1
|
Hetrick B, Siddiqui S, Spear M, Guo J, Liang H, Fu Y, Yang Z, Doyle-Meyers L, Pahar B, Veazey RS, Dufour J, Andalibi A, Ling B, Wu Y. Suppression of viral rebound by a Rev-dependent lentiviral particle in SIV-infected rhesus macaques. Gene Ther 2025; 32:16-24. [PMID: 39025983 PMCID: PMC11785524 DOI: 10.1038/s41434-024-00467-9] [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: 01/21/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Persistence of human immunodeficiency virus (HIV) reservoirs prevents viral eradication, and consequently HIV-infected patients require lifetime treatment with antiretroviral therapy (ART) [1-5]. Currently, there are no effective therapeutics to prevent HIV rebound upon ART cessation. Here we describe an HIV/SIV Rev-dependent lentiviral particle that can be administered to inhibit viral rebound [6-9]. Using simian immunodeficiency virus (SIV)-infected rhesus macaques as a model, we demonstrate that the administration of pre-assembled SIV Rev-dependent lentiviral particles into SIVmac239-infected Indian rhesus macaques can lead to reduction of viral rebound upon ART termination. One of the injected animals, KC50, controlled plasma and CNS viremia to an undetectable level most of the time for over two years after ART termination. Surprisingly, detailed molecular and immunological characterization revealed that viremia control was concomitant with the induction of neutralizing antibodies (nAbs) following the administration of the Rev-dependent vectors. This study emphasizes the importance of neutralizing antibodies (nAbs) for viremia control [10-15], and also provides proof of concept that the Rev-dependent vector can be used to target viral reservoirs, including the CNS reservoirs, in vivo. However, future large-scale in vivo studies are needed to understand the potential mechanisms of viremia control induced by the Rev-dependent vector.
Collapse
Affiliation(s)
- Brian Hetrick
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Summer Siddiqui
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Mark Spear
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Jia Guo
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Huizhi Liang
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Yajing Fu
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Zhijun Yang
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Lara Doyle-Meyers
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Bapi Pahar
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, MD, USA
| | - Ronald S Veazey
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Jason Dufour
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
| | - Ali Andalibi
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA
| | - Binhua Ling
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA, 70433, USA
- Host-Pathogen Interaction Program, Texas Biomedical Research Institute, 8715 W Military Dr., San Antonio, TX, 78227, USA
| | - Yuntao Wu
- Center for Infectious Disease Research, George Mason University, Manassas, VA, 20110, USA.
| |
Collapse
|
2
|
Tucci AR, da Rosa RM, Rosa AS, Augusto Chaves O, Ferreira VNS, Oliveira TKF, Coutinho Souza DD, Borba NRR, Dornelles L, Rocha NS, Mayer JCP, da Rocha JBT, Rodrigues OED, Miranda MD. Antiviral Effect of 5'-Arylchalcogeno-3-aminothymidine Derivatives in SARS-CoV-2 Infection. Molecules 2023; 28:6696. [PMID: 37764472 PMCID: PMC10537738 DOI: 10.3390/molecules28186696] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 09/04/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The understanding that zidovudine (ZDV or azidothymidine, AZT) inhibits the RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 and that chalcogen atoms can increase the bioactivity and reduce the toxicity of AZT has directed our search for the discovery of novel potential anti-coronavirus compounds. Here, the antiviral activity of selenium and tellurium containing AZT derivatives in human type II pneumocytes cell model (Calu-3) and monkey kidney cells (Vero E6) infected with SARS-CoV-2, and their toxic effects on these cells, was evaluated. Cell viability analysis revealed that organoselenium (R3a-R3e) showed lower cytotoxicity than organotellurium (R3f, R3n-R3q), with CC50 ≥ 100 µM. The R3b and R3e were particularly noteworthy for inhibiting viral replication in both cell models and showed better selectivity index. In Vero E6, the EC50 values for R3b and R3e were 2.97 ± 0.62 µM and 1.99 ± 0.42 µM, respectively, while in Calu-3, concentrations of 3.82 ± 1.42 µM and 1.92 ± 0.43 µM (24 h treatment) and 1.33 ± 0.35 µM and 2.31 ± 0.54 µM (48 h) were observed, respectively. The molecular docking calculations were carried out to main protease (Mpro), papain-like protease (PLpro), and RdRp following non-competitive, competitive, and allosteric inhibitory approaches. The in silico results suggested that the organoselenium is a potential non-competitive inhibitor of RdRp, interacting in the allosteric cavity located in the palm region. Overall, the cell-based results indicated that the chalcogen-zidovudine derivatives were more potent than AZT in inhibiting SARS-CoV-2 replication and that the compounds R3b and R3e play an important inhibitory role, expanding the knowledge about the promising therapeutic capacity of organoselenium against COVID-19.
Collapse
Affiliation(s)
- Amanda Resende Tucci
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Raquel Mello da Rosa
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Alice Santos Rosa
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Otávio Augusto Chaves
- CQC-IMS, Departamento de Química, Universidade de Coimbra, Rua Larga, 3004-535 Coimbra, Portugal
- Laboratório de Imunofarmacologia, Centro de Pesquisa, Inovação e Vigilância em COVID-19 e Emergências Sanitárias (CPIV), Instituto Oswaldo Cruz (IOC), Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-900, RJ, Brazil
| | - Vivian Neuza Santos Ferreira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
| | - Thamara Kelcya Fonseca Oliveira
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Daniel Dias Coutinho Souza
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| | - Nathalia Roberto Resende Borba
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
| | - Luciano Dornelles
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Nayra Salazar Rocha
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - João Candido Pilar Mayer
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - João B. Teixeira da Rocha
- Programa de Pós-Graduação em Bioquímica Toxicológica, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil;
| | - Oscar Endrigo D. Rodrigues
- LabSelen-NanoBio—Departamento de Química, Universidade Federal de Santa Maria, Santa Maria 97105-900, RS, Brazil; (R.M.d.R.); (L.D.); (N.S.R.); (J.C.P.M.)
| | - Milene Dias Miranda
- Laboratório de Morfologia e Morfogênese Viral, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil; (A.R.T.); (A.S.R.); (V.N.S.F.); (T.K.F.O.); (D.D.C.S.); (N.R.R.B.)
- Programa de Pós-Graduação em Biologia Celular e Molecular, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro 21041-250, RJ, Brazil
| |
Collapse
|
3
|
Drug Repurposing Approaches to Combating Viral Infections. J Clin Med 2020; 9:jcm9113777. [PMID: 33238464 PMCID: PMC7700377 DOI: 10.3390/jcm9113777] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 11/18/2020] [Accepted: 11/19/2020] [Indexed: 12/14/2022] Open
Abstract
Development of novel antiviral molecules from the beginning costs an average of $350 million to $2 billion per drug, and the journey from the laboratory to the clinic takes about 10–15 years. Utilization of drug repurposing approaches has generated substantial interest in order to overcome these drawbacks. A drastic reduction in the failure rate, which otherwise is ~92%, is achieved with the drug repurposing approach. The recent exploration of the drug repurposing approach to combat the COVID-19 pandemic has further validated the fact that it is more beneficial to reinvestigate the in-practice drugs for a new application instead of designing novel drugs. The first successful example of drug repurposing is zidovudine (AZT), which was developed as an anti-cancer agent in the 1960s and was later approved by the US FDA as an anti-HIV therapeutic drug in the late 1980s after fast track clinical trials. Since that time, the drug repurposing approach has been successfully utilized to develop effective therapeutic strategies against a plethora of diseases. Hence, an extensive application of the drug repurposing approach will not only help to fight the current pandemics more efficiently but also predict and prepare for newly emerging viral infections. In this review, we discuss in detail the drug repurposing approach and its advancements related to viral infections such as Human Immunodeficiency Virus (HIV) and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2).
