1
|
Vijayan K. K. V, De Paris K. Nonhuman primate models of pediatric viral diseases. Front Cell Infect Microbiol 2024; 14:1493885. [PMID: 39691699 PMCID: PMC11649651 DOI: 10.3389/fcimb.2024.1493885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024] Open
Abstract
Infectious diseases are the leading cause of death in infants and children under 5 years of age. In utero exposure to viruses can lead to spontaneous abortion, preterm birth, congenital abnormalities or other developmental defects, often resulting in lifelong health sequalae. The underlying biological mechanisms are difficult to study in humans due to ethical concerns and limited sample access. Nonhuman primates (NHP) are closely related to humans, and pregnancy and immune ontogeny in infants are very similar to humans. Therefore, NHP are a highly relevant model for understanding fetal and postnatal virus-host interactions and to define immune mechanisms associated with increased morbidity and mortality in infants. We will discuss NHP models of viruses causing congenital infections, respiratory diseases in early life, and HIV. Cytomegalovirus (CMV) remains the most common cause of congenital defects worldwide. Measles is a vaccine-preventable disease, yet measles cases are resurging. Zika is an example of an emerging arbovirus with devastating consequences for the developing fetus and the surviving infant. Among the respiratory viruses, we will discuss influenza and Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). We will finish with HIV as an example of a lifelong infection without a cure or vaccine. The review will highlight (i) the impact of viral infections on fetal and infant immune development, (ii) how differences in infant and adult immune responses to infection alter disease outcome, and emphasize the invaluable contribution of pediatric NHP infection models to the design of effective treatment and prevention strategies, including vaccines, for human infants.
Collapse
Affiliation(s)
- Vidya Vijayan K. K.
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, NC, United States
- Center for AIDS Research, University of North Carolina, Chapel Hill, NC, United States
- Children’s Research Institute, University of North Carolina, Chapel Hill, NC, United States
| |
Collapse
|
2
|
Tessema FA, Barenie RE, Avorn J, Kesselheim AS. Federal Funding For Discovery And Development Of Costly HIV Drugs Was Far More Than Previously Estimated. Health Aff (Millwood) 2023; 42:642-649. [PMID: 37126755 DOI: 10.1377/hlthaff.2022.01134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In July 2012, tenofovir disoproxil fumarate-emtricitabine (TDF-FTC, brand name Truvada) was approved by the Food and Drug Administration (FDA) to prevent HIV infection. To estimate the extent of the US government's direct financial contribution to the discovery and development of Truvada, we identified National Institutes of Health awards using FDA documents, peer-reviewed literature, patent records, court filings, and other publicly available materials. We classified seventy-three federal government awards to eleven researchers as being directly linked to the development and clinical testing of Truvada for prevention therapy, through which the US government spent an estimated $143 million. The substantial public funding raises questions about the high price charged by the drug's manufacturer, which reduced its affordability and limited its accessibility as HIV preventive therapy.
Collapse
Affiliation(s)
- Frazer A Tessema
- Frazer A. Tessema , Brigham and Women's Hospital and Harvard University, Boston, Massachusetts; and University of Chicago, Chicago, Illinois
| | - Rachel E Barenie
- Rachel E. Barenie, Brigham and Women's Hospital and Harvard University; and University of Tennessee, Memphis, Tennessee
| | - Jerry Avorn
- Jerry Avorn, Brigham and Women's Hospital and Harvard University
| | - Aaron S Kesselheim
- Aaron S. Kesselheim, Brigham and Women's Hospital and Harvard University
| |
Collapse
|
3
|
Lantz AM, Nicol MR. Translational Models to Predict Target Concentrations for Pre-Exposure Prophylaxis in Women. AIDS Res Hum Retroviruses 2022; 38:909-923. [PMID: 36097755 PMCID: PMC9805887 DOI: 10.1089/aid.2022.0057] [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] [Indexed: 01/25/2023] Open
Abstract
The HIV epidemic remains a significant public health burden. Women represent half of the global HIV epidemic, yet there is an urgent need for a variety of prevention options to meet the needs of more women. Pre-exposure prophylaxis (PrEP) is a valuable prevention tool that uses antiretrovirals before a potential HIV exposure to prevent virus transmission. Development of effective preventive drug regimens for women is dependent on convenient dosing schedules and routes of administration, and on identifying defined target concentrations in mucosal tissues that provide complete protection against HIV transmission. There is a critical need for a translational model that can accurately predict in vivo target concentrations that are completely protective against HIV infection. There is no gold-standard preclinical model to predict PrEP efficacy. In this study, we review the strengths and limitations of three different preclinical models and their utility in predicting target concentrations in the female genital tract: humanized mice, non-human primates, and the ex vivo tissue model.
Collapse
Affiliation(s)
- Alyssa M. Lantz
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| | - Melanie R. Nicol
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
4
|
Massud I, Cong ME, Ruone S, Holder A, Dinh C, Nishiura K, Khalil G, Pan Y, Lipscomb J, Johnson R, Deyounks F, Rooney JF, Babusis D, Park Y, McCallister S, Callebaut C, Heneine W, García-Lerma JG. Efficacy of Oral Tenofovir Alafenamide/Emtricitabine Combination or Single-Agent Tenofovir Alafenamide Against Vaginal Simian Human Immunodeficiency Virus Infection in Macaques. J Infect Dis 2020; 220:1826-1833. [PMID: 31362305 DOI: 10.1093/infdis/jiz383] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/18/2019] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Tenofovir alafenamide (TAF)-based regimens are being evaluated for pre-exposure prophylaxis (PrEP). We used a macaque model of repeated exposures to simian human immunodeficiency virus (SHIV) to investigate whether TAF alone or the combination of TAF and emtricitabine (FTC) can prevent vaginal infection. METHODS Pigtail macaques were exposed vaginally to SHIV162p3 once a week for up to 15 weeks. Animals received clinical doses of FTC/TAF (n = 6) or TAF (n = 9) orally 24 hours before and 2 hours after each weekly virus exposure. Infection was compared with 21 untreated controls. RESULTS Five of the 6 animals in the FTC/TAF and 4 of the 9 animals in the TAF alone group were protected against infection (P = .001 and P = .049, respectively). The calculated efficacy of FTC/TAF and TAF was 91% (95% confidence interval [CI], 34.9%-98.8%) and 57.8% (95% CI, -8.7% to 83.6%), respectively. Infection in FTC/TAF but not TAF-treated macaques was delayed relative to controls (P = .005 and P = .114). Median tenofovir diphosphate (TFV-DP) levels in peripheral blood mononuclear cells (PBMCs) were similar among infected and uninfected macaques receiving TAF PrEP (351 and 143 fmols/106 cells, respectively; P = .921). CONCLUSIONS Emtricitabine/TAF provided a level of protection against vaginal challenge similar to FTC/TFV disoproxil fumarate combination in the macaque model. Our results support the clinical evaluation of FTC/TAF for PrEP in women.
Collapse
Affiliation(s)
- Ivana Massud
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Mian-Er Cong
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Susan Ruone
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Angela Holder
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Chuong Dinh
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Kenji Nishiura
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - George Khalil
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Yi Pan
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jonathan Lipscomb
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Ryan Johnson
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Frank Deyounks
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | | - Yeojin Park
- Gilead Sciences, Inc., Foster City, California
| | | | | | - Walid Heneine
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - J Gerardo García-Lerma
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
5
|
Daly MB, Clayton AM, Ruone S, Mitchell J, Dinh C, Holder A, Jolly J, García-Lerma JG, Weed JL. Training rhesus macaques to take daily oral antiretroviral therapy for preclinical evaluation of HIV prevention and treatment strategies. PLoS One 2019; 14:e0225146. [PMID: 31730629 PMCID: PMC6857902 DOI: 10.1371/journal.pone.0225146] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 10/29/2019] [Indexed: 11/18/2022] Open
Abstract
Background Macaque models of simian or simian/human immunodeficiency virus (SIV or SHIV) infection are critical for the evaluation of antiretroviral (ARV)-based HIV treatment and prevention strategies. However, modelling human oral ARV administration is logistically challenging and fraught by limited adherence. Here, we developed a protocol for administering daily oral doses of ARVs to macaques with a high rate of compliance. Methods Parameters of positive reinforcement training (PRT), behavioral responses and optimal drug delivery foods were defined in 7 male rhesus macaques (Macaca mulatta). Animals were trained to sit in a specified cage location prior to receiving ARVs, emtricitabine (FTC) and tenofovir alafenamide (TAF), in a blended food mixture, which was followed immediately with a juice chaser. Consistency of daily oral adherence was evaluated in 4 trained macaques receiving clinically equivalent doses of FTC and TAF (20 and 1.5 mg/kg, respectively) in a short-term (1 month) and an extended (6 month) trial. Adherence was monitored using medication diaries and by quantifying intracellular FTC-triphosphate (FTC-TP) and tenofovir-diphosphate (TFV-DP) concentrations in peripheral mononuclear blood cells (PBMCs). Results Trained macaques quickly and consistently took daily oral ARVs for 1 month with an average 99.8% observed adherence. Intracellular concentrations of TFV-DP (median = 845.8 fmol/million cells [range, 620.8–1031.3]) and FTC-TP (median = 367.0 fmol/million cells [range, 289.5–413.5) in PBMCs were consistent with high adherence. Extended treatment with select subjects yielded similar observations for three months (99.5% adherence, 352/356 complete doses taken), although a sudden drop in adherence was observed after splenic biopsy surgery. Conclusions We demonstrate that trained macaques reliably adhere to a daily oral ARV regimen, although unexpected adherence issues are possible. Our approach, using clinical doses of oral FTC and TAF daily, further refines macaque models of HIV treatment and prevention by mimicking the human route and timing of ARV administration.
