1
|
Xu Y, Wu Y, Wu X, Zhang Y, Yang Y, Li D, Yang B, Gao K, Zhang Z, Dong C. Structural basis of human mpox viral DNA replication inhibition by brincidofovir and cidofovir. Int J Biol Macromol 2024; 270:132231. [PMID: 38735603 DOI: 10.1016/j.ijbiomac.2024.132231] [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: 01/24/2024] [Revised: 04/24/2024] [Accepted: 05/03/2024] [Indexed: 05/14/2024]
Abstract
Mpox virus has wildly spread over 108 non-endemic regions in the world since May 2022. DNA replication of mpox is performed by DNA polymerase machinery F8-A22-E4, which is known as a great drug target. Brincidofovir and cidofovir are reported to have broad-spectrum antiviral activity against poxviruses, including mpox virus in animal models. However, the molecular mechanism is not understood. Here we report cryogenic electron microscopy structures of mpox viral F8-A22-E4 in complex with a DNA duplex, or dCTP and the DNA duplex, or cidofovir diphosphate and the DNA duplex at resolution of 3.22, 2.98 and 2.79 Å, respectively. Our structural work and DNA replication inhibition assays reveal that cidofovir diphosphate is located at the dCTP binding position with a different conformation to compete with dCTP to incorporate into the DNA and inhibit DNA synthesis. Conformation of both F8-A22-E4 and DNA is changed from the pre-dNTP binding state to DNA synthesizing state after dCTP or cidofovir diphosphate is bound, suggesting a coupling mechanism. This work provides the structural basis of DNA synthesis inhibition by brincidofovir and cidofovir, providing a rational strategy for new therapeutical development for mpox virus and other pox viruses.
Collapse
Affiliation(s)
- Yunxia Xu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yaqi Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Xiaoying Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuanyuan Zhang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yaxue Yang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Danyang Li
- The Cryo-EM Center, Core Facility of Wuhan University, Wuhan University, Wuhan 430071, China
| | - Biao Yang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Kaiting Gao
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Zhengyu Zhang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China.
| | - Changjiang Dong
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China.
| |
Collapse
|
2
|
Rani I, Joshi H, Sharma U, Kaur J, Sood S, Ramniwas S, Chauhan A, Abdulabbas HS, Tuli HS. Potential use of cidofovir, brincidofovir, and tecovirimat drugs in fighting monkeypox infection: recent trends and advancements. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:2055-2065. [PMID: 37837475 DOI: 10.1007/s00210-023-02769-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 10/03/2023] [Indexed: 10/16/2023]
Abstract
Recent years have witnessed the rise of more recent pandemic outbreaks including COVID-19 and monkeypox. A multinational monkeypox outbreak creates a complex situation that necessitates countermeasures to the existing quo. The first incidence of monkeypox was documented in the 1970s, and further outbreaks led to a public health emergency of international concern. Yet as of right now, neither vaccines nor medicines are certain to treat monkeypox. Even the inability of conducting human clinical trials has prevented thousands of patients from receiving effective disease management. The current state of the disease's understanding, the treatment options available, financial resources, and lastly international policies to control an epidemic state are the major obstacles to controlling epidemics. The current review focuses on the epidemiology of monkeypox, scientific ideas, and available treatments, including potential monkeypox therapeutic methods. As a result, a thorough understanding of monkeypox literature will facilitate in the development of new therapeutic medications for the prevention and treatment of monkeypox.
Collapse
Affiliation(s)
- Isha Rani
- Department of Biochemistry, Maharishi Markandeshwar College of Medical Sciences and Research (MMCMSR), Sadopur, Ambala, 134007, India
| | - Hemant Joshi
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, 110067, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, Faculty of Engineering, The University of New South Wales, Sydney, 2052, Australia
| | - Shivani Sood
- GIOSTAR-USA, Global Institute of Stem Cell Therapy and Research, Mohali, 140308, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida, 201303, India
| | - Hadi Sajid Abdulabbas
- Department of Biology, College of Science, University of Babylon, Babylon, 51002, Iraq
| | - Hardeep Singh Tuli
- Department of Bio-Sciences and Technology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to Be University), Mullana, Ambala, 133207, India.
| |
Collapse
|
3
|
Tippin T, Faison S, Schuck V, Dunn J, Naderer O. Utility of Cytochrome P450 4F2 Genotyping to Assess Drug Interaction Risk for Brincidovovir, a Cytochrome P450 4F2 Substrate. Clin Pharmacol Drug Dev 2024; 13:288-296. [PMID: 38171911 DOI: 10.1002/cpdd.1356] [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: 08/10/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024]
Abstract
Smallpox was eradicated in 1980 but remains a biothreat due to the potential release of variola virus into the general population. Brincidofovir, the second medicine approved by the US Food and Drug Administration to treat smallpox, is metabolized by oxidative and hydrolytic pathways. The oxidative pathway is initiated by cytochrome P450 4F2 (CYP4F2), an enzyme lacking clinical probes for drug interaction studies. The aim of this work was to assess the impact of reduced activity CYP4F2 variants (rs2108622, C/T and T/T) on brincidofovir pharmacokinetics as a surrogate for drug inhibition. Genotyping was performed on blood from healthy participants receiving oral (n = 261) and intravenous (IV, n = 49) brincidofovir across 6 phase 1 trials. Plasma concentrations were measured by validated liquid chromatography tandem mass spectrometry methods. After oral administration, subjects with the lowest activity CYP4F2 genotype (T/T) had up to 36% higher AUCinf and 29% higher Cmax while subjects with the moderate activity CYP4F2 genotype (C/T) had similar Cmax and AUCinf compared to those with the wild-type genotype. Little to no increase in brincidofovir exposure parameters was observed following IV administration. Based on the lack of significant increases in brincidofovir plasma concentrations in subjects with low activity CYP4F2, a clinically meaningful drug-drug interaction is not expected with CYP4F2 inhibitor and brincidofovir coadministration.
Collapse
|
4
|
Subbaiah MAM, Rautio J, Meanwell NA. Prodrugs as empowering tools in drug discovery and development: recent strategic applications of drug delivery solutions to mitigate challenges associated with lead compounds and drug candidates. Chem Soc Rev 2024; 53:2099-2210. [PMID: 38226865 DOI: 10.1039/d2cs00957a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
The delivery of a drug to a specific organ or tissue at an efficacious concentration is the pharmacokinetic (PK) hallmark of promoting effective pharmacological action at a target site with an acceptable safety profile. Sub-optimal pharmaceutical or ADME profiles of drug candidates, which can often be a function of inherently poor physicochemical properties, pose significant challenges to drug discovery and development teams and may contribute to high compound attrition rates. Medicinal chemists have exploited prodrugs as an informed strategy to productively enhance the profiles of new chemical entities by optimizing the physicochemical, biopharmaceutical, and pharmacokinetic properties as well as selectively delivering a molecule to the site of action as a means of addressing a range of limitations. While discovery scientists have traditionally employed prodrugs to improve solubility and membrane permeability, the growing sophistication of prodrug technologies has enabled a significant expansion of their scope and applications as an empowering tool to mitigate a broad range of drug delivery challenges. Prodrugs have emerged as successful solutions to resolve non-linear exposure, inadequate exposure to support toxicological studies, pH-dependent absorption, high pill burden, formulation challenges, lack of feasibility of developing solid and liquid dosage forms, first-pass metabolism, high dosing frequency translating to reduced patient compliance and poor site-specific drug delivery. During the period 2012-2022, the US Food and Drug Administration (FDA) approved 50 prodrugs, which amounts to 13% of approved small molecule drugs, reflecting both the importance and success of implementing prodrug approaches in the pursuit of developing safe and effective drugs to address unmet medical needs.
Collapse
Affiliation(s)
- Murugaiah A M Subbaiah
- Department of Medicinal Chemistry, Biocon Bristol Myers Squibb R&D Centre, Biocon Park, Bommasandra Phase IV, Bangalore, PIN 560099, India.
| | - Jarkko Rautio
- School of Pharmacy, University of Eastern Finland, Kuopio, Finland
| | - Nicholas A Meanwell
- The Baruch S. Blumberg Institute, Doylestown, PA 18902, USA
- Department of Medicinal Chemistry, The College of Pharmacy, The University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Alagarsamy V, Shyam Sundar P, Raja Solomon V, Narendhar B, Sulthana MT, Rohitha K, Dhanwar S, Dharshini Aishwarya A, Murugesan S. Pharmacophore modelling-based drug repurposing approaches for monkeypox therapeutics. J Biomol Struct Dyn 2023; 41:10678-10689. [PMID: 36905675 DOI: 10.1080/07391102.2023.2188428] [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: 09/27/2022] [Accepted: 12/05/2022] [Indexed: 03/13/2023]
Abstract
Monkeypox is a zoonotic viral disease that mainly affects tropical rainforest regions of central and west Africa, with sporadic exportations to other places. Since there is no cure, treating monkeypox with an antiviral drug developed for smallpox is currently acceptable. Our study mainly focused on finding new therapeutics to target monkeypox from existing compounds or medications. It is a successful method for discovering or developing medicinal compounds with novel pharmacological or therapeutic applications. In this study, homology modelling developed the Monkeypox VarTMPK (IMNR) structure. Ligand-based pharmacophore was generated using the best docking pose of standard ticovirimat. Further, molecular docking analysis showed compounds, tetrahydroxycurcumin, procyanidin, rutin, vicenin-2, kaempferol 3-(6''-malonylglucoside) were the top five binding energy compounds against VarTMPK (1MNR). Furthermore, we carried out MD simulations for 100 ns for the six compounds, including reference based on the binding energies and interactions. MD studies revealed that as ticovirimat interacted with residues Lys17, Ser18, and Arg45, all the above five compounds interacted with the same amino acids at the active site during docking and simulation studies. Among all the compounds, ZINC4649679 (Tetrahydroxycurcumin) was shown to have the highest binding energy -9.7 kcal/mol and also observed stable protein-ligand complex during MD studies. ADMET profile estimation showed that the docked phytochemicals were safe. However, further biological assessment through a wet lab is essential to measure the efficacy and safety of the compounds.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- V Alagarsamy
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Hyderabad, India
| | - P Shyam Sundar
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Hyderabad, India
| | - V Raja Solomon
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Hyderabad, India
| | - B Narendhar
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Hyderabad, India
| | - M T Sulthana
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Hyderabad, India
| | - Kotha Rohitha
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Hyderabad, India
| | - Sangeeta Dhanwar
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Hyderabad, India
| | - A Dharshini Aishwarya
- Medicinal Chemistry Research Laboratory, MNR College of Pharmacy, Sangareddy, Hyderabad, India
| | - S Murugesan
- Department of Pharmacy, BITS, Pilani, Pilani, Rajasthan, India
| |
Collapse
|
6
|
Temrikar ZH, Golden JE, Jonsson CB, Meibohm B. Clinical and Translational Pharmacology Considerations for Anti-infectives Approved Under the FDA Animal Rule. Clin Pharmacokinet 2023; 62:943-953. [PMID: 37326917 PMCID: PMC10471120 DOI: 10.1007/s40262-023-01267-x] [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] [Accepted: 05/21/2023] [Indexed: 06/17/2023]
Abstract
The US Food and Drug Administration's Animal Rule provides a pathway for approval of drugs and biologics aimed to treat serious or life-threatening conditions wherein traditional clinical trials are either not ethical or feasible. In such a scenario, determination of safety and efficacy are based on integration of data on drug disposition and drug action collected from in vitro models, infected animals, and healthy volunteer human studies. The demonstration of clinical efficacy and safety in humans based on robust, well-controlled animal studies is filled with challenges. This review elaborates on the challenges in the translation of data from in vitro and animal models to human dosing for antimicrobials. In this context, it discusses precedents of drugs approved under the Animal Rule, along with the approaches and guidance undertaken by sponsors.
