1
|
Hill G, Gao H, Fan Y, Zhang R, Wang L, Zong Z, Zhou Y, Qin D, Hou J. Pharmacokinetics and Safety Study of HN0141, a Novel Anti-Human Cytomegalovirus Inhibitor, in Healthy Volunteers. Clin Pharmacol Drug Dev 2025. [PMID: 40377417 DOI: 10.1002/cpdd.1542] [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: 01/29/2025] [Accepted: 02/21/2025] [Indexed: 05/18/2025]
Abstract
HN0141 is a human cytomegalovirus (HCMV) DNA terminase complex inhibitor being developed for prophylaxis and/or preemptive treatment of HCMV infection and diseases. This study evaluated the safety, tolerability, and pharmacokinetics (PK) of HN0141, following oral administration in healthy volunteers. The double-blind, placebo-controlled Phase 1 study comprised Part 1, an escalating, single-dose study involving 50-, 100-, 200-, 300-, 400-, and 525-mg doses; and Part 2, a multiple-dose study over 7 days involving 50-, 100-, 200-, and 400-mg twice-daily doses. HN0141 was rapidly absorbed following oral doses, with median time to maximum concentration achieved within 1-2 hours across all doses. At doses ranging from 50 to 100 mg, the PK profile was reasonably linear, while at doses above 200 mg, the PK profile was more than proportional. The predicted therapeutic dose would be 50-200 mg twice daily, which is at a reasonable linear PK range. All exposures tested (up to 525-mg single dose and 400-mg twice-daily multiple doses) were within the no-observed-adverse-effect level defined in animal toxicity studies. There was a mild accumulation on systemic exposure at steady state, which was achieved within 72 hours of multiple twice-daily dosing. No serious or severe adverse events were reported. HN0141's PK profile favors the twice-daily dosing, which gives a smaller concentration variation during the treatment, and thus could bring a better treatment effect with sufficient convenience. These Phase 1 data in safety and PK profile warrant further drug development in both preemptive and prophylactic treatment in patients with HCMV.
Collapse
Affiliation(s)
- George Hill
- Clinical and Medical Science Department, Phaeno Therapeutics Co. Ltd, Hangzhou, China
| | - Hong Gao
- Clinical and Medical Science Department, Phaeno Therapeutics Co. Ltd, Hangzhou, China
| | - Yingzhe Fan
- Clinical and Medical Science Department, Phaeno Therapeutics Co. Ltd, Hangzhou, China
| | - Rui Zhang
- Clinical and Medical Science Department, Phaeno Therapeutics Co. Ltd, Hangzhou, China
| | - Lu Wang
- Phase I Trial Center, PKU Care Luzhong Hospital, Shandong, China
| | - Zhaoshan Zong
- Phase I Trial Center, PKU Care Luzhong Hospital, Shandong, China
| | - Yi Zhou
- Clinical and Medical Science Department, Phaeno Therapeutics Co. Ltd, Hangzhou, China
| | - Donghui Qin
- Clinical and Medical Science Department, Phaeno Therapeutics Co. Ltd, Hangzhou, China
| | - Jie Hou
- Phase I Trial Center, PKU Care Luzhong Hospital, Shandong, China
| |
Collapse
|
2
|
Lynch K, Märtson AG. The Importance of Drug Exposure in the Development of Cytomegalovirus Resistance. Int J Antimicrob Agents 2025:107537. [PMID: 40374080 DOI: 10.1016/j.ijantimicag.2025.107537] [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: 01/10/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/17/2025]
Abstract
Human cytomegalovirus (CMV) is a widespread pathogen which remains asymptomatic in healthy individuals. However, CMV disease can be life-threatening in immunocompromised individuals, particularly in transplant patients. This disease is routinely managed by antiviral agents including (val)ganciclovir, foscarnet, cidofovir, letermovir, and maribavir. Subtherapeutic antiviral drug exposure is a common occurrence and can lead to drug-resistant CMV development, a key contributor to disease progression. Breakthrough CMV often results in graft loss, end-organ failure, or death. By optimising intracellular exposure levels of antiviral therapies, it may be possible to improve patient outcomes. Therefore, this review aims to explore the relationship between antiviral exposure and the development of drug-resistant CMV. There are several challenges to achieving optimal concentrations of current and novel CMV therapies. Narrow therapeutic indices and toxicity profiles of current CMV therapeutics contribute to their subtherapeutic exposure and hence suboptimal clinical outcomes. Alternately, novel antivirals such as letermovir and maribavir offer improved pharmacokinetic profiles. However, these agents are associated with rapid resistance development. Overall, a distinct gap exists in understanding the relationship between antiviral exposure and resistance development. As a result, current clinical markers used to predict clinical efficacy lack reliability. In future, resistance development in relation to drug exposure should be included as a clinical trial endpoint to gain understanding of exposure-resistance relationships. With solid knowledge of these relationships, more predictive in vitro and in vivo markers of clinical efficacy can be identified. Additionally, pharmacokinetic-pharmacodynamic models and combination therapies should be further explored in to improve the management of CMV.
Collapse
Affiliation(s)
- Katie Lynch
- Leiden Academic Centre for Drug Research, Faculty of Science, Leiden University, The Netherlands
| | - Anne-Grete Märtson
- Leiden Academic Centre for Drug Research, Faculty of Science, Leiden University, The Netherlands.
| |
Collapse
|
3
|
Nuevalos Guaita M, Jimoh TO, Barrall EB, Atanasoff KE, Ehrlich ME, Gandy S, García-Ríos E, Perez Romero P, Duty JA, Tortorella D. Characterization of human cytomegalovirus infection dynamics in human microglia. J Gen Virol 2025; 106:002096. [PMID: 40299764 PMCID: PMC12041478 DOI: 10.1099/jgv.0.002096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/20/2025] [Indexed: 05/01/2025] Open
Abstract
Human cytomegalovirus (HCMV) is a β-herpesvirus that establishes asymptomatic infections in immunocompetent individuals but can cause severe or even life-threatening symptoms in immunocompromised patients. HCMV can replicate in a wide variety of cells through the engagement of diverse cell factors with the viral envelope protein gH/gL/gO (trimer) or gH/gL/UL128/UL130/UL131a (pentamer), allowing for systemic spread within the human host. This study explores HCMV infection tropism and dynamics in human microglia, demonstrating the susceptibility of microglia to both clinical and laboratory HCMV strains, albeit with lower efficacy for the laboratory strain, implying that both the gH/gL-trimer and -pentamer can mediate virus entry in microglia. The importance of the gH/gL pentamer for virus entry was demonstrated by the inhibition of virus infection upon pre-incubation with a soluble neuropilin-2 (NRP-2) entry factor. Further, we demonstrated that HCMV infection can be effectively inhibited by monoclonal antibodies specific for the gH/gL complexes and HCMV hyperimmunoglobulin. Lastly, we report that microglia infection can be prevented by newly characterized chemical entry inhibitors. Altogether, these findings underscore the potential of microglia as valuable models for studying HCMV neurotropism and strategies to block virus infection in cells that can impact neurological disorders.
Collapse
Affiliation(s)
- Marcos Nuevalos Guaita
- National Center for Microbiology, Instituto de Salud Carlos III. Majadahonda, Madrid, Spain
| | - Tajudeen O. Jimoh
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emma B. Barrall
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kristina E. Atanasoff
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Michelle E. Ehrlich
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pediatrics, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Sam Gandy
- Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry and Alzheimer’s Disease Research Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- James J Peters VA Medical Center, Bronx NY, USA
| | - Estéfani García-Ríos
- Instituto de Agroquímica y Tecnología de Alimentos (IATA), Consejo Superior de Investigaciones Científicas (CSIC), Valencia, Spain
| | - Pilar Perez Romero
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, 46556, USA
| | - J. Andrew Duty
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Therapeutic Antibody Development, Drug Discovery Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Domenico Tortorella
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
4
|
Putera I, La Distia Nora R, Dewi AC, Suhada DS, Cifuentes-González C, Rojas-Carabali W, Patnaik G, Mejia-Salgado G, Sitompul R, Edwar L, Susiyanti M, Aziza Y, Biswas J, Gupta V, de-la-Torre A, Agrawal R. Antiviral therapy for cytomegalovirus retinitis: A systematic review and meta-analysis. Surv Ophthalmol 2025; 70:215-231. [PMID: 39549781 DOI: 10.1016/j.survophthal.2024.11.004] [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] [Received: 01/04/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Cytomegalovirus retinitis (CMVR) is a significant cause of blindness in patients with advanced acquired immunodeficiency syndrome (AIDS). There are no established guidelines for its treatment, resulting in varied antiviral approaches. We pooled data from 59 studies (4501 patients) to evaluate treatment variations and outcomes (CRD42022321088). Overall pooled estimates showed visual acuity improvement at 18 % (95 % CI: 7-41 %), inflammation resolution at 90 % (95 % CI: 81-95 %), retinal detachment at 11 % (95 % CI: 8-14 %), and recurrence at 19 % (95 % CI: 11-31 %). The main antiviral treatment approaches identified were: (1) intravenous antivirals alone in 33 studies, (2) intravitreal antivirals alone in 26 studies, (3) oral antivirals alone in 3 studies, and (4) a combination of systemic (oral or intravenous[IV]) and intravitreal antivirals in 7 studies, with varying schemes and durations. Ganciclovir was the predominant antiviral, with intravenous administration being the most reported (in 23 studies), followed by intravitreal administration (in 20 studies). While visual acuity improvement was comparable, inflammation resolution tended to be higher with intravitreal than with IV antivirals, though not statistically significant (88 %, 95 % CI: 69-96 % vs 75 %, 95 % CI: 35-94 %, p = 0.38). Retinitis progression rate for IV ganciclovir was lower than for those without ganciclovir. Inflammation recurrence was significantly lower in antiretroviral (ART)-treated compared to non-ART-treated HIV/AIDS patients (10 % (95 % CI: 4-20 %) vs 33 % (95 % CI: 19-50 %), p < 0.01). Neutropenia, particularly with ganciclovir, was the most reported adverse effect (up to 50 %).
Collapse
Affiliation(s)
- Ikhwanuliman Putera
- Department of Ophthalmology, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia; Department of Ophthalmology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Internal Medicine, Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands.
| | - Rina La Distia Nora
- Department of Ophthalmology, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia; Department of Internal Medicine, Section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Arlin Chyntia Dewi
- Department of Ophthalmology, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia
| | - Ditta Shabrina Suhada
- Department of Ophthalmology, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia
| | - Carlos Cifuentes-González
- Neuroscience (NEUROS) Research Group, Neurovitae Research Center, Institute of Translational Medicine (IMT), Universidad Del Rosario Escuela de Medicina y Ciencias de la Salud, Bogotá, Colombia; Programme for Ocular Inflammation & Infection Translational Research, National Healthcare Group, Singapore, Singapore; National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | - William Rojas-Carabali
- Programme for Ocular Inflammation & Infection Translational Research, National Healthcare Group, Singapore, Singapore; Department of Bioinformatics, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore
| | | | - Germán Mejia-Salgado
- Neuroscience (NEUROS) Research Group, Neurovitae Research Center, Institute of Translational Medicine (IMT), Universidad Del Rosario Escuela de Medicina y Ciencias de la Salud, Bogotá, Colombia
| | - Ratna Sitompul
- Department of Ophthalmology, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia
| | - Lukman Edwar
- Department of Ophthalmology, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia
| | - Made Susiyanti
- Department of Ophthalmology, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia
| | - Yulia Aziza
- Department of Ophthalmology, Faculty of Medicine Universitas Indonesia - Cipto Mangunkusumo Kirana Eye Hospital, Jakarta, Indonesia
| | | | - Vishali Gupta
- Advanced Eye Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Alejandra de-la-Torre
- Neuroscience (NEUROS) Research Group, Neurovitae Research Center, Institute of Translational Medicine (IMT), Universidad Del Rosario Escuela de Medicina y Ciencias de la Salud, Bogotá, Colombia
| | - Rupesh Agrawal
- Programme for Ocular Inflammation & Infection Translational Research, National Healthcare Group, Singapore, Singapore; Department of Bioinformatics, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; National Institute for Health Research Biomedical Research Centre, Moorfields Eye Hospital, UK; UCL-Institute of Ophthalmology, UK; National Healthcare Group Eye Institute, Tan Tock Seng Hospital, Singapore; Duke NUS Medical School, Singapore; Singapore Eye Research Institute, Singapore
| |
Collapse
|
5
|
Kim KW, Kim MC. Cytomegalovirus-associated panenteritis in an immunocompetent 60-year-old woman treated with oral valganciclovir: a case report. J Int Med Res 2025; 53:3000605241311449. [PMID: 39852235 PMCID: PMC11760130 DOI: 10.1177/03000605241311449] [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] [Received: 09/13/2024] [Accepted: 12/16/2024] [Indexed: 01/26/2025] Open
Abstract
Cytomegalovirus (CMV) infection typically affects immunocompromised individuals. However, CMV-associated enteritis involving the entire small intestine is rare in immunocompetent patients. We report a case of a 60-year-old immunocompetent woman with a history of diabetes mellitus who presented with diarrhea for 3 weeks. Abdominal computed tomography and stool tests revealed panenteritis accompanied by protein-losing enteropathy. Enteroscopy revealed ulcerative mucosa throughout the small intestine, and subsequent biopsies confirmed CMV-associated enteritis. Initial treatment with intravenous ganciclovir was complicated by pancytopenia, leading to its discontinuation and a switch to oral valganciclovir. This adjustment led to significant clinical improvement and symptom resolution. This case highlights the importance of considering CMV-associated enteritis in immunocompetent patients presenting with unexplained gastrointestinal symptoms and demonstrates the effectiveness of oral valganciclovir following ganciclovir-induced pancytopenia.
