Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Nature ; 626(7997): 145-150, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38122820

RESUMEN

How likely is it to become infected by SARS-CoV-2 after being exposed? Almost everyone wondered about this question during the COVID-19 pandemic. Contact-tracing apps1,2 recorded measurements of proximity3 and duration between nearby smartphones. Contacts-individuals exposed to confirmed cases-were notified according to public health policies such as the 2 m, 15 min guideline4,5, despite limited evidence supporting this threshold. Here we analysed 7 million contacts notified by the National Health Service COVID-19 app6,7 in England and Wales to infer how app measurements translated to actual transmissions. Empirical metrics and statistical modelling showed a strong relation between app-computed risk scores and actual transmission probability. Longer exposures at greater distances had risk similar to that of shorter exposures at closer distances. The probability of transmission confirmed by a reported positive test increased initially linearly with duration of exposure (1.1% per hour) and continued increasing over several days. Whereas most exposures were short (median 0.7 h, interquartile range 0.4-1.6), transmissions typically resulted from exposures lasting between 1 h and several days (median 6 h, interquartile range 1.4-28). Households accounted for about 6% of contacts but 40% of transmissions. With sufficient preparation, privacy-preserving yet precise analyses of risk that would inform public health measures, based on digital contact tracing, could be performed within weeks of the emergence of a new pathogen.


Asunto(s)
COVID-19 , Trazado de Contacto , Aplicaciones Móviles , Salud Pública , Medición de Riesgo , Humanos , Trazado de Contacto/métodos , Trazado de Contacto/estadística & datos numéricos , COVID-19/epidemiología , COVID-19/transmisión , Pandemias , SARS-CoV-2 , Medicina Estatal , Factores de Tiempo , Inglaterra/epidemiología , Gales/epidemiología , Modelos Estadísticos , Composición Familiar , Salud Pública/métodos , Salud Pública/tendencias
2.
Nature ; 594(7863): 408-412, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33979832

RESUMEN

The COVID-19 pandemic has seen the emergence of digital contact tracing to help to prevent the spread of the disease. A mobile phone app records proximity events between app users, and when a user tests positive for COVID-19, their recent contacts can be notified instantly. Theoretical evidence has supported this new public health intervention1-6, but its epidemiological impact has remained uncertain7. Here we investigate the impact of the National Health Service (NHS) COVID-19 app for England and Wales, from its launch on 24 September 2020 to the end of December 2020. It was used regularly by approximately 16.5 million users (28% of the total population), and sent approximately 1.7 million exposure notifications: 4.2 per index case consenting to contact tracing. We estimated that the fraction of individuals notified by the app who subsequently showed symptoms and tested positive (the secondary attack rate (SAR)) was 6%, similar to the SAR for manually traced close contacts. We estimated the number of cases averted by the app using two complementary approaches: modelling based on the notifications and SAR gave an estimate of 284,000 (central 95% range of sensitivity analyses 108,000-450,000), and statistical comparison of matched neighbouring local authorities gave an estimate of 594,000 (95% confidence interval 317,000-914,000). Approximately one case was averted for each case consenting to notification of their contacts. We estimated that for every percentage point increase in app uptake, the number of cases could be reduced by 0.8% (using modelling) or 2.3% (using statistical analysis). These findings support the continued development and deployment of such apps in populations that are awaiting full protection from vaccines.


Asunto(s)
COVID-19/epidemiología , COVID-19/prevención & control , Trazado de Contacto/instrumentación , Trazado de Contacto/métodos , Aplicaciones Móviles/estadística & datos numéricos , Número Básico de Reproducción , COVID-19/mortalidad , COVID-19/transmisión , Inglaterra/epidemiología , Humanos , Mortalidad , Programas Nacionales de Salud , Cuarentena , Gales/epidemiología
3.
J Antimicrob Chemother ; 78(3): 656-664, 2023 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-36738248