Collapse
Affiliation(s)
- Michele B. Daly
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - April M. Clayton
- Comparative Medicine Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Susan Ruone
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - James Mitchell
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Chuong Dinh
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Angela Holder
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Julian Jolly
- Comparative Medicine Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - J. Gerardo García-Lerma
- Laboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (JGGL); (JLW)
| | - James L. Weed
- Comparative Medicine Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail: (JGGL); (JLW)
| |
Collapse
|
6
|
Janes H, Corey L, Ramjee G, Carpp LN, Lombard C, Cohen MS, Gilbert PB, Gray GE. Weighing the Evidence of Efficacy of Oral PrEP for HIV Prevention in Women in Southern Africa. AIDS Res Hum Retroviruses 2018; 34:645-656. [PMID: 29732896 PMCID: PMC6080090 DOI: 10.1089/aid.2018.0031] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
As oral tenofovir-based regimens for preexposure prophylaxis (PrEP) are adopted as standard of care for HIV prevention, their utilization in clinical trials among women in southern Africa will require an accurate estimate of oral PrEP efficacy in this population. This information is critical for women in choosing this prevention strategy, and in public health policy making. Estimates of the efficacy of oral PrEP regimens containing tenofovir have varied widely across trials that enrolled women, with some studies reporting high efficacy and others reporting no efficacy. Although poor adherence is strongly associated with lack of efficacy, other factors, such as mode of transmission (sexual vs. parenteral), predominant HIV subtype (C vs. non-C), intensity of exposure, and percentage of stable serodiscordant couples, may also contribute to the variation in efficacy estimates. In this article, we evaluate the evidence for PrEP efficacy in women and propose potential explanations for the observed differences in efficacy among studies. Our review emphasizes the need to continue to refine estimates of efficacy and effectiveness of tenofovir-based oral PrEP so as to best develop the next generation of HIV prevention tools, and to inform public policies directed toward HIV prevention.
Collapse
Affiliation(s)
- Holly Janes
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Lawrence Corey
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Medicine, University of Washington, Seattle, Washington
- Department of Laboratory Medicine, University of Washington, Seattle, Washington
| | - Gita Ramjee
- HIV Prevention Research Unit, South African Medical Research Council, Durban, South Africa
- Department of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Global Health, University of Washington, Seattle, Washington
| | - Lindsay N. Carpp
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Carl Lombard
- Biostatistics Unit, Medical Research Council of South Africa, Cape Town, South Africa
| | - Myron S. Cohen
- Institute for Global Health and Infectious Diseases, University of North Carolina, Chapel Hill, North Carolina
| | - Peter B. Gilbert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Glenda E. Gray
- Perinatal HIV Research Unit, University of the Witwatersrand, Chris Hani Baragwanath Academic Hospital, Johannesburg, South Africa
- Office of the President, South African Medical Research Council, Cape Town, South Africa
| |
Collapse
|
7
|
Xiao D, Ling KHJ, Custodio J, Majeed SR, Tarnowski T. Quantitation of intracellular triphosphate metabolites of antiretroviral agents in peripheral blood mononuclear cells (PBMCs) and corresponding cell count determinations: review of current methods and challenges. Expert Opin Drug Metab Toxicol 2018; 14:781-802. [DOI: 10.1080/17425255.2018.1500552] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Deqing Xiao
- Department of Clinical Pharmacology, Gilead Sciences, Inc, Foster City, CA, USA
| | - Kah Hiing John Ling
- Department of Clinical Pharmacology, Gilead Sciences, Inc, Foster City, CA, USA
| | - Joseph Custodio
- Department of Clinical Pharmacology, Gilead Sciences, Inc, Foster City, CA, USA
| | - Sophia R. Majeed
- Department of Clinical Pharmacology, Gilead Sciences, Inc, Foster City, CA, USA
| | - Thomas Tarnowski
- Department of Clinical Pharmacology, Gilead Sciences, Inc, Foster City, CA, USA
| |
Collapse
|
8
|
De Clercq E. Tenofovir alafenamide (TAF) as the successor of tenofovir disoproxil fumarate (TDF). Biochem Pharmacol 2016; 119:1-7. [PMID: 27133890 DOI: 10.1016/j.bcp.2016.04.015] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 04/27/2016] [Indexed: 02/06/2023]
Abstract
Tenofovir alafenamide (TAF) can be considered a new prodrug of tenofovir (TFV), as successor of tenofovir disoproxil fumarate (TDF). It is in vivo as potent against human immunodeficiency virus (HIV) at a 30-fold lower dose (10mg) than TDF (300mg). TAF has been approved in November 2015 (in the US and EU), as a single-tablet regimen (STR) containing 150mg elvitegravir (E), 150mg cobicistat (C), 200mg emtricitabine [(-)FTC] (F) and 10mg TAF, marketed as Genvoya®, on 01 March 2016 in the US as an STR containing 25mg rilpivirine (R), 200mg F and 25mg TAF, marketed as Odefsey®, and on 4 April 2016 in the US, as an STR containing 200mg F and 25mg TAF, marketed as Descovy®, for the treatment of HIV infections. STR combinations containing TAF and emtricitabine could be paired with a range of third agents, for example, darunavir and cobicistat. TAF has a much lower risk of kidney toxicity or bone density changes than TDF, and also offers long-term potential in the pre-exposure prophylaxis (PrEP) of HIV infections. TAF is specifically accumulated in lymphatic tissue, and in the liver, and hence also holds great potential for the treatment of hepatitis B virus (HBV) infections. Akin to TDF, TAF is converted intracellularly to TFV. Its active diphosphate metabolite (TFVpp) is targeted at the RNA-dependent DNA polymerase (reverse transcriptase) of either HIV or HBV.
Collapse
Affiliation(s)
- Erik De Clercq
- KU Leuven, Department of Microbiology and Immunology, Rega Institute for Medical Research, Minderbroedersstraat 10, B-3000 Leuven, Belgium.
| |
Collapse
|
9
|
Abstract
PURPOSE OF REVIEW To discuss nondaily preexposure prophylaxis (PrEP) modalities that may provide advantages compared with daily PrEP in cost and cumulative toxicity, but may have lower adherence forgiveness. RECENT FINDINGS Animal models have informed our understanding of early viral transmission events, which help guide event-driven PrEP dosing strategies. These models indicate early establishment of viral replication in rectal or cervicovaginal tissues, so event-driven PrEP should rapidly deliver high mucosal drug concentrations within hours of the potential exposure event. Macaque models have demonstrated the high biological efficacy for event-driven dosing of oral tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) against both vaginal and rectal virus transmission. In humans, the IPERGAY study demonstrated 86% efficacy for event-driven oral TDF/FTC dosing among men who have sex with men (MSM), while no similar efficacy data are available on women or heterosexual men. The HPTN 067 study showed that certain MSM populations adhere well to nondaily PrEP, whereas other populations of women adhere more poorly to nondaily versus daily regimens. Pharmacokinetic studies following oral TDF/FTC dosing in humans indicate that TFV-diphosphate (the active form of TFV) accumulates to higher concentrations in rectal versus cervicovaginal tissue, but nonadherence in trials complicates the interpretation of differential mucosal drug concentrations. SUMMARY Event-driven dosing for TFV-based PrEP has promise for HIV prevention in MSM. Future research of event-driven PrEP in women and heterosexual men should be guided by a better understanding of the importance of mucosal drug concentrations for PrEP efficacy and its sensitivity to adherence.
Collapse
Affiliation(s)
- Peter L Anderson
- aDepartment of Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado bLaboratory Branch, Division of HIV/AIDS Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, 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
|
Carryl H, Swang M, Lawrence J, Curtis K, Kamboj H, Van Rompay KKA, De Paris K, Burke MW. Of mice and monkeys: can animal models be utilized to study neurological consequences of pediatric HIV-1 infection? ACS Chem Neurosci 2015; 6:1276-89. [PMID: 26034832 PMCID: PMC4545399 DOI: 10.1021/acschemneuro.5b00044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Pediatric human immunodeficiency virus (HIV-1) infection remains a global health crisis. Children are much more susceptible to HIV-1 neurological impairments than adults, which can be exacerbated by coinfections. Neurological characteristics of pediatric HIV-1 infection suggest dysfunction in the frontal cortex as well as the hippocampus; limited MRI data indicate global cerebral atrophy, and pathological data suggest accelerated neuronal apoptosis in the cortex. An obstacle to pediatric HIV-1 research is a human representative model system. Host-species specificity of HIV-1 limits the ability to model neurological consequences of pediatric HIV-1 infection in animals. Several models have been proposed including neonatal intracranial injections of HIV-1 viral proteins in rats and perinatal simian immunodeficiency virus (SIV) infection of infant macaques. Nonhuman primate models recapitulate the complexity of pediatric HIV-1 neuropathogenesis while rodent models are able to elucidate the role specific viral proteins exert on neurodevelopment. Nonhuman primate models show similar behavioral and neuropathological characteristics to pediatric HIV-1 infection and offer a stage to investigate early viral mechanisms, latency reservoirs, and therapeutic interventions. Here we review the relative strengths and limitations of pediatric HIV-1 model systems.