Collapse
Affiliation(s)
- Zaid H Temrikar
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
| | - Jennifer E Golden
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI, USA
| | - Colleen B Jonsson
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA
- Department of Microbiology, Immunology, Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Regional Biocontainment Laboratory, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Bernd Meibohm
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Tennessee Health Science Center, 881 Madison Avenue, Memphis, TN, 38163, USA.
| |
Collapse
|
7
|
Grosenbach DW, Russo AT, Blum ED, Hruby DE. Emerging pharmacological strategies for treating and preventing mpox. Expert Rev Clin Pharmacol 2023; 16:843-854. [PMID: 37592723 DOI: 10.1080/17512433.2023.2249820] [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: 05/08/2023] [Revised: 08/10/2023] [Accepted: 08/16/2023] [Indexed: 08/19/2023]
Abstract
INTRODUCTION Since May 2022, there have been nearly 87,000 documented cases of mpox worldwide, with 119 deaths. Pharmacological interventions for mpox include the MVA-BN smallpox vaccine, tecovirimat, cidofovir, its pro-drug brincidofovir, and vaccinia immune globulin intravenous (VIGIV). AREAS COVERED The literature search and information gathering for this review included the PubMed database focusing on mpox and monkeypox, in combination with tecovirimat, brincidofovir, cidofovir, VIGIV, and smallpox vaccine. WHO.int, CDC.gov, FDA.gov, and ClinicalTrials.gov websites were accessed for the most recent information on the mpox outbreak. Mechanisms for deployment and access to treatment including expanded access, emergency use, and clinical trials will be discussed. Treatment outcomes with safety data will be presented. EXPERT OPINION The vaccine as a preventive measure, along with numerous treatment options, largely controlled the outbreak, although deployment of each could be improved upon to hasten and broaden access. More widespread coverage by the vaccine is necessary to prevent future resurgence of mpox. Tecovirimat has emerged as a safe frontline treatment for mpox, while brincidofovir use has been limited by safety concerns. VIGIV and cidofovir should be reserved for the most severe cases in which other options are not fully effective.
Collapse
|
8
|
Bhat S, Saha S, Garg T, Sehrawat H, Chopade BA, Gupta V. Insights into the challenging multi-country outbreak of Mpox: a comprehensive review. J Med Microbiol 2023; 72. [PMID: 37378642 DOI: 10.1099/jmm.0.001725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2023] Open
Abstract
Human monkeypox virus (hMpoxV) is of zoonotic origin and is closely related to the once-dreaded smallpox virus. It is largely endemic to the African continent but has moved out of the endemic regions as sporadic clusters in the past 20 years, raising concerns worldwide. Human Mpox is characterized by a mild to severe, self-limiting infection, with mortality ranging from less than 1% to up to 10% during different outbreaks caused by different clades of MpoxV. Bushmeat hunting is one of the primary reasons for its transmission from animals to humans. Various international and national health regulatory bodies are closely monitoring the disease and have laid down guidelines to manage and prevent hMpox cases. Emergency Use Status has been granted to Tecovirimat and Brincidofovir to treat severe cases and vaccination with the smallpox vaccine is recommended for high-risk group individuals. Strategies to repurpose and discover novel therapeutics and vaccines to control the outbreak are being researched. The current Mpox outbreak that has mainly affected men as approximately 96% of all cases are reported in men, is probably the result of a complex intersection of various factors. This necessitates a strong One Health response coordination involving human, animal and environmental health institutions. This review is an attempt to provide an all-inclusive overview of the biology, history, epidemiology, pathophysiology, diagnosis and management of hMpox in context to the recent 2022-2023 multi-country outbreak which is termed by WHO a 'Public Health Emergency of International Concern (PHEIC)'.
Collapse
Affiliation(s)
- Shreyas Bhat
- Department of Microbiology, Ram Lal Anand College, University of Delhi, New Delhi, India
| | - Sumana Saha
- Department of Microbiology, Ram Lal Anand College, University of Delhi, New Delhi, India
| | - Tanisha Garg
- Department of Microbiology, Ram Lal Anand College, University of Delhi, New Delhi, India
| | | | - Balu Ananda Chopade
- Department of Microbiology, Savitribai Phule Pune University, Pune 411007, Maharashtra, India
| | - Vandana Gupta
- Department of Microbiology, Ram Lal Anand College, University of Delhi, New Delhi, India
| |
Collapse
|
9
|
Xu Y, Wu Y, Zhang Y, Fan R, Yang Y, Li D, Zhu S, Yang B, Zhang Z, Dong C. Cryo-EM structures of human monkeypox viral replication complexes with and without DNA duplex. Cell Res 2023; 33:479-482. [PMID: 36973539 PMCID: PMC10235115 DOI: 10.1038/s41422-023-00796-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 03/06/2023] [Indexed: 03/29/2023] Open
Affiliation(s)
- Yunxia Xu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yaqi Wu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yuanyuan Zhang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Ruixin Fan
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yaxue Yang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Danyang Li
- The Cryo-EM Center, Core facility of Wuhan University, Wuhan University, Wuhan, Hubei, China
| | - Shimin Zhu
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Biao Yang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Zhengyu Zhang
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China.
| | - Changjiang Dong
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, State Key Laboratory of Virology, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
10
|
Abstract
BACKGROUND Mpox was declared a Public Health Emergency of International Concern (PHEIC) by the World Health Organization (WHO) on 23 July 2022, following the identification of thousands of cases in several non-endemic countries in previous months. There are currently no licenced therapeutics for treating mpox; however, some medications may be authorized for use in an outbreak. The efficacy and safety of possible therapeutic options has not been studied in humans with mpox. There is a need to investigate the evidence on safety and effectiveness of treatments for mpox in humans; should any therapeutic option be efficacious and safe, it may be approved for use around the world. OBJECTIVES There are two parts to this Cochrane Review: a review of evidence from randomized controlled trials (RCTs), and a narrative review of safety data from non-randomized studies. Randomized controlled trials review To systematically review the existing evidence on the effectiveness of therapeutics for mpox infection in humans compared to: a) another different therapeutic for mpox, or b) placebo, or c) supportive care, defined as the treatment of physical and psychological symptoms arising from the disease. Non-randomized studies review To assess the safety of therapeutics for mpox infection from non-randomized studies (NRS). SEARCH METHODS Randomized controlled trials review We searched the following databases up to 25 January 2023: MEDLINE (OVID), Embase (OVID), Biosis previews (Web of Science), CAB Abstracts (Web of science), and Cochrane CENTRAL (Issue 1 2023). We conducted a search of trial registries (Clinicaltrials.gov and International Clinical Trials Registry Platform (ICTRP)) on 25 January 2023. There were no date or language limits placed on the search. We undertook a call to experts in the field for relevant studies or ongoing trials to be considered for inclusion in the review. Non-randomized studies review We searched the following databases on 22 September 2022: Cochrane Central Register of Controlled Trials (CENTRAL; Issue 9 of 12, 2022), published in the Cochrane Library; MEDLINE (Ovid); Embase (Ovid); and Scopus (Elsevier). We also searched the WHO International Clinical Trials Registry Platform and ClinicalTrials.gov for trials in progress. SELECTION CRITERIA For the RCT review and the narrative review, any therapeutic for the treatment of mpox in humans was eligible for inclusion, including tecovirimat, brincidofovir, cidofovir, NIOCH-14, immunomodulators, and vaccine immune globulin. Randomized controlled trials review Studies were eligible for the main review if they were of randomized controlled design and investigated the effectiveness or safety of therapeutics in human mpox infection. Non-randomized studies review Studies were eligible for inclusion in the review of non-randomized studies if they were of non-randomized design and contained data concerning the safety of any therapeutic in human mpox infection. DATA COLLECTION AND ANALYSIS Randomized controlled trials review Two review authors independently applied study inclusion criteria to identify eligible studies. If we had identified any eligible studies, we planned to assess the risk of bias, and report results with 95% confidence intervals (CI). The critical outcomes were serious adverse events, development of disease-related complications, admission to hospital for non-hospitalized participants, pain as judged by any visual or numerical pain scale, level of virus detected in clinical samples, time to healing of all skin lesions, and mortality. We planned to perform subgroup analysis to explore whether the effect of the therapeutic on the planned outcomes was modified by disease severity and days from symptom onset to therapeutic administration. We also intended to explore the following subgroups of absolute effects: immunosuppression, age, and pre-existing skin disease. Non-randomized studies review One review author applied study inclusion criteria to identify eligible studies and extracted data. Studies of a non-randomized design containing data on the safety of therapeutics could not be meta-analyzed due to the absence of a comparator; we summarized these data narratively in an appendix. MAIN RESULTS Randomized controlled trials review We did not identify any completed RCTs investigating the effectiveness of therapeutics for treating mpox for the main review. We identified five ongoing trials that plan to assess the effectiveness of one therapeutic option, tecovirimat, for treating mpox in adults and children. One of these ongoing trials intends to include populations with, or at greater risk of, severe disease, which will allow an assessment of safety in more vulnerable populations. Non-randomized studies review Three non-randomized studies met the inclusion criteria for the narrative review, concerning data on the safety of therapeutics in mpox. Very low-certainty evidence from non-randomized studies of small numbers of people indicates no serious safety signals emerging for the use of tecovirimat in people with mpox infection, but a possible safety signal for brincidofovir. All three participants who received brincidofovir had raised alanine aminotransferase (ALT), but not bilirubin, suggesting mild liver injury. No study reported severe drug-induced liver injury with brincidofovir. AUTHORS' CONCLUSIONS Randomized controlled trials review This review found no evidence from randomized controlled trials concerning the efficacy and safety of therapeutics in humans with mpox. Non-randomized studies review Very low-certainty evidence from non-randomized studies indicates no serious safety signals emerging for the use of tecovirimat in people with mpox infection. In contrast, very low-certainty evidence raises a safety signal that brincidofovir may cause liver injury. This is also suggested by indirect evidence from brincidofovir use in smallpox. This warrants further investigation and monitoring. This Cochrane Review will be updated as new evidence becomes available to assist policymakers, health professionals, and consumers in making appropriate decisions for the treatment of mpox.