Collapse
Affiliation(s)
- Kye Whon Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Min Cheol Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| |
Collapse
|
6
|
Zheng JY, Huang SS, Ye JJ, Huang CT. Mpox: A narrative review on current knowledge. Biomed J 2024:100823. [PMID: 39681166 DOI: 10.1016/j.bj.2024.100823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/19/2024] [Accepted: 12/03/2024] [Indexed: 12/18/2024] Open
Affiliation(s)
- Jun-Yuan Zheng
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital at Kee-Lung, Kee-Lung, Taiwan.
| | - Shie-Shian Huang
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital at Kee-Lung, Kee-Lung, Taiwan.
| | - Jung-Jr Ye
- Division of Infectious Diseases, Department of Internal Medicine, Chang Gung Memorial Hospital at Kee-Lung, Kee-Lung, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Ching-Tai Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan; Division of Infectious Diseases, Department of Medicine, Chang Gung Memorial Hospital, Guishan, Taoyuan City, Taiwan.
| |
Collapse
|
7
|
Saqr A, Al-Kofahi M, Mohamed M, Dorr C, Remmel RP, Onyeaghala G, Oetting WS, Guan W, Mannon RB, Matas AJ, Israni A, Jacobson PA. Steroid-tacrolimus drug-drug interaction and the effect of CYP3A genotypes. Br J Clin Pharmacol 2024; 90:2837-2848. [PMID: 38994750 DOI: 10.1111/bcp.16172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 05/13/2024] [Accepted: 06/18/2024] [Indexed: 07/13/2024] Open
Abstract
AIMS Tacrolimus, metabolized by CYP3A4 and CYP3A5 enzymes, is susceptible to drug-drug interactions (DDI). Steroids induce CYP3A genes to increase tacrolimus clearance, but the effect is variable. We hypothesized that the extent of the steroid-tacrolimus DDI differs by CYP3A4/5 genotypes. METHODS Kidney transplant recipients (n = 2462) were classified by the number of loss of function alleles (LOF) (CYP3A5*3, *6 and *7 and CYP3A4*22) and steroid use at each tacrolimus trough in the first 6 months post-transplant. A population pharmacokinetic analysis was performed by nonlinear mixed-effect modelling (NONMEM) and stepwise covariate modelling to define significant covariates affecting tacrolimus clearance. A stochastic simulation was performed and translated into a Shiny application with the mrgsolve and Shiny packages in R. RESULTS Steroids were associated with modestly higher (3%-11.8%) tacrolimus clearance. Patients with 0-LOF alleles receiving steroids showed the greatest increase (11.8%) in clearance compared to no steroids, whereas those with 2-LOFs had a negligible increase (2.6%) in the presence of steroids. Steroid use increased tacrolimus clearance by 5% and 10.3% in patients with 1-LOF and 3/4-LOFs, respectively. CONCLUSIONS Steroids increase the clearance of tacrolimus but vary slightly by CYP3A genotype. This is important in individuals of African ancestry who are more likely to carry no LOF alleles, may more commonly receive steroid treatment, and will need higher tacrolimus doses.
Collapse
Affiliation(s)
- Abdelrahman Saqr
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Mahmoud Al-Kofahi
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
- Gilead Sciences, Inc., Foster City, California, USA
| | - Moataz Mohamed
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Casey Dorr
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rory P Remmel
- Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Guillaume Onyeaghala
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
| | - William S Oetting
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| | - Weihua Guan
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Roslyn B Mannon
- Division of Nephrology, Department of Internal Medicine, University of Nebraska, Omaha, Nebraska, USA
| | - Arthur J Matas
- Department of Surgery, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ajay Israni
- Hennepin Healthcare Research Institute, Minneapolis, Minnesota, USA
- Department of Medicine, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Epidemiology & Community Health, University of Minnesota, Minneapolis, Minnesota, USA
- Department of Medicine, Hennepin Healthcare, Minneapolis, Minnesota, USA
| | - Pamala A Jacobson
- Experimental and Clinical Pharmacology, University of Minnesota College of Pharmacy, Minneapolis, Minnesota, USA
| |
Collapse
|
8
|
He D, Kong D, Zeng Y, Han M, Zhang S, Li Z. Differences in safety profiles of anti-herpesvirus medications: a real-world pharmacovigilance study based on the FAERS database. Expert Opin Drug Saf 2024:1-10. [PMID: 39377277 DOI: 10.1080/14740338.2024.2412235] [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: 06/03/2024] [Revised: 08/24/2024] [Accepted: 08/27/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Anti-herpesvirus drug safety profiles have not been systematically compared. Understanding variations in adverse events (AEs) could provide reference for rational clinical use. METHODS We collected data on acyclovir, ganciclovir, valaciclovir, and foscarnet from the FDA Adverse Event Reporting System (FAERS) database from Q1 2004 to Q3 2023. Disproportionality analyses were conducted to evaluate the risk of AEs. RESULTS All drugs exhibited significant associations with hematotoxicity, with ganciclovir and foscarnet being more myelosuppressive. The correlation with renal impairment ranked as follows: foscarnet, ganciclovir, valaciclovir, and acyclovir (ROR = 16.72, 7.06, 3.51, and 2.02, respectively). Regarding hepatotoxicity, ganciclovir was associated with acute-on-chronic liver failure (ROR = 52.83), and foscarnet was associated with fulminant hepatitis (ROR = 49.91). In the nervous system, acyclovir showed the highest intensity of neurotoxicity (ROR = 14.95). Valaciclovir ranked first in toxic encephalopathy (ROR = 64.70). Foscarnet showed the highest intensity of status epilepticus (ROR = 6.45). Besides, acyclovir showed the strongest association with severe cutaneous adverse reactions (SCARs). CONCLUSIONS Our study revealed differences in safety profiles of four anti-herpesvirus medications. Ganciclovir exhibited the highest risk of hematotoxicity but appeared relatively safe in seizures and SCARs. Foscarnet was more likely to induce nephrotoxicity, seizures, and electrolyte imbalances than others. Acyclovir and valaciclovir were strongly associated with plasmacytosis, neurotoxicity, and SCARs.
Collapse
Affiliation(s)
- Dan He
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dexuan Kong
- Department of Pharmacy, Shengli Oilfield Central Hospital, Dongying, Shandong, China
| | - Yanbin Zeng
- Department of Pharmacy, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, Fujian, China
| | - Meifen Han
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shunguo Zhang
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiling Li
- Department of Pharmacy, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Obermaier B, Braun C, Hensen L, Ahmad O, Faul C, Lang P, Bethge W, Lengerke C, Vogel W. Adenovirus- and cytomegalovirus-specific adoptive T-cell therapy in the context of hematologic cell transplant or HIV infection - A single-center experience. Transpl Infect Dis 2024; 26:e14296. [PMID: 38830809 DOI: 10.1111/tid.14296] [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] [Received: 02/14/2024] [Revised: 04/17/2024] [Accepted: 05/05/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Reactivation of viral infections, in particular cytomegalovirus (CMV) and adenovirus (ADV), cause morbidity and non-relapse-mortality in states of immune deficiency, especially after allogeneic hematopoietic cell transplantation (allo-HCT). Against the background of few available pharmacologic antiviral agents, limited by toxicities and resistance, adoptive transfer of virus-specific T-cells (VST) is a promising therapeutic approach. METHODS We conducted a single-center retrospective analysis of adult patients treated with ADV- or CMV-specific T-cells in 2012-2022. Information was retrieved by review of electronic health records. Primary outcome was a response to VST by decreasing viral load or clinical improvement. Secondary outcomes included overall survival and safety of VST infusion, in particular association with graft-versus-host disease (GVHD). RESULTS Ten patients were included, of whom four were treated for ADV, five for CMV, and one for ADV-CMV-coinfection. Cells were derived from stem cell donors (6/10) or third-party donors (4/10). Response criteria were met by six of 10 patients (4/4 ADV, 2/5 CMV, and 0/1 ADV-CMV). Overall survival was 40%. No infusion related adverse events were documented. Aggravation of GVHD after adoptive immunotherapy was observed in two cases, however in temporal association with a conventional donor lymphocyte infusion and a stem cell boost, respectively. CONCLUSION In this cohort, CMV- and ADV-specific T-cell therapy appear to be safe and effective. We describe the first reported case of virus-specific T-cell therapy for CMV reactivation not associated with transplantation but with advanced HIV infection. This encourages further evaluation of adoptive immunotherapy beyond the context of allo-HCT.
Collapse
Affiliation(s)
- Benedikt Obermaier
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Christiane Braun
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Luca Hensen
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Osama Ahmad
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Christoph Faul
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Peter Lang
- Department of Pediatric Hematology and Oncology, University Children's Hospital Tuebingen, Tuebingen, Germany
| | - Wolfgang Bethge
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Claudia Lengerke
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| | - Wichard Vogel
- Department of Hematology, Oncology, Clinical Immunology, and Rheumatology, Center for Internal Medicine, University Hospital Tuebingen, Tuebingen, Germany
| |
Collapse
|
10
|
Opie J, Verburgh E, Bailly J, Mayne E, Louw V. Hematological Complications of Human Immunodeficiency Virus (HIV) Infection: An Update From an HIV-Endemic Setting. Open Forum Infect Dis 2024; 11:ofae162. [PMID: 38601746 PMCID: PMC11004791 DOI: 10.1093/ofid/ofae162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 03/17/2024] [Indexed: 04/12/2024] Open
Abstract
Medical professionals, particularly in regions with a high burden of human immunodeficiency virus (HIV), should be alert to the hematological complications of HIV, which may include cytopenias, malignancy, and coagulation disturbances. Patients may present with these conditions as the first manifestation of HIV infection. Hematological abnormalities are often multifactorial with opportunistic infections, drugs, malignancy, and HIV infection itself contributing to the clinical presentation, and the diagnosis should consider all these factors. Life-threatening hematological complications requiring urgent diagnosis and management include thrombotic thrombocytopenic purpura, superior mediastinal syndrome, spinal cord compression, and tumor lysis syndrome due to aggressive lymphoma. Antiretroviral therapy is the therapeutic backbone, including for patients with advanced HIV, in addition to specific therapy for the complication. This article reviews the impact of HIV on the hematological system and provides a clinical and diagnostic approach, including the role of a bone marrow biopsy, focusing on perspectives from sub-Saharan Africa.