RESUMEN

BACKGROUND: Next-generation sequencing (NGS) is gradually replacing Sanger sequencing (SS) as the primary method for HIV genotypic resistance testing. However, there are limited systematic data on comparability of these methods in a clinical setting for the presence of low-abundance drug resistance mutations (DRMs) and their dependency on the variant-calling thresholds. METHODS: To compare the HIV-DRMs detected by SS and NGS, we included participants enrolled in the Swiss HIV Cohort Study (SHCS) with SS and NGS sequences available with sample collection dates ≤7 days apart. We tested for the presence of HIV-DRMs and compared the agreement between SS and NGS at different variant-calling thresholds. RESULTS: We included 594 pairs of SS and NGS from 527 SHCS participants. Males accounted for 80.5% of the participants, 76.3% were ART naive at sample collection and 78.1% of the sequences were subtype B. Overall, we observed a good agreement (Cohen's kappa >0.80) for HIV-DRMs for variant-calling thresholds ≥5%. We observed an increase in low-abundance HIV-DRMs detected at lower thresholds [28/417 (6.7%) at 10%-25% to 293/812 (36.1%) at 1%-2% threshold]. However, such low-abundance HIV-DRMs were overrepresented in ART-naive participants and were in most cases not detected in previously sampled sequences suggesting high sequencing error for thresholds <3%. CONCLUSIONS: We found high concordance between SS and NGS but also a substantial number of low-abundance HIV-DRMs detected only by NGS at lower variant-calling thresholds. Our findings suggest that a substantial fraction of the low-abundance HIV-DRMs detected at thresholds <3% may represent sequencing errors and hence should not be overinterpreted in clinical practice.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , Seropositividad para VIH , VIH-1 , Masculino , Humanos , Infecciones por VIH/tratamiento farmacológico , Estudios de Cohortes , Farmacorresistencia Viral/genética , Carga Viral , Seropositividad para VIH/tratamiento farmacológico , Mutación , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Genotipo , Fármacos Anti-VIH/uso terapéutico
4.
PLoS Comput Biol ; 17(7): e1009146, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34252083

RESUMEN

SARS-CoV-2 has spread across the world, causing high mortality and unprecedented restrictions on social and economic activity. Policymakers are assessing how best to navigate through the ongoing epidemic, with computational models being used to predict the spread of infection and assess the impact of public health measures. Here, we present OpenABM-Covid19: an agent-based simulation of the epidemic including detailed age-stratification and realistic social networks. By default the model is parameterised to UK demographics and calibrated to the UK epidemic, however, it can easily be re-parameterised for other countries. OpenABM-Covid19 can evaluate non-pharmaceutical interventions, including both manual and digital contact tracing, and vaccination programmes. It can simulate a population of 1 million people in seconds per day, allowing parameter sweeps and formal statistical model-based inference. The code is open-source and has been developed by teams both inside and outside academia, with an emphasis on formal testing, documentation, modularity and transparency. A key feature of OpenABM-Covid19 are its Python and R interfaces, which has allowed scientists and policymakers to simulate dynamic packages of interventions and help compare options to suppress the COVID-19 epidemic.


Asunto(s)
COVID-19/prevención & control , Trazado de Contacto , Análisis de Sistemas , COVID-19/epidemiología , COVID-19/transmisión , COVID-19/virología , Prueba de COVID-19 , Vacunas contra la COVID-19/administración & dosificación , Brotes de Enfermedades , Humanos , Distanciamiento Físico , Cuarentena , SARS-CoV-2/aislamiento & purificación
5.
Philos Trans A Math Phys Eng Sci ; 380(2233): 20210304, 2022 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-35965459