Collapse
Affiliation(s)
- Tilly Fox
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Susan Gould
- Royal Liverpool University Hospital, Liverpool, UK
| | - Naveena Princy
- Department of Infectious Diseases, Christian Medical College Vellore, Vellore, India
| | - Tim Rowland
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Vittoria Lutje
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Rebecca Kuehn
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool, UK
| |
Collapse
|
11
|
Abstract
BACKGROUND The objective of this paper is to analyze the current status of monkeypox worldwide. In the face of this public health threat, our purpose is to elucidate the clinical characteristics and epidemiology of monkeypox, the developmental progress of monkeypox-related drugs and the vaccines available. DATA SOURCES The literature review was performed in databases including PubMed, Science Direct and Google Scholar up to July 2022. RESULTS Since May 2022, the World Health Organization has reported more than 45,000 confirmed cases from 92 nonendemic countries, including nine deaths. Although some women and children have been infected so far, most cases have occurred among men who have sex with other men, especially those with multiple sexual partners or anonymous sex. CONCLUSIONS Pediatric monkeypox infection has been associated with a higher likelihood of severe illness and mortality than in adults. Severe monkeypox illness in pediatrics often requires adjunctive antiviral therapy. It is crucial for all countries to establish sound monitoring and testing systems and be prepared with emergency preparedness.
Collapse
Affiliation(s)
- Ya-Mei Dou
- NHC Key Laboratory of Medical Virology and Viral Disease, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, 155 Changbai Road, ChangPing District, Beijing, 102206, China
| | - Hang Yuan
- NHC Key Laboratory of Medical Virology and Viral Disease, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, 155 Changbai Road, ChangPing District, Beijing, 102206, China
| | - Hou-Wen Tian
- NHC Key Laboratory of Medical Virology and Viral Disease, Chinese Center for Disease Control and Prevention, National Institute for Viral Disease Control and Prevention, 155 Changbai Road, ChangPing District, Beijing, 102206, China.
| |
Collapse
|
12
|
Sukhdeo S, Mishra S, Walmsley S. Human monkeypox: a comparison of the characteristics of the new epidemic to the endemic disease. BMC Infect Dis 2022; 22:928. [PMID: 36503476 PMCID: PMC9742013 DOI: 10.1186/s12879-022-07900-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/25/2022] [Indexed: 12/14/2022] Open
Abstract
In May 2022, a new global outbreak of mpox (formerly, human monkeypox) emerged that was declared a public health emergency of international concern by the World Health Organization on July 23, 2022. With new patterns of person-to-person spread within sexual networks in nonendemic countries and several differences from the classic disease course, we performed a comprehensive review of existing literature on human monkeypox to discuss epidemiology, modes of transmission, clinical presentation and asymptomatic infection, diagnostics, therapeutics, and vaccines with the primary aim to identify important areas for future research of this new epidemic form of the disease. A comprehensive literature search was performed of all published literature to August 15, 2022. Historically, in regions of monkeypox virus endemicity, human outbreaks have occurred related to discrete zoonotic events. The animal reservoir is unknown, but the virus has been isolated from rodents. Traditionally, transmission occurred by direct or indirect contact with an infected animal. In nonendemic countries affected in the 2022 outbreak, almost exclusive person-to-person spread has been observed, and most cases are connected to sexual networks of gay, bisexual, and other men who have sex with men. After an incubation period of approximately 13 days, in traditional human cases affected persons developed a febrile prodrome preceding a rash that started on the face and body, spread centrifugally to the palms and soles and healed monomorphically over two to four weeks. However, in the 2022 outbreak, the febrile illness is often absent or occurs after the onset of the rash. The rash presents primarily in the anogenital region and face before disseminating throughout the body, with lesions displaying regional pleomorphism. There is a paucity of data for the role of antiviral agents or vaccines. The epidemiology and clinical course of mpox has changed in the 2022 epidemic from that observed with the endemic disease. There is an urgent need to establish rapid and collaborative research platforms to diagnose, treat and prevent disease and inform important public health and other strategies to stop the spread of disease.
Collapse
Affiliation(s)
- Sharon Sukhdeo
- Department of Medicine, University of Toronto, Toronto, Canada.
| | - Sharmistha Mishra
- Division of Infectious Diseases, Department of Medicine, St. Michael's Hospital, MAP Centre for Urban Health Solutions, University of Toronto, Toronto, Canada
| | - Sharon Walmsley
- Department of Medicine, Division of Infectious Diseases, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
13
|
James J, A P, P K, Rani J, V S. An Update on the Pharmacological Aspects of Vaccines and Antivirals for the Management of Monkeypox. J Pharmacol Pharmacother 2022. [DOI: 10.1177/0976500x231156733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023] Open
Abstract
Monkeypox is a self-limiting zoonotic disease caused by the monkeypox virus belonging to the genus of orthopox viruses. Initially considered an ‘African disease’, this infection has crossed the boundaries to affect other continents and it has raised tremendous concerns among the general public as well as the medical fraternity all over the world, particularly because of the lack of specific vaccinations and drugs for the management of the illness. Epidemiological evaluation of the current infection has reported that it is mainly transmitted through sexual contact in bisexual men, mostly whites, and in those with pre-existing human immunodeficiency virus infection. The most common presentations were skin rash, anogenital lesions, or mucosal lesions along with systemic symptoms. It has been established that the vaccines and drugs approved for the management of smallpox could be used for the management of the current monkeypox outbreak. Vaccinia Immune Globulin (VIG) and vaccines like JYNNEOS and ACAM2000 and antiviral drugs like tecovirimat, cidofovir (CDV), and brincidofovir are being considered for those patients with serious diseases. It is imperative for physicians to understand the pharmacological aspects of these drugs for delivering better care to patients with monkeypox, which is eventually essential for the containment of this infection. This review covers updates on vaccines as well as drugs for the prevention and management of monkeypox.
Collapse
|
14
|
Huang Y, Mu L, Wang W. Monkeypox: epidemiology, pathogenesis, treatment and prevention. Signal Transduct Target Ther 2022; 7:373. [PMID: 36319633 PMCID: PMC9626568 DOI: 10.1038/s41392-022-01215-4] [Citation(s) in RCA: 117] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/18/2022] [Accepted: 09/27/2022] [Indexed: 11/15/2022] Open
Abstract
Monkeypox is a zoonotic disease that was once endemic in west and central Africa caused by monkeypox virus. However, cases recently have been confirmed in many nonendemic countries outside of Africa. WHO declared the ongoing monkeypox outbreak to be a public health emergency of international concern on July 23, 2022, in the context of the COVID-19 pandemic. The rapidly increasing number of confirmed cases could pose a threat to the international community. Here, we review the epidemiology of monkeypox, monkeypox virus reservoirs, novel transmission patterns, mutations and mechanisms of viral infection, clinical characteristics, laboratory diagnosis and treatment measures. In addition, strategies for the prevention, such as vaccination of smallpox vaccine, is also included. Current epidemiological data indicate that high frequency of human-to-human transmission could lead to further outbreaks, especially among men who have sex with men. The development of antiviral drugs and vaccines against monkeypox virus is urgently needed, despite some therapeutic effects of currently used drugs in the clinic. We provide useful information to improve the understanding of monkeypox virus and give guidance for the government and relative agency to prevent and control the further spread of monkeypox virus.
Collapse
Affiliation(s)
- Yong Huang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Mu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Wang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
15
|
Harapan H, Ophinni Y, Megawati D, Frediansyah A, Mamada SS, Salampe M, Bin Emran T, Winardi W, Fathima R, Sirinam S, Sittikul P, Stoian AM, Nainu F, Sallam M. Monkeypox: A Comprehensive Review. Viruses 2022; 14:2155. [PMID: 36298710 PMCID: PMC9612348 DOI: 10.3390/v14102155] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 09/22/2022] [Accepted: 09/27/2022] [Indexed: 11/16/2022] Open
Abstract
The 2022 multi-country monkeypox outbreak in humans has brought new public health adversity on top of the ongoing coronavirus disease 2019 (COVID-19) pandemic. The disease has spread to 104 countries throughout six continents of the world, with the highest burden in North America and Europe. The etiologic agent, monkeypox virus (MPXV), has been known since 1959 after isolation from infected monkeys, and virulence among humans has been reported since the 1970s, mainly in endemic countries in West and Central Africa. However, the disease has re-emerged in 2022 at an unprecedented pace, with particular concern on its human-to-human transmissibility and community spread in non-endemic regions. As a mitigation effort, healthcare workers, public health policymakers, and the general public worldwide need to be well-informed on this relatively neglected viral disease. Here, we provide a comprehensive and up-to-date overview of monkeypox, including the following aspects: epidemiology, etiology, pathogenesis, clinical features, diagnosis, and management. In addition, the current review discusses the preventive and control measures, the latest vaccine developments, and the future research areas in this re-emerging viral disease that was declared as a public health emergency of international concern.