Collapse
|
4
|
Gomes CB, Balaguez RA, Larroza A, Smaniotto TA, Domingues M, Casaril AM, Silva MS, Rodrigues OED, Savegnago L, Alves D. Organocatalysis in the Synthesis of 1,2,3‐Triazoyl‐zidovudine Derivatives: Synthesis and Preliminary Antioxidant Activity. ChemistrySelect 2020. [DOI: 10.1002/slct.202003355] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Carolina B. Gomes
- LASOL-CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Renata A. Balaguez
- LASOL-CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Allya Larroza
- LASOL-CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Thiago A. Smaniotto
- Programa de Pós-Graduação em Biotecnologia (PPGB) Universidade Federal de Pelotas - UFPel Pelotas RS Brazil
| | - Micaela Domingues
- Programa de Pós-Graduação em Biotecnologia (PPGB) Universidade Federal de Pelotas - UFPel Pelotas RS Brazil
| | - Angela M. Casaril
- Programa de Pós-Graduação em Biotecnologia (PPGB) Universidade Federal de Pelotas - UFPel Pelotas RS Brazil
| | - Márcio S. Silva
- LASOL-CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| | - Oscar E. D. Rodrigues
- LabSelen-NanoBio - Departamento de Química Universidade Federal de Santa Maria UFSM 97115-900 Santa Maria RS Brazil
| | - Lucielli Savegnago
- Programa de Pós-Graduação em Biotecnologia (PPGB) Universidade Federal de Pelotas - UFPel Pelotas RS Brazil
| | - Diego Alves
- LASOL-CCQFA Universidade Federal de Pelotas - UFPel P.O. Box 354 96010-900 Pelotas RS Brazil
| |
Collapse
|
5
|
Low ZY, Farouk IA, Lal SK. Drug Repositioning: New Approaches and Future Prospects for Life-Debilitating Diseases and the COVID-19 Pandemic Outbreak. Viruses 2020; 12:E1058. [PMID: 32972027 PMCID: PMC7551028 DOI: 10.3390/v12091058] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/02/2020] [Accepted: 08/21/2020] [Indexed: 02/06/2023] Open
Abstract
Traditionally, drug discovery utilises a de novo design approach, which requires high cost and many years of drug development before it reaches the market. Novel drug development does not always account for orphan diseases, which have low demand and hence low-profit margins for drug developers. Recently, drug repositioning has gained recognition as an alternative approach that explores new avenues for pre-existing commercially approved or rejected drugs to treat diseases aside from the intended ones. Drug repositioning results in lower overall developmental expenses and risk assessments, as the efficacy and safety of the original drug have already been well accessed and approved by regulatory authorities. The greatest advantage of drug repositioning is that it breathes new life into the novel, rare, orphan, and resistant diseases, such as Cushing's syndrome, HIV infection, and pandemic outbreaks such as COVID-19. Repositioning existing drugs such as Hydroxychloroquine, Remdesivir, Ivermectin and Baricitinib shows good potential for COVID-19 treatment. This can crucially aid in resolving outbreaks in urgent times of need. This review discusses the past success in drug repositioning, the current technological advancement in the field, drug repositioning for personalised medicine and the ongoing research on newly emerging drugs under consideration for the COVID-19 treatment.
Collapse
Affiliation(s)
- Zheng Yao Low
- School of Science, Monash University, Bandar Sunway, Subang Jaya 47500, Selangor Darul Ehsan, Malaysia; (Z.Y.L.); (I.A.F.)
| | - Isra Ahmad Farouk
- School of Science, Monash University, Bandar Sunway, Subang Jaya 47500, Selangor Darul Ehsan, Malaysia; (Z.Y.L.); (I.A.F.)
| | - Sunil Kumar Lal
- School of Science, Monash University, Bandar Sunway, Subang Jaya 47500, Selangor Darul Ehsan, Malaysia; (Z.Y.L.); (I.A.F.)
- Tropical Medicine & Biology Platform, Monash University, Subang Jaya 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
6
|
Ellinger B, Pohlmann D, Woens J, Jäkel FM, Reinshagen J, Stocking C, Prassolov VS, Fehse B, Riecken K. A High-Throughput HIV-1 Drug Screening Platform, Based on Lentiviral Vectors and Compatible with Biosafety Level-1. Viruses 2020; 12:E580. [PMID: 32466195 PMCID: PMC7290285 DOI: 10.3390/v12050580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 11/17/2022] Open
Abstract
HIV-1 infection is a complex, multi-step process involving not only viral, but also multiple cellular factors. To date, drug discovery methods have primarily focused on the inhibition of single viral proteins. We present an efficient and unbiased approach, compatible with biosafety level 1 (BSL-1) conditions, to identify inhibitors of HIV-1 reverse transcription, intracellular trafficking, nuclear entry and genome integration. Starting with a fluorescent assay setup, we systematically improved the screening methodology in terms of stability, efficiency and pharmacological relevance. Stability and throughput were optimized by switching to a luciferase-based readout. BSL-1 compliance was achieved without sacrificing pharmacological relevance by using lentiviral particles pseudo-typed with the mouse ecotropic envelope protein to transduce human PM1 T cells gene-modified to express the corresponding murine receptor. The cellular assay was used to screen 26,048 compounds selected for maximum diversity from a 200,640-compound in-house library. This yielded z' values greater than 0.8 with a hit rate of 3.3% and a confirmation rate of 50%. We selected 93 hits and enriched the collection with 279 similar compounds from the in-house library to identify promising structural features. The most active compounds were validated using orthogonal assay formats. The similarity of the compound profiles across the different platforms demonstrated that the reported lentiviral assay system is a robust and versatile tool for the identification of novel HIV-1 inhibitors.
Collapse
Affiliation(s)
- Bernhard Ellinger
- Department ScreeningPort, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 22525 Hamburg, Germany; (B.E.); (J.R.)
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Partner site Hamburg, 22525 Hamburg, Germany
| | - Daniel Pohlmann
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.P.); (J.W.); (F.M.J.); (C.S.)
| | - Jannis Woens
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.P.); (J.W.); (F.M.J.); (C.S.)
| | - Felix M. Jäkel
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.P.); (J.W.); (F.M.J.); (C.S.)
| | - Jeanette Reinshagen
- Department ScreeningPort, Fraunhofer Institute for Molecular Biology and Applied Ecology IME, 22525 Hamburg, Germany; (B.E.); (J.R.)
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases CIMD, Partner site Hamburg, 22525 Hamburg, Germany
| | - Carol Stocking
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.P.); (J.W.); (F.M.J.); (C.S.)
- Heinrich-Pette-Institute, Leibniz Institute for Experimental Virology, 20251 Hamburg, Germany
| | - Vladimir S. Prassolov
- Engelhardt-Institute of Molecular Biology, Russian Academy of Sciences, 117984 Moscow, Russia;
| | - Boris Fehse
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.P.); (J.W.); (F.M.J.); (C.S.)
- German Center for Infection Research (DZIF), Partner site Hamburg, 20246 Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf (UKE), 20246 Hamburg, Germany; (D.P.); (J.W.); (F.M.J.); (C.S.)
| |
Collapse
|
7
|
Abrams RPM, Solis J, Nath A. Therapeutic Approaches for Zika Virus Infection of the Nervous System. Neurotherapeutics 2017; 14:1027-1048. [PMID: 28952036 PMCID: PMC5722777 DOI: 10.1007/s13311-017-0575-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Zika virus has spread rapidly in the Americas and has caused devastation of human populations affected in these regions. The virus causes teratogenic effects involving the nervous system, and in adults and children can cause a neuropathy similar to Guillain-Barré syndrome, an anterior myelitis, or, rarely, an encephalitis. While major efforts have been undertaken to control mosquito populations that spread the virus and to develop a vaccine, drug development that directly targets the virus in an infected individual to prevent or treat the neurological manifestations is necessary. Rational and targeted drug development is possible since the viral life cycle and the structure of the key viral proteins are now well understood. While several groups have identified therapeutic candidates, their approaches differ in the types of screening processes and viral assays used. Animal studies are available for only a few compounds. Here we provide an exhaustive review and compare each of the classes of drugs discovered, the methods used for drug discovery, and their potential use in humans for the prevention or treatment of neurological complications of Zika virus infection.