Collapse
Affiliation(s)
- Jessica Opie
- Division of Haematology, Department of Pathology, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| | - Estelle Verburgh
- Division of Clinical Haematology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- Division of Clinical Haematology, Department of Medicine, Groote Schuur Hospital, Cape Town, South Africa
| | - Jenique Bailly
- Division of Haematology, Department of Pathology, University of Cape Town, Cape Town, South Africa
- National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| | - Elizabeth Mayne
- National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
- Division of Immunology, Department of Pathology, University of Cape Town, Cape Town, South Africa
| | - Vernon Louw
- Division of Clinical Haematology, Department of Medicine, University of Cape Town, Cape Town, South Africa
- Division of Clinical Haematology, Department of Medicine, Groote Schuur Hospital, Cape Town, South Africa
| |
Collapse
|
11
|
Schultz BG, Bullano M, Paratane D, Rajagopalan K. Cytomegalovirus related hospitalization costs among hematopoietic stem cell and solid organ transplant recipients treated with maribavir versus investigator-assigned therapy: A US-based study. Transpl Infect Dis 2024; 26:e14216. [PMID: 38221739 DOI: 10.1111/tid.14216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 10/19/2023] [Accepted: 12/02/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND Cytomegalovirus (CMV) infections among hematopoietic stem cell transplant (HSCT) and solid organ transplant (SOT) recipients impose a significant health care resource utilization (HCRU)-related economic burden. Maribavir (MBV), a novel anti-viral therapy (AVT), approved by the United States Food and Drug Administration for post-transplant CMV infections refractory (with/without resistance) to conventional AVTs has demonstrated lower hospital length of stay (LOS) versus investigator-assigned therapy (IAT; valgancilovir, ganciclovir, foscarnet, or cidofovir) in a phase 3 trial (SOLSTICE). This study estimated the HCRU costs of MBV versus IAT. METHODS An economic model was developed to estimate HCRU costs for patients treated with MBV or IAT. Mean per-patient-per-year (PPPY) HCRU costs were calculated using (i) annualized mean hospital LOS in SOLSTICE, and (ii) CMV-related direct costs from published literature. Probabilistic sensitivity analysis with Monte-Carlo simulations assessed model robustness. RESULTS Of 352 randomized patients receiving MBV (n = 235) or IAT (n = 117) for 8 weeks in SOLSTICE, 40% had HSCT and 60% had SOT. Mean overall PPPY HCRU costs of overall hospital-LOS were $67,205 (95% confidence interval [CI]: $33,767, $231,275) versus $145,501 (95% CI: $62,064, $589,505) for MBV and IAT groups, respectively. Mean PPPY ICU and non-ICU stay costs were: $32,231 (95% CI: $5,248, $184,524) versus $45,307 (95% CI: $3,957, $481,740) for MBV and IAT groups, and $82,237 (95% CI: $40,397, $156,945) MBV versus $228,329 (95% CI: $94,442, $517,476) for MBV and IAT groups, respectively. MBV demonstrated cost savings in over 99.99% of simulations. CONCLUSIONS This analysis suggests that Mean PPPY HCRU costs were 29%-64% lower with MBV versus other-AVTs.
Collapse
Affiliation(s)
- Bob G Schultz
- US Medical Affairs Outcomes Research, Takeda Pharmaceuticals U.S.A., Inc., Lexington, Massachusetts, USA
| | - Michael Bullano
- US Medical Affairs Outcomes Research, Takeda Pharmaceuticals U.S.A., Inc., Lexington, Massachusetts, USA
| | - Deepika Paratane
- Health Economics and Outcomes Research, Anlitiks, Inc., Windermere, Florida, USA
| | - Krithika Rajagopalan
- Health Economics and Outcomes Research, Anlitiks, Inc., Windermere, Florida, USA
| |
Collapse
|
12
|
Tverdek F, Escobar ZK, Liu C, Jain R, Lindsay J. Antimicrobials in patients with hematologic malignancies and recipients of hematopoietic cell transplantation and other cellular therapies. Transpl Infect Dis 2023; 25 Suppl 1:e14129. [PMID: 37594221 DOI: 10.1111/tid.14129] [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: 06/15/2023] [Revised: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND Appropriate use of antimicrobials for hematologic malignancy, hematopoietic stem cell transplant recipients, and other cellular therapies is vital, with infection causing significant morbidity and mortality in this unique population of immunocompromised hosts. However, often in this population the choice and management of antimicrobial therapy is complex. When selecting an antimicrobial agent, key considerations include the need for dose adjustments due to renal or hepatic impairment, managing drug interactions, the potential for additive drug toxicity among those receiving polypharmacy and therapeutic drug monitoring. Other factors include leveraging pharmacodynamic principles to enable optimization of directed therapy against challenging pathogens, as well as judicious use of antimicrobials to limit drug resistance and adverse drug reactions. SUMMARY This review summarizes the clinical considerations for commonly used antimicrobials in this setting, including antibacterial, antiviral, and antifungal agents.
Collapse
Affiliation(s)
- Frank Tverdek
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pharmacy, University of Washington Medicine, Seattle, Washington, USA
- School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Zahra Kassamali Escobar
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Pharmacy, University of Washington Medicine, Seattle, Washington, USA
- School of Pharmacy, University of Washington, Seattle, Washington, USA
| | - Catherine Liu
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Rupali Jain
- Department of Pharmacy, University of Washington Medicine, Seattle, Washington, USA
- School of Pharmacy, University of Washington, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Julian Lindsay
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- National Centre for Infection in Cancer and Transplantation (NCICT), Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Galletta TJ, Lane A, Lutzko C, Leemhuis T, Cancelas JA, Khoury R, Wang YM, Hanley PJ, Keller MD, Bollard CM, Davies SM, Grimley MS, Rubinstein JD. Third-Party and Patient-Specific Donor-Derived Virus-Specific T Cells Demonstrate Similar Efficacy and Safety for Management of Viral Infections after Hematopoietic Stem Cell Transplantation in Children and Young Adults. Transplant Cell Ther 2023; 29:305-310. [PMID: 36736781 DOI: 10.1016/j.jtct.2023.01.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/05/2023]
Abstract
Infections with double-stranded DNA viruses are a common complication after hematopoietic stem cell transplantation (HSCT) and cause significant morbidity and mortality in the post-transplantation period. Both donor-derived (DD) and third-party (TP) virus-specific T cells (VSTs) have shown efficacy and safety in viral management following HSCT in children and young adults. Owing to a greater degree of HLA matching between the recipient and stem cell donor, DD VSTs potentially persist longer in circulation compared to TP VSTs, because they are collected from a well-matched donor. However, TP VSTs are more easily accessible, particularly for smaller transplantation centers that do not have VST manufacturing capabilities, and more economical than creating a customized product for each transplant recipient. We conducted the present study to compare clinical efficacy and safety outcomes for DD VSTs and TP VSTs in a large cohort of pediatric and young adult HSCT recipients and to determine whether DD VSTs are associated with improved outcomes owing to potentially longer persistence in the recipient's circulation. This retrospective cohort study included 145 patients who received VSTs at Cincinnati Children's Hospital Medical Center (CCHMC) between 2017 and 2021 for the treatment of adenovirus, BK virus, cytomegalovirus, and/or Epstein-Barr virus. Viruses were detected using quantitative polymerase chain reaction. Patients received VSTs on a DD (NCT02048332) or TP (NCT02532452) protocol, and VST products for both protocols were manufactured in an identical fashion. The primary study outcome was clinical response to VSTs, evaluated 4 weeks after VST administration, defined as decrease in viral load to under the inclusion thresholds, or resolution of symptoms of invasive viral infection, without the need for additional conventional antiviral medication following VST administration. Secondary outcomes included graft-versus-host-disease, transplant-associated thrombotic microangiopathy, renal function, hospital length of stay, and overall survival at 30 days and 100 days after VST administration and 1 year after HSCT. Statistical analysis was performed using the Fisher exact test or chi-square test. An unpaired t test was used to compare continuous variables. The study group comprised 77 patients in the DD cohort and 68 patients in the TP cohort. Eighteen patients in the TP cohort underwent HSCT at CCHMC, and the other 50 underwent HSCT at other institutions and presented to CCHMC solely for VST administration. There was no statistically significant difference in clinical response rates between DD and TP cohorts (65.6% versus 62.7%; odds ratio [OR], 1.162; 95% confidence interval [CI], .619 to 2.164; P = .747). There were no significant differences in secondary outcomes between the 2 cohorts. The percentage of patients requiring multiple infusions for a clinical response did not differ significantly between the DD and TP cohorts (38.2% versus 32.5%; OR, .780; 95% CI, .345 to 1.805; P = .666). We found no significant difference in clinical response rate between DD VSTs and TP VSTs and a similar safety profile. Our data suggest that TP VSTs may be sufficient to control viral infection until immune reconstitution occurs despite the potential for more rapid VST clearance compared to DD VSTs. The lack of significant differences between DD VSTs and TP VSTs is an important finding, indicating that it is not necessary for every transplant center to manufacture customized DD VSTs, and that TP VSTs are a satisfactory substitute.
Collapse
Affiliation(s)
- Thomas J Galletta
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio.
| | - Adam Lane
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Carolyn Lutzko
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Divison of Experimental Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Hoxworth Blood Center, University of Cincinnati, Cincinnati, Ohio
| | - Thomas Leemhuis
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Hoxworth Blood Center, University of Cincinnati, Cincinnati, Ohio
| | - Jose A Cancelas
- Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Divison of Experimental Hematology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Hoxworth Blood Center, University of Cincinnati, Cincinnati, Ohio
| | - Ruby Khoury
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - YunZu M Wang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Hospital and Department of Pediatrics, The George Washington University, Washington, DC
| | - Michael D Keller
- Center for Cancer and Immunology Research, Children's National Hospital and Department of Pediatrics, The George Washington University, Washington, DC
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital and Department of Pediatrics, The George Washington University, Washington, DC
| | - Stella M Davies
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Michael S Grimley
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jeremy D Rubinstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio; Division of Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
14
|
Abstract
Maribavir was approved by the U.S. Food and Drug Administration in November 2021 for the treatment of adult and pediatric patients with post-transplant cytomegalovirus (CMV) infection/disease that is refractory to treatment (with or without genotypic resistance) with ganciclovir, valganciclovir, cidofovir, or foscarnet. Maribavir is an oral benzimidazole riboside with potent and selective multimodal anti-CMV activity. It utilizes a novel mechanism of action which confers activity against CMV strains that are resistant to traditional anti-CMV agents, and also offers a more favorable safety profile relative to the dose-limiting side effects of previously available therapies. Maribavir was initially studied as an agent for CMV prophylaxis in solid organ and hematopoietic stem cell recipients, but initial phase III trials failed to meet clinical efficacy endpoints. It has been more recently studied as a therapeutic agent at higher doses for refractory-resistant (R-R) CMV infections with favorable outcomes. After an overview of maribavir's chemistry and clinical pharmacology, this review will summarize clinical efficacy, safety, tolerability, and resistance data associated with maribavir therapy.
Collapse
|
15
|
Maillard M, Gong L, Nishii R, Yang JJ, Whirl-Carrillo M, Klein TE. PharmGKB summary: acyclovir/ganciclovir pathway. Pharmacogenet Genomics 2022; 32:201-208. [PMID: 35665708 PMCID: PMC9179945 DOI: 10.1097/fpc.0000000000000474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Affiliation(s)
- Maud Maillard
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Li Gong
- Departments of Biomedical Data Science
| | - Rina Nishii
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee
| | | | - Teri E Klein
- Departments of Biomedical Data Science
- Medicine (BMIR), Stanford University, Stanford, California, USA
| |
Collapse
|
16
|
Rubinstein JD, Lutzko C, Leemhuis T, Zhu X, Pham G, Ray L, Thomas S, Dourson C, Wilhelm J, Lane A, Cancelas JA, Lipps D, Ferrell J, Hanley PJ, Keller MD, Bollard CM, Wang YM, Davies SM, Nelson AS, Grimley MS. Scheduled administration of virus-specific T cells for viral prophylaxis after pediatric allogeneic stem cell transplant. Blood Adv 2022; 6:2897-2907. [PMID: 35108727 PMCID: PMC9092421 DOI: 10.1182/bloodadvances.2021006309] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 01/22/2022] [Indexed: 11/20/2022] Open
Abstract
Infections with double-stranded DNA viruses are a significant cause of morbidity and mortality in pediatric patients following allogeneic hematopoietic stem cell transplantation (HSCT). Virus-specific T-cell therapies (VSTs) have been shown to be an effective treatment for infections with adenovirus, BK virus, cytomegalovirus (CMV), and Epstein-Barr virus (EBV). To date, prophylactic regimens to prevent or mitigate these infections using conventional antiviral medications provide suboptimal response rates. Here we report on a clinical trial (NCT03883906) performed to assess the feasibility of rapid manufacturing and early infusion of quadrivalent VSTs generated from stem cell donors ("donor-derived VSTs") into allogeneic HSCT recipients with minimal or absent viremia. Patients were eligible to receive scheduled VSTs as early as 21 days after stem cell infusion. Twenty-three patients received scheduled VSTs. Twenty of 23 patients had no viremia at the time of infusion, while 3 patients had very low-level BK viremia. Two developed clinically significant graft-versus-host disease (GVHD), although this incidence was not outside of expected incidence early after HSCT, and both were successfully treated with systemic corticosteroids (n = 2). Five patients were deemed treatment failures. Three developed subsequent significant viremia/viral disease (n = 3). Eighteen patients did not fail treatment, 7 of whom did not develop any viremia, while 11 developed low-level, self-limited viremia that resolved without further intervention. No infusion reactions occurred. In conclusion, scheduled VSTs appear to be safe and potentially effective at limiting serious complications from viral infections after allogeneic transplantation. A randomized study comparing this scheduled approach to the use of VSTs to treat active viremia is ongoing.
