Mauro Schechter
Federal University of Rio de Janeiro
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Mauro Schechter.
The New England Journal of Medicine | 2010
Robert M. Grant; Javier R. Lama; Peter L. Anderson; Vanessa McMahan; Albert Liu; Lorena Vargas; Pedro Goicochea; Martin Casapia; Juan Vicente Guanira-Carranza; Maria Esther Ramirez-Cardich; Orlando Montoya-Herrera; Telmo Fernandez; Valdilea G. Veloso; Susan Buchbinder; Suwat Chariyalertsak; Mauro Schechter; Linda-Gail Bekker; Kenneth H. Mayer; Esper G. Kallas; K. Rivet Amico; Kathleen Mulligan; Lane R. Bushman; Robert J. Hance; Carmela Ganoza; Patricia Defechereux; Brian S. Postle; Furong Wang; J. Jeff McConnell; Jia-Hua Zheng; Jeanny Lee
BACKGROUND Antiretroviral chemoprophylaxis before exposure is a promising approach for the prevention of human immunodeficiency virus (HIV) acquisition. METHODS We randomly assigned 2499 HIV-seronegative men or transgender women who have sex with men to receive a combination of two oral antiretroviral drugs, emtricitabine and tenofovir disoproxil fumarate (FTC-TDF), or placebo once daily. All subjects received HIV testing, risk-reduction counseling, condoms, and management of sexually transmitted infections. RESULTS The study subjects were followed for 3324 person-years (median, 1.2 years; maximum, 2.8 years). Of these subjects, 10 were found to have been infected with HIV at enrollment, and 100 became infected during follow-up (36 in the FTC-TDF group and 64 in the placebo group), indicating a 44% reduction in the incidence of HIV (95% confidence interval, 15 to 63; P=0.005). In the FTC-TDF group, the study drug was detected in 22 of 43 of seronegative subjects (51%) and in 3 of 34 HIV-infected subjects (9%) (P<0.001). Nausea was reported more frequently during the first 4 weeks in the FTC-TDF group than in the placebo group (P<0.001). The two groups had similar rates of serious adverse events (P=0.57). CONCLUSIONS Oral FTC-TDF provided protection against the acquisition of HIV infection among the subjects. Detectable blood levels strongly correlated with the prophylactic effect. (Funded by the National Institutes of Health and the Bill and Melinda Gates Foundation; ClinicalTrials.gov number, NCT00458393.).
The Lancet | 2006
Paula Braitstein; Mwg Brinkhof; F Dabis; Mauro Schechter; Andrew Boulle; Paolo G. Miotti; Robin Wood; Christian Laurent; Eduardo Sprinz; Catherine Seyler; David R. Bangsberg; Eric Balestre; Jonathan A C Sterne; Margaret T May; Matthias Egger
BACKGROUND Highly active antiretroviral therapy (HAART) is being scaled up in developing countries. We compared baseline characteristics and outcomes during the first year of HAART between HIV-1-infected patients in low-income and high-income settings. METHODS 18 HAART programmes in Africa, Asia, and South America (low-income settings) and 12 HIV cohort studies from Europe and North America (high-income settings) provided data for 4810 and 22,217, respectively, treatment-naïve adult patients starting HAART. All patients from high-income settings and 2725 (57%) patients from low-income settings were actively followed-up and included in survival analyses. FINDINGS Compared with high-income countries, patients starting HAART in low-income settings had lower CD4 cell counts (median 108 cells per muL vs 234 cells per muL), were more likely to be female (51%vs 25%), and more likely to start treatment with a non-nucleoside reverse transcriptase inhibitor (NNRTI) (70%vs 23%). At 6 months, the median number of CD4 cells gained (106 cells per muL vs 103 cells per muL) and the percentage of patients reaching HIV-1 RNA levels lower than 500 copies/mL (76%vs 77%) were similar. Mortality was higher in low-income settings (124 deaths during 2236 person-years of follow-up) than in high-income settings (414 deaths during 20,532 person-years). The adjusted hazard ratio (HR) of mortality comparing low-income with high-income settings fell from 4.3 (95% CI 1.6-11.8) during the first month to 1.5 (0.7-3.0) during months 7-12. The provision of treatment free of charge in low-income settings was associated with lower mortality (adjusted HR 0.23; 95% CI 0.08-0.61). INTERPRETATION Patients starting HAART in resource-poor settings have increased mortality rates in the first months on therapy, compared with those in developed countries. Timely diagnosis and assessment of treatment eligibility, coupled with free provision of HAART, might reduce this excess mortality.