Collapse
Affiliation(s)
- Rachel P M Abrams
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Jamie Solis
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Avindra Nath
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
Denner J. Can Antiretroviral Drugs Be Used to Treat Porcine Endogenous Retrovirus (PERV) Infection after Xenotransplantation? Viruses 2017; 9:v9080213. [PMID: 28786944 PMCID: PMC5580470 DOI: 10.3390/v9080213] [Citation(s) in RCA: 42] [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: 07/06/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 01/09/2023] Open
Abstract
Porcine endogenous retroviruses (PERVs) are integrated in the genome of all pigs; they are released as infectious particles, and under certain conditions they can infect human cells. Therefore, they represent a risk when pigs are used as sources of cells, tissues, or organs for xenotransplantation. Xenotransplantation is under development due to the increasing shortage of human transplants. Whereas most porcine microorganisms which may be able to induce a disease (zoonosis) in the transplant recipient can be eliminated, this is not possible in the case of PERVs. Antiretroviral drugs which had been developed for the treatment of human immunodeficiency virus-1 (HIV-1) infections have been tested in vitro for their efficacy in inhibiting PERV replication. Inhibitors of the viral reverse transcriptase and of the integrase have been found effective. The most effective inhibitor of the reverse transcriptase was azidothymidine (AZT); the integrase inhibitors were the most potent inhibitors of PERV. Although in the past PERV transmission has not been observed after experimental or clinical xenotransplantation of pig cells or organs, and although PERVs may one day be inactivated in pigs by genome editing using CRISPR/Cas, knowing which antiretroviral drugs can effectively restrict PERV infection will still be important.
Collapse
Affiliation(s)
- Joachim Denner
- Robert Koch Fellow, Robert Koch Institute, Berlin, Germany.
| |
Collapse
|
9
|
Allen SJW, Krawczyk SH, McGee LR, Bischofberger N, Mulato AS, Cherrington JM. Inhibition of HIV-1 RNase H Activity by Nucleotide Dimers and Monomers. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/095632029600700107] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Nucleotide dimers and monomers were shown to inhibit human immunodeficiency virus type 1 (HIV) RNase H activity. Several effective inhibitors were identified and placed into three general groups based on biochemical characterization of their inhibition, The first group (group A) inhibited HIV RNase H and the closely related feline immunodeficiency virus (FIV) RNase H, but did not inhibit less related retroviral or cellular RNases H or HIV reverse transcriptase (RT). The second group (group B) inhibited the RNase H activity of several retroviruses as well as the reverse transcriptase function of HIV RT. The third group (group C) inhibited RNases H from retroviral and cellular sources but did not inhibit HIV RT. Kinetic analyses of HIV RNase H inhibition were conducted and all three types of inhibitors exhibited a competitive mode of inhibition with regard to substrate. The small nucleotides described here represent the most potent (Ki values from 0.57 to 16 μM) and selective inhibitors of HIV RNase H reported to date. Further structure - function analyses of these molecules may lead to the discovery of unique, potent antiretroviral therapeutics.
Collapse
Affiliation(s)
- S. J. W. Allen
- Gilead Sciences, Inc., 346 Lakeside Dr., Foster City, CA. 94404, USA
| | - S. H. Krawczyk
- Gilead Sciences, Inc., 346 Lakeside Dr., Foster City, CA. 94404, USA
| | - L. R. McGee
- Gilead Sciences, Inc., 346 Lakeside Dr., Foster City, CA. 94404, USA
| | - N. Bischofberger
- Gilead Sciences, Inc., 346 Lakeside Dr., Foster City, CA. 94404, USA
| | - A. S. Mulato
- Gilead Sciences, Inc., 346 Lakeside Dr., Foster City, CA. 94404, USA
| | - J. M. Cherrington
- Gilead Sciences, Inc., 346 Lakeside Dr., Foster City, CA. 94404, USA
| |
Collapse
|
10
|
Gogineni V, Schinazi RF, Hamann MT. Role of Marine Natural Products in the Genesis of Antiviral Agents. Chem Rev 2015; 115:9655-706. [PMID: 26317854 PMCID: PMC4883660 DOI: 10.1021/cr4006318] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Vedanjali Gogineni
- Department of Pharmacognosy, Pharmacology, Chemistry & Biochemistry, University of Mississippi, School of Pharmacy, University, Mississippi 38677, United States
| | - Raymond F. Schinazi
- Center for AIDS Research, Department of Pediatrics, Emory University/Veterans Affairs Medical Center, 1760 Haygood Drive NE, Atlanta, Georgia 30322, United States
| | - Mark T. Hamann
- Department of Pharmacognosy, Pharmacology, Chemistry & Biochemistry, University of Mississippi, School of Pharmacy, University, Mississippi 38677, United States
| |
Collapse
|
11
|
Metallocene-uracil conjugates: Synthesis and biological evaluation of novel mono-, di- and tri-nuclear systems. J Organomet Chem 2015. [DOI: 10.1016/j.jorganchem.2014.11.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
12
|
Yamamoto M, Onogi H, Kii I, Yoshida S, Iida K, Sakai H, Abe M, Tsubota T, Ito N, Hosoya T, Hagiwara M. CDK9 inhibitor FIT-039 prevents replication of multiple DNA viruses. J Clin Invest 2014; 124:3479-88. [PMID: 25003190 DOI: 10.1172/jci73805] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 05/23/2014] [Indexed: 01/09/2023] Open
Abstract
A wide range of antiviral drugs is currently available; however, drug-resistant viruses have begun to emerge and represent a potential public health risk. Here, we explored the use of compounds that inhibit or interfere with the action of essential host factors to prevent virus replication. In particular, we focused on the cyclin-dependent kinase 9 (CDK9) inhibitor, FIT-039, which suppressed replication of a broad spectrum of DNA viruses through inhibition of mRNA transcription. Specifically, FIT-039 inhibited replication of herpes simplex virus 1 (HSV-1), HSV-2, human adenovirus, and human cytomegalovirus in cultured cells, and topical application of FIT-039 ointment suppressed skin legion formation in a murine HSV-1 infection model. FIT-039 did not affect cell cycle progression or cellular proliferation in host cells. Compared with the general CDK inhibitor flavopiridol, transcriptome analyses of FIT-039-treated cells revealed that FIT-039 specifically inhibited CDK9. Given at concentrations above the inhibitory concentration, FIT-039 did not have a cytotoxic effect on mammalian cells. Importantly, administration of FIT-039 ameliorated the severity of skin lesion formation in mice infected with an acyclovir-resistant HSV-1, without noticeable adverse effects. Together, these data indicate that FIT-039 has potential as an antiviral agent for clinical therapeutics.
Collapse
MESH Headings
- Acyclovir/pharmacology
- Adenoviruses, Human/drug effects
- Adenoviruses, Human/physiology
- Animals
- Antiviral Agents/chemistry
- Antiviral Agents/pharmacology
- Antiviral Agents/toxicity
- Cyclin-Dependent Kinase 9/antagonists & inhibitors
- Cytomegalovirus/drug effects
- Cytomegalovirus/physiology
- DNA Viruses/drug effects
- DNA Viruses/genetics
- DNA Viruses/physiology
- Disease Models, Animal
- Drug Resistance, Viral
- Flavonoids/pharmacology
- HEK293 Cells
- HeLa Cells
- Herpes Simplex/drug therapy
- Herpes Simplex/pathology
- Herpes Simplex/virology
- Herpesvirus 1, Human/drug effects
- Herpesvirus 1, Human/physiology
- Herpesvirus 2, Human/drug effects
- Herpesvirus 2, Human/physiology
- Host-Pathogen Interactions/drug effects
- Host-Pathogen Interactions/genetics
- Humans
- Mice
- Mice, Inbred ICR
- Piperidines/pharmacology
- Protein Kinase Inhibitors/chemistry
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/toxicity
- Pyridines/chemistry
- Pyridines/pharmacology
- Pyridines/toxicity
- Rats
- Rats, Wistar
- Transcription, Genetic/drug effects
- Transcriptome/drug effects
- Virus Replication/drug effects
Collapse
|
13
|
Ziegler BE, Marta RA, Burt MB, Martens SM, Martens JK, McMahon TB. Structural investigation of protonated azidothymidine and protonated dimer. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2014; 25:176-185. [PMID: 24306778 DOI: 10.1007/s13361-013-0767-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 09/29/2013] [Accepted: 10/07/2013] [Indexed: 06/02/2023]
Abstract
Infrared multiple photon dissociation (IRMPD) spectroscopy experiments and quantum chemical calculations have been used to explore the possible structures of protonated azidothymidine and the corresponding protonated dimer. Many interesting differences between the protonated and neutral forms of azidothymidine were found, particularly associated with keto-enol tautomerization. Comparison of computational vibrational and the experimental IMRPD spectra show good agreement and give confidence that the dominant protonated species has been identified. The protonated dimer of azidothymidine exhibits three intramolecular hydrogen bonds. The IRMPD spectrum of the protonated dimer is consistent with the spectrum of the most stable computational structure. This work brings to light interesting keto-enol tautomerization and exocyclic hydrogen bonding involving azidothymidine and its protonated dimer. The fact that one dominant protonated species is observed in the gas phase, despite both the keto and enol structures being similar in energy, is proposed to be the direct result of the electrospray ionization process in which the dominant protonated dimer structure dissociates in the most energetically favorable way.