Collapse
Affiliation(s)
- Jeremy D. Rubinstein
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Oncology, and
| | - Carolyn Lutzko
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Experimental Hematology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Thomas Leemhuis
- Hoxworth Blood Center, University of Cincinnati, Cincinnati, OH
| | - Xiang Zhu
- Division of Experimental Hematology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Giang Pham
- Division of Experimental Hematology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Lorraine Ray
- Division of Experimental Hematology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Shawn Thomas
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Celeste Dourson
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Jamie Wilhelm
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Adam Lane
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Jose A. Cancelas
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Experimental Hematology, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
- Hoxworth Blood Center, University of Cincinnati, Cincinnati, OH
| | - Dakota Lipps
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Justin Ferrell
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Patrick J. Hanley
- Center for Cancer and Immunology Research, Children’s National Health System and Department of Pediatrics, The George Washington University, Washington, DC
| | - Michael D. Keller
- Center for Cancer and Immunology Research, Children’s National Health System and Department of Pediatrics, The George Washington University, Washington, DC
| | - Catherine M. Bollard
- Center for Cancer and Immunology Research, Children’s National Health System and Department of Pediatrics, The George Washington University, Washington, DC
| | - YunZu M. Wang
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Stella M. Davies
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Adam S. Nelson
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| | - Michael S. Grimley
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplant and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH; and
| |
Collapse
|
17
|
Scheuenpflug J, Kropeit D, Erb-Zohar K, Theis JGW, Stobernack HP, McCormick D, Zimmermann H, Rübsamen-Schaeff H. The Effect of Oral Letermovir Administration on the Pharmacokinetics of a Single Oral Dose of P-Glycoprotein Substrate Digoxin in Healthy Volunteers. Clin Pharmacol Drug Dev 2021; 11:6-15. [PMID: 34812580 DOI: 10.1002/cpdd.1043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/27/2021] [Indexed: 11/12/2022]
Abstract
Letermovir is a human cytomegalovirus (CMV) terminase inhibitor approved in the United States, Canada, Japan, and the European Union for prophylaxis of CMV infection and disease in CMV-seropositive, allogeneic, hematopoietic stem-cell transplant recipients. In vitro, letermovir is a substrate and potential modulator of P-glycoprotein. The potential of letermovir to alter the pharmacokinetics of digoxin (a P-glycoprotein substrate) upon coadministration in healthy subjects was therefore investigated in a phase 1 trial (EudraCT: 2011-004516-39). Oral letermovir 240 mg was administered twice daily for 12 days with a single oral digoxin 0.5-mg dose on day 7; after a washout period, oral digoxin 0.5 mg was administered on day 35 (sequence 1). The period order was reversed after a 28-day washout for sequence 2. Pharmacokinetics and safety were evaluated. The presence of steady-state letermovir reduced digoxin area under the plasma concentration-time curve from administration until last quantifiable measurement by 12% and maximum plasma concentration by 22% compared with digoxin alone; digoxin half-life and elimination rate remained similar in both conditions. The between-subject variability of digoxin maximum plasma concentration was higher with letermovir than without (42% vs 31%) and similar for digoxin area under the plasma concentration-time curve in both periods. No specific safety or tolerability concerns were identified. Overall, letermovir had no clinically relevant effect on concomitant administration with digoxin.
Collapse
Affiliation(s)
- Jürgen Scheuenpflug
- AiCuris Anti-infective Cures AG, Wuppertal, Germany.,Merck KGaA, Darmstadt, Germany
| | - Dirk Kropeit
- AiCuris Anti-infective Cures AG, Wuppertal, Germany
| | | | | | | | - David McCormick
- AiCuris Anti-infective Cures AG, Wuppertal, Germany.,DMPK Solutions Ltd, Nottinghamshire, UK
| | | | | |
Collapse
|
18
|
Kropeit D, McCormick D, Erb-Zohar K, Stobernack HP, Zimmermann H, Rübsamen-Schaeff H. Pharmacokinetics and Safety of Letermovir and Midazolam Coadministration in Healthy Subjects. Clin Pharmacol Drug Dev 2021; 11:16-24. [PMID: 34714967 DOI: 10.1002/cpdd.1027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 09/02/2021] [Indexed: 11/09/2022]
Abstract
Letermovir is a human cytomegalovirus (CMV) terminase inhibitor for the prophylaxis of CMV infection and disease in allogeneic hematopoietic stem-cell transplant recipients. In vitro studies have identified letermovir as a potential cytochrome P450 (CYP) 3A inhibitor. Thus, the effect of letermovir on the CYP3A isoenzyme-specific probe drug midazolam was investigated in a phase 1 trial. Healthy female subjects received single-dose intravenous (IV; 1 mg) and oral (2 mg) midazolam on days -4 and -2, respectively. Letermovir 240 mg once daily was administered on days 1 to 6, and further single doses of midazolam 1 mg IV and oral midazolam 2 mg were administered on days 4 and 6, respectively. Pharmacokinetics, tolerability, and safety were monitored throughout the trial. Following coadministration with letermovir, the least square means ratio for maximum plasma concentration and area under the plasma concentration-time curve from time 0 to the last measurable concentration was 172.4% and 225.3%, respectively, for oral midazolam, and 105.2% and 146.6%, respectively, for midazolam IV. The area under the plasma concentration-time curve from time 0 to the last measurable concentration ratio of midazolam to 1-hydroxymidazolam increased slightly in the presence of letermovir following IV (8.8-13.1; 49% increase) and oral (3.3-5.3; 59% increase) midazolam. Letermovir reached steady state, on average, by days 5 to 6. All treatments were generally well tolerated. Letermovir demonstrated moderate CYP3A inhibition.
Collapse
Affiliation(s)
- Dirk Kropeit
- AiCuris Anti-infective Cures AG, Wuppertal, Germany
| | - David McCormick
- AiCuris Anti-infective Cures AG, Wuppertal, Germany.,Current address: DMPK Solutions Ltd., Nottingham, Nottinghamshire, UK
| | | | | | | | | |
Collapse
|
19
|
Reddy S, Tyagi M, Behera S, Pappuru RR, Dave VP, Basu S, Agrawal H. Cytomegalovirus retinitis in patients of non-Hodgkin's lymphoma: clinical presentations and outcomes. J Ophthalmic Inflamm Infect 2021; 11:26. [PMID: 34611773 PMCID: PMC8492880 DOI: 10.1186/s12348-021-00257-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 08/09/2021] [Indexed: 01/10/2023] Open
Abstract
Background Cytomegalovirus (CMV) retinitis in patients with Non-Hodgkin’s Lymphoma (NHL) can occur even in the presence of high CD 4 counts and can behave differently when compared to CMV retinitis in human immunodeficiency (HIV) patients. It, therefore, becomes important to understand its varied presentations and the challenges in management of these cases. The aim of this study was to analyse the various patterns of presentations and outcomes of CMV Retinitis in patients with NHL. Study design A retrospective chart review of seven eyes of four patients of NHL presenting with CMV retinitis between June 2017 and May 2020 was done. Methods Clinical patterns of CMV Retinitis, CD4 counts at the time of presentation and the duration of treatment along with recurrences and time for recurrence of retinitis were assessed. Results Granular or indolent retinitis (6 out of 7 eyes) was the commonest form of CMV retinitis in patients of NHL. Three patients had a presenting CD4 count above 150 cells/mm3 and none of them were below 50 cells/mm3. Floaters were the commonest presenting complaint. All patients had vitritis and majority of the patients (3 out of 4) had anterior chamber (AC) inflammation. Two out of the 4 patients had a recurrence (mean time 33.8 days) after stopping the maintenance phase of ganciclovir and one patient had significant myelosuppression related to oral valganciclovir which required discontinuation of the drug. Conclusion CMV retinitis in NHL patients is usually of an indolent or granular type and can occur even in the presence of high CD4 counts as compared to patients with HIV. These patients may require a long term maintenance in view of frequent recurrences after discontinuation of treatment.
Collapse
Affiliation(s)
- Subhakar Reddy
- Uveitis and Ocular Immunology Services, LV Prasad Eye Institute, Hyderabad, -500034, India.,Smt Kanuri Santhamma Center for Vitreo-Retinal Diseases, L. V. Prasad Eye Institute, Hyderabad, -500034, India
| | - Mudit Tyagi
- Uveitis and Ocular Immunology Services, LV Prasad Eye Institute, Hyderabad, -500034, India. .,Smt Kanuri Santhamma Center for Vitreo-Retinal Diseases, L. V. Prasad Eye Institute, Hyderabad, -500034, India.
| | - Shashwat Behera
- Smt Kanuri Santhamma Center for Vitreo-Retinal Diseases, L. V. Prasad Eye Institute, Hyderabad, -500034, India
| | - Rajeev R Pappuru
- Uveitis and Ocular Immunology Services, LV Prasad Eye Institute, Hyderabad, -500034, India.,Smt Kanuri Santhamma Center for Vitreo-Retinal Diseases, L. V. Prasad Eye Institute, Hyderabad, -500034, India
| | - Vivek P Dave
- Smt Kanuri Santhamma Center for Vitreo-Retinal Diseases, L. V. Prasad Eye Institute, Hyderabad, -500034, India
| | - Soumyava Basu
- Uveitis and Ocular Immunology Services, LV Prasad Eye Institute, Hyderabad, -500034, India
| | - Hitesh Agrawal
- Uveitis and Ocular Immunology Services, LV Prasad Eye Institute, Hyderabad, -500034, India.,Smt Kanuri Santhamma Center for Vitreo-Retinal Diseases, L. V. Prasad Eye Institute, Hyderabad, -500034, India
| |
Collapse
|
20
|
Pediatric onco-nephrology: time to spread the word : Part I: early kidney involvement in children with malignancy. Pediatr Nephrol 2021; 36:2227-2255. [PMID: 33245421 DOI: 10.1007/s00467-020-04800-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 08/28/2020] [Accepted: 09/25/2020] [Indexed: 12/29/2022]
Abstract
Onco-nephrology has been a growing field within the adult nephrology scope of practice. Even though pediatric nephrologists have been increasingly involved in the care of children with different forms of malignancy, there has not been an emphasis on developing special expertise in this area. The fast pace of discovery in this field, including the development of new therapy protocols with their own kidney side effects and the introduction of the CD19-targeted chimeric antigen receptor T cell (CAR-T) therapy, has introduced new challenges for general pediatric nephrologists because of the unique effects of these treatments on the kidney. Moreover, with the improved outcomes in children receiving cancer therapy come an increased number of survivors at risk for chronic kidney disease related to both their cancer diagnosis and therapy. Therefore, it is time for pediatric onco-nephrology to take its spot on the expanding subspecialties map in pediatric nephrology.
Collapse
|
21
|
García-Ríos E, Nuévalos M, Mancebo FJ, Pérez-Romero P. Is It Feasible to Use CMV-Specific T-Cell Adoptive Transfer as Treatment Against Infection in SOT Recipients? Front Immunol 2021; 12:657144. [PMID: 33968058 PMCID: PMC8104120 DOI: 10.3389/fimmu.2021.657144] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 04/06/2021] [Indexed: 12/19/2022] Open
Abstract
During the last decade, many studies have demonstrated the role of CMV specific T-cell immune response on controlling CMV replication and dissemination. In fact, it is well established that transplanted patients lacking CMV-specific T-cell immunity have an increased occurrence of CMV replication episodes and CMV-related complications. In this context, the use of adoptive transfer of CMV-specific T-cells has been widely investigated and applied to Hematopoietic Stem Cell Transplant patients and may be useful as a therapeutic alternative, to reconstitute the CMV specific T-cell response and to control CMV viremia in patients receiving a transplantation. However, only few authors have explored the use of T-cell adoptive transfer in SOT recipients. We propose a novel review in which we provide an overview of the impact of using CMV-specific T-cell adoptive transfer on the control of CMV infection in SOT recipients, the different approaches to stimulate, isolate and expand CMV-specific T-cells developed over the years and a discussion of the possible use of CMV adoptive cellular therapy in this SOT population. Given the timeliness and importance of this topic, we believe that such an analysis will provide important insights into CMV infection and its treatment/prevention.
Collapse
Affiliation(s)
- Estéfani García-Ríos
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Marcos Nuévalos
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Francisco J Mancebo
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Pilar Pérez-Romero
- National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
| |
Collapse
|
22
|
Reddy S, Tyagi M, Behera S, Pappuru RR. Cytomegalovirus retinitis in a patient of granulomatosis with polyangiitis on long-term immunosuppressants. BMJ Case Rep 2021; 14:e236632. [PMID: 33542003 PMCID: PMC7868283 DOI: 10.1136/bcr-2020-236632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
A 69-year-old male patient presented to the retina clinic with a sudden decrease in vision in his right eye since 1 day. He was a known case of granulomatosis with polyangiitis and was on systemic immunosuppression for the past 3 years. The best-corrected visual acuity (BCVA) in his right eye was 20/60 and he has no perception of light in the left eye. Fundus examination revealed the presence of retinitis lesions in the right eye and total optic atrophy in the left eye. A vitreous biopsy was done and the PCR was found to be positive for cytomegalovirus (CMV). He was treated with intravitreal ganciclovir injections. Subsequently, the retinitis lesions regressed and BCVA in the right eye improved to 20/40.This case report elaborates on the risks of the development of opportunistic ocular infections in patients receiving long-term systemic immunosuppressants and the need for regular ocular examinations in such cases.