Collapse
Affiliation(s)
- Harapan Harapan
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
- Tropical Disease Centre, School of Medicine, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
- Department of Microbiology, School of Medicine, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
- Tsunami and Disaster Mitigation Research Center (TDMRC), Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
| | - Youdiil Ophinni
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0874, Japan
| | - Dewi Megawati
- Department of Veterinary Pathobiology, School of Veterinary Medicine, University of Missouri, Columbia, MO 65211, USA
- Department of Microbiology and Parasitology, School of Medicine, Universitas Warmadewa, Bali 80239, Indonesia
| | - Andri Frediansyah
- Research Group for Food Microbiology and Biotechnology, National Research and Innovation Agency (BRIN), Yogyakarta 55861, Indonesia
| | - Sukamto S. Mamada
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia
| | | | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
| | - Wira Winardi
- Department of Pulmonology and Respiratory Medicine, School of Medicine, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
| | - Raisha Fathima
- Medical Research Unit, School of Medicine, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
- Tropical Disease Centre, School of Medicine, Universitas Syiah Kuala, Banda Aceh 23111, Indonesia
| | - Salin Sirinam
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Pichamon Sittikul
- Department of Tropical Pediatrics, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Ana M. Stoian
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA 95616, USA
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar 90245, Indonesia
| | - Malik Sallam
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman 11942, Jordan
- Department of Translational Medicine, Faculty of Medicine, Lund University, 22184 Malmö, Sweden
| |
Collapse
|
16
|
Siegrist EA, Sassine J. Antivirals With Activity Against Mpox: A Clinically Oriented Review. Clin Infect Dis 2022; 76:155-164. [PMID: 35904001 PMCID: PMC9825831 DOI: 10.1093/cid/ciac622] [Citation(s) in RCA: 110] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/19/2022] [Accepted: 07/26/2022] [Indexed: 01/12/2023] Open
Abstract
Mpox virus is an emergent human pathogen. While it is less lethal than smallpox, it can still cause significant morbidity and mortality. In this review, we explore 3 antiviral agents with activity against mpox and other orthopoxviruses: cidofovir, brincidofovir, and tecovirimat. Cidofovir, and its prodrug brincidofovir, are inhibitors of DNA replication with a broad spectrum of activity against multiple families of double-stranded DNA viruses. Tecovirimat has more specific activity against orthopoxviruses and inhibits the formation of the extracellular enveloped virus necessary for cell-to-cell transmission. For each agent, we review basic pharmacology, data from animal models, and reported experience in human patients.
Collapse
Affiliation(s)
| | - Joseph Sassine
- Correspondence: J. Sassine, Infectious Diseases Section, Department of Medicine, The University of Oklahoma Health Sciences Center, 800 Stanton L. Young Blvd, Oklahoma City, OK 73104 ()
| |
Collapse
|
17
|
Lloyd M, Liu D, Lyu J, Fan J, Overhulse J, Kashemirov B, Prichard M, McKenna C, Moffat J. An acyclic phosphonate prodrug of HPMPC is effective against VZV in skin organ culture and mice. Antiviral Res 2022; 199:105275. [DOI: 10.1016/j.antiviral.2022.105275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/28/2022] [Accepted: 03/02/2022] [Indexed: 12/11/2022]
|
18
|
Garcia DR, Souza FR, Guimarães AP, Valis M, Pavelek Z, Kuca K, Ramalho TC, França TCC. In Silico Studies of Potential Selective Inhibitors of Thymidylate Kinase from Variola virus. Pharmaceuticals (Basel) 2021; 14:ph14101027. [PMID: 34681251 PMCID: PMC8537287 DOI: 10.3390/ph14101027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/22/2021] [Accepted: 09/30/2021] [Indexed: 11/17/2022] Open
Abstract
Continuing the work developed by our research group, in the present manuscript, we performed a theoretical study of 10 new structures derived from the antivirals cidofovir and ribavirin, as inhibitor prototypes for the enzyme thymidylate kinase from Variola virus (VarTMPK). The proposed structures were subjected to docking calculations, molecular dynamics simulations, and free energy calculations, using the molecular mechanics Poisson-Boltzmann surface area (MM-PBSA) method, inside the active sites of VarTMPK and human TMPK (HssTMPK). The docking and molecular dynamic studies pointed to structures 2, 3, 4, 6, and 9 as more selective towards VarTMPK. In addition, the free energy data calculated through the MM-PBSA method, corroborated these results. This suggests that these compounds are potential selective inhibitors of VarTMPK and, thus, can be considered as template molecules to be synthesized and experimentally evaluated against smallpox.
Collapse
Affiliation(s)
- Danielle R. Garcia
- Laboratory of Molecular Modeling Applied to Chemical and Biological Defense, Military Institute of Engineering, Praça General Tiburcio 80, Urca, Rio de Janeiro 22290-270, Brazil;
| | - Felipe R. Souza
- Department of Chemistry, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro 22541-041, Brazil;
| | - Ana P. Guimarães
- Department of Chemistry, Federal University of Viçosa, Avenida P. H. Rolfs, s/n, Centro, Viçosa 36570-000, MG, Brazil;
| | - Martin Valis
- Department of Neurology of the Medical Faculty of Charles University and University Hospital in Hradec Kralove, Sokolska 581, 50005 Hradec Kralove, Czech Republic; (M.V.); (Z.P.)
| | - Zbyšek Pavelek
- Department of Neurology of the Medical Faculty of Charles University and University Hospital in Hradec Kralove, Sokolska 581, 50005 Hradec Kralove, Czech Republic; (M.V.); (Z.P.)
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003 Hradec Kralove, Czech Republic;
- Biomedical Research Center, University Hospital in Hradec Kralove, Sokolska 581, 50005 Hradec Kralove, Czech Republic
- Correspondence: (K.K.); (T.C.C.F.)
| | - Teodorico C. Ramalho
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003 Hradec Kralove, Czech Republic;
- Laboratory of Computational Chemistry, Department of Chemistry, UFLA, Lavras 37200-000, MG, Brazil
| | - Tanos C. C. França
- Laboratory of Molecular Modeling Applied to Chemical and Biological Defense, Military Institute of Engineering, Praça General Tiburcio 80, Urca, Rio de Janeiro 22290-270, Brazil;
- Department of Chemistry, Faculty of Science, University of Hradec Kralove, Rokitanskeho 62, 50003 Hradec Kralove, Czech Republic;
- Correspondence: (K.K.); (T.C.C.F.)
| |
Collapse
|
19
|
Chan-Tack K, Harrington P, Bensman T, Choi SY, Donaldson E, O'Rear J, McMillan D, Myers L, Seaton M, Ghantous H, Cao Y, Valappil T, Birnkrant D, Struble K. Benefit-risk assessment for brincidofovir for the treatment of smallpox: U.S. Food and Drug Administration's Evaluation. Antiviral Res 2021; 195:105182. [PMID: 34582915 DOI: 10.1016/j.antiviral.2021.105182] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 11/18/2022]
Abstract
The development and approval of brincidofovir for the treatment of smallpox, a disease that was eradicated from the world over 40 years ago, has resulted in the second antiviral approved via the Medical Countermeasure Initiative (MCMi) to combat this disease. Approval of brincidofovir required a unique regulatory approach based on the FDA Animal Rule, and development was supported by many years of research and collaboration among academic investigators, the pharmaceutical industry and multiple government agencies. This article summarizes the FDA regulatory pathway and describes the challenges involved.
Collapse
Affiliation(s)
- Kirk Chan-Tack
- Division of Antivirals, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA.