Collapse
Affiliation(s)
- Blake E Ziegler
- Department of Chemistry, University of Waterloo, Waterloo, Ontario, N2L 3G1, Canada
| | | | | | | | | | | |
Collapse
|
14
|
Reynolds C, de Koning CB, Pelly SC, van Otterlo WAL, Bode ML. In search of a treatment for HIV--current therapies and the role of non-nucleoside reverse transcriptase inhibitors (NNRTIs). Chem Soc Rev 2012; 41:4657-70. [PMID: 22618809 DOI: 10.1039/c2cs35058k] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The human immunodeficiency virus (HIV) causes AIDS (acquired immune deficiency syndrome), a disease in which the immune system progressively deteriorates, making sufferers vulnerable to all manner of opportunistic infections. Currently, world-wide there are estimated to be 34 million people living with HIV, with the vast majority of these living in sub-Saharan Africa. Therefore, an important research focus is development of new drugs that can be used in the treatment of HIV/AIDS. This review gives an overview of the disease and addresses the drugs currently used for treatment, with specific emphasis on new developments within the class of allosteric non-nucleoside reverse transcriptase inhibitors (NNRTIs).
Collapse
Affiliation(s)
- Chevonne Reynolds
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Private Bag 3, PO WITS, 2050, South Africa
| | | | | | | | | |
Collapse
|
15
|
Castro AA, Aucélio RQ, Rey NA, Miguel EM, Farias PAM. Determination of the Antiretroviral Drug Zidovudine in Diluted Alkaline Electrolyte by Adsorptive Stripping Voltammetry at the Mercury Film Electrode. ACTA ACUST UNITED AC 2011. [DOI: 10.4236/ajac.2011.22027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
16
|
Stocking C, Grez M, Fehse B, von Laer D, Itoh K, Prassolov V, Nowock J, Kühlcke K, Just U, Schröder T, Klump H, Schiedlmeier B, Grassman E, Meyer J, Li Z, Schambach A, Modlich U, Kustikova O, Galla M, Bode J, Zander A, Baum C. Cell and virus genetics at the roots of gene therapy, retrovirology, and hematopoietic stem cell biology: Wolfram Ostertag (1937-2010). Hum Gene Ther 2010; 21:1501-3. [PMID: 21091034 DOI: 10.1089/hum.2010.1901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
17
|
Leandro KC, Moreira JC, Farias PAM. Determination of Zidovudine in Pharmaceuticals by Differential Pulse Voltammetry. ANAL LETT 2010. [DOI: 10.1080/00032711003687021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
18
|
Chromatographic separation of antiviral/anticancer nucleoside reverse transcriptase inhibitor drugs. Anal Bioanal Chem 2010; 398:295-312. [PMID: 20585942 DOI: 10.1007/s00216-010-3917-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 05/28/2010] [Accepted: 06/09/2010] [Indexed: 12/23/2022]
Abstract
This paper discusses the current methods used for quantitative determination of analogues of nucleotide reverse transcriptase inhibitors (NtRTIs) in body fluids, cells, and tissues. Nucleoside reverse transcriptase inhibitors (NRTIs) prodrugs given to AIDS/herpes/cancer patients conjugate with phosphates at the site of their action. Separation of phosphorylated NRTIs is generally performed by reversed-phase chromatography. After separation, plasma NRTIs can be detected using a variety of methods, including immunoassay through monitoring of UV absorbance, fluorescence, and mass spectrometry. The most recent development in the field of detection of plasma NtRTIs shows a tendency toward the use double- or triple-focusing mass spectrometry, the most specific and sensitive monitoring technique.
Collapse
|
19
|
Broder S. The development of antiretroviral therapy and its impact on the HIV-1/AIDS pandemic. Antiviral Res 2010; 85:1-18. [PMID: 20018391 PMCID: PMC2815149 DOI: 10.1016/j.antiviral.2009.10.002] [Citation(s) in RCA: 311] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Revised: 07/31/2009] [Accepted: 10/10/2009] [Indexed: 12/21/2022]
Abstract
In the last 25 years, HIV-1, the retrovirus responsible for the acquired immunodeficiency syndrome (AIDS), has gone from being an "inherently untreatable" infectious agent to one eminently susceptible to a range of approved therapies. During a five-year period, starting in the mid-1980s, my group at the National Cancer Institute played a role in the discovery and development of the first generation of antiretroviral agents, starting in 1985 with Retrovir (zidovudine, AZT) in a collaboration with scientists at the Burroughs-Wellcome Company (now GlaxoSmithKline). We focused on AZT and related congeners in the dideoxynucleoside family of nucleoside reverse transcriptase inhibitors (NRTIs), taking them from the laboratory to the clinic in response to the pandemic of AIDS, then a terrifying and lethal disease. These drugs proved, above all else, that HIV-1 infection is treatable, and such proof provided momentum for new therapies from many sources, directed at a range of viral targets, at a pace that has rarely if ever been matched in modern drug development. Antiretroviral therapy has brought about a substantial decrease in the death rate due to HIV-1 infection, changing it from a rapidly lethal disease into a chronic manageable condition, compatible with very long survival. This has special implications within the classic boundaries of public health around the world, but at the same time in certain regions may also affect a cycle of economic and civil instability in which HIV-1/AIDS is both cause and consequence. Many challenges remain, including (1) the life-long duration of therapy; (2) the ultimate role of pre-exposure prophylaxis (PrEP); (3) the cardiometabolic side-effects or other toxicities of long-term therapy; (4) the emergence of drug-resistance and viral genetic diversity (non-B subtypes); (5) the specter of new cross-species transmissions from established retroviral reservoirs in apes and Old World monkeys; and (6) the continued pace of new HIV-1 infections in many parts of the world. All of these factors make refining current therapies and developing new therapeutic paradigms essential priorities, topics covered in articles within this special issue of Antiviral Research. Fortunately, there are exciting new insights into the biology of HIV-1, its interaction with cellular resistance factors, and novel points of attack for future therapies. Moreover, it is a short journey from basic research to public health benefit around the world. The current science will lead to new therapeutic strategies with far-reaching implications in the HIV-1/AIDS pandemic. This article forms part of a special issue of Antiviral Research marking the 25th anniversary of antiretroviral drug discovery and development, Vol. 85, issue 1, 2010.