Collapse
Affiliation(s)
- Subhakar Reddy
- Smt Kanuri Santhamma Centre for Vitreoretinal Diseases, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Mudit Tyagi
- Smt Kanuri Santhamma Centre for Vitreoretinal Diseases, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Shashwat Behera
- Smt Kanuri Santhamma Centre for Vitreoretinal Diseases, LV Prasad Eye Institute, Hyderabad, Telangana, India
| | - Rajeev Reddy Pappuru
- Smt Kanuri Santhamma Centre for Vitreoretinal Diseases, LV Prasad Eye Institute, Hyderabad, Telangana, India
| |
Collapse
|
23
|
Prohn M, Viberg A, Zhang D, Dykstra K, Davis C, Macha S, Sabato P, de Alwis D, Iwamoto M, Fancourt C, Cho CR. Population pharmacokinetics of letermovir following oral and intravenous administration in healthy participants and allogeneic hematopoietic cell transplantation recipients. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2021; 10:255-267. [PMID: 33440077 PMCID: PMC7965833 DOI: 10.1002/psp4.12593] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 01/25/2023]
Abstract
Letermovir is indicated for prophylaxis of cytomegalovirus infection and disease in allogeneic hematopoietic stem cell transplant (HSCT) recipients. Two‐stage population pharmacokinetic (PK) modeling of letermovir was conducted to support dose rationale and evaluate the impact of intrinsic/extrinsic factors. Data from healthy phase I study participants over a wide dose range were modeled to evaluate the effects of selected intrinsic factors, including pharmacogenomics; next, phase III HSCT‐recipient data at steady‐state following clinical doses were modeled. The model in HSCT recipients adequately described letermovir PK following both oral or i.v. administration, and was consistent with the healthy participant model at steady‐state clinical doses. Intrinsic factor effects were not clinically meaningful. These staged analyses indicate that letermovir PK in HSCT recipients and healthy participants differ only with respect to bioavailability and absorption rate. The HSCT recipient model was suitable for predicting exposure for exposure–response analysis supporting final dose selection.
Collapse
Affiliation(s)
| | | | - Da Zhang
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | - Casey Davis
- Merck & Co., Inc., Kenilworth, New Jersey, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Carlson TJ, Gonzales-Luna AJ, Garey KW. Recent developments in antimicrobial therapy for gastrointestinal infections. Curr Opin Gastroenterol 2021; 37:30-36. [PMID: 33229860 DOI: 10.1097/mog.0000000000000696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
PURPOSE OF REVIEW This focused, narrative review summarizes human clinical trial data for direct-acting antimicrobials in development for the treatment of gastrointestinal infections that were published in the past 18 months (1 January 2019 to 30 June 2020). RECENT FINDINGS Antimicrobial agents for Clostridioides difficile infection (n = 6), cryptosporidiosis (n = 1), cytomegalovirus infection (n = 3) and Helicobacter pylori infection (n = 1) have completed and/or are undergoing human clinical trials. SUMMARY Although this review highlights significant advances in four disease states, many common gastrointestinal pathogens have no antimicrobials in human clinical trials, emphasizing the need for continued prioritization in this field of study.
Collapse
Affiliation(s)
- Travis J Carlson
- Department of Clinical Sciences, High Point University Fred Wilson School of Pharmacy, High Point, North Carolina
| | - Anne J Gonzales-Luna
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Kevin W Garey
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| |
Collapse
|
25
|
"Mini" bank of only 8 donors supplies CMV-directed T cells to diverse recipients. Blood Adv 2020; 3:2571-2580. [PMID: 31481503 DOI: 10.1182/bloodadvances.2019000371] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/19/2019] [Indexed: 12/20/2022] Open
Abstract
Cytomegalovirus (CMV) infections remain a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT), and standard antiviral therapies are associated with significant side effects and development of drug-resistant mutants. Adoptively transferred donor-derived CMV-specific T cells (CMVSTs) can provide an alternative treatment modality with few side effects but are not widely available due to their patient-specific nature. Here we report the establishment and use of a bank of CMVSTs derived from just 8 CMV-seropositive donors, with HLA types representing the diverse US population, as an "off-the-shelf" therapy to treat drug-refractory infections. To date, we have screened 29 patients for study participation and identified a suitable line, with ≥2 of 8 shared HLA antigens, for 28 (96.6%) patients with a median of 4 shared HLA antigens. Of these, 10 patients with persistent/refractory CMV infections or disease were eligible for treatment; a single infusion of cells produced 3 partial responses and 7 complete responses, for a cumulative response rate of 100% (95% confidence interval, 69.2-100) with no graft-versus-host disease, graft failure, or cytokine release syndrome. Potential wider use of the tested CMVSTs across transplant centers is made more feasible by our ability to produce sufficient material to generate cells for >2000 infusions from a single donor collection. Our data indicate that a "mini" bank of CMVSTs prepared from just 8 well-chosen third-party donors can supply the majority of patients with an appropriately matched line that produces safe and effective anti-CMV activity post-HSCT.
Collapse
|
26
|
Papanicolaou GA, Silveira FP, Langston AA, Pereira MR, Avery RK, Uknis M, Wijatyk A, Wu J, Boeckh M, Marty FM, Villano S. Maribavir for Refractory or Resistant Cytomegalovirus Infections in Hematopoietic-cell or Solid-organ Transplant Recipients: A Randomized, Dose-ranging, Double-blind, Phase 2 Study. Clin Infect Dis 2020; 68:1255-1264. [PMID: 30329038 PMCID: PMC6451997 DOI: 10.1093/cid/ciy706] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 10/03/2018] [Indexed: 12/27/2022] Open
Abstract
Background Cytomegalovirus (CMV) infections that are refractory or resistant (RR) to available antivirals ([val]ganciclovir, foscarnet, cidofovir) are associated with higher mortality in transplant patients. Maribavir is active against RR CMV strains. Methods Hematopoietic-cell or solid-organ transplant recipients ≥12 years old with RR CMV infections and plasma CMV deoxyribonucleic acid (DNA) ≥1000 copies/mL were randomized (1:1:1) to twice-daily dose-blinded maribavir 400, 800, or 1200 mg for up to 24 weeks. The primary efficacy endpoint was the proportion of patients with confirmed undetectable plasma CMV DNA within 6 weeks of treatment. Safety analyses included the frequency and severity of treatment-emergent adverse events (TEAEs). Results From July 2012 to December 2014, 120 patients were randomized and treated (40 per dose group): 80/120 (67%) patients achieved undetectable CMV DNA within 6 weeks of treatment (95% confidence interval, 57–75%), with rates of 70%, 63%, and 68%, respectively, for maribavir 400, 800, and 1200 mg twice daily. Recurrent on-treatment CMV infections occurred in 25 patients; 13 developed mutations conferring maribavir resistance. Maribavir was discontinued due to adverse events in 41/120 (34%) patients, and 17/41 discontinued due to CMV infections. During the study, 32 (27%) patients died, 4 due to CMV disease. Dysgeusia was the most common TEAE (78/120; 65%) and led to maribavir discontinuation in 1 patient. Absolute neutrophil counts <1000/µL were noted in 12/106 (11%) evaluable patients, with rates similar across doses. Conclusions Maribavir ≥400 mg twice daily was active against RR CMV infections in transplant recipients; no new safety signals were identified. Clinical Trials Registration NCT01611974.
Collapse
Affiliation(s)
| | - Fernanda P Silveira
- The Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | | | - Marcus R Pereira
- Department of Medicine, Columbia University Medical Center, New York, New York
| | | | - Marc Uknis
- Shire Pharmaceuticals, Wayne, Pennsylvania
| | | | - Jingyang Wu
- Shire Pharmaceuticals, Lexington, Massachusetts
| | - Michael Boeckh
- The Fred Hutchinson Cancer Research Center, Seattle, Washington
| | | | | |
Collapse
|
27
|
Ilic K, Song I, Wu J, Martin P. Evaluation of the Effect of Maribavir on Cardiac Repolarization in Healthy Participants: Thorough QT/QTc Study. Clin Transl Sci 2020; 13:1260-1270. [PMID: 32506738 PMCID: PMC7719377 DOI: 10.1111/cts.12814] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/29/2020] [Indexed: 12/27/2022] Open
Abstract
Maribavir is an orally bioavailable benzimidazole riboside in clinical development for treatment of cytomegalovirus infection in patients who undergo transplantation. Maribavir was evaluated in a thorough QT (TQT) study to determine any effects on cardiac repolarization. The effect of maribavir 100 and 1,200 mg oral doses on the baseline-adjusted and placebo-adjusted corrected QT (QTc) interval (delta delta QTc (ddQTc)) and other electrocardiogram (ECG) parameters was assessed in a randomized, phase I, placebo-controlled, four-period crossover study in healthy participants (men and women ages 18-50 years). Additionally, maribavir pharmacokinetics, safety, and tolerability were investigated. Moxifloxacin (400 mg) was used as a positive control to demonstrate the study's ability to detect QT prolongation. Digital 12-lead Holter ECG monitoring was performed over 22 hours following study drug administration. Individual, Fridericia's, and Bazett's QTc intervals were calculated. Of 52 randomized participants (29 ± 8.1 years old; 31 men (60%)), 50 (96%) completed the study. For both 100-mg and 1200-mg doses of maribavir, analysis of ddQTc demonstrated that the upper bound of the two-sided 90% confidence interval was below the 10-ms threshold at all time points. The concentration-effect analysis demonstrated no relationship between ddQTc and plasma concentrations of maribavir (and its metabolite). There were no clinically meaningful changes in heart rate and systolic blood pressure. The most common adverse event was dysgeusia; no serious adverse events were reported. This TQT study demonstrated that maribavir did not have impact on cardiac repolarization.
Collapse
Affiliation(s)
- Katarina Ilic
- Shire, a Takeda Company, Lexington, Massachusetts, USA
| | - Ivy Song
- Shire, a Takeda Company, Lexington, Massachusetts, USA
| | - Jingyang Wu
- Shire, a Takeda Company, Lexington, Massachusetts, USA
| | | |
Collapse
|
28
|
Zavras P, Su Y, Fang J, Stern A, Gupta N, Tang Y, Raval A, Giralt S, Perales MA, Jakubowski AA, Papanicolaou GA. Impact of Preemptive Therapy for Cytomegalovirus on Toxicities after Allogeneic Hematopoietic Cell Transplantation in Clinical Practice: A Retrospective Single-Center Cohort Study. Biol Blood Marrow Transplant 2020; 26:1482-1491. [PMID: 32315708 DOI: 10.1016/j.bbmt.2020.03.019] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022]
Abstract
(Val)ganciclovir (vGCV) or foscarnet (FCN) as preemptive therapy (PET) for cytomegalovirus (CMV) after allogeneic hematopoietic cell transplantation (HCT) is associated with myelosuppression and nephrotoxicity, respectively. We analyzed a cohort of CMV-seropositive (R+) HCT recipients managed preemptively at a single center. The objectives of our study were to (1) quantify the frequencies of neutropenia and acute kidney injury (AKI) through day +100 (D100) post-HCT and at PET discontinuation and (2) assess the impact of PET on neutropenia and AKI in multivariate models. This was a retrospective cohort study of adult CMV R+ recipients who underwent allo-HCT at Memorial Sloan Kettering Cancer Center from March 18, 2013, through December 31, 2017, and were managed with PET. Patients were grouped by receipt of PET (PET and no PET). Neutropenia and AKI were defined by Common Terminology Criteria for Adverse Events version 4. Frequencies of toxicities by D100 were compared between relevant groups. The impact of PET on toxicities was examined in univariate and multivariate Poisson/negative binomial regression models. Of 368 CMV R+ HCT recipients, 208 (56.5%) received PET. Neutropenia by D100 occurred in 41.8% and 28.6% patients in PET and no PET, respectively (P = .0009). PET increased the risk of neutropenia (adjusted relative risk = 1.81; 95% confidence interval [CI], 1.48 to 2.21; P < .0001) in multivariate analyses. AKI by D100 occurred in 12.0% and 7.8% patients in PET and no PET, respectively (P = .19). PET increased the risk of AKI by 2.75-fold (95% CI, 1.71 to 4.42; P < .0001). When PET recipients were grouped by first antiviral, neutropenia by D100 occurred in 34.8% and 48.9% of vGCV and FCN recipients, respectively, (P = .08), and AKI occurred in 13.0% and 34.0% of vGCV and FCN recipients, respectively (P = .001). At discontinuation of vGCV or FCN, neutropenia was present in 11.2% versus 2.1% patients, respectively (P = .08), and AKI was present in 1.9% of versus 12.8% patients respectively (P = .005). Preemptive therapy for CMV increased the risk of neutropenia and AKI in the first 100 days post-HCT by 1.8-fold and 2.8-fold, respectively. Our results underscore the need for safer antivirals for CMV management in HCT recipients.