| | - Patrick Harrington
- Division of Antivirals, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Timothy Bensman
- Office of Translational Sciences, Office of Clinical Pharmacology, Division of Clinical Pharmacology IV, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Su-Young Choi
- Office of Translational Sciences, Office of Clinical Pharmacology, Division of Clinical Pharmacology IV, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Eric Donaldson
- Division of Antivirals, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Julian O'Rear
- Division of Antivirals, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - David McMillan
- Division of Pharmacology/Toxicology for Infectious Diseases, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Laine Myers
- Division of Pharmacology/Toxicology for Infectious Diseases, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Mark Seaton
- Division of Pharmacology/Toxicology for Infectious Diseases, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Hanan Ghantous
- Division of Pharmacology/Toxicology for Infectious Diseases, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Yu Cao
- Division of Biometrics IV, Office of Biostatistics, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Thamban Valappil
- Division of Biometrics IV, Office of Biostatistics, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Debra Birnkrant
- Division of Antivirals, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| | - Kimberly Struble
- Division of Antivirals, Office of Infectious Diseases, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
20
|
Hutson CL, Kondas AV, Mauldin MR, Doty JB, Grossi IM, Morgan CN, Ostergaard SD, Hughes CM, Nakazawa Y, Kling C, Martin BE, Ellison JA, Carroll DS, Gallardo-Romero NF, Olson VA. Pharmacokinetics and Efficacy of a Potential Smallpox Therapeutic, Brincidofovir, in a Lethal Monkeypox Virus Animal Model. mSphere 2021; 6:e00927-20. [PMID: 33536322 PMCID: PMC7860987 DOI: 10.1128/msphere.00927-20] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 12/02/2020] [Indexed: 12/17/2022] Open
Abstract
Smallpox, caused by Variola virus (VARV), was eradicated in 1980; however, VARV bioterrorist threats still exist, necessitating readily available therapeutics. Current preparedness activities recognize the importance of oral antivirals and recommend therapeutics with different mechanisms of action. Monkeypox virus (MPXV) is closely related to VARV, causing a highly similar clinical human disease, and can be used as a surrogate for smallpox antiviral testing. The prairie dog MPXV model has been characterized and used to study the efficacy of antipoxvirus therapeutics, including recently approved TPOXX (tecovirimat). Brincidofovir (BCV; CMX001) has shown antiviral activity against double-stranded DNA viruses, including poxviruses. To determine the exposure of BCV following oral administration to prairie dogs, a pharmacokinetics (PK) study was performed. Analysis of BCV plasma concentrations indicated variability, conceivably due to the outbred nature of the animals. To determine BCV efficacy in the MPXV prairie dog model, groups of animals were intranasally challenged with 9 × 105 plaque-forming units (PFU; 90% lethal dose [LD90]) of MPXV on inoculation day 0 (ID0). Animals were divided into groups based on the first day of BCV treatment relative to inoculation day (ID-1, ID0, or ID1). A trend in efficacy was noted dependent upon treatment initiation (57% on ID-1, 43% on ID0, and 29% on ID1) but was lower than demonstrated in other animal models. Analysis of the PK data indicated that BCV plasma exposure (maximum concentration [Cmax]) and the time of the last quantifiable concentration (AUClast) were lower than in other animal models administered the same doses, indicating that suboptimal BCV exposure may explain the lower protective effect on survival.IMPORTANCE Preparedness activities against highly transmissible viruses with high mortality rates have been highlighted during the ongoing coronavirus disease 2019 (COVID-19) pandemic. Smallpox, caused by variola virus (VARV) infection, is highly transmissible, with an estimated 30% mortality. Through an intensive vaccination campaign, smallpox was declared eradicated in 1980, and routine smallpox vaccination of individuals ceased. Today's current population has little/no immunity against VARV. If smallpox were to reemerge, the worldwide results would be devastating. Recent FDA approval of one smallpox antiviral (tecovirimat) was a successful step in biothreat preparedness; however, orthopoxviruses can become resistant to treatment, suggesting the need for multiple therapeutics. Our paper details the efficacy of the investigational smallpox drug brincidofovir in a monkeypox virus (MPXV) animal model. Since brincidofovir has not been tested in vivo against smallpox, studies with the related virus MPXV are critical in understanding whether it would be protective in the event of a smallpox outbreak.
Collapse
Affiliation(s)
- Christina L Hutson
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Ashley V Kondas
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Mathew R Mauldin
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Jeffrey B Doty
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | | | - Clint N Morgan
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
- Oak Ridge Institute for Science and Education, CDC Fellowship Program, Oak Ridge, Tennessee, USA
| | | | - Christine M Hughes
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Yoshinori Nakazawa
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Chantal Kling
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Brock E Martin
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - James A Ellison
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Darin S Carroll
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Nadia F Gallardo-Romero
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Victoria A Olson
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| |
Collapse
|
21
|
Jockusch S, Tao C, Li X, Anderson TK, Chien M, Kumar S, Russo JJ, Kirchdoerfer RN, Ju J. A library of nucleotide analogues terminate RNA synthesis catalyzed by polymerases of coronaviruses that cause SARS and COVID-19. Antiviral Res 2020; 180:104857. [PMID: 32562705 PMCID: PMC7299870 DOI: 10.1016/j.antiviral.2020.104857] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 05/30/2020] [Accepted: 06/12/2020] [Indexed: 02/08/2023]
Abstract
SARS-CoV-2, a member of the coronavirus family, is responsible for the current COVID-19 worldwide pandemic. We previously demonstrated that five nucleotide analogues inhibit the SARS-CoV-2 RNA-dependent RNA polymerase (RdRp), including the active triphosphate forms of Sofosbuvir, Alovudine, Zidovudine, Tenofovir alafenamide and Emtricitabine. We report here the evaluation of a library of nucleoside triphosphate analogues with a variety of structural and chemical features as inhibitors of the RdRps of SARS-CoV and SARS-CoV-2. These features include modifications on the sugar (2' or 3' modifications, carbocyclic, acyclic, or dideoxynucleotides) or on the base. The goal is to identify nucleotide analogues that not only terminate RNA synthesis catalyzed by these coronavirus RdRps, but also have the potential to resist the viruses' exonuclease activity. We examined these nucleotide analogues for their ability to be incorporated by the RdRps in the polymerase reaction and to prevent further incorporation. While all 11 molecules tested displayed incorporation, 6 exhibited immediate termination of the polymerase reaction (triphosphates of Carbovir, Ganciclovir, Stavudine and Entecavir; 3'-OMe-UTP and Biotin-16-dUTP), 2 showed delayed termination (Cidofovir diphosphate and 2'-OMe-UTP), and 3 did not terminate the polymerase reaction (2'-F-dUTP, 2'-NH2-dUTP and Desthiobiotin-16-UTP). The coronaviruses possess an exonuclease that apparently requires a 2'-OH at the 3'-terminus of the growing RNA strand for proofreading. In this study, all nucleoside triphosphate analogues evaluated form Watson-Crick-like base pairs. The nucleotide analogues demonstrating termination either lack a 2'-OH, have a blocked 2'-OH, or show delayed termination. Thus, these nucleotide analogues are of interest for further investigation to evaluate whether they can evade the viral exonuclease activity. Prodrugs of five of these nucleotide analogues (Cidofovir, Abacavir, Valganciclovir/Ganciclovir, Stavudine and Entecavir) are FDA-approved medications for treatment of other viral infections, and their safety profiles are well established. After demonstrating potency in inhibiting viral replication in cell culture, candidate molecules can be rapidly evaluated as potential therapies for COVID-19.
Collapse
Affiliation(s)
- Steffen Jockusch
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemistry, Columbia University, New York, NY, 10027, USA
| | - Chuanjuan Tao
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Xiaoxu Li
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Thomas K Anderson
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Institute of Molecular Virology, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Minchen Chien
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Shiv Kumar
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - James J Russo
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA
| | - Robert N Kirchdoerfer
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, 53706, USA; Institute of Molecular Virology, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Jingyue Ju
- Center for Genome Technology and Biomolecular Engineering, Columbia University, New York, NY, 10027, USA; Department of Chemical Engineering, Columbia University, New York, NY, 10027, USA; Department of Pharmacology, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
22
|
Bugert JJ, Hucke F, Zanetta P, Bassetto M, Brancale A. Antivirals in medical biodefense. Virus Genes 2020; 56:150-167. [PMID: 32076918 PMCID: PMC7089181 DOI: 10.1007/s11262-020-01737-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 01/20/2020] [Indexed: 02/07/2023]
Abstract
The viruses historically implicated or currently considered as candidates for misuse in bioterrorist events are poxviruses, filoviruses, bunyaviruses, orthomyxoviruses, paramyxoviruses and a number of arboviruses causing encephalitis, including alpha- and flaviviruses. All these viruses are of concern for public health services when they occur in natural outbreaks or emerge in unvaccinated populations. Recent events and intelligence reports point to a growing risk of dangerous biological agents being used for nefarious purposes. Public health responses effective in natural outbreaks of infectious disease may not be sufficient to deal with the severe consequences of a deliberate release of such agents. One important aspect of countermeasures against viral biothreat agents are the antiviral treatment options available for use in post-exposure prophylaxis. These issues were adressed by the organizers of the 16th Medical Biodefense Conference, held in Munich in 2018, in a special session on the development of drugs to treat infections with viruses currently perceived as a threat to societies or associated with a potential for misuse as biothreat agents. This review will outline the state-of-the-art methods in antivirals research discussed and provide an overview of antiviral compounds in the pipeline that are already approved for use or still under development.
Collapse
Affiliation(s)
- J J Bugert
- Bundeswehr Institute for Microbiology, Neuherbergstraße 11, 80937, Munich, Germany.
| | - F Hucke
- Bundeswehr Institute for Microbiology, Neuherbergstraße 11, 80937, Munich, Germany
| | - P Zanetta
- Bundeswehr Institute for Microbiology, Neuherbergstraße 11, 80937, Munich, Germany
| | - M Bassetto
- Department of Chemistry, Swansea University, Swansea, SA2 8PP, UK
| | - A Brancale
- Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, CF10 3NB, UK
| |
Collapse
|
23
|
Sheean ME, Malikova E, Duarte D, Capovilla G, Fregonese L, Hofer MP, Magrelli A, Mariz S, Mendez-Hermida F, Nistico R, Leest T, Sipsas NV, Tsigkos S, Vitezic D, Larsson K, Sepodes B, Stoyanova-Beninska V. Nonclinical data supporting orphan medicinal product designations in the area of rare infectious diseases. Drug Discov Today 2019; 25:274-291. [PMID: 31704277 DOI: 10.1016/j.drudis.2019.10.015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/15/2019] [Accepted: 10/30/2019] [Indexed: 01/13/2023]
Abstract
This review provides an overview of nonclinical in vivo models that can be used to support orphan designation in selected rare infectious diseases in Europe, with the aim to inform and stimulate the planning of nonclinical development in this area of often neglected diseases.
