Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
PLoS Pathog ; 12(9): e1005885, 2016 09.
Article in English | MEDLINE | ID: mdl-27658293

ABSTRACT

Currently, there are mounting data suggesting that HIV-1 acquisition in women can be affected by the use of certain hormonal contraceptives. However, in non-human primate models, endogenous or exogenous progestin-dominant states are shown to increase acquisition. To gain mechanistic insights into this increased acquisition, we studied how mucosal barrier function and CD4+ T-cell and CD68+ macrophage density and localization changed in the presence of natural progestins or after injection with high-dose DMPA. The presence of natural or injected progestins increased virus penetration of the columnar epithelium and the infiltration of susceptible cells into a thinned squamous epithelium of the vaginal vault, increasing the likelihood of potential virus interactions with target cells. These data suggest that increasing either endogenous or exogenous progestin can alter female reproductive tract barrier properties and provide plausible mechanisms for increased HIV-1 acquisition risk in the presence of increased progestin levels.


Subject(s)
Host-Pathogen Interactions/drug effects , Macrophages/drug effects , Mucous Membrane/drug effects , Progestins/therapeutic use , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/drug effects , Vagina/drug effects , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/virology , Cervix Uteri/drug effects , Cervix Uteri/immunology , Cervix Uteri/metabolism , Cervix Uteri/virology , Delayed-Action Preparations , Female , Injections, Intramuscular , Lymphocyte Activation/drug effects , Macaca mulatta , Macaca nemestrina , Macrophage Activation/drug effects , Macrophages/immunology , Macrophages/metabolism , Macrophages/virology , Medroxyprogesterone Acetate/administration & dosage , Medroxyprogesterone Acetate/therapeutic use , Menstrual Cycle , Mucous Membrane/immunology , Mucous Membrane/metabolism , Mucous Membrane/virology , Progestins/administration & dosage , Progestins/metabolism , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/immunology , Simian Immunodeficiency Virus/physiology , Vagina/immunology , Vagina/metabolism , Vagina/virology , Virus Internalization/drug effects
2.
Vaccine ; 34(51): 6597-6609, 2016 12 12.
Article in English | MEDLINE | ID: mdl-27395563

ABSTRACT

The Brighton Collaboration Viral Vector Vaccines Safety Working Group (V3SWG) was formed to evaluate the safety of live, recombinant viral vaccines incorporating genes from heterologous viral and other microbial pathogens in their genome (so-called "chimeric virus vaccines"). Many such viral vector vaccines are now at various stages of clinical evaluation. Here, we introduce an attenuated form of recombinant vesicular stomatitis virus (rVSV) as a potential chimeric virus vaccine for HIV-1, with implications for use as a vaccine vector for other pathogens. The rVSV/HIV-1 vaccine vector was attenuated by combining two major genome modifications. These modifications acted synergistically to greatly enhance vector attenuation and the resulting rVSV vector demonstrated safety in sensitive mouse and non-human primate neurovirulence models. This vector expressing HIV-1 gag protein has completed evaluation in two Phase I clinical trials. In one trial the rVSV/HIV-1 vector was administered in a homologous two-dose regimen, and in a second trial with pDNA in a heterologous prime boost regimen. No serious adverse events were reported nor was vector detected in blood, urine or saliva post vaccination in either trial. Gag specific immune responses were induced in both trials with highest frequency T cell responses detected in the prime boost regimen. The rVSV/HIV-1 vector also demonstrated safety in an ongoing Phase I trial in HIV-1 positive participants. Additionally, clinical trial material has been produced with the rVSV vector expressing HIV-1 env, and Phase I clinical evaluation will initiate in the beginning of 2016. In this paper, we use a standardized template describing key characteristics of the novel rVSV vaccine vectors, in comparison to wild type VSV. The template facilitates scientific discourse among key stakeholders by increasing transparency and comparability of information. The Brighton Collaboration V3SWG template may also be useful as a guide to the evaluation of other recombinant viral vector vaccines.


Subject(s)
AIDS Vaccines/adverse effects , AIDS Vaccines/immunology , Drug Carriers , Vesiculovirus/genetics , AIDS Vaccines/genetics , Animals , Clinical Trials, Phase I as Topic , Drug Evaluation, Preclinical , Drug-Related Side Effects and Adverse Reactions/epidemiology , Drug-Related Side Effects and Adverse Reactions/pathology , Genetic Vectors , Humans , Primates , Risk Assessment , T-Lymphocytes/immunology , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/genetics , Vaccines, Synthetic/adverse effects , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , env Gene Products, Human Immunodeficiency Virus/genetics , env Gene Products, Human Immunodeficiency Virus/immunology , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
3.
PLoS One ; 10(4): e0120021, 2015.
Article in English | MEDLINE | ID: mdl-25853710