Collapse
Affiliation(s)
- Phaedon Zavras
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yiqi Su
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jiaqi Fang
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Anat Stern
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nitasha Gupta
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yuexin Tang
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Kenilworth, New Jersey
| | - Amit Raval
- Center for Observational and Real-World Evidence, Merck & Co., Inc., Kenilworth, New Jersey
| | - Sergio Giralt
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Miguel Angel Perales
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York
| | - Ann A Jakubowski
- Adult Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Genovefa A Papanicolaou
- Infectious Disease Service, Memorial Sloan Kettering Cancer Center, New York, New York; Weill Cornell Medical College, New York, New York.
| |
Collapse
|
29
|
Tsujimura M, Kusamori K, Nishikawa M. Rapid Regulation of Human Mesenchymal Stem Cell Proliferation Using Inducible Caspase-9 Suicide Gene for Safe Cell-Based Therapy. Int J Mol Sci 2019; 20:ijms20225759. [PMID: 31744061 PMCID: PMC6887989 DOI: 10.3390/ijms20225759] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/01/2019] [Accepted: 11/14/2019] [Indexed: 12/18/2022] Open
Abstract
The regulation of transplanted cell proliferation and function is important to achieve safe cell-based therapies. We previously reported that the proliferation and function of transplanted cells, which expressed the herpes simplex virus thymidine kinase (HSVtk) suicide gene, could be controlled by ganciclovir (GCV) administration. However, there are some concerns regarding the use of GCV. It is reported that the inducible caspase-9 (iC9) gene, a human caspase-9-derived genetically engineered suicide gene, rapidly induces cell apoptosis in the presence of apoptosis inducers, such as AP20187. In this study, we used a combination of the iC9 gene and AP20187 to achieve rapid regulation of transplanted cell proliferation. Cells from the human mesenchymal stem cell line UE7T-13 were transfected with the iC9 gene to obtain UE7T-13/iC9 cells. AP20187 significantly reduced the number of UE7T-13/iC9 cells within 24 h in a concentration-dependent manner. This reduction was much faster than the reduction of HSVtk-expressing UE7T-13 cells induced by GCV addition. Subcutaneous AP20187 administration rapidly reduced the luminescence signal from NanoLuc luciferase (Nluc)-expressing UE7T-13/iC9 cells transplanted into mice. These results indicate that the combined use of the iC9 gene and AP20187 is effective in rapidly regulating transplanted cell proliferation.
Collapse
Affiliation(s)
| | - Kosuke Kusamori
- Correspondence: ; Tel.: +81-4-7124-1501; Fax: +81-4-7121-4450
| | | |
Collapse
|
30
|
Generation and infusion of multi-antigen-specific T cells to prevent complications early after T-cell depleted allogeneic stem cell transplantation—a phase I/II study. Leukemia 2019; 34:831-844. [DOI: 10.1038/s41375-019-0600-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/21/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022]
|
31
|
Maertens J, Cordonnier C, Jaksch P, Poiré X, Uknis M, Wu J, Wijatyk A, Saliba F, Witzke O, Villano S. Maribavir for Preemptive Treatment of Cytomegalovirus Reactivation. N Engl J Med 2019; 381:1136-1147. [PMID: 31532960 DOI: 10.1056/nejmoa1714656] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND Maribavir is a benzimidazole riboside with activity against cytomegalovirus (CMV). The safety and efficacy of maribavir for preemptive treatment of CMV infection in transplant recipients is not known. METHODS In a phase 2, open-label, maribavir dose-blinded trial, recipients of hematopoietic-cell or solid-organ transplants (≥18 years of age, with CMV reactivation [1000 to 100,000 DNA copies per milliliter]) were randomly assigned to receive maribavir at a dose of 400, 800, or 1200 mg twice daily or the standard dose of valganciclovir for no more than 12 weeks. The primary efficacy end point was the percentage of patients with a response to treatment, defined as confirmed undetectable CMV DNA in plasma, within 3 weeks and 6 weeks after the start of treatment. The primary safety end point was the incidence of adverse events that occurred or worsened during treatment. RESULTS Of the 161 patients who underwent randomization, 159 received treatment, and 156 had postbaseline data available - 117 in the maribavir group and 39 in the valganciclovir group. The percentage of patients with postbaseline data available who had a response to treatment within 3 weeks was 62% among those who received maribavir and 56% among those who received valganciclovir. Within 6 weeks, 79% and 67% of patients, respectively, had a response (risk ratio, 1.20; 95% confidence interval, 0.95 to 1.51). The percentages of patients with a response to treatment were similar among the maribavir dose groups. Two patients who had a response to treatment had a recurrence of CMV infection within 6 weeks after starting maribavir at a dose of 800 mg twice daily; T409M resistance mutations in CMV UL97 protein kinase developed in both patients. The incidence of serious adverse events that occurred or worsened during treatment was higher in the maribavir group than in the valganciclovir group (52 of 119 patients [44%] vs. 13 of 40 [32%]). A greater percentage of patients in the maribavir group discontinued the trial medication because of an adverse event (27 of 119 [23%] vs. 5 of 40 [12%]). A higher incidence of gastrointestinal adverse events was reported with maribavir, and a higher incidence of neutropenia was reported with valganciclovir. CONCLUSIONS Maribavir at a dose of at least 400 mg twice daily had efficacy similar to that of valganciclovir for clearing CMV viremia among recipients of hematopoietic-cell or solid-organ transplants. A higher incidence of gastrointestinal adverse events - notably dysgeusia - and a lower incidence of neutropenia were found in the maribavir group. (Funded by ViroPharma/Shire Development; EudraCT number, 2010-024247-32.).
Collapse
Affiliation(s)
- Johan Maertens
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Catherine Cordonnier
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Peter Jaksch
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Xavier Poiré
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Marc Uknis
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Jingyang Wu
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Anna Wijatyk
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Faouzi Saliba
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Oliver Witzke
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| | - Stephen Villano
- From the Hematology Department, University Hospitals Leuven, KU Leuven, Leuven (J.M.), and the Section of Hematology, Cliniques Universitaires Saint-Luc, Brussels (X.P.) - both in Belgium; the Hematology Department, Henri Mondor Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) and University Paris-Est-Créteil, Créteil (C.C.), and AP-HP Hôpital Paul Brousse, Villejuif (F.S.) - all in France; the Medical University of Vienna, General Hospital, Vienna (P.J.); Shire, Wayne, PA (M.U., S.V.); Shire, Lexington, MA (J.W., A.W.); and the Department of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany (O.W.)
| |
Collapse
|
32
|
Andronova VL. [Modern ethiotropic chemotherapy of human cytomegalovirus infection: clinical effectiveness, molecular mechanism of action, drug resistance, new trends and prospects. Part 1.]. Vopr Virusol 2019; 63:202-211. [PMID: 30550096 DOI: 10.18821/0507-4088-2018-63-5-202-211] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/12/2017] [Indexed: 12/25/2022]
Abstract
Modern chemotherapy of cytomegalovirus (CMV) infections has a very limited arsenal of first-line drugs. These are preparations of ganciclovir (GCV) belonging to the class of modified nucleosides and its metabolic precursor ganciclovir valine ester. After three-step phosphorylation, GCV, as a structural analogue of the natural nucleotide, competes with it for binding to DNA polymerase and, due to its structural features, inhibits its activity. However, with prolonged use of GCV, mainly under conditions of immunosuppression, the virus develops drug resistance associated in most cases with changes in pUL97 catalyzing the first stage of GCV phosphorylation, as well as in the catalytic subunit of DNA polymerase. When variants of viruses resistant to GCV appear, second-line drugs are used: pyrophosphate analog of foscarnet and nucleotide cidofovir. Resistance to second-line drugs is due to mutations in the pol-gene and in a number of cases leads to multiresistance, which makes it impossible to use traditional anti-CMV drugs. In addition, the use of all of the above drugs is accompanied by the development of severe side effects. All of the above determines the need to search for new compounds that can effectively inhibit the reproduction of the virus, harmless to the macroorganism, convenient to use, overcoming the drug resistance barrier in viruses.As a result of the search in international databases (PubMed, MedLine, eLIBRARY.RU, ClinicalTrials.gov, etc.), the main trends in the search for new anti-CMV agents were identified. In the first part of the review, we concentrated on compounds that are modifications of known antiviral agents currently used in clinical practice, the most promising for the development of drug anti-CMV drugs.
Collapse
Affiliation(s)
- V L Andronova
- National Research Center for Epidemiology and Microbiology named after the honorary academician N.F. Gamaleya, Moscow, 123098, Russian Federation
| |
Collapse
|
33
|
Comment on: "Cytomegalovirus Corneal Endotheliitis After Descemet Membrane Endothelial Keratoplasty". Cornea 2019; 38:e48. [PMID: 31498245 DOI: 10.1097/ico.0000000000002079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
34
|
El Helou G, Razonable RR. Safety considerations with current and emerging antiviral therapies for cytomegalovirus infection in transplantation. Expert Opin Drug Saf 2019; 18:1017-1030. [PMID: 31478398 DOI: 10.1080/14740338.2019.1662787] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Introduction: Human cytomegalovirus (HCMV) is a major contributor of morbidity and mortality, and its management is essential for the successful outcome of solid organ and hematopoietic stem cell transplantation. Areas covered: This review discusses the safety profiles of currently available and emerging antiviral drugs and the other strategies for HCMV prevention and treatment after transplantation. Expert opinion: Strategies for management of HCMV rely largely on the use of antiviral agents that inhibit viral DNA polymerase (ganciclovir/valganciclovir, foscarnet, and cidofovir/brincidofovir) and viral terminase complex (letermovir), with different types and degrees of adverse effects. An investigational agent, maribavir, exerts its anti-CMV effect through UL97 inhibition, and its safety profile is under clinical evaluation. In choosing the antiviral medication to use, it is important to consider these safety profiles in addition to overall efficacy. In addition to antiviral drugs, reduction of immunosuppression is often generally needed in the management of HCMV infection, but with a potential risk of allograft rejection or graft-versus-host disease. The use of HCMV-specific or non-specific intravenous immunoglobulins remains debated, while adoptive HCMV-specific T cell therapy remains investigational, and associated with unique set of adverse effects.
Collapse
Affiliation(s)
- Guy El Helou
- Division of Infectious Diseases, Department of Medicine, and William J von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| | - Raymund R Razonable
- Division of Infectious Diseases, Department of Medicine, and William J von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| |
Collapse
|
35
|
Carter J, Alston CI, Oh J, Duncan LA, Esquibel Nemeno JG, Byfield SN, Dix RD. Mechanisms of AIDS-related cytomegalovirus retinitis. Future Virol 2019. [DOI: 10.2217/fvl-2019-0033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Human cytomegalovirus (HCMV) generates a significant clinical burden worldwide, particularly among the immune compromised. In approximately 30% of untreated HIV/AIDS patients without access or sufficient response to antiretroviral therapies, for example, HCMV causes a sight-threatening retinitis. To study the mechanisms of AIDS-related HCMV retinitis, our lab has for many years used a mouse model in which a mixture of mouse retroviruses induces murine AIDS after approximately 10 weeks, rendering otherwise resistant mice susceptible to opportunistic pathogens. This immunodeficiency combined with subretinal inoculation of murine cytomegalovirus yields a reproducible model of the human disease, facilitating the discovery of many clinically relevant virologic and immunologic mechanisms of retinal destruction which we summarize in this review.
Collapse
Affiliation(s)
- Jessica Carter
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, GA 30303, USA
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christine I Alston
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, GA 30303, USA
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jay Oh
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Lauren-Ashley Duncan
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | | | - Shauntelle N Byfield
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Richard D Dix
- Viral Immunology Center, Department of Biology, Georgia State University, Atlanta, GA 30303, USA
- Department of Ophthalmology, Emory Eye Center, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
36
|
Nehme Z, Pasquereau S, Herbein G. Control of viral infections by epigenetic-targeted therapy. Clin Epigenetics 2019; 11:55. [PMID: 30917875 PMCID: PMC6437953 DOI: 10.1186/s13148-019-0654-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/13/2019] [Indexed: 12/13/2022] Open
Abstract
Epigenetics is defined as the science that studies the modifications of gene expression that are not owed to mutations or changes in the genetic sequence. Recently, strong evidences are pinpointing toward a solid interplay between such epigenetic alterations and the outcome of human cytomegalovirus (HCMV) infection. Guided by the previous possibly promising experimental trials of human immunodeficiency virus (HIV) epigenetic reprogramming, the latter is paving the road toward two major approaches to control viral gene expression or latency. Reactivating HCMV from the latent phase ("shock and kill" paradigm) or alternatively repressing the virus lytic and reactivation phases ("block and lock" paradigm) by epigenetic-targeted therapy represent encouraging options to overcome latency and viral shedding or otherwise replication and infectivity, which could lead eventually to control the infection and its complications. Not limited to HIV and HCMV, this concept is similarly studied in the context of hepatitis B and C virus, herpes simplex virus, and Epstein-Barr virus. Therefore, epigenetic manipulations stand as a pioneering research area in modern biology and could constitute a curative methodology by potentially consenting the development of broad-spectrum antivirals to control viral infections in vivo.
