used to determine the drug susceptibilities of 36 HCMV clinical isolates to the BAY compounds and ganciclovir. Using either assay, both BAY compounds at a concentration of approximately 1 microM inhibited the replication of all 36 HCMV clinical isolates, including 11 ganciclovir-resistant clinical isolates, by 50%.
Collapse
Affiliation(s)
- J J McSharry
- Center for Immunology and Microbial Diseases, Albany Medical College, 47 New Scotland Ave., Albany, NY 12208, USA.
| | | | | | | | | | | | | |
Collapse
|