RESUMEN

The SARS-CoV-2 epidemic has been extended by the evolution of more transmissible viral variants. In autumn 2020, the B.1.177 lineage became the dominant variant in England, before being replaced by the B.1.1.7 (Alpha) lineage in late 2020, with the sweep occurring at different times in each region. This period coincided with a large number of non-pharmaceutical interventions (e.g. lockdowns) to control the epidemic, making it difficult to estimate the relative transmissibility of variants. In this paper, we model the spatial spread of these variants in England using a meta-population agent-based model which correctly characterizes the regional variation in cases and distribution of variants. As a test of robustness, we additionally estimated the relative transmissibility of multiple variants using a statistical model based on the renewal equation, which simultaneously estimates the effective reproduction number R. Relative to earlier variants, the transmissibility of B.1.177 is estimated to have increased by 1.14 (1.12-1.16) and that of Alpha by 1.71 (1.65-1.77). The vaccination programme starting in December 2020 is also modelled. Counterfactual simulations demonstrate that the vaccination programme was essential for reopening in March 2021, and that if the January lockdown had started one month earlier, up to 30 k (24 k-38 k) deaths could have been prevented. This article is part of the theme issue 'Technical challenges of modelling real-life epidemics and examples of overcoming these'.


Asunto(s)
COVID-19 , SARS-CoV-2 , COVID-19/epidemiología , Control de Enfermedades Transmisibles , Humanos , SARS-CoV-2/genética , Estaciones del Año
6.
Clin Infect Dis ; 72(1): 30-37, 2021 01 23.
Artículo en Inglés | MEDLINE | ID: mdl-31922537

RESUMEN

BACKGROUND: Phylogenetic analysis can be used to assess human immunodeficiency virus (HIV) transmission in populations. We inferred the direction of HIV transmission using whole-genome HIV sequences from couples with known linked infection and known transmission direction. METHODS: Complete next-generation sequencing (NGS) data were obtained for 105 unique index-partner sample pairs from 32 couples enrolled in the HIV Prevention Trials Network (HPTN) 052 study (up to 2 samples/person). Index samples were obtained up to 5.5 years before partner infection; partner samples were obtained near the time of seroconversion. The bioinformatics method, phyloscanner, was used to infer transmission direction. Analyses were performed using samples from individual sample pairs, samples from all couples (1 sample/person; group analysis), and all available samples (multisample group analysis). Analysis was also performed using NGS data from defined regions of the HIV genome (gag, pol, env). RESULTS: Using whole-genome NGS data, transmission direction was inferred correctly (index to partner) for 98 of 105 (93.3%) of the individual sample pairs, 99 of 105 (94.3%) sample pairs using group analysis, and 31 of the 32 couples (96.9%) using multisample group analysis. There were no cases where the incorrect transmission direction (partner to index) was inferred. The accuracy of the method was higher with greater time between index and partner sample collection. Pol region sequences performed better than env or gag sequences for inferring transmission direction. CONCLUSIONS: We demonstrate the potential of a phylogenetic method to infer the direction of HIV transmission between 2 individuals using whole-genome and pol NGS data.


Asunto(s)
Infecciones por VIH , VIH-1 , Infecciones por VIH/prevención & control , VIH-1/genética , Humanos , Filogenia
7.
J Clin Microbiol ; 58(10)2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32669382

RESUMEN

Viral genetic sequencing can be used to monitor the spread of HIV drug resistance, identify appropriate antiretroviral regimes, and characterize transmission dynamics. Despite decreasing costs, next-generation sequencing (NGS) is still prohibitively costly for routine use in generalized HIV epidemics in low- and middle-income countries. Here, we present veSEQ-HIV, a high-throughput, cost-effective NGS sequencing method and computational pipeline tailored specifically to HIV, which can be performed using leftover blood drawn for routine CD4 cell count testing. This method overcomes several major technical challenges that have prevented HIV sequencing from being used routinely in public health efforts; it is fast, robust, and cost-efficient, and generates full genomic sequences of diverse strains of HIV without bias. The complete veSEQ-HIV pipeline provides viral load estimates and quantitative summaries of drug resistance mutations; it also exploits information on within-host viral diversity to construct directed transmission networks. We evaluated the method's performance using 1,620 plasma samples collected from individuals attending 10 large urban clinics in Zambia as part of the HPTN 071-2 study (PopART Phylogenetics). Whole HIV genomes were recovered from 91% of samples with a viral load of >1,000 copies/ml. The cost of the assay (30 GBP per sample) compares favorably with existing VL and HIV genotyping tests, proving an affordable option for combining HIV clinical monitoring with molecular epidemiology and drug resistance surveillance in low-income settings.