Collapse
Affiliation(s)
- Zeina Nehme
- Department Pathogens & Inflammation-EPILAB, UPRES EA4266, University of Franche-Comté, University of Bourgogne Franche-Comté, 16 route de Gray, F-25030 Besançon cedex, France
- Université Libanaise, Beirut, Lebanon
| | - Sébastien Pasquereau
- Department Pathogens & Inflammation-EPILAB, UPRES EA4266, University of Franche-Comté, University of Bourgogne Franche-Comté, 16 route de Gray, F-25030 Besançon cedex, France
| | - Georges Herbein
- Department Pathogens & Inflammation-EPILAB, UPRES EA4266, University of Franche-Comté, University of Bourgogne Franche-Comté, 16 route de Gray, F-25030 Besançon cedex, France
- Department of Virology, CHRU Besancon, F-25030 Besançon, France
| |
Collapse
|
37
|
Nukui M, O'Connor CM, Murphy EA. The Natural Flavonoid Compound Deguelin Inhibits HCMV Lytic Replication within Fibroblasts. Viruses 2018; 10:v10110614. [PMID: 30405048 PMCID: PMC6265796 DOI: 10.3390/v10110614] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/02/2018] [Accepted: 11/03/2018] [Indexed: 12/12/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a ubiquitous herpesvirus for which there is no vaccine or cure. This viral infection, once acquired, is life-long, residing latently in hematopoietic cells. However, latently infected individuals with weakened immune systems often undergo HCMV reactivation, which can cause serious complications in immunosuppressed and immunocompromised patients. Current anti-viral therapies target late stages of viral replication, and are often met with therapeutic resistance, necessitating the development of novel therapeutics. In this current study, we identified a naturally-occurring flavonoid compound, deguelin, which inhibits HCMV lytic replication. Our findings reveal that nanomolar concentrations of deguelin significantly suppress the production of the infectious virus. Further, we show that deguelin inhibits the lytic cycle during the phase of the replication cycle consistent with early (E) gene and protein expression. Importantly, our data reveal that deguelin inhibits replication of a ganciclovir-resistant strain of HCMV. Together, our findings identify a novel, naturally occurring compound that may prove useful in the treatment of HCMV replication.
Collapse
Affiliation(s)
- Masatoshi Nukui
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Christine M O'Connor
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44106, USA.
| | - Eain A Murphy
- FORGE Life Science, Pennsylvania Biotechnology Center, Doylestown, PA 18901, USA.
| |
Collapse
|
38
|
Kim SJ, Huang YT, Foldi J, Lee YJ, Maloy M, Giralt SA, Jakubowski AA, Papanicolaou GA. Cytomegalovirus resistance in CD34 + -selected hematopoietic cell transplant recipients. Transpl Infect Dis 2018; 20:e12881. [PMID: 29570237 PMCID: PMC5988949 DOI: 10.1111/tid.12881] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Revised: 02/02/2018] [Accepted: 02/18/2018] [Indexed: 12/26/2022]
Abstract
BACKGROUND Cytomegalovirus (CMV) viremia after CD34+ -selected hematopoietic stem cell transplant (HCT) often requires prolonged antiviral therapy. We report rates and outcomes of resistant CMV in a contemporary cohort of CD34+ -selected HCT recipients managed preemptively. METHODS We retrospectively reviewed 220 consecutive, CMV-seropositive recipients (R+), who received CD34+ -selected HCT at Memorial Sloan Kettering Cancer Center between June 2010 and December 2014. Patients were monitored by quantitative CMV PCR and were treated preemptively. CMV resistance was tested by a genotypic assay. RESULTS One hundred and sixty-one (73%) patients developed CMV viremia and 47 (29% of viremic and 21% of total patients) had CMV resistance testing by one-year from HCT. CMV resistance was confirmed in 19 (12% of viremic and 9% of total) patients and was identified >3 months from HCT in 90% of patients. Twelve patients had mutations in UL97 only; the remaining 7 patients had mutations in UL54 only or UL54 and UL97. By 1 year from HCT, 11 of 19 (58%) patients with mutations had CMV end-organ disease. CMV-related mortality in patients with resistance was 42%. CONCLUSIONS Nine percent of CMV R+, CD34+ -selected HCT recipients had resistant CMV by 1 year from HCT. Of 19 patients with resistant CMV, 58% had CMV end-organ disease and 42% died of CMV. Effective strategies for CMV prevention and restoration of CMV immunity are needed for CD34+ -selected HCT.
Collapse
Affiliation(s)
- Seong Jin Kim
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yao-Ting Huang
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julia Foldi
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yeon Joo Lee
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College, Cornell University, New York, NY, USA
| | - Molly Maloy
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio A Giralt
- Weill Medical College, Cornell University, New York, NY, USA
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ann A Jakubowski
- Weill Medical College, Cornell University, New York, NY, USA
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Genovefa A Papanicolaou
- Infectious Disease Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Medical College, Cornell University, New York, NY, USA
| |
Collapse
|
39
|
Marshall WL, McCrea JB, Macha S, Menzel K, Liu F, van Schanke A, de Haes JIU, Hussaini A, Jordan HR, Drexel M, Kantesaria BS, Tsai C, Cho CR, Hulskotte EGJ, Butterton JR, Iwamoto M. Pharmacokinetics and Tolerability of Letermovir Coadministered With Azole Antifungals (Posaconazole or Voriconazole) in Healthy Subjects. J Clin Pharmacol 2018; 58:897-904. [DOI: 10.1002/jcph.1094] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Accepted: 01/15/2018] [Indexed: 11/10/2022]
Affiliation(s)
| | | | | | | | - Fang Liu
- Merck & Co, Inc; Kenilworth NJ USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Schlott F, Steubl D, Ameres S, Moosmann A, Dreher S, Heemann U, Hösel V, Busch DH, Neuenhahn M. Characterization and clinical enrichment of HLA-C*07:02-restricted Cytomegalovirus-specific CD8+ T cells. PLoS One 2018; 13:e0193554. [PMID: 29489900 PMCID: PMC5831000 DOI: 10.1371/journal.pone.0193554] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/13/2018] [Indexed: 12/31/2022] Open
Abstract
Human Cytomegalovirus (CMV) reactivation remains a major source of morbidity in patients after solid organ and hematopoietic stem cell transplantation (HSCT). Adoptive T cell therapy (ACT) with CMV-specific T cells is a promising therapeutic approach for HSCT recipients, but might be counteracted by CMV’s immune evasion strategies. HLA-C*07:02 is less susceptible to viral immune evasion suggesting HLA-C*07:02-restricted viral epitopes as promising targets for ACT. For a better understanding of HLA-C*07:02-restricted CMV-specific T cells we used recently generated reversible HLA-C*07:02/IE-1 multimers (Streptamers) recognizing a CMV-derived Immediate-Early-1 (IE-1) epitope and analyzed phenotypic and functional T cell characteristics. Initially, we detected very high frequencies of HLA-C*07:02/IE-1 multimer+ T cells (median = 11.35%), as well as robust functional responses after stimulation with IE-1 peptide (IFNγ+; median = 5.02%) in healthy individuals. However, MHC-multimer+ and IFNγ-secreting T cell frequencies showed a relatively weak correlation (r2 = 0.77), which could be attributed to an unexpected contribution of CMV-epitope-independent KIR2DL2/3-binding of HLA-C*07:02/IE-1 multimers. Therefore, we developed a MHC-multimer double-staining approach against a cancer epitope-specific HLA-C*07:02 multimer to identify truly HLA-C*07:02/IE-1 epitope-specific T cells. The frequencies of these truly HLA-C*07:02/IE-1 multimer+ T cells were still high (median = 6.86%) and correlated now strongly (r2 = 0.96) with IFNγ-secretion. Interestingly, HLA-C*07:02/IE-1-restricted T cells contain substantial numbers with a central memory T cell phenotype, indicating high expansion potential e.g. for ACT. In line with that, we developed a clinical enrichment protocol avoiding epitope-independent KIR-binding to make HLA-C*07:02/IE-1-restricted T cells available for ACT. Initial depletion of KIR-expressing CD8+ T cells followed by HLA-C*07:02/IE-1 Streptamer positive selection using paramagnetic labeling techniques allowed to enrich successfully HLA-C*07:02/IE-1-restricted T cells. Such specifically enriched populations of functional HLA-C*07:02/IE-1-restricted T cells with significant central memory T cell content could become a potent source for ACT.
Collapse
Affiliation(s)
- Fabian Schlott
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
- DZIF—National Centre for Infection Research, Munich, Germany
| | - Dominik Steubl
- Department of Nephrology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Stefanie Ameres
- DZIF Research Group "Host Control of Viral Latency and Reactivation" (HOCOVLAR), Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
| | - Andreas Moosmann
- DZIF—National Centre for Infection Research, Munich, Germany
- DZIF Research Group "Host Control of Viral Latency and Reactivation" (HOCOVLAR), Research Unit Gene Vectors, Helmholtz Zentrum München, Munich, Germany
| | - Stefan Dreher
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - Uwe Heemann
- DZIF—National Centre for Infection Research, Munich, Germany
- Department of Nephrology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Volker Hösel
- Technical University Munich, Chair of Biomathematics, Garching, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
- DZIF—National Centre for Infection Research, Munich, Germany
| | - Michael Neuenhahn
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
- DZIF—National Centre for Infection Research, Munich, Germany
- * E-mail:
| |
Collapse
|
41
|
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: 70] [Impact Index Per Article: 8.8] [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
|
42
|
Kropeit D, McCormick D, Erb-Zohar K, Moiseev VS, Kobalava ZD, Stobernack HP, Zimmermann H, Rübsamen-Schaeff H. Pharmacokinetics and safety of the anti-human cytomegalovirus drug letermovir in subjects with hepatic impairment. Br J Clin Pharmacol 2017; 83:2678-2686. [PMID: 28722153 DOI: 10.1111/bcp.13376] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 05/25/2017] [Accepted: 06/11/2017] [Indexed: 11/28/2022] Open
Abstract
AIMS Human cytomegalovirus constitutes a prevalent and serious threat to immunocompromised individuals and requires new treatments. Letermovir is a novel viral-terminase inhibitor that has demonstrated prophylactic/pre-emptive activity against human cytomegalovirus in Phase 2 and 3 transplant trials. As unchanged letermovir is primarily excreted via the liver by bile, this trial aimed to assess the effect of hepatic impairment on letermovir pharmacokinetics. METHODS Phase 1, open-label, parallel-group pharmacokinetic and safety comparison of multiple once-daily oral letermovir in female subjects with hepatic impairment and healthy matched controls. For 8 days, subjects with moderate hepatic impairment (n = 8) and their matched healthy controls (n = 9) received 60 mg letermovir/day and those with severe hepatic impairment (n = 8) and their matched healthy controls (n = 8) received 30 mg letermovir/day. Pharmacokinetic parameters were determined from blood samples. RESULTS For subjects with moderate hepatic impairment, maximal observed concentration at steady state (Css,max ) and the area under the concentration vs. time curve over a dosing interval at steady state (AUCτ,ss ) for total letermovir were 1.37-fold (90% confidence interval: 0.87, 2.17) and 1.59-fold (0.98, 2.57) higher, respectively, than in healthy subjects. For subjects with severe hepatic impairment, Css,max and AUCτ,ss values of total letermovir were 2.34-fold (1.91, 2.88) and 3.82-fold (2.94, 4.97) higher, respectively, compared with healthy subjects. CONCLUSIONS Moderate hepatic impairment increased exposure to letermovir <2-fold, while severe hepatic impairment increased letermovir exposure approximately 4-fold as compared with healthy subjects. Letermovir 60/30 mg/day was generally well-tolerated in subjects with hepatic impairment.