Collapse
Affiliation(s)
- Heather Carryl
- Department of Physiology & Biophysics, College of Medicine, Howard University, Washington, D.C. 20059, United States
| | - Melanie Swang
- Department of Biology, Howard University, Washington, D.C. 20059, United States
| | - Jerome Lawrence
- Department of Biology, Howard University, Washington, D.C. 20059, United States
| | - Kimberly Curtis
- Department of Physiology & Biophysics, College of Medicine, Howard University, Washington, D.C. 20059, United States
| | - Herman Kamboj
- Department of Physiology & Biophysics, College of Medicine, Howard University, Washington, D.C. 20059, United States
| | - Koen K. A. Van Rompay
- California National Primate Research Center, University of California at Davis, Davis, California 95616, United States
| | - Kristina De Paris
- Department of Microbiology and Immunology and Center for AIDS Research School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - Mark W. Burke
- Department of Physiology & Biophysics, College of Medicine, Howard University, Washington, D.C. 20059, United States
| |
Collapse
|
12
|
Castel AD, Magnus M, Greenberg AE. Pre-exposure prophylaxis for human immunodeficiency virus: the past, present, and future. Infect Dis Clin North Am 2014; 28:563-83. [PMID: 25455314 DOI: 10.1016/j.idc.2014.08.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
This article presents an overview of pre-exposure prophylaxis (PrEP) for human immunodeficiency virus (HIV) prevention. The authors describe the past animal and human research that has been conducted that informs our current understanding of PrEP; summarize ongoing research in the area, including describing new regimens and delivery mechanisms being studied for PrEP; and highlight key issues that must be addressed in order to implement and optimize the use of this HIV prevention tool.
Collapse
Affiliation(s)
- Amanda D Castel
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, The George Washington University, 950 New Hampshire Avenue, Northwest, 5th Floor, Washington, DC 20052, USA.
| | - Manya Magnus
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, The George Washington University, 950 New Hampshire Avenue, Northwest, 5th Floor, Washington, DC 20052, USA
| | - Alan E Greenberg
- Department of Epidemiology and Biostatistics, Milken Institute School of Public Health, The George Washington University, 950 New Hampshire Avenue, Northwest, 5th Floor, Washington, DC 20052, USA
| |
Collapse
|
13
|
Jiang J, Yang X, Ye L, Zhou B, Ning C, Huang J, Liang B, Zhong X, Huang A, Tao R, Cao C, Chen H, Liang H. Pre-exposure prophylaxis for the prevention of HIV infection in high risk populations: a meta-analysis of randomized controlled trials. PLoS One 2014; 9:e87674. [PMID: 24498350 PMCID: PMC3912017 DOI: 10.1371/journal.pone.0087674] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Accepted: 12/28/2013] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Nearly ten randomized controlled trials (RCTs) of pre-exposure prophylaxis (PrEP) have been completed or are ongoing worldwide to evaluate the effectiveness of PrEP in HIV transmission among HIV-uninfected high risk populations. The purpose of this study was to evaluate the role of PrEP to prevent HIV transmission through a Mata-analysis. METHODS A comprehensive computerized literature search was carried out in PubMed, EMbase, Ovid, Web of Science, Science Direct, Wan Fang, CNKI and related websites to collect relevant articles (from their establishment date to August 30, 2013). The search terms were "pre-exposure prophylaxis", "high risk population", "HIV infection", "reduction", "relative risk" and "efficacy". We included any RCT assessing PrEP for the prevention of HIV infection in high risk populations. Interventions of the studies were continuously daily or intermittent doses of single or compound antiretrovirals (ARVs) before HIV exposure or during HIV exposure. A meta-analysis was conducted using Stata 10.0. A random-effects method was used to calculate the pooled relative risk (RR) and 95% confidence intervals (CI) for all studies included. RESULTS Seven RCTs involving 14,804 individuals in high risk populations were eligible for this study. The number of subjects in the experimental groups was 8,195, with HIV infection rate of 2.03%. The number of subjects in the control groups was 6,609, with HIV infection rate of 4.07%. The pooled RR was 0.53 (95% CI = 0.40 ∼ 0.71, P<0.001). The re-analyzed pooled RR were 0.61 (95% CI = 0.48 ∼ 0.77, P<0.001), 0.49 (95% CI = 0.38 ∼ 0.63, P<0.001), respectively, by excluding the largest study or two studies without statistical significance. Publication bias analysis revealed a symmetry funnel plot. The fail-safe number was 1,022. CONCLUSION These results show that PrEP is an effective strategy for reducing new HIV infections in high risk populations.
Collapse
Affiliation(s)
- Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoyi Yang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Bo Zhou
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
| | - Chuanyi Ning
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiegang Huang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Bingyu Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaoni Zhong
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Ailong Huang
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Renchuan Tao
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
| | - Cunwei Cao
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
| | - Hui Chen
- Geriatrics Digestion Department of Internal Medicine, The First Affiliated Hospital of GuangXi Medical University, Nanning, Guangxi, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, China
- School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
14
|
Ye L, Wei S, Zou Y, Yang X, Abdullah AS, Zhong X, Ruan Y, Lin X, Li M, Wu D, Jiang J, Xie P, Huang J, Liang B, Zhou B, Su J, Liang H, Huang A. HIV pre-exposure prophylaxis interest among female sex workers in Guangxi, China. PLoS One 2014; 9:e86200. [PMID: 24465956 PMCID: PMC3899205 DOI: 10.1371/journal.pone.0086200] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 12/08/2013] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Acceptability of pre-exposure prophylaxis (PrEP) and willingness to participate in a clinical trial for both safety and efficacy of PrEP were investigated among female sex workers (FSWs) in Guangxi, China. METHODS A cross-sectional study was performed in three cities in Guangxi. Structured, self-administered questionnaires were used to assess the acceptability of PrEP and the willingness to participate in a clinical trial. Multivariable logistic regression models were fitted to identify predictors. RESULTS Among 405 participants, 15.1% had heard of PrEP. If PrEP was deemed to be effective, safe and provided for free, 85.9% reported that they would accept it, and 54.3% of those who accepted PrEP said that they would participate in a clinical trial. The increased acceptability of PrEP was associated with working in male dominated venues, higher income, a poor family relationship, better HIV/AIDS knowledge, not realizing HIV risk from unfamiliar clients, not being forced to use condoms by the gatekeepers, consistent use of condoms, and use of drugs to prevent STD infection. The increased willingness to participate in a clinical trial was associated with a poor family relationship, better HIV/AIDS knowledge, not realizing HIV risk from unfamiliar clients, a willingness to adhere to daily PreP use, and not being concerned about discrimination by others. The main reason for rejecting PrEP or participating in a clinical trial was the concern about the side effects of PrEP. CONCLUSIONS Acceptability of PrEP among Guangxi FSWs is relatively high, indicating that PrEP intervention programs may be feasible for Chinese FSWs. Given the fact that most of the participants had never heard of PrEP before, and that family, gatekeepers, and social discrimination could significantly affect its acceptability, a comprehensive mix of multiple interventions is necessary for the successful implementation of a PrEP program among this population in Guangxi.
Collapse
Affiliation(s)
- Li Ye
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Suosu Wei
- The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi, China
| | - Yunfeng Zou
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaobo Yang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Abu S. Abdullah
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
- Department of Medicine, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, United Staes of America
| | - Xiaoni Zhong
- School of Public Health, Chongqing Medical University, Chongqing, China
| | - Yuhua Ruan
- State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Xinqin Lin
- Nanning Center for Disease Control and Prevention, Nanning, Guangxi, China
| | - Mingqiang Li
- Liuzhou Center for Disease Control and Prevention, Liuzhou, Guangxi, China
| | - Deren Wu
- Beihai Center for Disease Control and Prevention, Beihai, Guangxi, China
| | - Junjun Jiang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Peiyan Xie
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiegang Huang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Bingyu Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Bo Zhou
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Jinming Su
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Hao Liang
- Guangxi Key Laboratory of AIDS Prevention and Treatment, School of Public Health, Guangxi Medical University, Nanning, Guangxi, China
| | - Ailong Huang
- School of Public Health, Chongqing Medical University, Chongqing, China
| |
Collapse
|
15
|
Plosker GL. Emtricitabine/tenofovir disoproxil fumarate: a review of its use in HIV-1 pre-exposure prophylaxis. Drugs 2013; 73:279-91. [PMID: 23444256 DOI: 10.1007/s40265-013-0024-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The fixed-dose combination of emtricitabine (FTC) 200 mg and tenofovir disoproxil fumarate (TDF) 300 mg (Truvada(®)), administered orally once daily, is widely used as part of first-line regimens for the treatment of HIV-1 infection. Recently, once-daily administration of FTC/TDF was approved in the USA for pre-exposure prophylaxis in conjunction with safer sex practices to reduce the risk of sexually acquired HIV-1 in high-risk adults who are not infected. To date, results of four large, randomized, double-blind, placebo-controlled, multicentre trials with FTC/TDF as pre-exposure prophylaxis have been published. Three studies showed statistically significant reductions in the number of individuals with emergent HIV-1 infection when FTC/TDF was compared with placebo over the ≈1- to 2-year study periods. Efficacy (i.e. risk reduction relative to placebo) was 44 % in the iPrEx trial in men who have sex with men, 75 % in the Partners PrEP study in heterosexual HIV-1-serodiscordant couples and 62 % in the TDF2 trial in heterosexual men and women. The fourth study (FEM-PrEP) in heterosexual women did not show a statistically significant difference between FTC/TDF and placebo, although low adherence rates reported in this trial may have been a factor. No unexpected adverse events were reported in the trials. However, since pre-exposure prophylaxis involves long-term administration of drugs to healthy individuals, it is important to monitor the long-term safety of FTC/TDF (e.g. renal function, bone mineral density) in this setting. Other notable considerations include adherence, cost and the potential for development of drug resistance. Interim guidelines are available for prescribing FTC/TDF as pre-exposure prophylaxis. If used appropriately in selected high-risk individuals, pre-exposure prophylaxis with FTC/TDF represents an important additional strategy to reduce the spread of HIV-1 infection, which continues to be a significant global concern.
Collapse
Affiliation(s)
- Greg L Plosker
- Adis, 41 Centorian Drive, Private Bag 65901, Mairangi Bay, North Shore, 0754, Auckland, New Zealand.
| |
Collapse
|
16
|
Abstract
The impressive advances in antiretroviral (ARV) therapy of chronic human immunodeficiency virus (HIV) infections during the last decade and the availability of potent ARV drugs have fueled interest in using chemoprophylaxis as a novel HIV prevention strategy. Preexposure prophylaxis (PrEP) refers to the use of ARV drugs in HIV-negative persons to prevent HIV infection. The rationale for PrEP builds on the success of ARV prophylaxis in preventing mother-to-child transmission of HIV and on a large body of animal studies that show the efficacy of PrEP against mucosal and parenteral infection. We focus on oral administration of ARV drugs for prevention of HIV infection. Identifying an effective prophylactic pill that individuals can take outside the setting of sexual intercourse precludes the necessity to disclose such use to their partners, thereby empowering those who might not be in a position to negotiate with their partners. Several human clinical trials evaluating the efficacy of daily regimens of the HIV reverse-transcriptase (RT) inhibitors tenofovir disoproxil fumarate (TDF) or Truvada (TDF and emtricitabine [FTC]) are under way among high-risk populations. The results of one trial among men who have sex with men showed that daily Truvada was safe and effective, providing the first support for oral PrEP as a prevention strategy. Here we outline the preclinical and clinical research on oral PrEP, pharmacologic considerations, and future directions and challenges.
Collapse
Affiliation(s)
- Walid Heneine
- Laboratory Branch, Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia 30333, USA.
| | | |
Collapse
|
17
|
Abstract
PURPOSE OF REVIEW Oral and topical pre-exposure prophylaxis (PrEP) with antiretroviral drugs are novel biomedical interventions recently found to prevent HIV transmission among high-risk populations. In this review, we outline lessons learned from animal studies and discuss next steps in preclinical PrEP research including the study of new PrEP modalities, pharmacologic correlates of protection, and biological factors that may modulate PrEP efficacy. RECENT FINDINGS Studies using macaque or humanized mice models of mucosal simian immunodeficiency virus (SIV), HIV, or simian/human immunodeficiency virus (SHIV) transmission have provided efficacy data against rectal and vaginal infection. A multitude of oral and topical PrEP regimens including drugs such as tenofovir (TFV), tenofovir disoproxil fumarate (TDF) and emtricitabine (FTC) were tested against either wild-type or drug-resistant viruses. These models have also helped define prophylactic windows of protection of nondaily dosing and are being used increasingly to study pharmacokinetic and pharmacodynamic relationships. SUMMARY As human data from PrEP trials validate animal models or help fine tune them, it is expected that these models will play increasingly important roles in PrEP development as the field extends into new drug classes and combinations, episodic dosing, and novel long-acting drug formulations. By providing both efficacy and pharmacologic information these models can define correlates and mechanisms of protection, inform dose selection, and advance the most promising PrEP candidates and dosing modalities.
Collapse
|
18
|
Lu C, Jia Y, Chen L, Ding Y, Yang J, Chen M, Song Y, Sun X, Wen A. Pharmacokinetics and food interaction of a novel prodrug of tenofovir, tenofovir dipivoxil fumarate, in healthy volunteers. J Clin Pharm Ther 2012; 38:136-40. [PMID: 23278367 DOI: 10.1111/jcpt.12023] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/09/2012] [Indexed: 11/28/2022]
Affiliation(s)
- C. Lu
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Y. Jia
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - L. Chen
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Y. Ding
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - J. Yang
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - M. Chen
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - Y. Song
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - X. Sun
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| | - A. Wen
- Department of Pharmacy; Xijing Hospital; Fourth Military Medical University; Xi'an China
| |
Collapse
|
19
|
Van Rompay KKA, Jayashankar K. Animal models of HIV transmission through breastfeeding and pediatric HIV infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 743:89-108. [PMID: 22454344 DOI: 10.1007/978-1-4614-2251-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Affiliation(s)
- Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, CA 95616, USA.
| | | |
Collapse
|
20
|
De Clercq E. Tenofovir: Quo Vadis Anno 2012 (Where Is It Going in the Year 2012)
? Med Res Rev 2012; 32:765-85. [PMID: 22581627 DOI: 10.1002/med.21267] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Erik De Clercq
- Rega Institute for Medical Research; KU Leuven; Minderbroedersstraat; 10, B-3000 Leuven Belgium
| |
Collapse
|
21
|
Lu C, Jia Y, Yang J, Song Y, Liu W, Ding Y, Sun X, Wen A. Relative Bioavailability Study of a Novel Prodrug of Tenofovir, Tenofovir Dipivoxil Fumarate, in Healthy Male Fasted Volunteers. Clin Drug Investig 2012; 32:333-8. [DOI: 10.2165/11599910-000000000-00000] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
22
|
Gengiah TN, Baxter C, Mansoor LE, Kharsany AB, Abdool Karim SS. A drug evaluation of 1% tenofovir gel and tenofovir disoproxil fumarate tablets for the prevention of HIV infection. Expert Opin Investig Drugs 2012; 21:695-715. [PMID: 22394224 PMCID: PMC3460694 DOI: 10.1517/13543784.2012.667072] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
INTRODUCTION More than a million people acquire HIV infection annually. Pre-exposure prophylaxis (PrEP) using antiretrovirals is currently being investigated for HIV prevention. Oral and topical formulations of tenofovir have undergone preclinical and clinical testing to assess acceptability, safety and effectiveness in preventing HIV infection. AREAS COVERED The tenofovir drug development pathway from compound discovery, preclinical animal model testing and human testing were reviewed for safety, tolerability and efficacy. Tenofovir is well tolerated and safe when used both systemically or applied topically for HIV prevention. High drug concentrations at the site of HIV transmission and concomitant low systemic drug concentrations are achieved with vaginal application. Coitally applied gel may be the favored prevention option for women compared with the tablets, which may be more suitable for prevention in men and sero-discordant couples. However, recent contradictory effectiveness outcomes in women need to be better understood. EXPERT OPINION Emerging evidence has brought new hope that antiretrovirals can potentially change the course of the HIV epidemic when used as early treatment for prevention, as topical or oral PrEP. Although some trial results appear conflicting, behavioral factors, adherence to dosing and pharmacokinetic properties of the different tenofovir formulations and dosing approaches offer plausible explanations for most of the variations in effectiveness observed in different trials.
Collapse
Affiliation(s)
- Tanuja N Gengiah
- Centre for the AIDS Program of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Doris Duke Medical Research Institute, Nelson R Mandela School of Medicine, Congella, Durban, South Africa.
| | | | | | | | | |
Collapse
|
23
|
Uckun FM, Cahn P, Qazi S, D'Cruz O. Stampidine as a promising antiretroviral drug candidate for pre-exposure prophylaxis against sexually transmitted HIV/AIDS. Expert Opin Investig Drugs 2012; 21:489-500. [PMID: 22360744 DOI: 10.1517/13543784.2012.664635] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Pre-exposure prophylaxis (PrEP) is an evolving new approach to prevention of sexually transmitted HIV-1 that employs antiretroviral (ARV) agents prior to potential HIV-1 exposure in an attempt to reduce the likelihood of HIV-1 infection postexposure. The identification of new ARV agents with potent activity against multidrug-resistant HIV remains an unmet and urgent challenge in the field of PrEP. AREAS COVERED This article reviews the preclinical and early clinical activity and safety profile of stampidine, a novel antiretroviral (ARV) drug candidate that exhibits remarkable subnanomolar to low nanomolar in vitro antiretroviral potency against genotypically and phenotypically nucleoside reverse transcriptase inhibitor (NRTI)-resistant primary clinical HIV isolates, non-nucleoside RT-resistant HIV-1 isolates. Stampidine has a favorable pharmacokinetic profile in mice, rats, dogs and cats with 25 or 50 mg/kg tolerable dose levels yielding micromolar plasma concentrations that are 1000-fold higher than its in vitro IC(50) value against HIV. Stampidine has a favorable, safety profile in mice, rats, dogs and cats and it showed significant in vivo ARV activity in HIV-infected Hu-PBL-SCID mice as well as FIV-infected domestic cats. Furthermore, it did not cause any maternal toxicity, developmental toxicity or teratogenicity in rabbits treated at 10 - 40 mg/kg/day dose levels. In a recently completed first-in-human Phase I clinical trial, stampidine did not cause dose-limiting toxicity at single dose levels ranging from 5 to 25 mg/kg. EXPERT OPINION The favorable safety and activity profile of stampidine warrants its further development as a promising next-generation PrEP candidate to prevent the sexual transmission of HIV-1. The discovery of stampidine as a potent antiretroviral agent represents a significant step forward in the development of effective therapeutic as well as preventive strategies against HIV/AIDS.
Collapse
Affiliation(s)
- Fatih M Uckun
- Developmental Therapeutics Program, Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Los Angeles, CA 90027, USA
| | | | | | | |
Collapse
|
24
|
Van Rompay KK. The use of nonhuman primate models of HIV infection for the evaluation of antiviral strategies. AIDS Res Hum Retroviruses 2012; 28:16-35. [PMID: 21902451 DOI: 10.1089/aid.2011.0234] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Several nonhuman primate models are used in HIV/AIDS research. In contrast to natural host models, infection of macaques with virulent simian immunodeficiency virus (SIV) isolates results in a disease (simian AIDS) that closely resembles HIV infection and AIDS. Although there is no perfect animal model, and each of the available models has its limitations, a carefully designed study allows experimental approaches that are not feasible in humans, but that can provide better insights in disease pathogenesis and proof-of-concept of novel intervention strategies. In the early years of the HIV pandemic, nonhuman primate models played a minor role in the development of antiviral strategies. Since then, a better understanding of the disease and the development of better compounds and assays to monitor antiviral effects have increased the usefulness and relevance of these animal models in the preclinical development of HIV vaccines, microbicides, and antiretroviral drugs. Several strategies that were first discovered to have efficacy in nonhuman primate models are now increasingly used in humans. Recent trends include the use of nonhuman primate models to explore strategies that could reduce viral reservoirs and, ultimately, attempt to cure infection. Ongoing comparison of results obtained in nonhuman primate models with those observed in human studies will lead to further validation and improvement of these animal models so they can continue to advance our scientific knowledge and guide clinical trials.
Collapse
Affiliation(s)
- Koen K.A. Van Rompay
- California National Primate Research Center, University of California, Davis, California
| |
Collapse
|
25
|
Melchjorsen J, Risør MW, Søgaard OS, O'Loughlin KL, Chow S, Paludan SR, Ellermann-Eriksen S, Hedley DW, Minderman H, Østergaard L, Tolstrup M. Tenofovir selectively regulates production of inflammatory cytokines and shifts the IL-12/IL-10 balance in human primary cells. J Acquir Immune Defic Syndr 2011; 57:265-75. [PMID: 21471820 DOI: 10.1097/qai.0b013e3182185276] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVES In this study, we aimed to investigate the possible immune modulatory effects of HIV nucleoside reverse transcriptase inhibitors during secondary infections and inflammation, focusing on inflammatory cytokine responses and the interleukin (IL)-12/IL-10 balance. METHODS We investigated the in vitro effect of tenofovir and zidovudine (AZT) on production of proinflammatory cytokines in monocytes and human peripheral blood mononuclear cells (PBMCs). Stimulation panels included Toll-Like receptor (TLR) ligands; the inflammation mediator tumor necrosis factor-α; and the pathogens cytomegalovirus, Neisseria meningitides, Escherichia coli, and Streptococcus pneumoniae. Cytokine levels were measured using enzyme-linked immunosorbent assay and luminex technology. RNA levels were assessed using real-time polymerase chain reaction. Activity of mitogen-activated protein kinase and NF-κB signaling was evaluated using flow cytometry and multispectral imaging cytometry, respectively. RESULTS Tenofovir decreased and AZT increased both IL-8 and CCL3 production from monocytes after stimulation with TLR ligands, tumor necrosis factor-α, or live pathogens. Similarly, tenofovir decreased CCL3 levels in human PBMCs. Furthermore, tenofovir strongly decreased induction of IL-10 but increased levels of IL-12. AZT did not affect IL-12 or IL-10 levels. The observed drug-induced changes in cytokine production were independent from transcriptional regulation through the mitogen-activated protein kinase and nuclear factor kappa B pathways. CONCLUSIONS Our data suggest divergent effects of tenofovir and AZT on proinflammatory responses in monocytes (CCL3 and IL-8) and PBMCs (CCL3). Moreover, tenofovir shifts the IL-10/IL-12 balance after cell stimulation with TLR ligands or infection with live bacteria, thus suggesting that the choice of nucleoside reverse transcriptase inhibitor affects overall inflammation and early immune responses against secondary pathogens.
Collapse
Affiliation(s)
- Jesper Melchjorsen
- Department of Infectious Diseases, Aarhus University Hospital Skejby, Aarhus, Denmark.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Martin M, Vanichseni S, Suntharasamai P, Sangkum U, Chuachoowong R, Mock PA, Leethochawalit M, Chiamwongpaet S, Kittimunkong S, van Griensven F, McNicholl JM, Paxton L, Choopanya K, for the Bangkok Tenofovir Study Group. Enrollment characteristics and risk behaviors of injection drug users participating in the Bangkok Tenofovir Study, Thailand. PLoS One 2011; 6:e25127. [PMID: 21969870 PMCID: PMC3182181 DOI: 10.1371/journal.pone.0025127] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Accepted: 08/25/2011] [Indexed: 11/30/2022] Open
Abstract
Background The Bangkok Tenofovir Study was launched in 2005 to determine if pre-exposure prophylaxis with tenofovir will reduce the risk of HIV infection among injecting drug users (IDUs). We describe recruitment, screening, enrollment, and baseline characteristics of study participants and contrast risk behavior of Tenofovir Study participants with participants in the 1999–2003 AIDSVAX B/E Vaccine Trial. Methods The Bangkok Tenofovir Study is an ongoing, phase-3, randomized, double-blind, placebo-controlled, HIV pre-exposure prophylaxis trial of daily oral tenofovir. The Tenofovir Study and the Vaccine Trial were conducted among IDUs at 17 drug-treatment clinics in Bangkok. Tenofovir Study sample size was based on HIV incidence in the Vaccine Trial. Standardized questionnaires were used to collect demographic, risk behavior, and incarceration data. The Tenofovir Study is registered with ClinicalTrials.gov, number-NCT00119106. Results From June 2005 through July 2010, 4094 IDUs were screened and 2413 enrolled in the Bangkok Tenofovir Study. The median age of enrolled participants was 31 years (range, 20–59), 80% were male, and 63% reported they injected drugs during the 3 months before enrollment. Among those who injected, 53% injected methamphetamine, 37% midazolam, and 35% heroin. Tenofovir Study participants were less likely to inject drugs, inject daily, or share needles (all, p<0.001) than Vaccine Trial participants. Discussion The Bangkok Tenofovir Study has been successfully launched and is fully enrolled. Study participants are significantly less likely to report injecting drugs and sharing needles than participants in the 1999–2003 AIDSVAX B/E Vaccine Trial suggesting HIV incidence will be lower than expected. In response, the Bangkok Tenofovir Study enrollment was increased from 1600 to 2400 and the study design was changed from a defined 1-year follow-up period to an endpoint-driven design. Trial results demonstrating whether or not daily oral tenofovir reduces the risk of HIV infection among IDUs are expected in 2012.
Collapse
Affiliation(s)
- Michael Martin
- Thailand Ministry of Public Health – US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
- * E-mail:
| | | | | | | | - Rutt Chuachoowong
- Thailand Ministry of Public Health – US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Philip A. Mock
- Thailand Ministry of Public Health – US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | | | | | | | - Frits van Griensven
- Thailand Ministry of Public Health – US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
| | - Janet M. McNicholl
- Thailand Ministry of Public Health – US Centers for Disease Control and Prevention Collaboration, Nonthaburi, Thailand
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Lynn Paxton
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | | | | |
Collapse
|
27
|
Pharmacokinetics and safety of single-dose tenofovir disoproxil fumarate and emtricitabine in HIV-1-infected pregnant women and their infants. Antimicrob Agents Chemother 2011; 55:5914-22. [PMID: 21896911 DOI: 10.1128/aac.00544-11] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Tenofovir (TFV) is effective in preventing simian immunodeficiency virus (SIV) transmission in a macaque model, is available as the oral agent tenofovir disoproxil fumarate (TDF), and may be useful in the prevention of mother-to-child transmission of human immunodeficiency virus (HIV). We conducted a trial of TDF and TDF-emtricitabine (FTC) in HIV-infected pregnant women and their infants. Women received a single dose of either 600 mg TDF, 900 mg TDF, or 900 mg TDF-600 mg FTC at labor onset or prior to a cesarean section. Infants received no drug or a single dose of TDF at 4 mg/kg of body weight or of TDF at 4 mg/kg plus FTC at 3 mg/kg as soon as possible after birth. All regimens were safe and well tolerated. Maternal areas under the serum concentration-time curve (AUC) and concentrations at the end of sampling after 24 h (C(24)) were similar between the two doses of TDF; the maximum concentrations of the drugs in serum (C(max)) and cord blood concentrations were higher in women delivering via cesarean section than in those who delivered vaginally (P = 0.04 and 0.046, respectively). The median ratio of the TFV concentration in cord blood to that in the maternal plasma at delivery was 0.73 (range, 0.26 to 1.95). Without TDF administration, infants had a median TFV concentration of 12 ng/ml 12 h after birth. Following administration of a single dose of TDF at 4 mg/kg, infant TFV concentrations fell below the targeted level, 50 ng/ml, by 24 h postdose. In HIV-infected pregnant women and their infants, 600 mg of TDF is acceptable as a single dose during labor. Low concentrations at birth support infant dosing as soon after birth as possible. Rapidly decreasing TFV levels in infants suggest that multiple or higher doses of TDF will be necessary to maintain concentrations that are effective for viral suppression.
Collapse
|
28
|
Abstract
Reducing the incidence of HIV remains one of our greatest public health challenges. However, there is growing optimism that preexposure prophylaxis (PrEP) could have a major impact on preventing incident HIV infection. Recently presented data on the use of oral PrEP in men who have sex with men (MSM) have provided proof-of-principle for this strategy. Additional clinical trials are evaluating whether PrEP provides similar protection to risk groups other than MSM, such as heterosexual persons and injection drug users. Still unanswered questions include optimal dosing strategies, long-term safety, maximizing adherence and minimizing costs, addressing drug resistance in the face of PrEP failure, optimizing access, and assessing effects on risk behavior. Future implementation will be guided by the results of clinical trials in progress. This article provides a review of the data on the potential strengths and limitations of PrEP as an HIV prevention strategy, identifies challenges to implementation of this approach, and outlines knowledge gaps.
Collapse
Affiliation(s)
- Theodoros Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
29
|
Romanelli F, Murphy B. Systemic preexposure prophylaxis for human immunodeficiency virus infection. Pharmacotherapy 2011; 30:1021-30. [PMID: 20874040 DOI: 10.1592/phco.30.10.1021] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Antiretroviral therapy has significantly improved the typical course of human immunodeficiency virus (HIV) infection in industrialized nations, and life expectancies associated with the infection have increased. However, infection rates have generally remained unchanged, with increases noted among certain subpopulations. The use of systemic preexposure prophylaxis for HIV infection has been proposed as an intervention to reduce the risk of disease transmission in at-risk individuals. The basis of this prophylaxis involves the orchestrated use of antiretrovirals in uninfected individuals either continuously or just before high-risk situations, such as perinatal and occupational exposure to HIV, in order to reduce the likelihood of successful HIV infection. Data from the use of antiretrovirals to prevent HIV infection in these scenarios support the concept of preexposure prophylaxis. Preliminary animal studies have focused on the use of antiretrovirals to prevent simian immunodeficiency virus infection in macaque monkeys, and these data have provided support for the potential efficacy of preexposure prophylaxis for HIV in humans. Limited human data are available, however, but studies are ongoing. Clinical trials have focused on the use of tenofovir disoproxil fumarate either alone or in combination with emtricitabine. Tenofovir-emtricitabine-based regimens may be ideal, given the drugs' pharmacodynamic and pharmacokinetic properties. Some investigators have surveyed at-risk individuals to assess their knowledge of preexposure prophylaxis and whether they used or intended to use this prevention strategy. Routine use of preexposure prophylaxis and even knowledge of its existence appear to be very limited. If efficacy is proved, use of preexposure prophylaxis faces several ethical issues. Ultimately, its success will depend on proof of cost-effectiveness. Until the many questions concerning optimal use of preexposure prophylaxis for HIV are answered, however, its use should be limited to research-related clinical investigations.
Collapse
Affiliation(s)
- Frank Romanelli
- Colleges of Pharmacy, University of Kentucky, Lexington, Kentucky 40536, USA.
| | | |
Collapse
|
30
|
Anderson PL, Kiser JJ, Gardner EM, Rower JE, Meditz A, Grant RM. Pharmacological considerations for tenofovir and emtricitabine to prevent HIV infection. J Antimicrob Chemother 2011; 66:240-50. [PMID: 21118913 PMCID: PMC3019086 DOI: 10.1093/jac/dkq447] [Citation(s) in RCA: 150] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The use of antiretroviral medications in HIV-negative individuals as pre-exposure prophylaxis (PrEP) is a promising approach to prevent HIV infection. Tenofovir disoproxil fumarate (TDF) and emtricitabine exhibit desirable properties for PrEP including: favourable pharmacokinetics that support infrequent dosing; few major drug-drug or drug-food interactions; an excellent clinical safety record; and pre-clinical evidence for efficacy. Several large, randomized, controlled clinical trials are evaluating the safety and efficacy of TDF and emtricitabine for this new indication. A thorough understanding of variability in drug response will help determine future investigations in the field and/or implementation into clinical care. Because tenofovir and emtricitabine are nucleos(t)ide analogues, the HIV prevention and toxicity effects depend on the triphosphate analogue formed intracellularly. This review identifies important cellular pharmacology considerations for tenofovir and emtricitabine, which include drug penetration into relevant tissues and cell types, race/ethnicity/pharmacogenetics, gender, cellular activation state and appropriate episodic or alternative dosing strategies based on pharmacokinetic principles. The current state of knowledge in these areas is summarized and the future utility of intracellular pharmacokinetics/pharmacodynamics for the PrEP field is discussed.
Collapse
Affiliation(s)
- Peter L Anderson
- Department of Pharmaceutical Sciences, University of Colorado Denver, Aurora, CO, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Myers GM, Mayer KH. Oral preexposure anti-HIV prophylaxis for high-risk U.S. populations: current considerations in light of new findings. AIDS Patient Care STDS 2011; 25:63-71. [PMID: 21284497 DOI: 10.1089/apc.2010.0222] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
This article reviews the status of current research evaluating oral preexposure prophylaxis (PrEP) for prevention of HIV infection in high-risk populations. In animal model studies, the use of antiretrovirals has been shown to be effective in preventing HIV acquisition. Early-phase PrEP studies have established safety in humans. Currently, more than 20,000 men and women will soon be enrolled in studies of oral or topical chemoprophylaxis, testing a variety of drug delivery methods including tenofovir disoproxil fumarate (TDF) gel applied vaginally or rectally, as well as oral PrEP using TDF by itself or coformulated with emtricitabine (FTC). The largest global PrEP trial in men who have sex with men (MSM), known as iPrEx has demonstrated that oral chemoprophylaxis can decrease HIV incidence in this population. Although TDF/FTC PrEP was generally well tolerated, side effects such as nausea, as well as mild and reversible renal abnormalities were increased among the men who received active medication, suggesting that PrEP users will need ongoing PrEP clinical monitoring. The prophylactic benefits of TDF/FTC were substantially attenuated by nonadherence, indicating that effective PrEP implementation programs will need to focus on this behavioral variable, in addition to safer sex counseling. This article considers biological, policy, and practical implications of large-scale oral PrEP implementation.
Collapse
Affiliation(s)
- Gavin M. Myers
- Alpert Medical School of Brown University, Department of Community Health, Providence, Rhode Island
| | - Kenneth H. Mayer
- Alpert Medical School of Brown University, Department of Community Health, Providence, Rhode Island
- The Miriam Hospital, Providence, Rhode Island
- Fenway Institute, Boston, Massachusetts
| |
Collapse
|
32
|
Neff CP, Ndolo T, Tandon A, Habu Y, Akkina R. Oral pre-exposure prophylaxis by anti-retrovirals raltegravir and maraviroc protects against HIV-1 vaginal transmission in a humanized mouse model. PLoS One 2010; 5:e15257. [PMID: 21203568 PMCID: PMC3006206 DOI: 10.1371/journal.pone.0015257] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 11/02/2010] [Indexed: 12/31/2022] Open
Abstract
Sexual HIV-1 transmission by vaginal route is the most predominant mode of viral transmission, resulting in millions of new infections every year. In the absence of an effective vaccine, there is an urgent need to develop other alternative methods of pre-exposure prophylaxis (PrEP). Many novel drugs that are currently approved for clinical use also show great potential to prevent viral sexual transmission when administered systemically. A small animal model that permits rapid preclinical evaluation of potential candidates for their systemic PrEP efficacy will greatly enhance progress in this area of investigation. We have previously shown that RAG-hu humanized mouse model permits HIV-1 mucosal transmission via both vaginal and rectal routes and displays CD4 T cell loss typical to that seen in the human. Thus far systemic PrEP studies have been primarily limited to RT inhibitors exemplified by tenofovir and emtricitabine. In these proof-of-concept studies we evaluated two new classes of clinically approved drugs with different modes of action namely, an integrase inhibitor raltegravir and a CCR5 inhibitor maraviroc as potential systemically administered chemo-prophylactics. Our results showed that oral administration of either of these drugs fully protects against vaginal HIV-1 challenge in the RAG-hu mouse model. Based on these results both these drugs show great promise for further development as orally administered PrEPs.
Collapse
Affiliation(s)
- C. Preston Neff
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Thomas Ndolo
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Apurva Tandon
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Yuichiro Habu
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
| | - Ramesh Akkina
- Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, Colorado, United States of America
- * E-mail:
| |
Collapse
|
33
|
García-Lerma JG, Cong ME, Mitchell J, Youngpairoj AS, Zheng Q, Masciotra S, Martin A, Kuklenyik Z, Holder A, Lipscomb J, Pau CP, Barr JR, Hanson DL, Otten R, Paxton L, Folks TM, Heneine W. Intermittent prophylaxis with oral truvada protects macaques from rectal SHIV infection. Sci Transl Med 2010; 2:14ra4. [PMID: 20371467 DOI: 10.1126/scitranslmed.3000391] [Citation(s) in RCA: 142] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
HIV continues to spread globally, mainly through sexual contact. Despite advances in treatment and care, preventing transmission with vaccines or microbicides has proven difficult. A promising strategy to avoid transmission is prophylactic treatment with antiretroviral drugs before exposure to HIV. Clinical trials evaluating the efficacy of daily treatment with the reverse transcriptase inhibitors tenofovir disoproxil fumarate (TDF) or Truvada (TDF plus emtricitabine) are under way. We hypothesized that intermittent prophylactic treatment with long-acting antiviral drugs would be as effective as daily dosing in blocking the earliest stages of viral replication and preventing mucosal transmission. We tested this hypothesis by intermittently giving prophylactic Truvada to macaque monkeys and then exposing them rectally to simian-human immunodeficiency virus (SHIV) once a week for 14 weeks. A simple regimen with an oral dose of Truvada given 1, 3, or 7 days before exposure followed by a second dose 2 hours after exposure was as protective as daily drug administration, possibly because of the long intracellular persistence of the drugs. In addition, a two-dose regimen initiated 2 hours before or after virus exposure was effective, and full protection was obtained by doubling the Truvada concentration in both doses. We saw no protection if the first dose was delayed until 24 hours after exposure, underscoring the importance of blocking initial replication in the mucosa. Our results show that intermittent prophylactic treatment with an antiviral drug can be highly effective in preventing SHIV infection, with a wide window of protection. They strengthen the possibility of developing feasible, cost-effective strategies to prevent HIV transmission in humans.
Collapse
Affiliation(s)
- J Gerardo García-Lerma
- Division of HIV/AIDS Prevention, National Center for HIV, Hepatitis, STD, and Prevention, Centers for Disease Control and Prevention, 1600 Clifton Road, Atlanta, GA 30329, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Vaccines and microbicides preventing HIV-1, HSV-2, and HPV mucosal transmission. J Invest Dermatol 2009; 130:352-61. [PMID: 19829304 DOI: 10.1038/jid.2009.227] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
HIV-1, herpes simplex virus type 2 (HSV-2), and human papillomavirus (HPV), among other sexually transmitted infections, represent a major burden for global health. Initial insights into the mucosal transmission of these viral pathogens have raised optimism with regard to the rapid generation of protective vaccines. Nevertheless, setbacks for HIV-1 and HSV-2 vaccines have seriously challenged the initial enthusiasm. Recently, two new vaccines that efficiently prevented HPV infection have renewed the hope that vaccinal prevention of viral mucosal sexually transmitted infections is possible. HIV-1 and HSV-2 differ from HPV, and each virus needs to be tackled with a distinct approach. However, vaccines are not the only possible answer. Topically applied agents (microbicides) are an attractive alternative in the prevention of HIV-1 and HSV-2 mucosal transmission. Progress in understanding the mechanisms of genital transmission of HIV-1 and HSV-2 is required for successful vaccine or microbicide candidates to emerge from current approaches.
Collapse
|
35
|
Van Rompay KKA. Evaluation of antiretrovirals in animal models of HIV infection. Antiviral Res 2009; 85:159-75. [PMID: 19622373 DOI: 10.1016/j.antiviral.2009.07.008] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 07/07/2009] [Accepted: 07/13/2009] [Indexed: 01/07/2023]
Abstract
Animal models of HIV infection have played an important role in the development of antiretroviral drugs. Although each animal model has its limitations and never completely mimics HIV infection of humans, a carefully designed study allows experimental approaches that are not feasible in humans, but that can help to better understand disease pathogenesis and to provide proof-of-concept of novel intervention strategies. While rodent and feline models are useful for initial screening, further testing is best done in non-human primate models, such as simian immunodeficiency virus (SIV) infection of macaques, because they share more similarities with HIV infection of humans. In the early years of the HIV pandemic, non-human primate models played a relatively minor role in the antiretroviral drug development process. Since then, a better understanding of the disease and the development of better drugs and assays to monitor antiviral efficacy have increased the usefulness of the animal models. In particular, non-human primate models have provided proof-of-concept for (i) the benefits of chemoprophylaxis and early treatment, (ii) the preclinical efficacy of novel drugs such as tenofovir, (iii) the virulence and clinical significance of drug-resistant viral mutants, and (iv) the role of antiviral immune responses during drug therapy. Ongoing comparison of results obtained in animal models with those observed in human studies will further validate and improve these animal models so they can continue to help advance our scientific knowledge and to guide clinical trials. 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)
- Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, CA 95616, USA.
| |
Collapse
|
36
|
The effect of ongoing exposure dynamics in dose response relationships. PLoS Comput Biol 2009; 5:e1000399. [PMID: 19503605 PMCID: PMC2685010 DOI: 10.1371/journal.pcbi.1000399] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2008] [Accepted: 05/04/2009] [Indexed: 11/20/2022] Open
Abstract
Characterizing infectivity as a function of pathogen dose is integral to
microbial risk assessment. Dose-response experiments usually administer doses to
subjects at one time. Phenomenological models of the resulting data, such as the
exponential and the Beta-Poisson models, ignore dose timing and assume
independent risks from each pathogen. Real world exposure to pathogens, however,
is a sequence of discrete events where concurrent or prior pathogen arrival
affects the capacity of immune effectors to engage and kill newly arriving
pathogens. We model immune effector and pathogen interactions during the period
before infection becomes established in order to capture the dynamics generating
dose timing effects. Model analysis reveals an inverse relationship between the
time over which exposures accumulate and the risk of infection. Data from one
time dose experiments will thus overestimate per pathogen infection risks of
real world exposures. For instance, fitting our model to one time dosing data
reveals a risk of 0.66 from 313 Cryptosporidium parvum
pathogens. When the temporal exposure window is increased 100-fold using the
same parameters fitted by our model to the one time dose data, the risk of
infection is reduced to 0.09. Confirmation of this risk prediction requires data
from experiments administering doses with different timings. Our model
demonstrates that dose timing could markedly alter the risks generated by
airborne versus fomite transmitted pathogens. We model the relationship between the temporal patterns of pathogen exposure and
infection take off within people. Since different routes of transmission (e.g.,
airborne versus surface transfer routes) may result in different temporal
patterns of exposure, this model helps to better compare the risks of
transmission from one person to another through these different routes. Previous
models assumed that the risk of infection is the same whether pathogens are
inoculated all at once or over one day. Our model, in contrast, captures how one
pathogen affects the potential of immunity to keep concurrently or subsequently
arriving particles from initiating an infection. Since the pattern of timing of
airborne and surface spread pathogen arrivals differ, our model shows that each
airborne pathogen could carry less risk than each surface transmitted pathogen.
Unfortunately, data to fully fit our model are not currently available.
Therefore new experiments will have to be conducted where doses are given across
different temporal windows.
Collapse
|
37
|
Schulte R, Suh YS, Sauermann U, Ochieng W, Sopper S, Kim KS, Ahn SS, Park KS, Stolte-Leeb N, Hunsmann G, Sung YC, Stahl-Hennig C. Mucosal prior to systemic application of recombinant adenovirus boosting is more immunogenic than systemic application twice but confers similar protection against SIV-challenge in DNA vaccine-primed macaques. Virology 2009; 383:300-9. [DOI: 10.1016/j.virol.2008.10.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 08/21/2008] [Accepted: 10/08/2008] [Indexed: 10/21/2022]
|
38
|
HIV Postexposure Prophylaxis Use Among Ontario Female Adolescent Sexual Assault Victims: A Prospective Analysis. Sex Transm Dis 2008; 35:973-8. [DOI: 10.1097/olq.0b013e3181824f3c] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Van Rompay KKA, Durand-Gasselin L, Brignolo LL, Ray AS, Abel K, Cihlar T, Spinner A, Jerome C, Moore J, Kearney BP, Marthas ML, Reiser H, Bischofberger N. Chronic administration of tenofovir to rhesus macaques from infancy through adulthood and pregnancy: summary of pharmacokinetics and biological and virological effects. Antimicrob Agents Chemother 2008; 52:3144-60. [PMID: 18573931 PMCID: PMC2533487 DOI: 10.1128/aac.00350-08] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2008] [Revised: 04/21/2008] [Accepted: 06/16/2008] [Indexed: 02/07/2023] Open
Abstract
The reverse transcriptase (RT) inhibitor tenofovir (TFV) is highly effective in the simian immunodeficiency virus (SIV) macaque model of human immunodeficiency virus infection. The current report describes extended safety and efficacy data on 32 animals that received prolonged (>or=1- to 13-year) daily subcutaneous TFV regimens. The likelihood of renal toxicity (proximal renal tubular dysfunction [PRTD]) correlated with plasma drug concentrations, which depended on the dosage regimen and age-related changes in drug clearance. Below a threshold area under the concentration-time curve for TFV in plasma of approximately 10 microg x h/ml, an exposure severalfold higher than that observed in humans treated orally with 300 mg TFV disoproxil fumarate (TDF), prolonged TFV administration was not associated with PRTD based on urinalysis, serum chemistry analyses, bone mineral density, and clinical observations. At low-dose maintenance regimens, plasma TFV concentrations and intracellular TFV diphosphate concentrations were similar to or slightly higher than those observed in TDF-treated humans. No new toxicities were identified. The available evidence does not suggest teratogenic effects of prolonged low-dose TFV treatment; by the age of 10 years, one macaque, on TFV treatment since birth, had produced three offspring that were healthy by all criteria up to the age of 5 years. Despite the presence of viral variants with a lysine-to-arginine substitution at codon 65 (K65R) of RT in all 28 SIV-infected animals, 6 animals suppressed viremia to undetectable levels for as long as 12 years of TFV monotherapy. In conclusion, these findings illustrate the safety and sustained benefits of prolonged TFV-containing regimens throughout development from infancy to adulthood, including pregnancy.
Collapse
Affiliation(s)
- Koen K A Van Rompay
- California National Primate Research Center, University of California, Davis, California 95616, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cranage M, Sharpe S, Herrera C, Cope A, Dennis M, Berry N, Ham C, Heeney J, Rezk N, Kashuba A, Anton P, McGowan I, Shattock R. Prevention of SIV rectal transmission and priming of T cell responses in macaques after local pre-exposure application of tenofovir gel. PLoS Med 2008; 5:e157; discussion e157. [PMID: 18684007 PMCID: PMC2494562 DOI: 10.1371/journal.pmed.0050157] [Citation(s) in RCA: 129] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2007] [Accepted: 06/09/2008] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The rectum is particularly vulnerable to HIV transmission having only a single protective layer of columnar epithelium overlying tissue rich in activated lymphoid cells; thus, unprotected anal intercourse in both women and men carries a higher risk of infection than other sexual routes. In the absence of effective prophylactic vaccines, increasing attention is being given to the use of microbicides and preventative antiretroviral (ARV) drugs. To prevent mucosal transmission of HIV, a microbicide/ARV should ideally act locally at and near the virus portal of entry. As part of an integrated rectal microbicide development programme, we have evaluated rectal application of the nucleotide reverse transcriptase (RT) inhibitor tenofovir (PMPA, 9-[(R)-2-(phosphonomethoxy) propyl] adenine monohydrate), a drug licensed for therapeutic use, for protective efficacy against rectal challenge with simian immunodeficiency virus (SIV) in a well-established and standardised macaque model. METHODS AND FINDINGS A total of 20 purpose-bred Indian rhesus macaques were used to evaluate the protective efficacy of topical tenofovir. Nine animals received 1% tenofovir gel per rectum up to 2 h prior to virus challenge, four macaques received placebo gel, and four macaques remained untreated. In addition, three macaques were given tenofovir gel 2 h after virus challenge. Following intrarectal instillation of 20 median rectal infectious doses (MID50) of a noncloned, virulent stock of SIVmac251/32H, all animals were analysed for virus infection, by virus isolation from peripheral blood mononuclear cells (PBMC), quantitative proviral DNA load in PBMC, plasma viral RNA (vRNA) load by sensitive quantitative competitive (qc) RT-PCR, and presence of SIV-specific serum antibodies by ELISA. We report here a significant protective effect (p = 0.003; Fisher exact probability test) wherein eight of nine macaques given tenofovir per rectum up to 2 h prior to virus challenge were protected from infection (n = 6) or had modified virus outcomes (n = 2), while all untreated macaques and three of four macaques given placebo gel were infected, as were two of three animals receiving tenofovir gel after challenge. Moreover, analysis of lymphoid tissues post mortem failed to reveal sequestration of SIV in the protected animals. We found a strong positive association between the concentration of tenofovir in the plasma 15 min after rectal application of gel and the degree of protection in the six animals challenged with virus at this time point. Moreover, colorectal explants from non-SIV challenged tenofovir-treated macaques were resistant to infection ex vivo, whereas no inhibition was seen in explants from the small intestine. Tissue-specific inhibition of infection was associated with the intracellular detection of tenofovir. Intriguingly, in the absence of seroconversion, Gag-specific gamma interferon (IFN-gamma)-secreting T cells were detected in the blood of four of seven protected animals tested, with frequencies ranging from 144 spot forming cells (SFC)/10(6) PBMC to 261 spot forming cells (SFC)/10(6) PBMC. CONCLUSIONS These results indicate that colorectal pretreatment with ARV drugs, such as tenofovir, has potential as a clinically relevant strategy for the prevention of HIV transmission. We conclude that plasma tenofovir concentration measured 15 min after rectal administration may serve as a surrogate indicator of protective efficacy. This may prove to be useful in the design of clinical studies. Furthermore, in vitro intestinal explants served as a model for drug distribution in vivo and susceptibility to virus infection. The finding of T cell priming following exposure to virus in the absence of overt infection is provocative. Further studies would reveal if a combined modality microbicide and vaccination strategy is feasible by determining the full extent of local immune responses induced and their protective potential.
Collapse
Affiliation(s)
- Martin Cranage
- Centre for Infection, Division of Cellular & Molecular Medicine, St George's University of London, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
García-Lerma JG, Otten RA, Qari SH, Jackson E, Cong ME, Masciotra S, Luo W, Kim C, Adams DR, Monsour M, Lipscomb J, Johnson JA, Delinsky D, Schinazi RF, Janssen R, Folks TM, Heneine W. Prevention of rectal SHIV transmission in macaques by daily or intermittent prophylaxis with emtricitabine and tenofovir. PLoS Med 2008; 5:e28. [PMID: 18254653 PMCID: PMC2225435 DOI: 10.1371/journal.pmed.0050028] [Citation(s) in RCA: 275] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2007] [Accepted: 12/18/2007] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND In the absence of an effective vaccine, HIV continues to spread globally, emphasizing the need for novel strategies to limit its transmission. Pre-exposure prophylaxis (PrEP) with antiretroviral drugs could prove to be an effective intervention strategy if highly efficacious and cost-effective PrEP modalities are identified. We evaluated daily and intermittent PrEP regimens of increasing antiviral activity in a macaque model that closely resembles human transmission. METHODS AND FINDINGS We used a repeat-exposure macaque model with 14 weekly rectal virus challenges. Three drug treatments were given once daily, each to a different group of six rhesus macaques. Group 1 was treated subcutaneously with a human-equivalent dose of emtricitabine (FTC), group 2 received orally the human-equivalent dosing of both FTC and tenofovir-disoproxil fumarate (TDF), and group 3 received subcutaneously a similar dosing of FTC and a higher dose of tenofovir. A fourth group of six rhesus macaques (group 4) received intermittently a PrEP regimen similar to group 3 only 2 h before and 24 h after each weekly virus challenge. Results were compared to 18 control macaques that did not receive any drug treatment. The risk of infection in macaques treated in groups 1 and 2 was 3.8- and 7.8-fold lower than in untreated macaques (p = 0.02 and p = 0.008, respectively). All six macaques in group 3 were protected. Breakthrough infections had blunted acute viremias; drug resistance was seen in two of six animals. All six animals in group 4 that received intermittent PrEP were protected. CONCLUSIONS This model suggests that single drugs for daily PrEP can be protective but a combination of antiretroviral drugs may be required to increase the level of protection. Short but potent intermittent PrEP can provide protection comparable to that of daily PrEP in this SHIV/macaque model. These findings support PrEP trials for HIV prevention in humans and identify promising PrEP modalities.
Collapse
Affiliation(s)
- J. Gerardo García-Lerma
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Ron A Otten
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Shoukat H Qari
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Eddie Jackson
- Division of Scientific Resources, National Center for Preparedness, Detection, and Control of Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Mian-er Cong
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Silvina Masciotra
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Wei Luo
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Caryn Kim
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Debra R Adams
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Michael Monsour
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jonathan Lipscomb
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Jeffrey A Johnson
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - David Delinsky
- Emory University School of Medicine/Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Raymond F Schinazi
- Emory University School of Medicine/Veterans Affairs Medical Center, Decatur, Georgia, United States of America
| | - Robert Janssen
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Thomas M Folks
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Walid Heneine
- Division of HIV/AIDS Prevention, National Center for HIV/AIDS, Viral Hepatitis, STD, and TB Prevention, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| |
Collapse
|
42
|
Jagannathan P, Landovitz R, Roland ME. Postexposure prophylaxis after sexual exposure to HIV. ACTA ACUST UNITED AC 2007. [DOI: 10.2217/17469600.1.1.35] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Increasing numbers of international, national and state/provincial health organizations in the developed and developing world recommend postexposure prophylaxis (PEP) following potential sexual exposure to HIV. The evidence for these policies is extrapolated from occupational healthcare worker, perinatal prophylaxis and animal studies. There is no direct evidence of PEP efficacy after sexual exposures. Studies addressing potential increases in risk behavior, toxicities and cost–effectiveness are encouraging. Considerations for offering PEP include the timing and characteristics of the exposure and the HIV status of the source and exposed individuals. PEP includes 28 days of a combination antiretroviral drug regimen containing two or more drugs and associated laboratory testing, counseling and referrals. PEP service delivery challenges include ensuring adequate access to services, optimizing PEP adherence and facilitating follow-up HIV testing and counseling. Men who have sex with men, sexual assault survivors, children and adolescents, and individuals in resource-limited settings present unique needs and challenges.
Collapse
Affiliation(s)
- Prasanna Jagannathan
- University of California, San Francisco, California, Department of General Internal Medicine, San Francisco General Hospital Ward 13, 1001 Potrero Avenue, San Francisco, CA 94110, USA
| | - Raphael Landovitz
- University of California, Los Angeles, Center for Clinical AIDS Research and Education, David Geffen School of Medicine, 9 911 W Pico Blvd., Suite 980 Los Angeles, CA 90035, USA
| | - Michelle E Roland
- University of California. San Francisco, Positive Health Program, Ward 84, San Francisco General Hospital, 995 Potrero Avenue, San Francisco, CA 94110, USA
| |
Collapse
|
43
|
Nikolic DS, Garcia E, Piguet V. Microbicides and other topical agents in the prevention of HIV and sexually transmitted infections. Expert Rev Anti Infect Ther 2007; 5:77-88. [PMID: 17266456 DOI: 10.1586/14787210.5.1.77] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
According to information from UNAIDS, more than 42 million individuals are living with HIV worldwide. Most infected individuals live in developing countries where the availability of antiretroviral agents is still limited. As this pandemic is increasing largely through mucosal transmission, new methods of prevention are urgently needed. If available, agents that block HIV prior to or early after contact with mucosal epithelia would decrease the incidence of HIV infection and, therefore, potentially save millions of lives over the next few decades. Topically applied microbicides acting against HIV-1 can be subdivided into four subgroups, including agents directly inhibiting pathogens, agents acting on genital pH, agents blocking pathogen entry and replication inhibitors. In addition, microbicides might also allow fighting against other sexually transmitted infections, such as herpes simplex viruses. With concerted efforts directed towards developing efficient microbicides, topical anti-infective compounds may well become a new weapon against sexually transmitted infections, including HIV, in everyday clinical practice.
Collapse
Affiliation(s)
- Damjan S Nikolic
- Department of Dermatology and Venereology, University Hospital and Medical School of Geneva, Geneva, Switzerland.
| | | | | |
Collapse
|
44
|
Abstract
PURPOSE OF REVIEW HIV postexposure prophylaxis is often recommended following potential sexual exposure to HIV. Recent data address the effectiveness of postexposure prophylaxis and prevention counseling, cost-effectiveness, antiretroviral options, challenges with nonoccupational postexposure prophylaxis among adolescents and children and following sexual assault in high HIV prevalence areas, and a successful program in Amsterdam. RECENT FINDINGS Postexposure prophylaxis is not completely protective. Seroconversion may result from antiretroviral failure or from ongoing exposures. Postexposure prophylaxis associated risk reduction counseling results in reductions in subsequent risk behavior. Programs that target outreach and limit prescriptions to those with exposure sources who are at risk of being HIV infected are cost-effective. Less restrictive guidelines result in more prescriptions for low-risk exposures; this practice is not cost-effective. The ideal antiretrovirals for postexposure prophylaxis use have not been established. Tenofovir has several attractive properties. Developing systems to support the effective delivery of postexposure prophylaxis among children and adolescents and following sexual assault in high HIV prevalence, resource limited settings is challenging. SUMMARY Numerous national and international guidelines recommend postexposure prophylaxis following potential sexual exposure to HIV. Maximizing adherence and minimizing subsequent HIV exposures will be critical to enhancing the effectiveness of this HIV prevention intervention.
Collapse
Affiliation(s)
- Michelle E Roland
- Positive Health (AIDS) Program at San Francisco General Hospital, University of California, San Francisco, California 94110, USA.
| |
Collapse
|