The New England Journal of Medicine | 2008
Roy T. Steigbigel; David A. Cooper; Princy Kumar; Joseph E. Eron; Mauro Schechter; Martin Markowitz; Mona Loutfy; Jeffrey L. Lennox; José M. Gatell; Jürgen K. Rockstroh; Christine Katlama; Patrick Yeni; Adriano Lazzarin; Bonaventura Clotet; Jing Zhao; Joshua Chen; Desmond Ryan; Rand R. Rhodes; John A. Killar; Lucinda R. Gilde; Kim M. Strohmaier; Anne Meibohm; Michael D. Miller; Daria J. Hazuda; Michael L. Nessly; Mark J. DiNubile; Robin Isaacs; Bach Yen Nguyen; Hedy Teppler
BACKGROUND Raltegravir (MK-0518) is an inhibitor of human immunodeficiency virus type 1 (HIV-1) integrase active against HIV-1 susceptible or resistant to older antiretroviral drugs. METHODS We conducted two identical trials in different geographic regions to evaluate the safety and efficacy of raltegravir, as compared with placebo, in combination with optimized background therapy, in patients infected with HIV-1 that has triple-class drug resistance in whom antiretroviral therapy had failed. Patients were randomly assigned to raltegravir or placebo in a 2:1 ratio. RESULTS In the combined studies, 699 of 703 randomized patients (462 and 237 in the raltegravir and placebo groups, respectively) received the study drug. Seventeen of the 699 patients (2.4%) discontinued the study before week 16. Discontinuation was related to the study treatment in 13 of these 17 patients: 7 of the 462 raltegravir recipients (1.5%) and 6 of the 237 placebo recipients (2.5%). The results of the two studies were consistent. At week 16, counting noncompletion as treatment failure, 355 of 458 raltegravir recipients (77.5%) had HIV-1 RNA levels below 400 copies per milliliter, as compared with 99 of 236 placebo recipients (41.9%, P<0.001). Suppression of HIV-1 RNA to a level below 50 copies per milliliter was achieved at week 16 in 61.8% of the raltegravir recipients, as compared with 34.7% of placebo recipients, and at week 48 in 62.1% as compared with 32.9% (P<0.001 for both comparisons). Without adjustment for the length of follow-up, cancers were detected in 3.5% of raltegravir recipients and in 1.7% of placebo recipients. The overall frequencies of drug-related adverse events were similar in the raltegravir and placebo groups. CONCLUSIONS In HIV-infected patients with limited treatment options, raltegravir plus optimized background therapy provided better viral suppression than optimized background therapy alone for at least 48 weeks. (ClinicalTrials.gov numbers, NCT00293267 and NCT00293254.)