ABSTRACT

BACKGROUND: Personal lubricant use is common during anal intercourse. Some water-based products with high osmolality and low pH can damage genital and rectal tissues, and the polymer polyquaternium 15 (PQ15) can enhance HIV replication in vitro. This has raised concerns that lubricants with such properties may increase STD/HIV infection risk, although in vivo evidence is scarce. We use a macaque model to evaluate rectal cytotoxicity and SHIV infection risk after use of a highly osmolar (>8,000 mOsm/kg) water-based lubricant with pH of 4.4, and containing PQ15. METHODS: Cytotoxicity was documented by measuring inflammatory cytokines and epithelial tissue sloughing during six weeks of repeated, non-traumatic lubricant or control buffer applications to rectum and anus. We measured susceptibility to SHIVSF162P3 infection by comparing virus doses needed for rectal infection in twenty-one macaques treated with lubricant or control buffer 30 minutes prior to virus exposure. RESULTS: Lubricant increased pro-inflammatory cytokines and tissue sloughing while control buffer (phosphate buffered saline; PBS) did not. However, the estimated AID50 (50% animal infectious dose) was not different in lubricant- and control buffer-treated macaques (p = 0.4467; logistic regression models). CONCLUSIONS: Although the test lubricant caused acute cytotoxicity in rectal tissues, it did not increase susceptibility to infection in this macaque model. Thus neither the lubricant-induced type/extent of inflammation nor the presence of PQ15 affected infection risk. This study constitutes a first step in the in vivo evaluation of lubricants with regards to HIV transmission.


Subject(s)
Lubricants/chemistry , Lubricants/toxicity , Rectum/drug effects , Rectum/virology , Simian Acquired Immunodeficiency Syndrome , Simian Immunodeficiency Virus/drug effects , Animals , Epithelium/drug effects , Female , Hemorrhage/chemically induced , Hydrogen-Ion Concentration , Lubricants/administration & dosage , Macaca fascicularis , Microbiota/drug effects , Osmolar Concentration , Rectum/cytology , Rectum/microbiology , Risk , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/physiology , Time Factors , Viremia/chemically induced , Virus Shedding/drug effects , Water/chemistry
4.
AIDS Res Hum Retroviruses ; 30(11): 1072-81, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24914761

ABSTRACT

Vaccination and preexposure prophylaxis (PrEP) with antiretrovirals have shown only partial protection from HIV-1 infection in human trials. Oral Truvada (emtricitabine/tenofovir disoproxil fumarate) is FDA approved as PrEP but partial adherence reduces efficacy. If combined as biomedical preventions (CBP), an HIV vaccine could protect when PrEP adherence is low and PrEP could prevent vaccine breakthroughs. The efficacy of combining oral PrEP with an HIV vaccine has not been evaluated in humans. We determined the efficacy of combining a DNA/virus-like particle (VLP) vaccine with partially effective intermittent PrEP in Indian rhesus macaques (RM). Eight RM received intramuscular inoculations of five DNA plasmids encoding four HIV-1 Clade B primary isolate Envs and SIVmac239 Gag (at weeks 0 and 4), followed by intramuscular and intranasal inoculations of homologous Gag VLPs and four Env VLPs (at weeks 12, 16, and 53). At week 61, we initiated weekly rectal exposures with heterologous SHIV162p3 (10 TCID50) along with oral Truvada (TDF, 22 mg/kg; FTC 20 mg/kg) dosing 2 h before and 22 h after each exposure. This PrEP regimen previously demonstrated 50% efficacy. Five controls (no vaccine, no PrEP) received weekly SHIV162p3. All controls were infected after a median of four exposures; the mean peak plasma viral load (VL) was 3.9×10(7) vRNA copies/ml. CBP protected seven of eight (87.5%) RM. The one infected CBP RM had a reduced peak VL of 8.8×10(5) copies/ml. SHIV exposures during PrEP amplified Gag and Env antibody titers in protected RM. These results suggest that combining oral PrEP with HIV vaccines could enhance protection against HIV-1 infection.


Subject(s)
Anti-HIV Agents/administration & dosage , Deoxycytidine/analogs & derivatives , Organophosphorus Compounds/administration & dosage , Pre-Exposure Prophylaxis/methods , SAIDS Vaccines/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Vaccination/methods , Vaccines, Virus-Like Particle/immunology , Administration, Intranasal , Administration, Oral , Animals , Antibodies, Viral/blood , Chemoprevention/methods , Combined Modality Therapy/methods , Deoxycytidine/administration & dosage , Drug Combinations , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Female , HIV-1/genetics , HIV-1/immunology , Injections, Intramuscular , Macaca mulatta , Male , SAIDS Vaccines/administration & dosage , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology , Treatment Outcome , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vaccines, Virus-Like Particle/administration & dosage , Viral Load
5.
Antimicrob Agents Chemother ; 58(5): 2665-74, 2014 May.
Article in English | MEDLINE | ID: mdl-24566178

ABSTRACT

Vaginal rapidly disintegrating tablets (RDTs) containing tenofovir (TFV) or TFV and emtricitabine (FTC) were evaluated for safety and pharmacokinetics in pigtailed macaques. Two separate animal groups (n = 4) received TFV (10 mg) or TFV-FTC (10 mg each) RDTs, administered near the cervix. A third group (n = 4) received 1 ml TFV gel. Blood plasma, vaginal tissue biopsy specimens, and vaginal fluids were collected before and after product application at 0, 0.5, 1, 4, and 24 h. A disintegration time of <30 min following vaginal application of the RDTs was noted, with negligible effects on local inflammatory cytokines, vaginal pH, and microflora. TFV pharmacokinetics were generally similar for both RDTs and gel, with peak median concentrations in vaginal tissues and vaginal secretions being on the order of 10(4) to 10(5) ng/g (147 to 571 µM) and 10(6) ng/g (12 to 34 mM), respectively, at 1 to 4 h postdose. At 24 h, however, TFV vaginal tissue levels were more sustained after RDT dosing, with median TFV concentrations being approximately 1 log higher than those with gel dosing. FTC pharmacokinetics after combination RDT dosing were similar to those of TFV, with peak median vaginal tissue and fluid levels being on the order of 10(4) ng/g (374 µM) and 10(6) ng/g (32 mM), respectively, at 1 h postdose with levels in fluid remaining high at 24 h. RDTs are a promising alternative vaginal dosage form, delivering TFV and/or FTC at levels that would be considered inhibitory to simian-human immunodeficiency virus in the macaque vaginal microenvironment over a 24-h period.