Collapse
Affiliation(s)
- Maria E Sheean
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands; Max-Delbrück Center for Molecular Medicine in Helmholz Association, Berlin, Germany.
| | - Eva Malikova
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; State Institute for Drug Control, Bratislava, Slovak Republic; Comenius University, Department of Pharmacology and Toxicology, Bratislava, Slovak Republic
| | - Dinah Duarte
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; INFARMED - Autoridade Nacional do Medicamento, Lisbon, Portugal
| | - Giuseppe Capovilla
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; C. Poma Hospital, Mantova, Italy; Fondazione Poliambulanza, Brescia, Italy
| | - Laura Fregonese
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Matthias P Hofer
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Armando Magrelli
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; National Center for Drug Research and Evaluation, Istituto Superiore di Sanità, Rome, Italy
| | - Segundo Mariz
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Fernando Mendez-Hermida
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; Agencia Española de Medicamentos y Productos Sanitarios, Madrid, Spain
| | - Robert Nistico
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; Malta Medicines Authority, San Ġwann, Malta
| | - Tim Leest
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; The Federal Agency for Medicines and Health Products, Brussels, Belgium
| | - Nikolaos V Sipsas
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Stelios Tsigkos
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Dinko Vitezic
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; University of Rijeka Medical School and University Hospital Centre Rijeka, Rijeka, Croatia
| | - Kristina Larsson
- Orphan Medicines Office, European Medicines Agency, Amsterdam, The Netherlands
| | - Bruno Sepodes
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; INFARMED - Autoridade Nacional do Medicamento, Lisbon, Portugal; Universidade de Lisboa - Faculdade de Farmácia, Lisbon, Portugal
| | - Violeta Stoyanova-Beninska
- Committee of Orphan Medicinal Products, European Medicines Agency, Amsterdam, The Netherlands; Medicines Evaluation Board, Utrecht, The Netherlands
| |
Collapse
|
24
|
Rodrigues Garcia D, Rodrigues de Souza F, Paula Guimarães A, Castro Ramalho T, Palermo de Aguiar A, Celmar Costa França T. Design of inhibitors of thymidylate kinase from Variola virus as new selective drugs against smallpox: part II. J Biomol Struct Dyn 2019; 37:4569-4579. [PMID: 30488769 PMCID: PMC9491145 DOI: 10.1080/07391102.2018.1554510] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/21/2023]
Abstract
Acknowledging the importance of studies toward the development of measures against terrorism and bioterrorism, this study aims to contribute to the design of new prototypes of potential drugs against smallpox. Based on a former study, nine synthetic feasible prototypes of selective inhibitors for thymidylate kinase from Variola virus (VarTMPK) were designed and submitted to molecular docking, molecular dynamics simulations and binding energy calculations. The compounds are simplifications of two more complex scaffolds, with a guanine connected to an amide or alcohol through a spacer containing ether and/or amide groups, formerly suggested as promising for the design of selective inhibitors of VarTMPK. Our study showed that, despite the structural simplifications, the compounds presented effective energy values in interactions with VarTMPK and HssTMPK and that the guanine could be replaced by a simpler imidazole ring linked to a -NH2 group, without compromising the affinity for VarTMPK. It was also observed that a positive charge in the imidazole ring is important for the selectivity toward VarTMPK and that an amide group in the spacer does not contribute to selectivity. Finally, prototype 3 was pointed as the most promising to be synthesized and experimentally evaluated. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Danielle Rodrigues Garcia
- Laboratory of Molecular Modeling Applied to Chemical and Biological Defense, Military Institute of Engineering, Rio de Janeiro, RJ, Brazil
| | - Felipe Rodrigues de Souza
- Laboratory of Molecular Modeling Applied to Chemical and Biological Defense, Military Institute of Engineering, Rio de Janeiro, RJ, Brazil
| | | | - Teodorico Castro Ramalho
- Laboratory of Computational Chemistry, Department of Chemistry, UFLA, Lavras, MG, Brazil
- Faculty of Informatics and Management, Center for Basic and Applied Research, University of Hradec Králové, Hradec Králove, Czech Republic
| | | | - Tanos Celmar Costa França
- Laboratory of Molecular Modeling Applied to Chemical and Biological Defense, Military Institute of Engineering, Rio de Janeiro, RJ, Brazil
- Faculty of Informatics and Management, Center for Basic and Applied Research, University of Hradec Králové, Hradec Králove, Czech Republic
| |
Collapse
|
25
|
Conrad SJ, Liu J. Poxviruses as Gene Therapy Vectors: Generating Poxviral Vectors Expressing Therapeutic Transgenes. Methods Mol Biol 2019; 1937:189-209. [PMID: 30706397 DOI: 10.1007/978-1-4939-9065-8_11] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Treatments with poxvirus vectors can have long-lasting immunological impact in the host, and thus they have been extensively studied to treat diseases and for vaccine development. More importantly, the oncolytic properties of poxviruses have led to their development as cancer therapeutics. Two poxviruses, vaccinia virus (VACV) and myxoma virus (MYXV), have been extensively studied as virotherapeutics with promising results. Vaccinia virus vectors have advanced to the clinic and have been tested as oncolytic therapeutics for several cancer types with successes in phase I/II clinical trials. In addition to oncolytic applications, MYXV has been explored for additional applications including immunotherapeutics, purging of cancer progenitor cells, and treatments for graft-versus-host diseases. These novel therapeutic applications have encouraged its advancement into clinical trials. To meet the demands of different treatment needs, VACV and MYXV can be genetically engineered to express therapeutic transgenes. The engineering process used in poxvirus vectors can be very different from that of other DNA virus vectors (e.g., the herpesviruses). This chapter is intended to serve as a guide to those wishing to engineer poxvirus vectors for therapeutic transgene expression and to produce viral preparations for preclinical studies.
Collapse
Affiliation(s)
- Steven J Conrad
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA
| | - Jia Liu
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences (UAMS), Little Rock, AR, USA. .,The Center for Microbial Pathogenesis and Host Inflammatory Responses, University of Arkansas for Medical Sciences, Little Rock, AR, USA.
| |
Collapse
|
26
|
Ananthula HK, Parker S, Touchette E, Buller RM, Patel G, Kalman D, Salzer JS, Gallardo-Romero N, Olson V, Damon IK, Moir-Savitz T, Sallans L, Werner MH, Sherwin CM, Desai PB. Preclinical pharmacokinetic evaluation to facilitate repurposing of tyrosine kinase inhibitors nilotinib and imatinib as antiviral agents. BMC Pharmacol Toxicol 2018; 19:80. [PMID: 30514402 PMCID: PMC6278073 DOI: 10.1186/s40360-018-0270-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Accepted: 11/13/2018] [Indexed: 11/28/2022] Open
Abstract
Background Several tyrosine kinase inhibitors (TKIs) developed as anti-cancer drugs, also have anti-viral activity due to their ability to disrupt productive replication and dissemination in infected cells. Consequently, such drugs are attractive candidates for “repurposing” as anti-viral agents. However, clinical evaluation of therapeutics against infectious agents associated with high mortality, but low or infrequent incidence, is often unfeasible. The United States Food and Drug Administration formulated the “Animal Rule” to facilitate use of validated animal models for conducting anti-viral efficacy studies. Methods To enable such efficacy studies of two clinically approved TKIs, nilotinib, and imatinib, we first conducted comprehensive pharmacokinetic (PK) studies in relevant rodent and non-rodent animal models. PK of these agents following intravenous and oral dosing were evaluated in C57BL/6 mice, prairie dogs, guinea pigs and Cynomolgus monkeys. Plasma samples were analyzed using an LC-MS/MS method. Secondarily, we evaluated the utility of allometry-based inter-species scaling derived from previously published data to predict the PK parameters, systemic clearance (CL) and the steady state volume of distribution (Vss) of these two drugs in prairie dogs, an animal model not tested thus far. Results Marked inter-species variability in PK parameters and resulting oral bioavailability was observed. In general, elimination half-lives of these agents in mice and guinea pigs were much shorter (1–3 h) relative to those in larger species such as prairie dogs and monkeys. The longer nilotinib elimination half-life in prairie dogs (i.v., 6.5 h and oral, 7.5 h), facilitated multiple dosing PK and safety assessment. The allometry-based predicted values of the Vss and CL were within 2.0 and 2.5-fold, respectively, of the observed values. Conclusions Our results suggest that prairie dogs and monkeys may be suitable rodent and non-rodent species to perform further efficacy testing of these TKIs against orthopoxvirus infections. The use of rodent models such as C57BL/6 mice and guinea pigs for assessing pre-clinical anti-viral efficacy of these two TKIs may be limited due to short elimination and/or low oral bioavailability. Allometry-based correlations, derived from existing literature data, may provide initial estimates, which may serve as a useful guide for pre-clinical PK studies in untested animal models.
Collapse
Affiliation(s)
| | - Scott Parker
- Department of Molecular Microbiology and Immunology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Erin Touchette
- Department of Molecular Microbiology and Immunology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - R Mark Buller
- Department of Molecular Microbiology and Immunology, School of Medicine, Saint Louis University, St. Louis, MO, USA
| | - Gopi Patel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Daniel Kalman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | - Victoria Olson
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Inger K Damon
- Centers for Disease Control and Prevention, Atlanta, GA, USA
| | | | - Larry Sallans
- Mass Spectrometry Facility, University of Cincinnati, Cincinnati, OH, USA
| | | | - Catherine M Sherwin
- Division Clinical Pharmacology, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Pankaj B Desai
- James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
27
|
Perry MR, Warren R, Merchlinsky M, Houchens C, Rogers JV. Rabbitpox in New Zealand White Rabbits: A Therapeutic Model for Evaluation of Poxvirus Medical Countermeasures Under the FDA Animal Rule. Front Cell Infect Microbiol 2018; 8:356. [PMID: 30345258 PMCID: PMC6182097 DOI: 10.3389/fcimb.2018.00356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/18/2018] [Indexed: 11/18/2022] Open
Abstract
The elimination of smallpox as an endemic disease and the obvious ethical problems with clinical challenge requires the efficacy evaluation of medical countermeasures against smallpox using the FDA Animal Rule. This approach requires the evaluation of antiviral efficacy in an animal model whose infection recapitulates the human disease sufficiently well enough to provide predictive value of countermeasure effectiveness. The narrow host range of variola virus meant that no other animal species was sufficiently susceptible to variola to manifest a disease with predictive value. To address this dilemma, the FDA, after a public forum with virologists in December 2011, suggested the development of two animal models infected with the cognate orthopoxvirus, intradermal infection of rabbits and intranasal infection of mice, to supplement the non-human primate models in use. In this manuscript, we describe the development of an intradermal challenge model of New Zealand White rabbits with rabbitpox virus (RPXV) for poxvirus countermeasure evaluation. Lethality of RPXV was demonstrated in both 9 and 16-weeks old rabbits with an LD50 < 10 PFU. The natural history of RPXV infection was documented in both ages of rabbits by monitoring the time to onset of abnormal values in clinical data at a lethal challenge of 300 PFU. All infected animals became viremic, developed a fever, exhibited weight loss, developed secondary lesions, and were euthanized after 7 or 8 days. The 16-weeks RPXV-infected animals exhibiting similar clinical signs with euthanasia applied about a day later than for 9-weeks old rabbits. For all animals, the first two unambiguous indicators of infection were detection of viral copies by quantitative polymerase chain reaction and fever at 2 and 3 days following challenge, respectively. These biomarkers provide clinically-relevant trigger(s) for initiating therapy. The major advantage for using 16-weeks NZW rabbits is that older rabbits were more robust and less subject to stress-induced death allowing more reproducible studies.