Collapse
Affiliation(s)
- Samuel Broder
- Celera Corporation, 1401 Harbor Bay Pkwy, Alameda, CA 94502-7070, USA.
| |
Collapse
|
20
|
Revoltella RP, Trombi L, Petrini M, Grassi B, Manara G, Mese ED. Low-Frequency Electromagnetic Fields Do Not Affect Cell Growth, Erythroid Differentiation, and Virus Production in Variant Lines of Untreated and Dimethyl Sulfoxide-Treated Friend Erythroleukemia Cells. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/15368379309012868] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
21
|
|
22
|
Jagetia GC, Aruna R. Correlation of micronuclei-induction with the cell survival in HeLa cells treated with a base analogue, azidothymidine (AZT) before exposure to different doses of gamma-radiation. Toxicol Lett 2003; 139:33-43. [PMID: 12595156 DOI: 10.1016/s0378-4274(02)00439-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The effect of 0.1 microM azidothymidine (AZT) a pyrimidine analogue has been studied on the growth kinetics, cell survival and micronuclei formation in HeLa cells exposed to 0, 0.25, 0.5, 1, 2 and 3 Gy of 60Co gamma-radiation. The AZT pretreatment resulted in a significant decline in the cell growth kinetics, cell survival and cell proliferation indices when compared with the PBS+irradiation group at 20, 30 and 40 h post-irradiation. Conversely, the frequency of micronucleated binucleate cells (MNBNC) elevated in a dose dependent manner in both PBS+irradiation and AZT+irradiation group. This elevation in MNBNC-induction was significantly higher in the latter when compared with the former group at all post-irradiation scoring time periods studied. The dose-response relationship for micronuclei induction for both the PBS+irrradiation and AZT+irradiation groups was linear. The biological response was studied by correlating the cell survival with MNBNC-induction. The cell survival and MNBNC-induction showed a close but inverse relationship and this relationship gave a best fit on the linear quadratic model.
Collapse
|
23
|
Vogt B, Roscher S, Abel B, Hildinger M, Lamarre A, Baum C, von Laer D. Lack of superinfection interference in retroviral vector producer cells. Hum Gene Ther 2001; 12:359-65. [PMID: 11242528 DOI: 10.1089/10430340150503984] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Vesicular stomatitis virus G protein (VSV-G)-pseudotyped retroviral vectors have become more feasible for clinical gene transfer protocols since stable tetracycline (tet)-regulated packaging cell lines have become available. Here, we analyzed superinfection interference in VSV-G-pseudotyped and classic amphotropic packaging cell lines. No superinfection interference was observed in VSV-G-pseudotyped packaging cell lines. Thus, integrated retroviral vector genomes accumulated during culture. Similar results were obtained with the amphotropic packaging cells, but to a lesser degree. In addition, VSV-G packaging cells were susceptible to infection with vector particles devoid of envelope proteins, which are produced by these cells in high titers when VSV-G expression is suppressed by tetracycline. For both packaging systems, superinfection could be blocked by azidothymidine (AZT). With regard to safety, this study suggests that in clinical protocols amphotropic producer clones should be tested for superinfection interference and VSV-G packaging cells should always be cultured in the presence of AZT.
Collapse
Affiliation(s)
- B Vogt
- Heinrich-Pette-Institute für Experimentelle Virologie und Immunologie, Universität Hamburg, 20251 Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
24
|
|
25
|
Ott DE, Hewes SM, Alvord WG, Henderson LE, Arthur LO. Inhibition of Friend virus replication by a compound that reacts with the nucleocapsid zinc finger: anti-retroviral effect demonstrated in vivo. Virology 1998; 243:283-92. [PMID: 9568028 DOI: 10.1006/viro.1998.9062] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The zinc finger structure that is found in the nucleocapsid protein of nearly all retroviruses has been proposed as a target for antiviral therapy. Since compounds that chemically attack the cysteines of the finger have been shown to inactivate both human immunodeficiency virus type 1 (HIV-1) and murine leukemia virus (MuLV) in vitro, 14 of these compounds were tested in an MuLV-induced Friend disease model to assess their ability to inhibit retroviral replication in vivo. Of the 14 compounds tested, only Aldrithiol-2 clearly exhibited anti-retroviral activity as measured indirectly by the delay of Friend disease onset (P < 0.05). These results were confirmed by quantitative competitive polymerase chain reaction studies which monitored viral spread by measuring the level of viral DNA in the peripheral blood mononuclear cells of treated mice. Comparison of treated mice with untreated mice revealed that Aldrithiol-2 produced a greater than 2-log reduction in virus levels. These results functionally demonstrate that a zinc finger-attacking compound can inhibit viral replication in vivo. Since only 1 of the 14 compounds studied was effective, this study also shows the importance of in vivo testing of these types of antiviral compounds in an animal model. Given the strict conservation of the metal-coordinating cysteine structure within HIV-1 and MuLV zinc fingers, our results support the proposal that anti-retroviral drugs which target the nucleocapsid zinc finger may be clinically useful against HIV-1.
Collapse
Affiliation(s)
- D E Ott
- AIDS Vaccine Program, SAIC/Frederick, National Cancer Institute, Maryland 21702-1201, USA.
| | | | | | | | | |
Collapse
|
26
|
Arts EJ, Wainberg MA. Mechanisms of nucleoside analog antiviral activity and resistance during human immunodeficiency virus reverse transcription. Antimicrob Agents Chemother 1996; 40:527-40. [PMID: 8851566 PMCID: PMC163153 DOI: 10.1128/aac.40.3.527] [Citation(s) in RCA: 91] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Affiliation(s)
- E J Arts
- McGill University AIDS Centre, Montreal, Quebec, Canada
| | | |
Collapse
|
27
|
Corcuera T, Alonso MJ, Picazo A, Gómez F, Roldán M, Abad M, Muñoz E, López-Bravo A. Hepatic Morphological alterations induced by zidovudine (ZDV) in an experimental model. Pathol Res Pract 1996; 192:182-7. [PMID: 8692720 DOI: 10.1016/s0344-0338(96)80218-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Zidovudine (AZT) inhibits HIV replication. Many studies have demonstrated its toxic myopathic effect in both HIV-positive patients treated with the drug and experimental animal models. So far hepatic lesions induced by AZT have not been reported. In our study, an experimental rat model was used in which the rats were administered AZT (1 mg/ml) in drinking water; histological and ultrastructural alterations were observed in the liver of treated animals and compared with the findings in control animals. The histological alterations detected were turbid swelling, vacuolar degeneration and microvacuolar fatty degeneration of panlobular distribution; these lesions were progressively greater as the duration of treatment increased. The ultrastructural alterations detected involved the mitochondria (similar to those described in cardiac muscle), smooth and rough endoplasmic reticulum (SER and RER), and the accumulation of fat and glycogen in the hepatocytes of treated animals. The histopathological and ultrastructural findings in our experimental model suggest hepatotoxicity induced by AZT or its catabolites in treated, as compared to control animals.
Collapse
Affiliation(s)
- T Corcuera
- Department of Pathology, C.I.C., Instituto de salud Carlos III, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Greely HT. Conflicts in the biotechnology industry. THE JOURNAL OF LAW, MEDICINE & ETHICS : A JOURNAL OF THE AMERICAN SOCIETY OF LAW, MEDICINE & ETHICS 1995; 23:354-359. [PMID: 8715055 DOI: 10.1111/j.1748-720x.1995.tb01377.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
True revolutions turn the entire world upside down, in ways expected and surprising, profound and mundane. The revolution spawned by advances in molecular biology is no exception. Most of the attention has gone, deservedly, to the possible effects of these advances on medicine, on society, and on our understanding of what it means to be human. But the revolution has already had effects—large and small, good and bad—in other areas. This paper analyzes one aspect of the industry created by that revolution in molecular biology–biotechnology. Specifically, it surveys the various kinds of conflicting interests, both real and perceived, that develop among commercial enterprises, government, and institutions in biotechnology; and it examines the legal implications and public policy concerns of these conflicting interests.The paper focuses on three different kinds of conflicting interests that confront private and public enterprises competing or collaborating in the biotechnology industry: (1) those among businesses involved within the industry; (2) those in relationships between industry and government; and (3) those in relationships between industry and universities. These types of conflicts raise very different issues, but each stems from circumstances unique to the young biotechnology industry.