Subject(s)
Adenine/analogs & derivatives , Deoxycytidine/analogs & derivatives , Organophosphonates/administration & dosage , Organophosphonates/pharmacokinetics , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacokinetics , Adenine/administration & dosage , Adenine/pharmacokinetics , Administration, Intravaginal , Animals , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacokinetics , Emtricitabine , Female , Macaca , Tenofovir
6.
J Med Primatol ; 43(5): 349-59, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24372425

ABSTRACT

BACKGROUND: Long-acting, hormonal contraception may increase HIV risk. Copper intrauterine devices (IUDs) could serve as non-hormonal alternatives. We pilot a pigtail macaque model for evaluating HIV susceptibility factors during copper IUD use. METHODS: Frameless and flexible GyneFix(®) copper IUDs were surgically implanted into three SHIVSF 162p3 -positive macaques via hysterotomy and monitored for up to 4 months. Four macaques served as non-IUD controls. RESULTS: All animals retained the devices without complications. No consistent change in vaginal viral RNA or inflammatory cytokines was seen. Two animals had altered menstrual cycles and experienced marked thinning of vaginal epithelium after IUD insertion. Histological examination of uterine tissue at necropsy revealed endometrial ulceration and lymphocytic inflammation with glandular loss at sites of direct IUD contact. CONCLUSIONS: Although the need for insertion surgery could limit its usefulness, this model will allow studies on copper IUDs and SHIV shedding, disease progression, and HIV susceptibility factors.


Subject(s)
HIV Infections/prevention & control , Intrauterine Devices, Copper/adverse effects , Macaca nemestrina , Simian Acquired Immunodeficiency Syndrome/prevention & control , Animals , Contraception , Disease Models, Animal , Disease Susceptibility/immunology , Disease Susceptibility/physiopathology , Disease Susceptibility/virology , Female , HIV/physiology , HIV Infections/immunology , HIV Infections/physiopathology , HIV Infections/virology , Humans , Macaca nemestrina/immunology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/physiopathology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/isolation & purification , Uterus/immunology , Virus Shedding
7.
Proc Natl Acad Sci U S A ; 110(40): 16145-50, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24043812

ABSTRACT

Topical preexposure prophylaxis interrupts HIV transmission at the site of mucosal exposure. Intermittently dosed vaginal gels containing the HIV-1 reverse transcriptase inhibitor tenofovir protected pigtailed macaques depending on the timing of viral challenge relative to gel application. However, modest or no protection was observed in clinical trials. Intravaginal rings (IVRs) may improve efficacy by providing long-term sustained drug delivery leading to constant mucosal antiretroviral concentrations and enhancing adherence. Although a few IVRs have entered the clinical pipeline, 100% efficacy in a repeated macaque vaginal challenge model has not been achieved. Here we describe a reservoir IVR technology that delivers the tenofovir prodrug tenofovir disoproxil fumarate (TDF) continuously over 28 d. With four monthly ring changes in this repeated challenge model, TDF IVRs generated reproducible and protective drug levels. All TDF IVR-treated macaques (n = 6) remained seronegative and simian-HIV RNA negative after 16 weekly vaginal exposures to 50 tissue culture infectious dose SHIV162p3. In contrast, 11/12 control macaques became infected, with a median of four exposures assuming an eclipse of 7 d from infection to virus RNA detection. Protection was associated with tenofovir levels in vaginal fluid [mean 1.8 × 10(5) ng/mL (range 1.1 × 10(4) to 6.6 × 10(5) ng/mL)] and ex vivo antiviral activity of cervicovaginal lavage samples. These observations support further advancement of TDF IVRs as well as the concept that extended duration drug delivery devices delivering topical antiretrovirals could be effective tools in preventing the sexual transmission of HIV in humans.


Subject(s)
Adenine/analogs & derivatives , Drug Delivery Systems/methods , HIV/drug effects , Lentivirus Infections/prevention & control , Organophosphonates/pharmacology , Simian Immunodeficiency Virus/drug effects , Adenine/administration & dosage , Adenine/pharmacology , Administration, Intravaginal , Animals , Delayed-Action Preparations , Female , Macaca mulatta , Organophosphonates/administration & dosage , Tenofovir
8.
AIDS Res Hum Retroviruses ; 29(7): 1091-4, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23461569