Asunto(s)
Fármacos Anti-VIH , Infecciones por VIH , VIH-1 , Fármacos Anti-VIH/uso terapéutico , Farmacorresistencia Viral/genética , Genómica , Infecciones por VIH/diagnóstico , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/epidemiología , Humanos , Carga Viral , Zambia
8.
PLoS Biol ; 15(6): e2001855, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28604782

RESUMEN

HIV-1 set-point viral load-the approximately stable value of viraemia in the first years of chronic infection-is a strong predictor of clinical outcome and is highly variable across infected individuals. To better understand HIV-1 pathogenesis and the evolution of the viral population, we must quantify the heritability of set-point viral load, which is the fraction of variation in this phenotype attributable to viral genetic variation. However, current estimates of heritability vary widely, from 6% to 59%. Here we used a dataset of 2,028 seroconverters infected between 1985 and 2013 from 5 European countries (Belgium, Switzerland, France, the Netherlands and the United Kingdom) and estimated the heritability of set-point viral load at 31% (CI 15%-43%). Specifically, heritability was measured using models of character evolution describing how viral load evolves on the phylogeny of whole-genome viral sequences. In contrast to previous studies, (i) we measured viral loads using standardized assays on a sample collected in a strict time window of 6 to 24 months after infection, from which the viral genome was also sequenced; (ii) we compared 2 models of character evolution, the classical "Brownian motion" model and another model ("Ornstein-Uhlenbeck") that includes stabilising selection on viral load; (iii) we controlled for covariates, including age and sex, which may inflate estimates of heritability; and (iv) we developed a goodness of fit test based on the correlation of viral loads in cherries of the phylogenetic tree, showing that both models of character evolution fit the data well. An overall heritability of 31% (CI 15%-43%) is consistent with other studies based on regression of viral load in donor-recipient pairs. Thus, about a third of variation in HIV-1 virulence is attributable to viral genetic variation.


Asunto(s)
Variación Genética , Genoma Viral , Infecciones por VIH/microbiología , Seropositividad para VIH/microbiología , VIH-1/genética , Proteínas del Virus de la Inmunodeficiencia Humana/genética , Modelos Genéticos , Adulto , Anciano , Estudios de Cohortes , Europa (Continente) , Evolución Molecular , Femenino , Estudio de Asociación del Genoma Completo , Infecciones por VIH/sangre , Seropositividad para VIH/sangre , VIH-1/crecimiento & desarrollo , VIH-1/aislamiento & purificación , VIH-1/patogenicidad , Proteínas del Virus de la Inmunodeficiencia Humana/sangre , Proteínas del Virus de la Inmunodeficiencia Humana/metabolismo , Humanos , Masculino , Persona de Mediana Edad , Filogenia , Sistema de Registros , Seroconversión , Carga Viral , Virulencia
10.
Mol Biol Evol ; 35(3): 719-733, 2018 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-29186559

RESUMEN

A central feature of pathogen genomics is that different infectious particles (virions and bacterial cells) within an infected individual may be genetically distinct, with patterns of relatedness among infectious particles being the result of both within-host evolution and transmission from one host to the next. Here, we present a new software tool, phyloscanner, which analyses pathogen diversity from multiple infected hosts. phyloscanner provides unprecedented resolution into the transmission process, allowing inference of the direction of transmission from sequence data alone. Multiply infected individuals are also identified, as they harbor subpopulations of infectious particles that are not connected by within-host evolution, except where recombinant types emerge. Low-level contamination is flagged and removed. We illustrate phyloscanner on both viral and bacterial pathogens, namely HIV-1 sequenced on Illumina and Roche 454 platforms, HCV sequenced with the Oxford Nanopore MinION platform, and Streptococcus pneumoniae with sequences from multiple colonies per individual. phyloscanner is available from https://github.com/BDI-pathogens/phyloscanner.