Collapse
Affiliation(s)
- Mark R Perry
- Battelle, Biomedical Research Center, West Jefferson, OH, United States
| | - Richard Warren
- U.S. Department of Health and Human Services, Biomedical Advanced Research and Development Authority, Washington, DC, United States
| | - Michael Merchlinsky
- U.S. Department of Health and Human Services, Biomedical Advanced Research and Development Authority, Washington, DC, United States
| | - Christopher Houchens
- U.S. Department of Health and Human Services, Biomedical Advanced Research and Development Authority, Washington, DC, United States
| | | |
Collapse
|
28
|
A Randomized, Double-Blind, Placebo-Controlled Phase 3 Trial of Oral Brincidofovir for Cytomegalovirus Prophylaxis in Allogeneic Hematopoietic Cell Transplantation. Biol Blood Marrow Transplant 2018; 25:369-381. [PMID: 30292744 PMCID: PMC8196624 DOI: 10.1016/j.bbmt.2018.09.038] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 09/27/2018] [Indexed: 12/16/2022]
Abstract
Cytomegalovirus (CMV) infection is a common complication of allogeneic hematopoietic cell transplantation (HCT). In this trial, we randomized adult CMV-seropositive HCT recipients without CMV viremia at screening 2:1 to receive brincidofovir or placebo until week 14 post-HCT. Randomization was stratified by center and risk of CMV infection. Patients were assessed weekly through week 15 and every third week thereafter through week 24 post-HCT. Patients who developed clinically significant CMV infection (CS-CMVi; CMV viremia requiring preemptive therapy or CMV disease) discontinued the study drug and began anti-CMV treatment. The primary endpoint was the proportion of patients with CS-CMVi through week 24 post-HCT; patients who discontinued the trial or with missing data were imputed as primary endpoint events. Between August 2013 and June 2015, 452 patients were randomized at a median of 15 days after HCT and received study drug. The proportion of patients who developed CS-CMVi or were imputed as having a primary endpoint event through week 24 was similar between brincidofovir-treated patients and placebo recipients (155 of 303 [51.2%] versus 78 of 149 [52.3%]; odds ratio, .95 [95% confidence interval, .64 to 1.41]; P = .805); fewer brincidofovir recipients developed CMV viremia through week 14 compared with placebo recipients (41.6%; P < .001). Serious adverse events were more frequent among brincidofovir recipients (57.1% versus 37.6%), driven by acute graft-versus-host disease (32.3% versus 6.0%) and diarrhea (6.9% versus 2.7%). Week 24 all-cause mortality was 15.5% among brincidofovir recipients and 10.1% among placebo recipients. Brincidofovir did not reduce CS-CMVi by week 24 post-HCT and was associated with gastrointestinal toxicity.
Collapse
|
29
|
Koban R, Neumann M, Daugs A, Bloch O, Nitsche A, Langhammer S, Ellerbrok H. A novel three-dimensional cell culture method enhances antiviral drug screening in primary human cells. Antiviral Res 2018; 150:20-29. [DOI: 10.1016/j.antiviral.2017.12.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/20/2017] [Accepted: 12/06/2017] [Indexed: 12/12/2022]
|
30
|
Narayanan N, Lacy CR, Cruz JE, Nahass M, Karp J, Barone JA, Hermes-DeSantis ER. Disaster Preparedness: Biological Threats and Treatment Options. Pharmacotherapy 2018; 38:217-234. [PMID: 29236288 DOI: 10.1002/phar.2068] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Biological disasters can be natural, accidental, or intentional. Biological threats have made a lasting impact on civilization. This review focuses on agents of clinical significance, bioterrorism, and national security, specifically Category A agents (anthrax, botulism, plague, tularemia, and smallpox), as well as briefly discusses other naturally emerging infections of public health significance, Ebola virus (also a Category A agent) and Zika virus. The role of pharmacists in disaster preparedness and disaster response is multifaceted and important. Their expertise includes clinical knowledge, which can aid in drug information consultation, patient-specific treatment decision making, and development of local treatment plans. To fulfill this role, pharmacists must have a comprehensive understanding of medical countermeasures for these significant biological threats across all health care settings. New and reemerging infectious disease threats will continue to challenge the world. Pharmacists will be at the forefront of preparedness and response, sharing knowledge and clinical expertise with responders, official decision makers, and the general public.
Collapse
Affiliation(s)
- Navaneeth Narayanan
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, New Jersey.,Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey
| | - Clifton R Lacy
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey.,Rutgers School of Communication and Information, New Brunswick, New Jersey
| | - Joseph E Cruz
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Department of Pharmacy, Englewood Hospital and Medical Center, Englewood, New Jersey
| | - Meghan Nahass
- Alpert Medical School, Brown University, Providence, Rhode Island
| | - Jonathan Karp
- University of Vermont (at the time of writing), Burlington, Vermont
| | - Joseph A Barone
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| | - Evelyn R Hermes-DeSantis
- Department of Pharmacy Practice and Administration, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, New Jersey.,Department of Pharmacy, Robert Wood Johnson University Hospital, New Brunswick, New Jersey
| |
Collapse
|
31
|
Delaune D, Iseni F, Ferrier-Rembert A, Peyrefitte CN, Ferraris O. The French Armed Forces Virology Unit: A Chronological Record of Ongoing Research on Orthopoxvirus. Viruses 2017; 10:E3. [PMID: 29295488 PMCID: PMC5795416 DOI: 10.3390/v10010003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/20/2017] [Accepted: 12/21/2017] [Indexed: 01/04/2023] Open
Abstract
Since the official declaration of smallpox eradication in 1980, the general population vaccination has ceased worldwide. Therefore, people under 40 year old are generally not vaccinated against smallpox and have no cross protection against orthopoxvirus infections. This naïve population may be exposed to natural or intentional orthopoxvirus emergences. The virology unit of the Institut de Recherche Biomédicale des Armées (France) has developed research programs on orthopoxviruses since 2000. Its missions were conceived to improve the diagnosis capabilities, to foster vaccine development, and to develop antivirals targeting specific viral proteins. The role of the virology unit was asserted in 2012 when the responsibility of the National Reference Center for the Orthopoxviruses was given to the unit. This article presents the evolution of the unit activity since 2000, and the past and current research focusing on orthopoxviruses.
Collapse
Affiliation(s)
- Déborah Delaune
- Unité de virologie, Centre National de Référence-Laboratoire Expert Orthopoxvirus, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France.
| | - Frédéric Iseni
- Unité de virologie, Centre National de Référence-Laboratoire Expert Orthopoxvirus, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France.
| | - Audrey Ferrier-Rembert
- Unité de virologie, Centre National de Référence-Laboratoire Expert Orthopoxvirus, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France.
| | - Christophe N Peyrefitte
- Unité de virologie, Centre National de Référence-Laboratoire Expert Orthopoxvirus, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France.
| | - Olivier Ferraris
- Unité de virologie, Centre National de Référence-Laboratoire Expert Orthopoxvirus, Institut de Recherche Biomédicale des Armées, 91220 Brétigny-sur-Orge, France.
| |
Collapse
|
32
|
Foster SA, Parker S, Lanier R. The Role of Brincidofovir in Preparation for a Potential Smallpox Outbreak. Viruses 2017; 9:v9110320. [PMID: 29773767 PMCID: PMC5707527 DOI: 10.3390/v9110320] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Revised: 10/20/2017] [Accepted: 10/26/2017] [Indexed: 11/16/2022] Open
Abstract
Smallpox (variola) virus is considered a Category A bioterrorism agent due to its ability to spread rapidly and the high morbidity and mortality rates associated with infection. Current recommendations recognize the importance of oral antivirals and call for having at least two smallpox antivirals with different mechanisms of action available in the event of a smallpox outbreak. Multiple antivirals are recommended due in large part to the propensity of viruses to become resistant to antiviral therapy, especially monotherapy. Advances in synthetic biology heighten concerns that a bioterror attack with variola would utilize engineered resistance to antivirals and potentially vaccines. Brincidofovir, an oral antiviral in late stage development, has proven effective against orthopoxviruses in vitro and in vivo, has a different mechanism of action from tecovirimat (the only oral smallpox antiviral currently in the US Strategic National Stockpile), and has a resistance profile that reduces concerns in the scenario of a bioterror attack using genetically engineered smallpox. Given the devastating potential of smallpox as a bioweapon, preparation of a multi-pronged defense that accounts for the most obvious bioengineering possibilities is strategically imperative.
Collapse
Affiliation(s)
| | - Scott Parker
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, St. Louis, MO 63104, USA.
| | | |
Collapse
|
33
|
Olson VA, Shchelkunov SN. Are We Prepared in Case of a Possible Smallpox-Like Disease Emergence? Viruses 2017; 9:E242. [PMID: 32962316 PMCID: PMC5618008 DOI: 10.3390/v9090242] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 08/22/2017] [Accepted: 08/23/2017] [Indexed: 12/16/2022] Open
Abstract
Smallpox was the first human disease to be eradicated, through a concerted vaccination campaign led by the World Health Organization. Since its eradication, routine vaccination against smallpox has ceased, leaving the world population susceptible to disease caused by orthopoxviruses. In recent decades, reports of human disease from zoonotic orthopoxviruses have increased. Furthermore, multiple reports of newly identified poxviruses capable of causing human disease have occurred. These facts raise concerns regarding both the opportunity for these zoonotic orthopoxviruses to evolve and become a more severe public health issue, as well as the risk of Variola virus (the causative agent of smallpox) to be utilized as a bioterrorist weapon. The eradication of smallpox occurred prior to the development of the majority of modern virological and molecular biological techniques. Therefore, there is a considerable amount that is not understood regarding how this solely human pathogen interacts with its host. This paper briefly recounts the history and current status of diagnostic tools, vaccines, and anti-viral therapeutics for treatment of smallpox disease. The authors discuss the importance of further research to prepare the global community should a smallpox-like virus emerge.