The New England Journal of Medicine | 2008
David A. Cooper; Roy T. Steigbigel; José M. Gatell; Jürgen K. Rockstroh; Christine Katlama; Patrick Yeni; Adriano Lazzarin; Bonaventura Clotet; Princy Kumar; Joseph E. Eron; Mauro Schechter; Martin Markowitz; Mona Loutfy; Jeffrey L. Lennox; Jing Zhao; Joshua Chen; Desmond Ryan; Rand R. Rhodes; John A. Killar; Lucinda R. Gilde; Kim M. Strohmaier; Anne Meibohm; Michael D. Miller; Daria J. Hazuda; Michael L. Nessly; Mark J. DiNubile; Robin Isaacs; Hedy Teppler; Bach Yen Nguyen
BACKGROUND We evaluated the efficacy of raltegravir and the development of viral resistance in two identical trials involving patients who were infected with human immunodeficiency virus type 1 (HIV-1) with triple-class drug resistance and in whom antiretroviral therapy had failed. METHODS We conducted subgroup analyses of the data from week 48 in both studies according to baseline prognostic factors. Genotyping of the integrase gene was performed in raltegravir recipients who had virologic failure. RESULTS Virologic responses to raltegravir were consistently superior to responses to placebo, regardless of the baseline values of HIV-1 RNA level; CD4 cell count; genotypic or phenotypic sensitivity score; use or nonuse of darunavir, enfuvirtide, or both in optimized background therapy; or demographic characteristics. Among patients in the two studies combined who were using both enfuvirtide and darunavir for the first time, HIV-1 RNA levels of less than 50 copies per milliliter were achieved in 89% of raltegravir recipients and 68% of placebo recipients. HIV-1 RNA levels of less than 50 copies per milliliter were achieved in 69% and 80% of the raltegravir recipients and in 47% and 57% of the placebo recipients using either darunavir or enfuvirtide for the first time, respectively. At 48 weeks, 105 of the 462 raltegravir recipients (23%) had virologic failure. Genotyping was performed in 94 raltegravir recipients with virologic failure. Integrase mutations known to be associated with phenotypic resistance to raltegravir arose during treatment in 64 patients (68%). Forty-eight of these 64 patients (75%) had two or more resistance-associated mutations. CONCLUSIONS When combined with an optimized background regimen in both studies, a consistently favorable treatment effect of raltegravir over placebo was shown in clinically relevant subgroups of patients, including those with baseline characteristics that typically predict a poor response to antiretroviral therapy: a high HIV-1 RNA level, low CD4 cell count, and low genotypic or phenotypic sensitivity score. (ClinicalTrials.gov numbers, NCT00293267 and NCT00293254.)
Lancet Infectious Diseases | 2014
Robert M. Grant; Peter L. Anderson; Vanessa McMahan; Albert Liu; K. Rivet Amico; Megha Mehrotra; Sybil Hosek; Carlos Mosquera; Martin Casapia; Orlando Montoya; Susan Buchbinder; Valdilea G. Veloso; Kenneth H. Mayer; Suwat Chariyalertsak; Linda-Gail Bekker; Esper G. Kallas; Mauro Schechter; Juan V. Guanira; Lane R. Bushman; David N. Burns; James F. Rooney; David V. Glidden
BACKGROUND The effect of HIV pre-exposure prophylaxis (PrEP) depends on uptake, adherence, and sexual practices. We aimed to assess these factors in a cohort of HIV-negative people at risk of infection. METHODS In our cohort study, men and transgender women who have sex with men previously enrolled in PrEP trials (ATN 082, iPrEx, and US Safety Study) were enrolled in a 72 week open-label extension. We measured drug concentrations in plasma and dried blood spots in seroconverters and a random sample of seronegative participants. We assessed PrEP uptake, adherence, sexual practices, and HIV incidence. Statistical methods included Poisson models, comparison of proportions, and generalised estimating equations. FINDINGS We enrolled 1603 HIV-negative people, of whom 1225 (76%) received PrEP. Uptake was higher among those reporting condomless receptive anal intercourse (416/519 [81%] vs 809/1084 [75%], p=0·003) and having serological evidence of herpes (612/791 [77%] vs 613/812 [75%] p=0·03). Of those receiving PrEP, HIV incidence was 1·8 infections per 100 person-years, compared with 2·6 infections per 100 person-years in those who concurrently did not choose PrEP (HR 0·51, 95% CI 0·26-1·01, adjusted for sexual behaviours), and 3·9 infections per 100 person-years in the placebo group of the previous randomised phase (HR 0·49, 95% CI 0·31-0·77). Among those receiving PrEP, HIV incidence was 4·7 infections per 100 person-years if drug was not detected in dried blood spots, 2·3 infections per 100 person-years if drug concentrations suggested use of fewer than two tablets per week, 0·6 per 100 person-years for use of two to three tablets per week, and 0·0 per 100 person-years for use of four or more tablets per week (p<0·0001). PrEP drug concentrations were higher among people of older age, with more schooling, who reported non-condom receptive anal intercourse, who had more sexual partners, and who had a history of syphilis or herpes. INTERPRETATION PrEP uptake was high when made available free of charge by experienced providers. The effect of PrEP is increased by greater uptake and adherence during periods of higher risk. Drug concentrations in dried blood spots are strongly correlated with protective benefit. FUNDING US National Institutes of Health.