11.
Phys Chem Chem Phys ; 18(20): 13770-80, 2016 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-26688116

RESUMEN

Proteins possess qualities of robustness and adaptability to perturbations such as mutations, but occasionally fail to withstand them, resulting in loss of function. Herein, the structural impact of mutations is investigated independently of the functional impact. Primarily, we aim at understanding the mechanisms of structural robustness pre-requisite for functional integrity. The structural changes due to mutations propagate from the site of mutation to residues much more distant than typical scales of chemical interactions, following a cascade mechanism. This can trigger dramatic changes or subtle ones, consistent with a loss of function and disease or the emergence of new functions. Robustness is enhanced by changes producing alternative structures, in good agreement with the view that proteins are dynamic objects fulfilling their functions from a set of conformations. This result, robust alternative structures, is also coherent with epistasis or rescue mutations, or more generally, with non-additive mutational effects and compensatory mutations. To achieve this study, we developed the first algorithm, referred to as Amino Acid Rank (AAR), which follows the structural changes associated with mutations from the site of the mutation to the entire protein structure and quantifies the changes so that mutations can be ranked accordingly. Assessing the paths of changes opens the possibility of assuming secondary mutations for compensatory mechanisms.


Asunto(s)
Mutación , Proteínas/química , Algoritmos , Aminoácidos/química , Simulación por Computador , Humanos , Modelos Moleculares , Conformación Proteica , Proteínas/genética
12.
Nat Commun ; 14(1): 858, 2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36813770

RESUMEN

The NHS COVID-19 app was launched in England and Wales in September 2020, with a Bluetooth-based contact tracing functionality designed to reduce transmission of SARS-CoV-2. We show that user engagement and the app's epidemiological impacts varied according to changing social and epidemic characteristics throughout the app's first year. We describe the interaction and complementarity of manual and digital contact tracing approaches. Results of our statistical analyses of anonymised, aggregated app data include that app users who were recently notified were more likely to test positive than app users who were not recently notified, by a factor that varied considerably over time. We estimate that the app's contact tracing function alone averted about 1 million cases (sensitivity analysis 450,000-1,400,000) during its first year, corresponding to 44,000 hospital cases (SA 20,000-60,000) and 9,600 deaths (SA 4600-13,000).


Asunto(s)
COVID-19 , Aplicaciones Móviles , Humanos , SARS-CoV-2 , Medicina Estatal , Gales , Trazado de Contacto/métodos , Inglaterra
13.
Virus Evol ; 8(1): veac022, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35402002

RESUMEN

Set-point viral load (SPVL), a common measure of human immunodeficiency virus (HIV)-1 virulence, is partially determined by viral genotype. Epidemiological evidence suggests that this viral property has been under stabilising selection, with a typical optimum for the virus between 104 and 105 copies of viral RNA per ml. Here we aimed to detect transmission fitness differences between viruses from individuals with different SPVLs directly from phylogenetic trees inferred from whole-genome sequences. We used the local branching index (LBI) as a proxy for transmission fitness. We found that LBI is more sensitive to differences in infectiousness than to differences in the duration of the infectious state. By analysing subtype-B samples from the Bridging the Evolution and Epidemiology of HIV in Europe project, we inferred a significant positive relationship between SPVL and LBI up to approximately 105 copies/ml, with some evidence for a peak around this value of SPVL. This is evidence of selection against low values of SPVL in HIV-1 subtype-B strains, likely related to lower infectiousness, and perhaps a peak in the transmission fitness in the expected range of SPVL. The less prominent signatures of selection against higher SPVL could be explained by an inherent limit of the method or the deployment of antiretroviral therapy.

14.
Science ; 375(6580): 540-545, 2022 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-35113714

RESUMEN

We discovered a highly virulent variant of subtype-B HIV-1 in the Netherlands. One hundred nine individuals with this variant had a 0.54 to 0.74 log10 increase (i.e., a ~3.5-fold to 5.5-fold increase) in viral load compared with, and exhibited CD4 cell decline twice as fast as, 6604 individuals with other subtype-B strains. Without treatment, advanced HIV-CD4 cell counts below 350 cells per cubic millimeter, with long-term clinical consequences-is expected to be reached, on average, 9 months after diagnosis for individuals in their thirties with this variant. Age, sex, suspected mode of transmission, and place of birth for the aforementioned 109 individuals were typical for HIV-positive people in the Netherlands, which suggests that the increased virulence is attributable to the viral strain. Genetic sequence analysis suggests that this variant arose in the 1990s from de novo mutation, not recombination, with increased transmissibility and an unfamiliar molecular mechanism of virulence.