Collapse
|
29
|
Balzarini J. Metabolism and mechanism of antiretroviral action of purine and pyrimidine derivatives. PHARMACY WORLD & SCIENCE : PWS 1994; 16:113-26. [PMID: 8032337 DOI: 10.1007/bf01880662] [Citation(s) in RCA: 140] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Unlike herpes viruses, human immunodeficiency virus and other retroviruses do not encode specific enzymes required for the metabolism of the purine or pyrimidine nucleotides to their corresponding 5'-triphosphates. Therefore, 2',3'-dideoxynucleosides and acyclic nucleoside phosphonates must be phosphorylated and metabolized by host cell kinases and other enzymes of purine and/or pyrimidine metabolism. Different animal species (or even different cell types within one animal species) may differ in the efficiency of conversion of these drugs to their antivirally active metabolite(s). Three 2',3'-dideoxynucleosides are officially licensed for clinical use [i.e., zidovudine (3'-azido-2',3'-dideoxythymidine, AZT), didanosine (2',3'-dideoxyinosine, DDI) and zalcitabine (2',3'-dideoxycytidine, DDC)]. A number of other 2',3'-dideoxynucleoside analogues [among them stavudine (2',3'-didehydro-2',3'-dideoxythymidine, D4T), 2',3'-dideoxy-3'-thiacytidine (3TC), 2',3'-dideoxy-5-fluoro-3'-thiacytidine (FTC) and the acyclic nucleoside phosphonate 9-(2-phosphonylmethoxyethyl)adenine (PMEA)] are currently under clinical investigation and are candidate compounds for eventual licensing as anti-AIDS drugs. The metabolic pathways, antimetabolic effects and mechanism of antiviral action of these nucleoside analogues will be discussed.
Collapse
Affiliation(s)
- J Balzarini
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Belgium
| |
Collapse
|
30
|
Schinazi RF. Competitive inhibitors of human immunodeficiency virus reverse transcriptase. ACTA ACUST UNITED AC 1993. [DOI: 10.1007/bf02171660] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
31
|
Affiliation(s)
- M S Hirsch
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
| | | |
Collapse
|
32
|
Abstract
Part I of this article reviewed the targets against which anti-HIV drugs can be directed, problems in assessing active compounds (e.g. resistance development and use of surrogate end-points). and nucleoside analogues effective against HIV reverse transcriptase. Intensive research is currently being undertaken in laboratories and hospitals to design and evaluate new inhibitors of HIV. In this work, combining different drugs is one important approach, both to decrease toxicity and to offset the rate of resistance development, which seems to be a major problem associated with therapy directed against the ever-changing HIV. Therapeutic vaccines and immunomodulators are other modalities being actively evaluated against HIV and AIDS, although this effort has not yet yielded any licensed treatment. It appears likely that new antiviral drugs and immunotherapies will be forthcoming during the next 5 years, that they will be used in a variety of combinations, and that the treatment options available for opportunistic infections will increase. These developments should improve the survival and the quality of life of patients with HIV infection.
Collapse
Affiliation(s)
- E Sandström
- Department of Dermatovenerology, Södersjukhuset, Stockholm, Sweden
| | | |
Collapse
|
33
|
De Clercq E. Antiviral agents: characteristic activity spectrum depending on the molecular target with which they interact. Adv Virus Res 1993; 42:1-55. [PMID: 8430518 DOI: 10.1016/s0065-3527(08)60082-2] [Citation(s) in RCA: 130] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The target protein (enzyme) with which antiviral agents interact determines their antiviral activity spectrum. Based on their activity spectrum, antiviral compounds could be divided into the following classes: (1) sulfated polysaccharides (i.e., dextran sulfate), which interact with the viral envelope glycoproteins and are inhibitory to a broad variety of enveloped viruses (i.e., retro-, herpes-, rhabdo-, and arenaviruses): (2) SAH hydrolase inhibitors (i.e., neplanocin A derivatives), which are particularly effective against poxvirus, (-)RNA viruses (paramyxovirus, rhabdovirus), and (+/-)RNA virus (reovirus); (3) OMP decarboxylase inhibitors (i.e., pyrazofurin) and CTP synthetase inhibitors (i.e., cyclopentenylcytosine), which are active against a broad range of DNA, (+)RNA, (-)RNA, and (+/-)RNA viruses; (4) IMP dehydrogenase inhibitors (i.e., ribavirin), which are also active against various (+)RNA and (-)RNA viruses and, in particular, ortho- and paramyxoviruses; (5) acyclic guanosine analogs (i.e., ganciclovir) and carbocyclic guanosine analogs (i.e., cyclobut-G), which are particularly active against herpesviruses (i.e., HSV-1, HSV-2, VZV, CMV); (6) thymidine analogs (i.e., BVDU, BVaraU), which are specifically active against HSV-1 and VZV because of their preferential phosphorylation by the virus-encoded thymidine kinase; (7) acyclic nucleoside phosphonates (i.e., HPMPA, HPMPC, PMEA, FPMPA), which, depending on the structure of the acyclic side chain, span an activity spectrum from DNA viruses (papova-, adeno-, herpes-, hepadna-, and poxvirus) to retroviruses (HIV); (8) dideoxynucleoside analogs (i.e., AZT, DDC), which act as chain terminators in the reverse transcriptase reaction and thus block the replication of retroviruses as well as hepadnaviruses; and (9) the TIBO, HEPT, and other TIBO-like compounds, which interact specifically with the reverse transcriptase of HIV-1 and thus block the replication of HIV-1, but not of HIV-2 or any other retrovirus.
Collapse
Affiliation(s)
- E De Clercq
- Rega Institute for Medical Research, Katholieke Universiteit Leuven, Belgium
| |
Collapse
|
34
|
Affiliation(s)
- R F Schinazi
- Emory University School of Medicine, Department of Pediatrics, Atlanta, GA 30322
| | | | | |
Collapse
|
35
|
Abstract
A myriad of chemical derivatives has been shown to inhibit in vitro replication of the AIDS virus at concentrations that are nontoxic to the host cells. The majority of these agents acts by either (i) inhibiting enzymes such as reverse transcriptase (RT), protease, or glucosidase, (ii) arresting expression of genes or gene products, or (iii) inhibiting viral processes such as giant cell (syncytia) formation or viral binding to the target cell. The nucleoside RT inhibitors are the most widely studied agents at both the preclinical and the clinical levels. Their inability to cure AIDS has stimulated the discovery of several novel nonnucleoside RT inhibitors, possessing varied structures and demonstrating activity at nanomolar concentrations. These agents demonstrate a unique mode of binding to RT and show a high specificity for HIV-1. Protease inhibitors, soluble CD4 derivatives, oligonucleotides, and many anionic derivatives also demonstrate potent anti-HIV-1 activities. These derivatives possess mechanisms of action different to the nucleosides and exhibit selectivity as exemplified by their high in vitro therapeutic indices. This article discusses the structural parameters that govern activity in these agents, the pros and cons regarding the development of these compounds as putative anti-AIDS agents, and the future promise of searching for newer agents directed at novel targets to inhibit the AIDS virus.
Collapse
Affiliation(s)
- P Mohan
- Department of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, University of Illinois, Chicago 60680
| |
Collapse
|
36
|
Ruprecht RM, Koch JA, Sharma PL, Armany RS. Development of antiviral treatment strategies in murine models. AIDS Res Hum Retroviruses 1992; 8:997-1011. [PMID: 1323985 DOI: 10.1089/aid.1992.8.997] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Murine models with type C murine leukemia viruses have been used to develop major new prophylactic and therapeutic strategies in vaccination, drug therapy of acute virus exposure and chronic viremia, combination therapy, prevention of maternal transmission, and therapy targeted to the central nervous system. Transgenic mice expressing either the whole human immunodeficiency virus type 1 (HIV-1) provirus or subgenomic sequences allow the in vivo analysis of selected HIV-1 functions. The full replicative cycle of HIV-1 can be studied in human/mouse chimerae which were created by transplanting human hematolymphoid cells into SCID mice. The chimeric SCID mouse models have been used successfully to evaluate anti-HIV-1 drugs. The role of the various murine retrovirus systems in the development of anti-HIV-1 and anti-AIDS therapies is summarized.