ABSTRACT

Infections following repeated, low-dose (RLD), mucal S(H)IV exposures of macaques are used to model sexual HIV exposures for biomedical prevention testing. Different susceptibilities among animals can complicate study designs. In rhesus macaques, TRIM5 alleles Q, CypA, and TFP are resistance factors for infection with some S(H)IV strains, but not for SIVmac239 due to its capsid properties. SIVmac239-derived SHIVSF162P3 has been demonstrated to reproducibly infect mucosally in vaginal and rectal RLD models. To further test the suitability of SHIVSF162P3 for RLD models, we studied the influence of the TRIM5 genotype on susceptibility to rectal RLD infection and on plasma viremia by analyzing 43 male Indian rhesus macaques from control arms of completed studies. The median number of exposures required for infection was three (Q/Q, n=4) (TRIM5 alleles, number of macaques, respectively), four (Q/CypA, n=7), three (TFP/Q, n=15), three (TFP/TFP, n=15), and two (TFP/CypA, n=2); TRIM5(CypA/CypA) was not represented in our study. Median peak viremia (log10 viral copies/ml) in infected animals was 7.4 (Q/Q, n=4), 7.2 (Q/CypA, n=6), 7.3 (TFP/Q, n=13), 7.1 (TFP/TFP, n=15), and 6.5 (TFP/CypA; n=2). Neither susceptibility nor peak viremia was significantly different (log rank test, Kruskal-Wallis test, respectively). Rhesus macaques' susceptibility to RLD SHIVSF162P3 is independent of the TRIM5 TFP, CypA, and Q alleles, with the limitation that the power to detect any impact of CypA/CypA and TFP/CypA genotypes was nonexistent or low, due to absence or infrequency, respectively. The finding that TRIM5 alleles do not restrict mucosal infection or ensuing replication rates suggests that SHIVSF162P3 is indeed suitable for RLD experimentation.


Subject(s)
Carrier Proteins/genetics , Macaca mulatta/genetics , Macaca mulatta/virology , Simian Immunodeficiency Virus/pathogenicity , Alleles , Animals , Disease Models, Animal , Genetic Predisposition to Disease , Genotype , HIV/pathogenicity , HIV Infections/transmission , Host-Pathogen Interactions/genetics , Intestinal Mucosa/virology , Male , Rectum/virology , Simian Acquired Immunodeficiency Syndrome/etiology , Simian Acquired Immunodeficiency Syndrome/genetics , Simian Acquired Immunodeficiency Syndrome/transmission , Viremia/etiology , Viremia/genetics
9.
Antimicrob Agents Chemother ; 56(11): 5952-60, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22964245

ABSTRACT

Antiretroviral-based microbicides applied topically to the vagina may play an important role in protecting women from HIV infection. Incorporation of the nucleoside reverse transcriptase inhibitor tenofovir (TFV) into intravaginal rings (IVRs) for sustained mucosal delivery may lead to increased microbicide product adherence and efficacy compared with those of conventional vaginal formulations. Formulations of a novel "pod IVR" platform spanning a range of IVR drug loadings and daily release rates of TFV were evaluated in a pig-tailed macaque model. The rings were safe and exhibited sustained release at controlled rates over 28 days. Vaginal secretion TFV levels were independent of IVR drug loading and were able to be varied over 1.5 log units by changing the ring configuration. Mean TFV levels in vaginal secretions were 72.4 ± 109 µg ml(-1) (slow releasing) and 1.84 ± 1.97 mg ml(-1) (fast releasing). The mean TFV vaginal tissue concentration from the slow-releasing IVRs was 76.4 ± 54.8 µg g(-1) and remained at steady state 7 days after IVR removal, consistent with the long intracellular half-life of TFV. Intracellular tenofovir diphosphate (TFV-DP), the active moiety in defining efficacy, was measured in vaginal lymphocytes collected in the study using the fast-releasing IVR formulation. Mean intracellular TFV-DP levels of 446 ± 150 fmol/10(6) cells fall within a range that may be protective of simian-human immunodeficiency virus strain SF162p3 (SHIV(SF162p3)) infection in nonhuman primates. These data suggest that TFV-releasing IVRs based on the pod design have potential for the prevention of transmission of human immunodeficiency virus type 1 (HIV-1) and merit further clinical investigation.


Subject(s)
Adenine/analogs & derivatives , Contraceptive Devices, Female/veterinary , Delayed-Action Preparations/pharmacokinetics , Organophosphonates/pharmacokinetics , Reverse Transcriptase Inhibitors/pharmacokinetics , Vagina/metabolism , Adenine/pharmacokinetics , Administration, Intravaginal , Animals , Cytokines/metabolism , Female , Half-Life , Lymphocytes/chemistry , Macaca nemestrina , Tenofovir , Vagina/cytology , Vagina/drug effects
10.
J Infect Dis ; 206(5): 770-9, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22740713