Science Translational Medicine | 2012
Peter L. Anderson; David V. Glidden; Albert Liu; Susan Buchbinder; Javier R. Lama; Juan V. Guanira; Vanessa McMahan; Lane R. Bushman; Martin Casapia; Orlando Montoya-Herrera; Valdilea G. Veloso; Kenneth H. Mayer; Suwat Chariyalertsak; Mauro Schechter; Linda-Gail Bekker; Esper G. Kallas; Robert M. Grant
PrEP drug concentrations associated with ≥90% reduction in HIV acquisition in men who have sex with men are achieved with daily dosing. PrEParing to Stop HIV Acquisition Pre-exposure prophylaxis (PrEP) using the antiretroviral drugs emtricitabine (FTC) and tenofovir disoproxil fumarate (TDF) is a recently proven strategy for preventing HIV acquisition. These drugs require phosphorylation in mononuclear cells to the pharmacologically active triphosphate moieties, called emtricitabine-triphosphate (FTC-TP) and tenofovir-diphosphate (TFV-DP). The iPrEx study was a randomized placebo-controlled trial of daily oral doses of FTC-TDF as PrEP in HIV-negative men who have sex with men. Participants all received a comprehensive package of HIV prevention services. HIV infections were reduced by 44% overall in the FTC-TDF arm relative to placebo. HIV risk was reduced by more than 90% among those having detectable drug in blood, indicating that adherence was a powerful determinant of drug efficacy at preventing HIV acquisition. A new study by Anderson et al. estimates specific drug concentrations and adherence levels associated with protection from HIV-1 acquisition in the iPrEx trial. A regression analysis predicted that a TFV-DP concentration of 16 fmol/106 peripheral blood mononuclear cells (PBMCs) (95% confidence interval, 3 to 28) was associated with a 90% reduction in HIV acquisition relative to placebo in the iPrEx study. To determine the number of tablets required to achieve this drug concentration, TFV-DP concentrations from another study called STRAND were used to establish expected TFV-DP concentrations. TFV-DP was detected in the blood at all dosing levels in all participants with a median (interquartile range) TFV-DP concentration of 11 fmol/106 PBMCs (6 to 13) after two doses per week, 32 fmol/106 PBMCs (25 to 39) after four doses per week, and 42 fmol/106 PBMCs (31 to 47) after seven doses per week. When the iPrEx study’s regression model was used to analyze the STRAND TFV-DP concentrations, the predicted HIV risk reductions were 76, 96, and 99% for two, four, and seven doses per week, respectively. These findings suggest that PrEP using oral FTC-TDF tablets is a robust intervention for preventing HIV acquisition among men who have sex with men. Drug concentrations associated with protection from HIV-1 acquisition have not been determined. We evaluated drug concentrations among men who have sex with men in a substudy of the iPrEx trial (1). In this randomized placebo-controlled trial, daily oral doses of emtricitabine/tenofovir disoproxil fumarate were used as pre-exposure prophylaxis (PrEP) in men who have sex with men. Drug was detected less frequently in blood plasma and in viable cryopreserved peripheral blood mononuclear cells (PBMCs) in HIV-infected cases at the visit when HIV was first discovered compared with controls at the matched time point of the study (8% versus 44%; P < 0.001) and in the 90 days before that visit (11% versus 51%; P < 0.001). An intracellular concentration of the active form of tenofovir, tenofovir-diphosphate (TFV-DP), of 16 fmol per million PBMCs was associated with a 90% reduction in HIV acquisition relative to the placebo arm. Directly observed dosing in a separate study, the STRAND trial, yielded TFV-DP concentrations that, when analyzed according to the iPrEx model, corresponded to an HIV-1 risk reduction of 76% for two doses per week, 96% for four doses per week, and 99% for seven doses per week. Prophylactic benefits were observed over a range of doses and drug concentrations, suggesting ways to optimize PrEP regimens for this population.
The Lancet | 2010
Frederick L. Altice; Adeeba Kamarulzaman; Vincent Soriano; Mauro Schechter; Gerald Friedland
HIV-infected drug users have increased age-matched morbidity and mortality compared with HIV-infected people who do not use drugs. Substance-use disorders negatively affect the health of HIV-infected drug users, who also have frequent medical and psychiatric comorbidities that complicate HIV treatment and prevention. Evidence-based treatments are available for the management of substance-use disorders, mental illness, HIV and other infectious complications such as viral hepatitis and tuberculosis, and many non-HIV-associated comorbidities. Tuberculosis co-infection in HIV-infected drug users, including disease caused by drug-resistant strains, is acquired and transmitted as a consequence of inadequate prescription of antiretroviral therapy, poor adherence, and repeated interfaces with congregate settings such as prisons. Medication-assisted therapies provide the strongest evidence for HIV treatment and prevention efforts, yet are often not available where they are needed most. Antiretroviral therapy, when prescribed and adherence is at an optimum, improves health-related outcomes for HIV infection and many of its comorbidities, including tuberculosis, viral hepatitis, and renal and cardiovascular disease. Simultaneous clinical management of multiple comorbidities in HIV-infected drug users might result in complex pharmacokinetic drug interactions that must be adequately addressed. Moreover, interventions to improve adherence to treatment, including integration of health services delivery, are needed. Multifaceted, interdisciplinary approaches are urgently needed to achieve parity in health outcomes in HIV-infected drug users.
Bulletin of The World Health Organization | 2008
Martin W. G. Brinkhof; François Dabis; Landon Myer; David R. Bangsberg; Andrew Boulle; Denis Nash; Mauro Schechter; Christian Laurent; Olivia Keiser; Margaret T May; Eduardo Sprinz; Matthias Egger; Xavier Anglaret
OBJECTIVE To analyse the early loss of patients to antiretroviral therapy (ART) programmes in resource-limited settings. METHODS Using data on 5491 adult patients starting ART (median age 35 years, 46% female) in 15 treatment programmes in Africa, Asia and South America with (3) 12 months of follow-up, we investigated risk factors for no follow-up after treatment initiation, and loss to follow-up or death in the first 6 months. FINDINGS Overall, 211 patients (3.8%) had no follow-up, 880 (16.0%) were lost to follow-up and 141 (2.6%) were known to have died in the first 6 months. The probability of no follow-up was higher in 2003-2004 than in 2000 or earlier (odds ratio, OR: 5.06; 95% confidence interval, CI: 1.28-20.0), as was loss to follow-up (hazard ratio, HR: 7.62; 95% CI: 4.55-12.8) but not recorded death (HR: 1.02; 95% CI: 0.44-2.36). Compared with a baseline CD4-cell count (3) 50 cells/microl, a count < 25 cells/microl was associated with a higher probability of no follow-up (OR: 2.49; 95% CI: 1.43-4.33), loss to follow-up (HR: 1.48; 95% CI: 1.23-1.77) and death (HR: 3.34; 95% CI: 2.10-5.30). Compared to free treatment, fee-for-service programmes were associated with a higher probability of no follow-up (OR: 3.71; 95% CI: 0.97-16.05) and higher mortality (HR: 4.64; 95% CI: 1.11-19.41). CONCLUSION Early patient losses were increasingly common when programmes were scaled up and were associated with a fee for service and advanced immunodeficiency at baseline. Measures to maximize ART programme retention are required in resource-poor countries.