Asunto(s)
Infecciones por VIH/virología , VIH-1/patogenicidad , Adulto , Fármacos Anti-VIH/uso terapéutico , Recuento de Linfocito CD4 , Evolución Molecular , Femenino , Genoma Viral , Genotipo , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Infecciones por VIH/transmisión , VIH-1/genética , VIH-1/fisiología , Humanos , Masculino , Mutación , Países Bajos , Filogenia , Carga Viral , Virulencia
15.
Lancet Digit Health ; 2(12): e658-e666, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33078140

RESUMEN

Background: In May 2020, the UK National Health Service (NHS) Test and Trace programme was launched in England in response to the COVID-19 pandemic. The programme was first rolled out on the Isle of Wight and included version 1 of the NHS contact tracing app. The aim of the study was to make a preliminary assessment of the epidemiological impact of the Test and Trace programme using publicly available data. Methods: We used COVID-19 daily case data from Public Health England to infer incidence of new infections and estimate the reproduction number (R) for each of the 150 Upper-Tier Local Authorities (UTLAs) in England and nationally, before and after the launch of the Test and Trace programme on the Isle of Wight. We used Bayesian and maximum-likelihood methods to estimate R and compared the Isle of Wight with other UTLAs using a synthetic control method. Findings: We observed significant decreases in incidence and R on the Isle of Wight immediately after the launch of the Test and Trace programme. The Isle of Wight had a marked reduction in R, from 1·3 before the Test and Trace programme to 0·5 after by one of our measures, and went from having the third highest R before the Test and Trace programme, to the twelfth lowest afterwards compared with other UTLAs. Interpretation: Our results show that the epidemic on the Isle of Wight was controlled quickly and effectively after the launch of Test and Trace. Our findings highlight the need for further research to determine the causes of the reduction in the spread of the disease, as these could be translated into local and national non-pharmaceutical intervention strategies in the period before a treatment or vaccination for COVID-19 becomes available. Funding: Li Ka Shing Foundation and UK Economic and Social Research Council.


Asunto(s)
Prueba de COVID-19/métodos , COVID-19/epidemiología , Trazado de Contacto/métodos , Islas/epidemiología , Adolescente , Adulto , Factores de Edad , Anciano , COVID-19/diagnóstico , COVID-19/prevención & control , Prueba de COVID-19/estadística & datos numéricos , Niño , Preescolar , Trazado de Contacto/estadística & datos numéricos , Inglaterra/epidemiología , Humanos , Lactante , Recién Nacido , Funciones de Verosimilitud , Persona de Mediana Edad , Medicina Estatal , Tiocarbamatos , Reino Unido/epidemiología , Adulto Joven
16.
Science ; 368(6491)2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32234805

RESUMEN

The newly emergent human virus SARS-CoV-2 (severe acute respiratory syndrome-coronavirus 2) is resulting in high fatality rates and incapacitated health systems. Preventing further transmission is a priority. We analyzed key parameters of epidemic spread to estimate the contribution of different transmission routes and determine requirements for case isolation and contact tracing needed to stop the epidemic. Although SARS-CoV-2 is spreading too fast to be contained by manual contact tracing, it could be controlled if this process were faster, more efficient, and happened at scale. A contact-tracing app that builds a memory of proximity contacts and immediately notifies contacts of positive cases can achieve epidemic control if used by enough people. By targeting recommendations to only those at risk, epidemics could be contained without resorting to mass quarantines ("lockdowns") that are harmful to society. We discuss the ethical requirements for an intervention of this kind.