ABSTRACT

BACKGROUND: The impact of pre-exposure prophylaxis (PrEP) with antiretrovirals on breakthrough HIV or SHIV infection is not fully documented. We addressed the hypothesis that SHIV(SF162P3) infection despite active PrEP results in altered early immune parameters, compared with untreated infection. METHODS: Eleven rhesus macaques were infected during repeated, rectal, low-dose SHIV(SF162P3) exposures while receiving concurrent oral PrEP (Truvada [n = 2] or GS7340 [n = 4]) or as untreated controls (n = 5). We measured SHIV RNA, inflammatory cytokines, CD4 cells, and SHIV-specific and memory T cells until 20 weeks after peak viremia. RESULTS: SHIV infection during PrEP resulted in 100-fold lower peak viremia and lower IL-15, IL-18, and IL-1Ra levels, compared with controls (P < .05; Wilcoxon rank-sum test). Unlike controls, PrEP-treated macaques showed no significant CD4 cell count reduction during acute infection and developed more SHIV-specific central memory T cells, relative to controls. After in vivo CD8 cell depletion, viral load increased to similar levels, indicating that CD8 cells were critical for viral control in both groups. CONCLUSIONS: PrEP with antiretrovirals has beneficial effects on early SHIV infection even when infection is not prevented. Although long-term immune control could not be examined in this SHIV infection model, our results suggest that PrEP results in improved early disease parameters in breakthrough infections.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacology , CD4-Positive T-Lymphocytes/immunology , Deoxycytidine/analogs & derivatives , Organophosphorus Compounds/pharmacology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Adenine/pharmacology , Alanine , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/virology , Cytokines/blood , Cytokines/immunology , Deoxycytidine/pharmacology , Drug Combinations , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Longitudinal Studies , Macaca mulatta , Male , RNA, Viral/chemistry , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Simian Acquired Immunodeficiency Syndrome/virology , Statistics, Nonparametric , Tenofovir/analogs & derivatives , Viremia/immunology , Viremia/virology
11.
Antimicrob Agents Chemother ; 56(3): 1291-9, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22155820

ABSTRACT

The potent antiretroviral pyrimidinediones IQP-0528 (PYD1) and IQP-0532 (PYD2) were formulated in polyurethane intravaginal rings (IVRs) as prophylactic drug delivery systems to prevent the sexual transmission of HIV-1. To aid in the selection of a pyrimidinedione candidate and the optimal loading of the drug in the IVR delivery system, four pyrimidinedione IVR formulations (PYD1 at 0.5 wt% [PYD1(0.5 wt%)], PYD1(1 wt%), PYD2(4 wt%), and PYD2(14 wt%)) were evaluated in pigtail macaques over 28 days for safety and pyrimidinedione vaginal biodistribution. Kinetic analysis of vaginal proinflammatory cytokines, native microflora, and drug levels suggested that all formulations were safe, but only the high-loaded PYD2(14 wt%) IVR demonstrated consistently high pyrimidinedione vaginal fluid and tissue levels over the 28-day study. This formulation delivered drug in excess of 10 µg/ml to vaginal fluid and 1 µg/g to vaginal tissue, a level over 1,000 times the in vitro 50% effective concentration. The in vitro release of PYD1 and PYD2 under nonsink conditions correlated well with in vivo release, both in amount and in kinetic profile, and therefore may serve as a more biologically relevant means of evaluating release in vitro than typically employed sink conditions. Lastly, the pyrimidinediones in the IVR formulation were chemically stable after 90 days of storage at elevated temperature, and the potent nanomolar-level antiviral activity of both molecules was retained after in vitro release. Altogether, these results point to the successful IVR formulation and vaginal biodistribution of the pyrimidinediones and demonstrate the usefulness of the pigtail macaque model in evaluating and screening antiretroviral IVR formulations prior to preclinical and clinical evaluation.


Subject(s)
Anti-HIV Agents/pharmacokinetics , HIV Infections/prevention & control , HIV-1/drug effects , Pyrimidinones/pharmacokinetics , Vagina/drug effects , Administration, Intravaginal , Animals , Anti-HIV Agents/therapeutic use , Cell Line , Contraceptive Devices, Female , Cytokines/biosynthesis , Cytokines/immunology , Drug Stability , Female , HIV Infections/immunology , HIV Infections/virology , HIV-1/physiology , Humans , Inhibitory Concentration 50 , Macaca nemestrina , Polyurethanes , Pyrimidinones/therapeutic use , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/virology , Tissue Distribution , Vagina/immunology , Vagina/virology , Virus Replication/drug effects
12.
J Med Primatol ; 40(4): 214-23, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21781129

ABSTRACT

BACKGROUND: Sexually transmitted infections (STIs) are associated with an increased risk of HIV infection. To model the interaction between STIs and HIV infection, we evaluated the capacity of the pigtail macaque model to sustain triple infection with Trichomonas vaginalis, Chlamydia trachomatis, and SHIV(SF162P3). METHODS: Seven SHIV(SF162P3) -infected pigtail macaques were inoculated with T. vaginalis only (n = 2), C. trachomatis only (n = 1), both T. vaginalis and C. trachomatis (n = 2), or control media (no STI; n = 2). Infections were confirmed by culture and/or nucleic acid testing. Genital mucosa was visualized by colposcopy. RESULTS: Characteristic gynecologic signs were observed for both STIs, but not in control animals. Manifestations were most prominent at days 7-10 post-infection. STIs persisted between 4 and 6 weeks and were cleared with antibiotics. CONCLUSIONS: These pilot studies demonstrate the first successful STI-SHIV triple infection of pigtail macaques, with clinical presentation of genital STI symptoms similar to those observed in humans.


Subject(s)
Chlamydia Infections/pathology , Disease Models, Animal , HIV Infections/complications , Sexually Transmitted Diseases/complications , Simian Acquired Immunodeficiency Syndrome/complications , Trichomonas Vaginitis/pathology , Animals , Cervix Uteri/microbiology , Cervix Uteri/parasitology , Cervix Uteri/pathology , Chlamydia Infections/blood , Chlamydia Infections/complications , Chlamydia trachomatis , Colposcopy , Female , HIV Infections/blood , HIV Infections/virology , Macaca nemestrina , Pilot Projects , Sexually Transmitted Diseases/blood , Sexually Transmitted Diseases/microbiology , Sexually Transmitted Diseases/parasitology , Simian Acquired Immunodeficiency Syndrome/blood , Simian Immunodeficiency Virus , Trichomonas Vaginitis/blood , Trichomonas Vaginitis/complications , Trichomonas vaginalis , Uterine Cervical Diseases/blood , Uterine Cervical Diseases/complications , Uterine Cervical Diseases/microbiology , Uterine Cervical Diseases/parasitology , Vagina/microbiology , Vagina/parasitology , Vagina/pathology
13.
J Virol ; 85(15): 7933-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21632769

ABSTRACT

Daily preexposure prophylaxis (PrEP) with Truvada (emtricitabine [FTC] and tenofovir disoproxil fumarate [TDF]) is a novel HIV prevention strategy recently found to reduce HIV incidence among men who have sex with men. We used a macaque model of HIV transmission to investigate if Truvada maintains prophylactic efficacy against an FTC-resistant isolate containing the M184V mutation. Five macaques received a dose of Truvada 3 days before exposing them rectally to the simian/human immunodeficiency virus mutant SHIV162p3(M184V), followed by a second dose 2 h after exposure. Five untreated animals were used as controls. Virus exposures were done weekly for up to 14 weeks. Despite the high (>100-fold) level of FTC resistance conferred by M184V, all five treated animals were protected from infection, while the five untreated macaques were infected (P = 0.0008). Our results show that Truvada maintains high prophylactic efficacy against an FTC-resistant isolate. Increased susceptibility to tenofovir due to M184V and other factors, including residual antiviral activity by FTC and/or reduced virus fitness due to M184V, may all have contributed to the observed protection.


Subject(s)
Antiviral Agents/pharmacology , Deoxycytidine/analogs & derivatives , HIV/drug effects , Mutation , Organophosphorus Compounds/pharmacology , Simian Acquired Immunodeficiency Syndrome/transmission , Simian Immunodeficiency Virus/drug effects , Animals , Cell Line , Deoxycytidine/pharmacology , Drug Combinations , Drug Resistance, Viral , Emtricitabine , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , HIV/genetics , Humans , Macaca mulatta , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/genetics
14.
J Acquir Immune Defic Syndr ; 57(4): 261-4, 2011 Aug 01.
Article in English | MEDLINE | ID: mdl-21546848

ABSTRACT

Fluctuations in susceptibility to HIV or SHIV during the menstrual cycle are currently not fully documented. To address this, the time point of infection was determined in 19 adult female pigtail macaques vaginally challenged during their undisturbed menstrual cycles with repeated, low-dose SHIV(SF162P3) exposures. Eighteen macaques (95%) first displayed viremia in the follicular phase, as compared with 1 macaque (5%) in the luteal phase (P < 0.0001). Due to a viral eclipse phase, we estimated a window of most frequent virus transmission between days 24 and 31 of the menstrual cycle, in the late luteal phase. Thus, susceptibility to vaginal SHIV infection is significantly elevated in the second half of the menstrual cycle when progesterone levels are high and when local immunity may be low. Such susceptibility windows have been postulated before but not definitively documented. Our data support the findings of higher susceptibility to HIV in women during progesterone-dominated periods including pregnancy and contraceptive use.


Subject(s)
Luteal Phase/physiology , Simian Acquired Immunodeficiency Syndrome/transmission , Vagina/virology , Animals , Disease Susceptibility , Female , HIV-1 , Macaca nemestrina , Pregnancy , Simian Immunodeficiency Virus , Viral Load , Viremia
15.
PLoS One ; 6(4): e19295, 2011 Apr 26.
Article in English | MEDLINE | ID: mdl-21541293

ABSTRACT

Pre-exposure prophylaxis (PrEP) with anti-viral drugs is currently in clinical trials for the prevention of HIV infection. Induction of adaptive immune responses to virus exposures during anti-viral drug administration, i.e., a "chemo-vaccination" effect, could contribute to PrEP efficacy. To study possible chemo-vaccination, we monitored humoral and cellular immune responses in nine rhesus macaques undergoing up to 14 weekly, low-dose SHIV(SF162P3) rectal exposures. Six macaques concurrently received PrEP with intermittent, oral Truvada; three were no-PrEP controls. PrEP protected 4 macaques from infection. Two of the four showed evidence of chemo-vaccination, because they developed anti-SHIV CD4(+) and CD8(+) T cells; SHIV-specific antibodies were not detected. Control macaques showed no anti-SHIV immune responses before infection. Chemo-vaccination-induced T cell responses were robust (up to 3,940 SFU/10(6) PBMCs), predominantly central memory cells, short-lived (≤22 weeks), and appeared intermittently and with changing specificities. The two chemo-vaccinated macaques were virus-challenged again after 28 weeks of rest, after T cell responses had waned. One macaque was not protected from infection. The other macaque concurrently received additional PrEP. It remained uninfected and T cell responses were boosted during the additional virus exposures. In summary, we document and characterize PrEP-induced T cell chemo-vaccination. Although not protective after subsiding in one macaque, chemo-vaccination-induced T cells warrant more comprehensive analysis during peak responses for their ability to prevent or to control infections after additional exposures. Our findings highlight the importance of monitoring these responses in clinical PrEP trials and suggest that a combination of vaccines and PrEP potentially might enhance efficacy.


Subject(s)
Deoxycytidine/analogs & derivatives , Mucous Membrane/virology , Organophosphorus Compounds/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Simian Acquired Immunodeficiency Syndrome/prevention & control , Simian Immunodeficiency Virus/immunology , Vaccination , Administration, Oral , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/virology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/drug effects , Cytokines/biosynthesis , Deoxycytidine/administration & dosage , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Drug Combinations , Emtricitabine, Tenofovir Disoproxil Fumarate Drug Combination , Epitopes/immunology , Macaca mulatta/immunology , Macaca mulatta/virology , Male , Mucous Membrane/drug effects , Mucous Membrane/immunology , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/pharmacology , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Acquired Immunodeficiency Syndrome/virology , Simian Immunodeficiency Virus/drug effects
16.
Future Virol ; 6(11)2011 Nov.
Article in English | MEDLINE | ID: mdl-37965646

ABSTRACT

An estimated 1.2 million persons in the USA are infected with HIV, of whom approximately 20% are unaware they are infected. HIV testing and knowledge of HIV serostatus have important individual and public health benefits, including reduction of morbidity, mortality and HIV transmission. Although testing is the necessary first step to prevention, more than half of the US adult population has never been tested for HIV. However, this proportion is increasing due to revised national recommendations to make HIV testing a routine part of healthcare, expansion of testing efforts at local, state and national levels, and progress in the development and adoption of new testing technologies. In this article, we describe the essential role of HIV testing as a public health prevention strategy, examine recent advances in HIV testing technologies and testing implementation, and identify future directions for HIV testing in the USA.

17.
Retrovirology ; 7: 57, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20594299

ABSTRACT

BACKGROUND: XMRV, a xenotropic murine leukemia virus (MuLV)-related virus, was recently identified by PCR testing in 67% of persons with chronic fatigue syndrome (CFS) and in 3.7% of healthy persons from the United States. To investigate the association of XMRV with CFS we tested blood specimens from 51 persons with CFS and 56 healthy persons from the US for evidence of XMRV infection by using serologic and molecular assays. Blinded PCR and serologic testing were performed at the US Centers for Disease Control and Prevention (CDC) and at two additional laboratories. RESULTS: Archived blood specimens were tested from persons with CFS defined by the 1994 international research case definition and matched healthy controls from Wichita, Kansas and metropolitan, urban, and rural Georgia populations. Serologic testing at CDC utilized a Western blot (WB) assay that showed excellent sensitivity to MuLV and XMRV polyclonal or monoclonal antibodies, and no reactivity on sera from 121 US blood donors or 26 HTLV-and HIV-infected sera. Plasma from 51 CFS cases and plasma from 53 controls were all WB negative. Additional blinded screening of the 51 cases and 53 controls at the Robert Koch Institute using an ELISA employing recombinant Gag and Env XMRV proteins identified weak seroreactivity in one CFS case and a healthy control, which was not confirmed by immunofluorescence. PCR testing at CDC employed a gag and a pol nested PCR assay with a detection threshold of 10 copies in 1 ug of human DNA. DNA specimens from 50 CFS patients and 56 controls and 41 US blood donors were all PCR-negative. Blinded testing by a second nested gag PCR assay at the Blood Systems Research Institute was also negative for DNA specimens from the 50 CFS cases and 56 controls. CONCLUSIONS: We did not find any evidence of infection with XMRV in our U.S. study population of CFS patients or healthy controls by using multiple molecular and serologic assays. These data do not support an association of XMRV with CFS.


Subject(s)
Blood/virology , Fatigue Syndrome, Chronic/etiology , Fatigue Syndrome, Chronic/virology , Gammaretrovirus/isolation & purification , Adolescent , Adult , Antibodies, Viral/blood , Blotting, Western , Case-Control Studies , Child , DNA, Viral/blood , Enzyme-Linked Immunosorbent Assay , Female , Georgia , Humans , Kansas , Male , Middle Aged , Polymerase Chain Reaction , Sensitivity and Specificity , Young Adult
18.
J Virol Methods ; 164(1-2): 43-50, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19941901

ABSTRACT

Propagation-defective vesicular stomatitis virus (VSV) vectors that encode a truncated G protein (VSV-Gstem) or lack the G gene entirely (VSV-DeltaG) are attractive vaccine vectors because they are immunogenic, cannot replicate and spread after vaccination, and do not express many of the epitopes that elicit neutralizing anti-VSV immunity. To consider advancing non-propagating VSV vectors towards clinical assessment, scalable technology that is compliant with human vaccine manufacturing must be developed to produce clinical trial material. Accordingly, two propagation methods were developed for VSV-Gstem and VSV-DeltaG vectors encoding HIV gag that have the potential to support large-scale production. One method is based on transient expression of G protein after electroporating plasmid DNA into Vero cells and the second is based on a stable Vero cell line that contains a G gene controlled by a heat shock-inducible transcription unit. Both methods reproducibly supported production of 1 x 10(7) to 1 x 10(8) infectious units (I.U.s) of vaccine vector per milliliter. Results from these studies also showed that optimization of the G gene is necessary for abundant G protein expression from electroporated plasmid DNA or from DNA integrated in the genome of a stable cell line, and that the titers of VSV-Gstem vectors generally exceeded VSV-DeltaG.


Subject(s)
Genetic Vectors , Membrane Glycoproteins/deficiency , Vesiculovirus/growth & development , Vesiculovirus/genetics , Viral Envelope Proteins/deficiency , Animals , Chlorocebus aethiops , Membrane Glycoproteins/biosynthesis , Molecular Sequence Data , RNA, Viral/genetics , Sequence Analysis, DNA , Vero Cells , Viral Envelope Proteins/biosynthesis , Virus Cultivation/methods , gag Gene Products, Human Immunodeficiency Virus/genetics
19.
Vaccine ; 27(22): 2930-9, 2009 May 14.
Article in English | MEDLINE | ID: mdl-19428903

ABSTRACT

Recombinant vesicular stomatitis viruses (rVSVs) are being developed as potential HIV-1 vaccine candidates. To characterize the in vivo replication and dissemination of rVSV vectors in mice, high doses of a highly attenuated vector expressing HIV-1 Gag, rVSV(IN)-N4CT9-Gag1, and a prototypic reference virus, rVSV(IN)-HIVGag5, were delivered intramuscularly (IM), intranasally (IN), or intravenously (IV). We used quantitative, real-time RT-PCR (Q-PCR) and standard plaque assays to measure the temporal dissemination of these viruses to various tissues. Following IM inoculation, both viruses were detected primarily at the injection site as well as in draining lymph nodes; neither virus induced significant weight loss, pathologic signs, or evidence of neuroinvasion. In contrast, following IN inoculation, the prototypic virus was detected in all tissues tested and caused significant weight loss leading to death. IN administration of rVSV(IN)-N4CT9-Gag1 resulted in detection in numerous tissues (brain, lung, nasal turbinates, and lymph nodes) albeit in significantly reduced levels, which caused little or no weight loss nor any mortality. Following IV inoculation, both prototypic and attenuated viruses were detected by Q-PCR in all tissues tested. In contrast to the prototype, rVSV(IN)-N4CT9-Gag1 viral loads were significantly lower in all organs tested, and no infectious virus was detected in the brain following IV inoculation, despite the presence of viral RNA. These studies demonstrated significant differences in the biodistribution patterns of and the associated pathogenicity engendered by the prototypic and attenuated vectors in a highly susceptible host.


Subject(s)
AIDS Vaccines/administration & dosage , AIDS Vaccines/pharmacokinetics , Genetic Vectors , Vesiculovirus/growth & development , Vesiculovirus/genetics , gag Gene Products, Human Immunodeficiency Virus/genetics , AIDS Vaccines/adverse effects , Administration, Intranasal , Animals , Female , Injections, Intramuscular , Injections, Intravenous , Mice , Mice, Inbred BALB C , RNA, Viral/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/adverse effects , Vaccines, Attenuated/pharmacokinetics , Viral Plaque Assay
20.
J Virol ; 82(1): 207-19, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17942549

ABSTRACT

Recombinant vesicular stomatitis virus (rVSV) has shown great potential as a new viral vector for vaccination. However, the prototypic rVSV vector described previously was found to be insufficiently attenuated for clinical evaluation when assessed for neurovirulence in nonhuman primates. Here, we describe the attenuation, neurovirulence, and immunogenicity of rVSV vectors expressing human immunodeficiency virus type 1 Gag. These rVSV vectors were attenuated by combinations of the following manipulations: N gene translocations (N4), G gene truncations (CT1 or CT9), noncytopathic M gene mutations (Mncp), and positioning of the gag gene into the first position of the viral genome (gag1). The resulting N4CT1-gag1, N4CT9-gag1, and MncpCT1-gag1 vectors demonstrated dramatically reduced neurovirulence in mice following direct intracranial inoculation. Surprisingly, in spite of a very high level of attenuation, the N4CT1-gag1 and N4CT9-gag1 vectors generated robust Gag-specific immune responses following intramuscular immunization that were equivalent to or greater than immune responses generated by the more virulent prototypic vectors. MncpCT1-gag1 also induced Gag-specific immune responses following intramuscular immunization that were equivalent to immune responses generated by the prototypic rVSV vector. Placement of the gag gene in the first position of the VSV genome was associated with increased in vitro expression of Gag protein, in vivo expression of Gag mRNA, and enhanced immunogenicity of the vector. These findings demonstrate that through directed manipulation of the rVSV genome, vectors that have reduced neurovirulence and enhanced immunogenicity can be made.


Subject(s)
AIDS Vaccines/immunology , Genetic Vectors , HIV-1/genetics , Vesiculovirus/genetics , Viral Vaccines/immunology , AIDS Vaccines/genetics , Animals , Cytokines/biosynthesis , HIV Antibodies/blood , Injections, Intramuscular , Lethal Dose 50 , Mice , Mice, Inbred BALB C , Point Mutation , Recombination, Genetic , Sequence Deletion , T-Lymphocytes, Cytotoxic/immunology , Translocation, Genetic , Vaccines, Attenuated/genetics , Vaccines, Attenuated/immunology , Viral Vaccines/genetics , Virulence , gag Gene Products, Human Immunodeficiency Virus/genetics , gag Gene Products, Human Immunodeficiency Virus/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...