Collapse
Affiliation(s)
- Victoria A. Olson
- Poxvirus and Rabies Branch, Division of High Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA
| | - Sergei N. Shchelkunov
- Department of Genomic Research and Development of DNA Diagnostics of Poxviruses, State Research Center of Virology and Biotechnology VECTOR, Koltsovo, 630559 Novosibirsk Region, Russia
- Department of Molecular Biology, Novosibirsk State University, 630090 Novosibirsk, Russia
| |
Collapse
|
34
|
Grossi IM, Foster SA, Gainey MR, Krile RT, Dunn JA, Brundage T, Khouri JM. Efficacy of delayed brincidofovir treatment against a lethal rabbitpox virus challenge in New Zealand White rabbits. Antiviral Res 2017; 143:278-286. [PMID: 28392420 DOI: 10.1016/j.antiviral.2017.04.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/04/2017] [Indexed: 10/19/2022]
Abstract
In the event of a bioterror attack with variola virus (smallpox), exposure may only be identified following onset of fever. To determine if antiviral therapy with brincidofovir (BCV; CMX001) initiated at, or following, onset of fever could prevent severe illness and death, a lethal rabbitpox model was used. BCV is in advanced development as an antiviral for the treatment of smallpox under the US Food and Drug Administration's 'Animal Rule'. This pivotal study assessed the efficacy of immediate versus delayed treatment with BCV following onset of symptomatic disease in New Zealand White rabbits intradermally inoculated with a lethal rabbitpox virus (RPXV), strain Utrecht. Infected rabbits with confirmed fever were randomized to blinded treatment with placebo, BCV, or BCV delayed by 24, 48, or 72 h. The primary objective evaluated the survival benefit with BCV treatment. The assessment of reduction in the severity and progression of clinical events associated with RPXV were secondary objectives. Clinically and statistically significant reductions in mortality were observed when BCV was initiated up to 48 h following the onset of fever; survival rates were 100%, 93%, and 93% in the immediate treatment, 24-h, and 48-h delayed treatment groups, respectively, versus 48% in the placebo group (p < 0.05 for each vs. placebo). Significant improvements in clinical and virologic parameters were also observed. These findings provide a scientific rationale for therapeutic intervention with BCV in the event of a smallpox outbreak when vaccination is contraindicated or when diagnosis follows the appearance of clinical signs and symptoms.
Collapse
|
35
|
Short-term clinical safety profile of brincidofovir: A favorable benefit-risk proposition in the treatment of smallpox. Antiviral Res 2017; 143:269-277. [PMID: 28093339 DOI: 10.1016/j.antiviral.2017.01.009] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 01/06/2017] [Accepted: 01/11/2017] [Indexed: 11/20/2022]
Abstract
Brincidofovir (BCV, CMX001) is an orally available, long-acting, broad-spectrum antiviral that has been evaluated in healthy subjects in Phase I studies and in hematopoietic cell transplant recipients and other immunocompromised patients in Phase II/III clinical trials for the prevention and treatment of cytomegalovirus and adenovirus infections. BCV has also shown in vitro activity against orthopoxviruses such as variola (smallpox) virus, and is under advanced development as a treatment for smallpox under the US FDA's 'Animal Rule'. The anticipated treatment regimen for smallpox is a total weekly dose of 200 mg administered orally for 3 consecutive weeks. To assess the benefit-to-risk profile of BCV for the treatment of smallpox, we evaluated short-term safety data associated with comparable doses from Phase I studies and from adult and pediatric subjects in the cytomegalovirus and adenovirus clinical programs. When administered at doses and durations similar to that proposed for the treatment of smallpox, BCV was generally well tolerated in both adults and pediatric subjects. The most common adverse events were mild gastrointestinal events and asymptomatic, transient, and reversible elevations in serum transaminases. The data presented herein indicate a favorable safety profile for BCV for the treatment of smallpox, and support its continued development for this indication.
Collapse
|
36
|
Tippin TK, Morrison ME, Brundage TM, Momméja-Marin H. Brincidofovir Is Not a Substrate for the Human Organic Anion Transporter 1: A Mechanistic Explanation for the Lack of Nephrotoxicity Observed in Clinical Studies. Ther Drug Monit 2016; 38:777-786. [PMID: 27851688 PMCID: PMC5113238 DOI: 10.1097/ftd.0000000000000353] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/05/2016] [Indexed: 01/02/2023]
Abstract
BACKGROUND Brincidofovir (BCV) is an orally bioavailable lipid conjugate of cidofovir (CDV) with increased in vitro potency relative to CDV against all 5 families of double-stranded DNA viruses that cause human disease. After intravenous (IV) administration of CDV, the organic anion transporter 1 (OAT1) transports CDV from the blood into the renal proximal tubule epithelial cells with resulting dose-limiting nephrotoxicity. OBJECTIVE To study whether OAT1 transports BCV and to evaluate the pharmacokinetic and renal safety profile of oral BCV compared with IV CDV. METHODS The cellular uptake of BCV and its major metabolites was assessed in vitro. Renal function at baseline and during and after treatment in subjects in BCV clinical studies was examined. RESULTS In OAT1-expressing cells, uptake of BCV and its 2 major metabolites (CMX103 and CMX064) was the same as in mock-transfected control cells and was not inhibited by the OAT inhibitor probenecid. In human pharmacokinetic studies, BCV administration at therapeutic doses resulted in detection of CDV as a circulating metabolite; peak CDV plasma concentrations after oral BCV administration in humans were <1% of those observed after IV CDV administration at therapeutic doses. Analysis of renal function and adverse events from 3 BCV clinical studies in immunocompromised adult and pediatric subjects indicated little to no evidence of associated nephrotoxicity. Over 80% of subjects who switched from CDV or foscarnet to BCV experienced an improvement in renal function as measured by maximum on-treatment estimated glomerular filtration rate. CONCLUSIONS The lack of BCV uptake through OAT1, together with lower CDV concentrations after oral BCV compared with IV CDV administration, likely explains the superior renal safety profile observed in immunocompromised subjects receiving BCV compared with CDV.
Collapse
|
37
|
Dunning J, Kennedy SB, Antierens A, Whitehead J, Ciglenecki I, Carson G, Kanapathipillai R, Castle L, Howell-Jones R, Pardinaz-Solis R, Grove J, Scott J, Lang T, Olliaro P, Horby PW, for the RAPIDE-BCV trial team. Experimental Treatment of Ebola Virus Disease with Brincidofovir. PLoS One 2016; 11:e0162199. [PMID: 27611077 PMCID: PMC5017617 DOI: 10.1371/journal.pone.0162199] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/15/2016] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND The nucleotide analogue brincidofovir was developed to prevent and treat infections caused by double-stranded DNA viruses. Based on in vitro data suggesting an antiviral effect against Ebola virus, brincidofovir was included in the World Health Organisation list of agents that should be prioritised for clinical evaluation in patients with Ebola virus disease (EVD) during the West African epidemic. METHODS AND FINDINGS In this single-arm phase 2 trial conducted in Liberia, patients with laboratory-confirmed EVD (two months of age or older, enrolment bodyweight ≥50 kg) received oral brincidofovir 200 mg as a loading dose on day 0, followed by 100 mg brincidofovir on days 3, 7, 10, and 14. Bodyweight-adjusted dosing was used for patients weighing <50 kg at enrolment. The primary outcome was survival at Day 14 after the first dose of brincidofovir. Four patients were enrolled between 01 January 2015 and 31 January 2015. The trial was stopped following the decision by the manufacturer to terminate their program of development of brincidofovir for EVD. No Serious Adverse Reactions or Suspected Unexpected Serious Adverse Reactions were identified. All enrolled subjects died of an illness consistent with EVD. CONCLUSIONS Due to the small sample size it was not possible to determine the efficacy of brincidofovir for the treatment of EVD. The premature termination of the trial highlights the need to establish better practices for preclinical in-vitro and animal screening of therapeutics for potentially emerging epidemic infectious diseases prior to their use in patients. TRIAL REGISTRATION Pan African Clinical Trials Registry PACTR201411000939962.
Collapse
Affiliation(s)
- Jake Dunning
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Stephen B. Kennedy
- University of Liberia-Pacific Institute for Research and Evaluation (UL-PIRE) Africa Center, University of Liberia, Monrovia, Liberia
| | | | - John Whitehead
- Department of Mathematics and Statistics, Lancaster University, Lancaster, United Kingdom
| | - Iza Ciglenecki
- Médecins Sans Frontières, Operational Center, Geneva, Switzerland
| | - Gail Carson
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Lyndsey Castle
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Rebecca Howell-Jones
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Raul Pardinaz-Solis
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Jennifer Grove
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Janet Scott
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Trudie Lang
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | - Piero Olliaro
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
- UNICEF-UNDP-World Bank-WHO Special Programme for Research and Training in Tropical Diseases, Geneva, Switzerland
| | - Peter W. Horby
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
38
|
Thornton PJ, Kadri H, Miccoli A, Mehellou Y. Nucleoside Phosphate and Phosphonate Prodrug Clinical Candidates. J Med Chem 2016; 59:10400-10410. [DOI: 10.1021/acs.jmedchem.6b00523] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Peter J. Thornton
- School
of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
- School
of Chemistry, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Hachemi Kadri
- School
of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Ageo Miccoli
- School
of Chemistry, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| | - Youcef Mehellou
- School
of Pharmacy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
- School
of Chemistry, College of Engineering and Physical Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, U.K
| |
Collapse
|