Collapse
Affiliation(s)
- R M Ruprecht
- Laboratory of Viral Pathogenesis, Dana-Farber Cancer Institute, Boston, MA
| | | | | | | |
Collapse
|
37
|
The gene for erythropoietin receptor is expressed in multipotential hematopoietic and embryonal stem cells: evidence for differentiation stage-specific regulation. Mol Cell Biol 1992. [PMID: 1312671 DOI: 10.1128/mcb.12.4.1815] [Citation(s) in RCA: 50] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The principal regulator of erythropoiesis is the glycoprotein erythropoietin, which interacts with a specific cell surface receptor (EpoR). A study aimed at analyzing EpoR gene regulation has shown that both pluripotent embryonal stem cells and early multipotent hematopoietic cells express EpoR transcripts. Commitment to nonerythroid lineages (e.g., macrophage or lymphocytic) results in the shutdown of EpoR gene expression, whereas commitment to the erythroid lineage is concurrent with or followed by dramatic increases in EpoR transcription. To determine whether gene activity could be correlated with chromatin alterations, DNase-hypersensitive sites (HSS) were mapped. Two major HSS located in the promoter region and within the first intron of the EpoR gene are present in all embryonal stem and hematopoietic cells tested, the intensities of which correlate well with EpoR expression levels. In addition, a third major HSS also located within the first intron of the EpoR gene is uniquely present in erythroid cells that express high levels of EpoR. Transfection assays show that sequences surrounding this major HSS impart erythroid cell-specific enhancer activity to a heterologous promoter and that this activity is at least in part mediated by GATA-1. These data, together with concordant expression levels of GATA-1 and EpoR in both early multipotent hematopoietic and committed erythroid cells, support a regulatory role of the erythroid cell-specific transcription factor GATA-1 in EpoR transcription in these cells. However, the lack of significant levels of GATA-1 expression in embryonal stem cells implies an alternative regulatory mechanism of EpoR transcription in cells not committed to the hematopoietic lineage.
Collapse
|
38
|
Heberlein C, Fischer KD, Stoffel M, Nowock J, Ford A, Tessmer U, Stocking C. The gene for erythropoietin receptor is expressed in multipotential hematopoietic and embryonal stem cells: evidence for differentiation stage-specific regulation. Mol Cell Biol 1992; 12:1815-26. [PMID: 1312671 PMCID: PMC369625 DOI: 10.1128/mcb.12.4.1815-1826.1992] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
The principal regulator of erythropoiesis is the glycoprotein erythropoietin, which interacts with a specific cell surface receptor (EpoR). A study aimed at analyzing EpoR gene regulation has shown that both pluripotent embryonal stem cells and early multipotent hematopoietic cells express EpoR transcripts. Commitment to nonerythroid lineages (e.g., macrophage or lymphocytic) results in the shutdown of EpoR gene expression, whereas commitment to the erythroid lineage is concurrent with or followed by dramatic increases in EpoR transcription. To determine whether gene activity could be correlated with chromatin alterations, DNase-hypersensitive sites (HSS) were mapped. Two major HSS located in the promoter region and within the first intron of the EpoR gene are present in all embryonal stem and hematopoietic cells tested, the intensities of which correlate well with EpoR expression levels. In addition, a third major HSS also located within the first intron of the EpoR gene is uniquely present in erythroid cells that express high levels of EpoR. Transfection assays show that sequences surrounding this major HSS impart erythroid cell-specific enhancer activity to a heterologous promoter and that this activity is at least in part mediated by GATA-1. These data, together with concordant expression levels of GATA-1 and EpoR in both early multipotent hematopoietic and committed erythroid cells, support a regulatory role of the erythroid cell-specific transcription factor GATA-1 in EpoR transcription in these cells. However, the lack of significant levels of GATA-1 expression in embryonal stem cells implies an alternative regulatory mechanism of EpoR transcription in cells not committed to the hematopoietic lineage.
Collapse
Affiliation(s)
- C Heberlein
- Heinrich-Pette-Institut für Experimentelle Virologie und Immunologie, Universität Hamburg, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Yarchoan R, Broder S. Correlations between the in vitro and in vivo activity of anti-HIV agents: implications for future drug development. JOURNAL OF ENZYME INHIBITION 1992; 6:99-111. [PMID: 1285306 DOI: 10.3109/14756369209041358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Some 10 years after the first recognition of acquired immunodeficiency syndrome (AIDS) as a new syndrome, we have identified a number of molecular targets to interrupt the replicative cycle of human immunodeficiency virus (HIV), the causative agent. A number of dideoxynucleosides have been identified as having anti-HIV activity in vitro, and several of these have been found to have clinical activity in patients. In contrast, while a number of agents have been found to block viral binding to the target cell in vitro, these agents have generally not shown clear-cut evidence of clinical activity. Agents which act at a variety of steps in the HIV replicative cycle are now under development, and it is likely that we will have an increased armamentarium to fight this disease in the near future.
Collapse
Affiliation(s)
- R Yarchoan
- Medicine Branch, National Cancer Institute, NIH, Bethesda, MD 20892
| | | |
Collapse
|
40
|
Lusso P, Di Marzo Veronese F, Ensoli B, Franchini G, Jemma C, Derocco SE, Kalyanaraman VS, Gallo RC. Response
: Pseudotypes in HIV-Infected Mice. Science 1990; 250:1153-4. [PMID: 17840195 DOI: 10.1126/science.250.4984.1153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
41
|
Fischer KD, Nowock J. The T----C substitution at -198 of the A gamma-globin gene associated with the British form of HPFH generates overlapping recognition sites for two DNA-binding proteins. Nucleic Acids Res 1990; 18:5685-93. [PMID: 1699206 PMCID: PMC332301 DOI: 10.1093/nar/18.19.5685] [Citation(s) in RCA: 30] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Defects in the developmental changes of human hemoglobin production characterized by the continued expression of fetal globin during adult life are classified as hereditary persistence of fetal hemoglobin (HPFH). Among the various molecular lesions associated with this phenotype, the non-deletion forms with point mutations in the promoter region are thought to provide mechanistic clues for gamma-globin gene regulation. The natural occurrence of four different base substitutions mapping within six nucleotides of a homopurine.homopyrimidine motif in the upstream promoter region demarcate a potential control element. To assess its importance for transcriptional activity, we compared the -202 (C----G), -198 (T----C) and -196 (C----T) HPFH mutations with the normal sequence in binding studies with nuclear proteins from erythroid and non-erythroid cells. Wildtype DNA and HPFH mutations at -202 or -196 showed only a weak protein interaction of unclear functional significance. In contrast, -198 (T----C) generated overlapping, high-affinity binding sites for two ubiquitous nuclear proteins. One cognate protein was identified as the transcription factor Sp1. The second one was termed NF-G.C as it interacted strongly with the homopolymer poly(dG).poly(dC). The generation of additional recognition sites for trans-acting factors by the -198 HPFH mutation correlated with a modest increase in promoter activity in vitro specifically with nuclear extracts from erythroid cells. The activation appears to be mediated by binding of Sp1, but it requires interaction with an erythroid-specific factor, most likely GF-1. Templates containing the -196 HPFH mutation showed a transcriptional activity identical to wildtype. This suggests that despite the topological proximity of the mutations, the HPFH phenotype may be established by different mechanisms.
Collapse
Affiliation(s)
- K D Fischer
- Heinrich-Pette-Institut für Experimentelle Virologie und Immunologie, Universität Hamburg
| | | |
Collapse
|
42
|
Broder S. Clinical applications of 3'-azido-2',3'-dideoxythymidine (AZT) and related dideoxynucleosides. Med Res Rev 1990; 10:419-39. [PMID: 2243510 DOI: 10.1002/med.2610100403] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- S Broder
- National Cancer Institute, Bethesda, Maryland 20892
| |
Collapse
|
43
|
Abstract
The development of antiretroviral therapy against acquired immunodeficiency syndrome (AIDS) has been an intense research effort since the discovery of the causative agent, human immunodeficiency virus (HIV). A large array of drugs and biologic substances can inhibit HIV replication in vitro. Nucleoside analogs--particularly those belonging to the dideoxynucleoside family--can inhibit reverse transcriptase after anabolic phosphorylation. 3'-Azido-2',3'-dideoxythymidine (AZT) was the first such drug tested in individuals with AIDS, and considerable knowledge of structure-activity relations has emerged for this class of drugs. However, virtually every step in the replication of HIV could serve as a target for a new therapeutic intervention. In the future, non-nucleoside-type drugs will likely become more important in the experimental therapy of AIDS, and antiretroviral therapy will exert major effects against the morbidity and mortality caused by HIV.