Collapse
Affiliation(s)
- Dirk Kropeit
- AiCuris Anti-infective Cures GmbH, Wuppertal, Germany
| | | | | | - Valentin S Moiseev
- Centre for Applied Clinical Pharmacology, Clinical Hospital #3, GOUVPO Russian Peoples' Friendship University, Moscow, Russia
| | - Zhanna D Kobalava
- Centre for Applied Clinical Pharmacology, City Clinical Hospital #64, GOUVPO Russian Peoples' Friendship University, Moscow, Russia
| | | | | | | |
Collapse
|
43
|
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
|
44
|
Chan ST, Logan AC. The clinical impact of cytomegalovirus infection following allogeneic hematopoietic cell transplantation: Why the quest for meaningful prophylaxis still matters. Blood Rev 2017; 31:173-183. [DOI: 10.1016/j.blre.2017.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 12/16/2016] [Accepted: 01/31/2017] [Indexed: 11/28/2022]
|
45
|
Arend KC, Lenarcic EM, Vincent HA, Rashid N, Lazear E, McDonald IM, Gilbert TSK, East MP, Herring LE, Johnson GL, Graves LM, Moorman NJ. Kinome Profiling Identifies Druggable Targets for Novel Human Cytomegalovirus (HCMV) Antivirals. Mol Cell Proteomics 2017; 16:S263-S276. [PMID: 28237943 PMCID: PMC5393402 DOI: 10.1074/mcp.m116.065375] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 02/23/2017] [Indexed: 11/06/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a significant cause of disease in immune-compromised adults and immune naïve newborns. No vaccine exists to prevent HCMV infection, and current antiviral therapies have toxic side effects that limit the duration and intensity of their use. There is thus an urgent need for new strategies to treat HCMV infection. Repurposing existing drugs as antivirals is an attractive approach to limit the time and cost of new antiviral drug development. Virus-induced changes in infected cells are often driven by changes in cellular kinase activity, which led us to hypothesize that defining the complement of kinases (the kinome), whose abundance or expression is altered during infection would identify existing kinase inhibitors that could be repurposed as new antivirals. To this end, we applied a kinase capture technique, multiplexed kinase inhibitor bead-mass spectrometry (MIB-MS) kinome, to quantitatively measure perturbations in >240 cellular kinases simultaneously in cells infected with a laboratory-adapted (AD169) or clinical (TB40E) HCMV strain. MIB-MS profiling identified time-dependent increases and decreases in MIB binding of multiple kinases including cell cycle kinases, receptor tyrosine kinases, and mitotic kinases. Based on the kinome data, we tested the antiviral effects of kinase inhibitors and other compounds, several of which are in clinical use or development. Using a novel flow cytometry-based assay and a fluorescent reporter virus we identified three compounds that inhibited HCMV replication with IC50 values of <1 μm, and at doses that were not toxic to uninfected cells. The most potent inhibitor of HCMV replication was OTSSP167 (IC50 <1.2 nm), a MELK inhibitor, blocked HCMV early gene expression and viral DNA accumulation, resulting in a >3 log decrease in virus replication. These results show the utility of MIB-MS kinome profiling for identifying existing kinase inhibitors that can potentially be repurposed as novel antiviral drugs.
Collapse
Affiliation(s)
- Kyle C Arend
- From the ‡Department of Microbiology & Immunology
- ¶Lineberger Comprehensive Cancer Center
| | - Erik M Lenarcic
- From the ‡Department of Microbiology & Immunology
- ¶Lineberger Comprehensive Cancer Center
| | - Heather A Vincent
- From the ‡Department of Microbiology & Immunology
- ¶Lineberger Comprehensive Cancer Center
| | - Naim Rashid
- ¶Lineberger Comprehensive Cancer Center
- ‖Department of Biostatistics
| | - Eric Lazear
- From the ‡Department of Microbiology & Immunology
- ¶Lineberger Comprehensive Cancer Center
| | | | | | | | - Laura E Herring
- §Department of Pharmacology
- **UNC Michael Hooker Proteomics Core Facility University of North Carolina, Chapel Hill, 27599 North Carolina
| | | | - Lee M Graves
- §Department of Pharmacology
- **UNC Michael Hooker Proteomics Core Facility University of North Carolina, Chapel Hill, 27599 North Carolina
| | - Nathaniel J Moorman
- From the ‡Department of Microbiology & Immunology,
- ¶Lineberger Comprehensive Cancer Center
| |
Collapse
|
46
|
Moazen B, Ebrahimi E, Nejatollahi F. Single Chain Antibodies Against gp55 of Human Cytomegalovirus (HCMV) for Prophylaxis and Treatment of HCMV Infections. Jundishapur J Microbiol 2016; 9:e16241. [PMID: 27217918 PMCID: PMC4870390 DOI: 10.5812/jjm.16241] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 07/18/2014] [Accepted: 07/23/2014] [Indexed: 02/06/2023] Open
Abstract
Background: Immunotherapy is a promising prospective new treatment for cytomegalovirus (CMV) infections. Neutralizing effects have been reported using monoclonal antibodies. Recombinant single chain antibodies (scFvs) due to their advantages over monoclonal antibodies are potential alternatives and provide valuable clinical agents. Objectives: The aim of this study was to select specific single chain antibodies against gp55 of CMV and to evaluate their neutralizing effects. In the present study, we selected specific single chain antibodies against glycoprotein 55 (gp55) of CMV for their use in treatment and diagnosis. Materials and Methods: Single chain antibodies specific against an epitope located in the C-terminal part of gp55 were selected from a phage antibody display library. After four rounds of panning, twenty clones were amplified by the polymerase chain reaction (PCR) and fingerprinted by MvaI restriction enzyme. The reactivities of the specific clones were tested by the enzyme-linked immunosorbent assay (ELISA) and the neutralizing effects were evaluated by the plaque reduction assay. Results: Fingerprinting of selected clones revealed three specific single chain antibodies (scFv1, scFv2 and scFv3) with frequencies 25%, 20 and 20%. The clones produced positive ELISA with the corresponding peptide. The percentages of plaque reduction for scFv1, scFv2 and scFv3 were 23.7, 68.8 and 11.6, respectively. Conclusions: Gp55 of human CMV is considered as an important candidate for immunotherapy. In this study, we selected three specific clones against gp55. The scFvs reacted only with the corresponding peptide in a positive ELISA. The scFv2 with 68.8% neutralizing effect showed the potential to be considered for prophylaxis and treatment of CMV infections, especially in solid organ transplant recipients, for whom treatment of CMV is urgently needed. The scFv2 with neutralizing effect of 68.8%, has the potential to be considered for treatment of these patients. The specific scFv1 and scFv3 with lower neutralizing effects can be used for diagnostic purposes.
Collapse
Affiliation(s)
- Bahareh Moazen
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Elahe Ebrahimi
- Department of Immunology, Recombinant Antibody Laboratory, Shiraz University of Medical Sciences, Shiraz, IR Iran
| | - Foroogh Nejatollahi
- Shiraz HIV/AIDS Research Center, Shiraz University of Medical Sciences, Shiraz, IR Iran
- Department of Immunology, Recombinant Antibody Laboratory, Shiraz University of Medical Sciences, Shiraz, IR Iran
- Corresponding author: Foroogh Nejatollahi, Department of Immunology, Recombinant Antibody Laboratory, Shiraz University of Medical Sciences, Shiraz, IR Iran. Tel: +98-7112351575, Fax: +98-7112351575, E-mail:
| |
Collapse
|
47
|
Anemia in children following renal transplantation-results from the ESPN/ERA-EDTA Registry. Pediatr Nephrol 2016; 31:325-33. [PMID: 26385862 DOI: 10.1007/s00467-015-3201-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Revised: 08/13/2015] [Accepted: 08/24/2015] [Indexed: 10/23/2022]
Abstract
BACKGROUND Our aim was to determine the prevalence of sub-target hemoglobin (Hb) levels in children with a renal allograft and to identify potential determinants associated with these Hb levels. METHODS Data from 3669 children with a functioning renal allograft, aged <18 years between 1 January 2000 and 31 December 2012, from 20 European countries were retrieved from the ESPN/ERA-EDTA Registry, providing 16,170 Hb measurements. RESULTS According to the NKF/KDOQI classification and the UK-NICE guidelines, 49.8 and 7.8% of the patients, respectively, were anemic. Hb levels were strongly associated with graft function, with Hb levels of 12.6 g/dl in children with chronic kidney disease (CKD) stage 1, declining to 10.7 g/dl in children with CKD stage 5 (P < 0.001). Higher Hb levels were associated with the use of tacrolimus compared to ciclosporin (0.14 g/dl; 95% confidence interval 0.02-0.27; P = 0.002). Low Hb levels were associated with an increased risk of graft failure (P = 0.01) or combined graft failure and death (P < 0.01), but not with death alone (not significant). CONCLUSIONS Anemia is present in a significant proportion of European pediatric kidney transplant recipients and is associated with renal allograft dysfunction and type of immunosuppressants used. In our patient cohort, higher Hb levels were associated with better graft and patient survival and less hypertension.
Collapse
|
48
|
Treatment of Cytomegalovirus Infection with Cidofovir and CMV Immune Globulin in a Lung Transplant Recipient. Case Rep Transplant 2016; 2016:4560745. [PMID: 26885432 PMCID: PMC4738720 DOI: 10.1155/2016/4560745] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 12/11/2015] [Accepted: 12/13/2015] [Indexed: 12/14/2022] Open
Abstract
Cytomegalovirus (CMV) infection after lung transplantation is associated with increased risk for pneumonitis and bronchiolitis obliterans as well as allograft rejection and opportunistic infections. Ganciclovir is the mainstay of prophylaxis and treatment but CMV infections can be unresponsive. Apart from direct antiviral drugs, CMV immunoglobulin (CMVIG) preparations may be considered but are only licensed for prophylaxis. A CMV-seronegative 42-year-old man with cystic fibrosis received a lung from a CMV-seropositive donor. Intravenous ganciclovir prophylaxis was delayed until day 12 due to acute postoperative renal failure and was accompanied by five doses of CMVIG (10 g). By day 16, CMV-DNA was detectable and rising; CMV-specific T-cells were undetectable. Switch from ganciclovir to foscarnet prompted a transient decrease in CMV viral load, but after increasing again to reach 3600 copies/mL foscarnet was changed to intravenous cidofovir and CMVIG was restarted. CMV load continued to fluctuate and declined slowly, whereas CMV-specific T-cells were detected five months later and increased thereafter. At last follow-up, the patient was in very good clinical condition with no evidence of bronchiolitis obliterans. No side effects of this treatment were observed. In this hard-to-treat case, the combination of cidofovir with off-label use of CMVIG contributed to a successful outcome.
Collapse
|
49
|
Martin-Gandul C, Mueller NJ, Pascual M, Manuel O. The Impact of Infection on Chronic Allograft Dysfunction and Allograft Survival After Solid Organ Transplantation. Am J Transplant 2015; 15:3024-40. [PMID: 26474168 DOI: 10.1111/ajt.13486] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 07/14/2015] [Accepted: 08/06/2015] [Indexed: 01/25/2023]
Abstract
Infectious diseases after solid organ transplantation (SOT) are a significant cause of morbidity and reduced allograft and patient survival; however, the influence of infection on the development of chronic allograft dysfunction has not been completely delineated. Some viral infections appear to affect allograft function by both inducing direct tissue damage and immunologically related injury, including acute rejection. In particular, this has been observed for cytomegalovirus (CMV) infection in all SOT recipients and for BK virus infection in kidney transplant recipients, for community-acquired respiratory viruses in lung transplant recipients, and for hepatitis C virus in liver transplant recipients. The impact of bacterial and fungal infections is less clear, but bacterial urinary tract infections and respiratory tract colonization by Pseudomonas aeruginosa and Aspergillus spp appear to be correlated with higher rates of chronic allograft dysfunction in kidney and lung transplant recipients, respectively. Evidence supports the beneficial effects of the use of antiviral prophylaxis for CMV in improving allograft function and survival in SOT recipients. Nevertheless, there is still a need for prospective interventional trials assessing the potential effects of preventive and therapeutic strategies against bacterial and fungal infection for reducing or delaying the development of chronic allograft dysfunction.
Collapse
Affiliation(s)
- C Martin-Gandul
- Transplantation Center, University Hospital and University of Lausanne, Lausanne, Switzerland.,Infectious Diseases Service, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - N J Mueller
- Division of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - M Pascual
- Transplantation Center, University Hospital and University of Lausanne, Lausanne, Switzerland
| | - O Manuel
- Transplantation Center, University Hospital and University of Lausanne, Lausanne, Switzerland.,Infectious Diseases Service, University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
50
|
Arasaratnam RJ, Leen AM. Adoptive T cell therapy for the treatment of viral infections. ANNALS OF TRANSLATIONAL MEDICINE 2015; 3:278. [PMID: 26605324 DOI: 10.3978/j.issn.2305-5839.2015.10.12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Reuben J Arasaratnam
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX 77030, USA
| | - Ann M Leen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital and Texas Children's Hospital, Houston, TX 77030, USA
| |
Collapse
|