Asunto(s)
Betacoronavirus , Teléfono Celular , Trazado de Contacto/métodos , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/transmisión , Aplicaciones Móviles , Pandemias/prevención & control , Neumonía Viral/prevención & control , Neumonía Viral/transmisión , Algoritmos , Enfermedades Asintomáticas , Número Básico de Reproducción , COVID-19 , China/epidemiología , Trazado de Contacto/ética , Infecciones por Coronavirus/epidemiología , Epidemias/prevención & control , Humanos , Control de Infecciones , Aplicaciones Móviles/ética , Modelos Teóricos , Neumonía Viral/epidemiología , Probabilidad , Cuarentena , SARS-CoV-2 , Factores de Tiempo
17.
Lancet HIV ; 7(3): e173-e183, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31953184

RESUMEN

BACKGROUND: International and global organisations advocate targeting interventions to areas of high HIV prevalence (ie, hotspots). To better understand the potential benefits of geo-targeted control, we assessed the extent to which HIV hotspots along Lake Victoria sustain transmission in neighbouring populations in south-central Uganda. METHODS: We did a population-based survey in Rakai, Uganda, using data from the Rakai Community Cohort Study. The study surveyed all individuals aged 15-49 years in four high-prevalence Lake Victoria fishing communities and 36 neighbouring inland communities. Viral RNA was deep sequenced from participants infected with HIV who were antiretroviral therapy-naive during the observation period. Phylogenetic analysis was used to infer partial HIV transmission networks, including direction of transmission. Reconstructed networks were interpreted through data for current residence and migration history. HIV transmission flows within and between high-prevalence and low-prevalence areas were quantified adjusting for incomplete sampling of the population. FINDINGS: Between Aug 10, 2011, and Jan 30, 2015, data were collected for the Rakai Community Cohort Study. 25 882 individuals participated, including an estimated 75·7% of the lakeside population and 16·2% of the inland population in the Rakai region of Uganda. 5142 participants were HIV-positive (2703 [13·7%] in inland and 2439 [40·1%] in fishing communities). 3878 (75·4%) people who were HIV-positive did not report antiretroviral therapy use, of whom 2652 (68·4%) had virus deep-sequenced at sufficient quality for phylogenetic analysis. 446 transmission networks were reconstructed, including 293 linked pairs with inferred direction of transmission. Adjusting for incomplete sampling, an estimated 5·7% (95% credibility interval 4·4-7·3) of transmissions occurred within lakeside areas, 89·2% (86·0-91·8) within inland areas, 1·3% (0·6-2·6) from lakeside to inland areas, and 3·7% (2·3-5·8) from inland to lakeside areas. INTERPRETATION: Cross-community HIV transmissions between Lake Victoria hotspots and surrounding inland populations are infrequent and when they occur, virus more commonly flows into rather than out of hotspots. This result suggests that targeted interventions to these hotspots will not alone control the epidemic in inland populations, where most transmissions occur. Thus, geographical targeting of high prevalence areas might not be effective for broader epidemic control depending on underlying epidemic dynamics. FUNDING: The Bill & Melinda Gates Foundation, the National Institute of Allergy and Infectious Diseases, the National Institute of Mental Health, the National Institute of Child Health and Development, the Division of Intramural Research of the National Institute for Allergy and Infectious Diseases, the World Bank, the Doris Duke Charitable Foundation, the Johns Hopkins University Center for AIDS Research, and the President's Emergency Plan for AIDS Relief through the Centers for Disease Control and Prevention.


Asunto(s)
Infecciones por VIH/epidemiología , Infecciones por VIH/transmisión , Adolescente , Adulto , Femenino , Infecciones por VIH/virología , VIH-1/clasificación , VIH-1/genética , VIH-1/aislamiento & purificación , VIH-1/fisiología , Humanos , Masculino , Persona de Mediana Edad , Filogenia , Prevalencia , Características de la Residencia/estadística & datos numéricos , Uganda/epidemiología , Adulto Joven
18.
Virus Evol ; 5(1): vey038, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30723550