PLOS Medicine | 2004
Frank van Leth; Prahpan Phanuphak; Erik Stroes; Brian Gazzard; Pedro Cahn; François Raffi; Robin Wood; Mark Bloch; Christine Katlama; John J. P. Kastelein; Mauro Schechter; Robert L. Murphy; Andrzej Horban; David B. Hall; Joep M. A. Lange; Peter Reiss
ABSTRACT Background Patients infected with HIV-1 initiating antiretroviral therapy (ART) containing a non-nucleoside reverse transcriptase inhibitor (NNRTI) show presumably fewer atherogenic lipid changes than those initiating most ARTs containing a protease inhibitor. We analysed whether lipid changes differed between the two most commonly used NNRTIs, nevirapine (NVP) and efavirenz (EFV). Methods and Findings Prospective analysis of lipids and lipoproteins was performed in patients enrolled in the NVP and EFV treatment groups of the 2NN study who remained on allocated treatment during 48 wk of follow-up. Patients were allocated to NVP (n = 417), or EFV (n = 289) in combination with stavudine and lamivudine. The primary endpoint was percentage change over 48 wk in high-density lipoprotein cholesterol (HDL-c), total cholesterol (TC), TC:HDL-c ratio, non-HDL-c, low-density lipoprotein cholesterol, and triglycerides. The increase of HDL-c was significantly larger for patients receiving NVP (42.5%) than for patients receiving EFV (33.7%; p = 0.036), while the increase in TC was lower (26.9% and 31.1%, respectively; p = 0.073), resulting in a decrease of the TC:HDL-c ratio for patients receiving NVP (−4.1%) and an increase for patients receiving EFV (+5.9%; p < 0.001). The increase of non-HDL-c was smaller for patients receiving NVP (24.7%) than for patients receiving EFV (33.6%; p = 0.007), as were the increases of triglycerides (20.1% and 49.0%, respectively; p < 0.001) and low-density lipoprotein cholesterol (35.0% and 40.0%, respectively; p = 0.378). These differences remained, or even increased, after adjusting for changes in HIV-1 RNA and CD4+ cell levels, indicating an effect of the drugs on lipids over and above that which may be explained by suppression of HIV-1 infection. The increases in HDL-c were of the same order of magnitude as those seen with the use of the investigational HDL-c-increasing drugs. Conclusion NVP-containing ART shows larger increases in HDL-c and decreases in TC:HDL-c ratio than an EFV-containing regimen. Based on these findings, protease-inhibitor-sparing regimens based on non-nucleoside reverse transcriptase inhibitor, particularly those containing NVP, may be expected to result in a reduced risk of coronary heart disease.
AIDS | 2013
Alison Rodger; Rebecca Lodwick; Mauro Schechter; Steven G. Deeks; Janaki Amin; Richard Gilson; Roger Paredes; Elzbieta Bakowska; Frederik N. Engsig; Andrew N. Phillips
Background:Due to the success of antiretroviral therapy (ART), it is relevant to ask whether death rates in optimally treated HIV are higher than the general population. The objective was to compare mortality rates in well controlled HIV-infected adults in the SMART and ESPRIT clinical trials with the general population. Methods:Non-IDUs aged 20–70 years from the continuous ART control arms of ESPRIT and SMART were included if the person had both low HIV plasma viral loads (⩽400 copies/ml SMART, ⩽500 copies/ml ESPRIT) and high CD4+ T-cell counts (≥350 cells/&mgr;l) at any time in the past 6 months. Standardized mortality ratios (SMRs) were calculated by comparing death rates with the Human Mortality Database. Results:Three thousand, two hundred and eighty individuals [665 (20%) women], median age 43 years, contributed 12 357 person-years of follow-up. Sixty-two deaths occurred during follow up. Commonest cause of death was cardiovascular disease (CVD) or sudden death (19, 31%), followed by non-AIDS malignancy (12, 19%). Only two deaths (3%) were AIDS-related. Mortality rate was increased compared with the general population with a CD4+ cell count between 350 and 499 cells/&mgr;l [SMR 1.77, 95% confidence interval (CI) 1.17–2.55]. No evidence for increased mortality was seen with CD4+ cell counts greater than 500 cells/&mgr;l (SMR 1.00, 95% CI 0.69–1.40). Conclusion:In HIV-infected individuals on ART, with a recent undetectable viral load, who maintained or had recovery of CD4+ cell counts to at least 500 cells/&mgr;l, we identified no evidence for a raised risk of death compared with the general population.