Collapse
Affiliation(s)
- H Mitsuya
- National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | |
Collapse
|
44
|
Abstract
The replication of human immunodeficiency virus (HIV) can be suppressed in vivo by drugs chosen on the basis of their selective in vitro antiviral activity. Such suppression can confer prolonged survival and improved quality of life in patients with already established HIV infection. The clinical benefits indicate that targeted therapy for acquired immunodeficiency syndrome based on the emerging knowledge of replicative cycle of HIV is an attainable goal.
Collapse
Affiliation(s)
- H Mitsuya
- Division of Cancer Treatment National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | | | |
Collapse
|
45
|
Yarchoan R, Mitsuya H, Myers CE, Broder S. Clinical pharmacology of 3'-azido-2',3'-dideoxythymidine (zidovudine) and related dideoxynucleosides. N Engl J Med 1989; 321:726-38. [PMID: 2671731 DOI: 10.1056/nejm198909143211106] [Citation(s) in RCA: 270] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- R Yarchoan
- Clinical Oncology Program, National Cancer Institute, Bethesda, MD 20892
| | | | | | | |
Collapse
|
46
|
Abstract
A rapid expansion of our knowledge of drugs that intervene with human immunodeficiency virus (HIV) infection has taken place. This review covers known and potential anti-HIV targets, including receptor blocking agents, membrane stabilisers, reverse transcriptase inhibitors and chain terminators, RNases, agents altering activation, assembly, budding or regulation of transcription and translation, post-transcriptional modifications and other areas. Important or promising agents, such as zidovudine (ZVD; azidothymidine, AZT), dideoxycytidine, dideoxyinosine, foscarnet, interferons, imuthiol, isoprinosine and others that are either on the market or in advanced clinical trials are emphasised. Four years after the discovery of the aetiological agent, the first drug, zidovudine, has been registered. Many questions about this drug remain, however, owing to the haste with which it was developed. An unprecedented number of other compounds are under evaluation, making it difficult to assess the relative merits of the different compounds and thus set priorities for their development. The point has been reached where a better economical and intellectual framework is necessary so that researchers and physicians are not overwhelmed by the difficulties of conducting clinical trials during the epidemic and have a reasonable chance of keeping up with laboratory developments.
Collapse
Affiliation(s)
- E Sandström
- Department of Dermatovenereology, Södersjukhuset, Stockholm, Sweden
| |
Collapse
|
47
|
Affiliation(s)
- C S Crumpacker
- Division of Infectious Diseases, Beth Israel Hospital, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
48
|
Patel BA, Chu CK, Boudinot FD. Pharmacokinetics and saturable renal tubular secretion of zidovudine in rats. J Pharm Sci 1989; 78:530-4. [PMID: 2778651 DOI: 10.1002/jps.2600780704] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The purpose of this study was to assess the effects of dose on the pharmacokinetics of zidovudine (3'-azido-3'-deoxythymidine; AZT) in rats. Zidovudine (AZT) was administered intravenously at doses of 10, 50, 100, and 250 mg/kg. Plasma and urine AZT concentrations were determined by HPLC. Plasma AZT concentrations declined rapidly with a terminal half-life ranging from 0.76 h at a dose of 10 mg/kg to 1.58 h at 250 mg/kg. Total clearance (CLT) was similar at the doses of 10 and 50 mg/kg, with values of 2.80 and 2.73 L/h/kg, respectively. However, there was a trend toward nonlinearity at the dose of 100 mg/kg (CLT = 2.13 L/h/kg) and a significant decrease in CLT (1.22 L/h/kg) at the dose of 250 mg/kg. Nonrenal clearance remained unaffected by dose with a mean value of 0.98 L/h/kg. Renal clearance (CLR) was similar at the doses of 10 and 50 mg/kg, with values of 1.89 and 1.37 L/h/kg, respectively. However, significant decreases in CLR were observed at the doses of 100 (CLR = 1.30 L/h/kg) and 250 mg/kg (CLR = 0.57 L/h/kg). The maximum transport capacity (Tmax) and the Michaelis-Menten constant (Km) for renal tubular secretion obtained after simultaneously fitting plasma concentration-time profiles at the four doses to a renal clearance model were 215.5 +/- 82.1 mg/h and 119.3 +/- 80.5 mg/L, respectively, thereby yielding an unbound secretory intrinsic clearance (CLus,int) of 1.81 L/h. The high Tmax and Km values account for the high CLR of AZT and explain the linearity of CLR over a wide range of AZT plasma concentrations.(ABSTRACT TRUNCATED AT 250 WORDS)
Collapse
Affiliation(s)
- B A Patel
- Department of Pharmaceutics, College of Pharmacy, University of Georgia, Athens 30602
| | | | | |
Collapse
|
49
|
Langtry HD, Campoli-Richards DM. Zidovudine. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic efficacy. Drugs 1989; 37:408-50. [PMID: 2661194 DOI: 10.2165/00003495-198937040-00003] [Citation(s) in RCA: 90] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Zidovudine (azidothymidine) is a thymidine analogue antiretroviral drug active against human immunodeficiency virus (HIV). In acquired immunodeficiency syndrome (AIDS) and AIDS-related complex (ARC) patients, orally and intravenously administered zidovudine is effective in reducing the incidence of opportunistic infections and neoplasms, increasing helper T lymphocyte numbers, and improving survival rates and quality of life. Adverse effects include serious haematological abnormalities and severe headache, abdominal discomfort, nausea, myalgia and insomnia. In addition, neutropenia and other anaemias frequently limit zidovudine therapy and may result in a need for multiple blood transfusions, dose reductions or withdrawal of the drug. However, despite these problems and the lack of information about some aspects of zidovudine use, zidovudine provides a major hope for HIV-infected patients, and it has rapidly become the standard therapy for improving the quality and duration of the lives of AIDS and ARC patients.
Collapse
Affiliation(s)
- H D Langtry
- ADIS Drug Information Services, Auckland, New Zealand
| | | |
Collapse
|
50
|
Abstract
3'-Azido-2'3'-dideoxythymidine (AZT), a thymidine analogue with potent antiretroviral activity against human immunodeficiency virus (HIV) in vitro, has been shown to confer a clinical benefit in patients with advanced acquired immune deficiency syndrome (AIDS). Other 2',3'-dideoxynucleosides, e.g., 2',3'-dideoxy-cytidine and 2',3'-dideoxyadenosine, block the infectivity of HIV against helper/inducer T cells in vitro and protect the cell against the cytopathic effect of the virus. The majority of these antiretroviral agents were synthesized more than twenty years ago as analogues of physiologically important deoxynucleosides in the quest of a more effective cancer chemotherapy. None of the synthetic analogues manifested significant activity when screened against L1210 leukemia in BDF mice despite the fact that the phosphorylation reactions, crucial to the activation of the 2',3'-dideoxynucleosides, are catalyzed by kinases of appropriate target cells. However, the 2',3'-dideoxynucleoside triphosphates, relative to corresponding deoxynucleotides, have a reduced affinity for DNA polymerase alpha, an enzyme that has key DNA synthetic and repair functions in the life of a cell. In contrast, HIV reverse transcriptase, like host cellular DNA polymerase beta (a repair enzyme) and gamma (a mitochondrial enzyme), is much more susceptible to the inhibitory effects of the dideoxynucleotides. This would explain the activity of the fradulent nucleotides at low concentrations against pathogenic retroviruses vis à vis the low cytoxic activity observed with these agents as anti-leukemia drugs.
Collapse
Affiliation(s)
- J P Horwitz
- Department of Chemistry, Michigan Cancer Foundation, Detroit
| |
Collapse
|