RESUMEN

Understanding which HIV-1 variants are most likely to be transmitted is important for vaccine design and predicting virus evolution. Since most infections are founded by single variants, it has been suggested that selection at transmission has a key role in governing which variants are transmitted. We show that the composition of the viral population within the donor at the time of transmission is also important. To support this argument, we developed a probabilistic model describing HIV-1 transmission in an untreated population, and parameterised the model using both within-host next generation sequencing data and population-level epidemiological data on heterosexual transmission. The most basic HIV-1 transmission models cannot explain simultaneously the low probability of transmission and the non-negligible proportion of infections founded by multiple variants. In our model, transmission can only occur when environmental conditions are appropriate (e.g. abrasions are present in the genital tract of the potential recipient), allowing these observations to be reconciled. As well as reproducing features of transmission in real populations, our model demonstrates that, contrary to expectation, there is not a simple link between the number of viral variants and the number of viral particles founding each new infection. These quantities depend on the timing of transmission, and infections can be founded with small numbers of variants yet large numbers of particles. Including selection, or a bias towards early transmission (e.g. due to treatment), acts to enhance this conclusion. In addition, we find that infections initiated by multiple variants are most likely to have derived from donors with intermediate set-point viral loads, and not from individuals with high set-point viral loads as might be expected. We therefore emphasise the importance of considering viral diversity in donors, and the timings of transmissions, when trying to discern the complex factors governing single or multiple variant transmission.

19.
Nat Commun ; 10(1): 1411, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926780

RESUMEN

To prevent new infections with human immunodeficiency virus type 1 (HIV-1) in sub-Saharan Africa, UNAIDS recommends targeting interventions to populations that are at high risk of acquiring and passing on the virus. Yet it is often unclear who and where these 'source' populations are. Here we demonstrate how viral deep-sequencing can be used to reconstruct HIV-1 transmission networks and to infer the direction of transmission in these networks. We are able to deep-sequence virus from a large population-based sample of infected individuals in Rakai District, Uganda, reconstruct partial transmission networks, and infer the direction of transmission within them at an estimated error rate of 16.3% [8.8-28.3%]. With this error rate, deep-sequence phylogenetics cannot be used against individuals in legal contexts, but is sufficiently low for population-level inferences into the sources of epidemic spread. The technique presents new opportunities for characterizing source populations and for targeting of HIV-1 prevention interventions in Africa.


Asunto(s)
Epidemias , Infecciones por VIH/epidemiología , Infecciones por VIH/transmisión , VIH-1/fisiología , Secuenciación de Nucleótidos de Alto Rendimiento , Filogenia , Adolescente , Adulto , África/epidemiología , Secuencia de Bases , Humanos , Persona de Mediana Edad , Adulto Joven
20.
Methods Mol Biol ; 1746: 55-61, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29492886

RESUMEN

We describe a detailed protocol for the manual workup of blood (plasma/serum) samples from individuals infected with the human immunodeficiency virus type 1 (HIV-1) for deep sequence analysis of the viral genome. The study optimizing the assay was performed in the context of the BEEHIVE (Bridging the Evolution and Epidemiology of HIV in Europe) project, which analyzes complete viral genomes from more than 3000 HIV-1-infected Europeans with high-throughput deep sequencing techniques. The goal of the BEEHIVE project is to determine the contribution of viral genetics to virulence. Recently we performed a pilot experiment with 125 patient plasma samples to identify the method that is most suitable for isolation of HIV-1 viral RNA for subsequent long-amplicon deep sequencing. We reported that manual isolation with the QIAamp Viral RNA Mini Kit (Qiagen) provides superior results over robotically extracted RNA. The latter approach used the MagNA Pure 96 System in combination with the MagNA Pure 96 Instrument (Roche Diagnostics), the QIAcube robotic system (Qiagen), or the mSample Preparation Systems RNA kit with automated extraction by the m2000sp system (Abbott Molecular). Here we present a detailed protocol for the labor-intensive manual extraction method that yielded the best results.


Asunto(s)
ADN Viral/genética , Genoma Viral , Infecciones por VIH/sangre , VIH-1/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , ADN Viral/sangre , Infecciones por VIH/diagnóstico , Infecciones por VIH/virología , VIH-1/aislamiento & purificación , Humanos , Juego de Reactivos para Diagnóstico , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA