您好!欢迎来到首都医科大学附属北京胸科医院官方网站!

高分SCI文摘

您所在的位置: 首页 >>胸科图书馆 >>信息推送 >>高分SCI文摘 >>2019年

高分SCI文摘

2019年

No.9

来源:tushuguan 发布时间:2019-07-17 浏览次数:
字号: + - 14

Medical Abstracts

Keyword: tuberculosis

1. Nat Med. 2019 Jun;25(6):977-987. doi: 10.1038/s41591-019-0441-3. Epub 2019 May

20.

IFN-γ-independent immune markers of Mycobacterium tuberculosis exposure.

Lu LL(1)(2), Smith MT(3), Yu KKQ(3), Luedemann C(2), Suscovich TJ(2), Grace

PS(2), Cain A(2), Yu WH(2)(4), McKitrick TR(5), Lauffenburger D(4), Cummings

RD(5), Mayanja-Kizza H(6), Hawn TR(3), Boom WH(7), Stein CM(7)(8), Fortune

SM(1)(2), Seshadri C(9), Alter G(10).

Author information:

(1)Department of Immunology and Infectious Diseases, Harvard TH Chan School of

Public Health, Boston, MA, USA.

(2)Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.

(3)Department of Medicine, University of Washington, Seattle, WA, USA.

Erratum in

Nat Med. 2019 Jun 20;:.

Exposure to Mycobacterium tuberculosis (Mtb) results in heterogeneous clinical

outcomes including primary progressive tuberculosis and latent Mtb infection

(LTBI). Mtb infection is identified using the tuberculin skin test and

interferon-γ (IFN-γ) release assay IGRA, and a positive result may prompt

chemoprophylaxis to prevent progression to tuberculosis. In the present study, we

report on a cohort of Ugandan individuals who were household contacts of patients

with TB. These individuals were highly exposed to Mtb but tested negative disease

by IFN-γ release assay and tuberculin skin test, 'resisting' development of

classic LTBI. We show that 'resisters' possess IgM, class-switched IgG antibody

responses and non-IFN-γ T cell responses to the Mtb-specific proteins ESAT6 and

CFP10, immunologic evidence of exposure to Mtb. Compared to subjects with classic

LTBI, 'resisters' display enhanced antibody avidity and distinct Mtb-specific IgG

Fc profiles. These data reveal a distinctive adaptive immune profile among

Mtb-exposed subjects, supporting an expanded definition of the host response to

Mtb exposure, with implications for public health and the design of clinical

trials.

DOI: 10.1038/s41591-019-0441-3

PMCID: PMC6559862 [Available on 2019-11-20]

PMID: 31110348 [Indexed for MEDLINE]

2. Nat Rev Immunol. 2019 May 21. doi: 10.1038/s41577-019-0174-z. [Epub ahead of

print]

Moving tuberculosis vaccines from theory to practice.

Andersen P(1)(2), Scriba TJ(3).

Author information:

(1)Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark.

pa@ssi.dk.

(2)Department of Immunology and Microbiology, University of Copenhagen,

Copenhagen, Denmark. pa@ssi.dk.

(3)South African Tuberculosis Vaccine Initiative, Institute of Infectious Disease

and Molecular Medicine and Division of Immunology, Department of Pathology,

University of Cape Town, Cape Town, South Africa. thomas.scriba@uct.ac.za.

Tuberculosis (TB) vaccine research has reached a unique point in time.

Breakthrough findings in both the basic immunology of Mycobacterium tuberculosis

infection and the clinical development of TB vaccines suggest, for the first time

since the discovery of the Mycobacterium bovis bacillus Calmette-Guérin (BCG)

vaccine more than a century ago, that a novel, efficacious TB vaccine is

imminent. Here, we review recent data in the light of our current understanding

of the immunology of TB infection and discuss the identification of biomarkers

for vaccine efficacy and the next steps in the quest for an efficacious vaccine

that can control the global TB epidemic.

DOI: 10.1038/s41577-019-0174-z

PMID: 31114037

3. Lancet Infect Dis. 2019 May 30. pii: S1473-3099(19)30001-5. doi:

10.1016/S1473-3099(19)30001-5. [Epub ahead of print]

Novel lipoarabinomannan point-of-care tuberculosis test for people with HIV: a

diagnostic accuracy study.

Broger T(1), Sossen B(2), du Toit E(3), Kerkhoff AD(4), Schutz C(2), Ivanova

Reipold E(1), Ward A(2), Barr DA(5), Macé A(1), Trollip A(6), Burton R(7),

Ongarello S(1), Pinter A(8), Lowary TL(9), Boehme C(1), Nicol MP(3), Meintjes

G(10), Denkinger CM(11).

Author information:

(1)FIND, Geneva, Switzerland.

(2)Department of Medicine, Faculty of Health Sciences, University of Cape Town,

Cape Town, South Africa; Wellcome Center for Infectious Diseases Research in

Africa, Institute of Infectious Disease and Molecular Medicine, University of

Cape Town, Cape Town, South Africa.

(3)Division of Medical Microbiology, University of Cape Town, Cape Town, South

Africa; National Health Laboratory Service, Cape Town, South Africa.

BACKGROUND: Most tuberculosis-related deaths in people with HIV could be

prevented with earlier diagnosis and treatment. The only commercially available

tuberculosis point-of-care test (Alere Determine TB LAM Ag [AlereLAM]) has

suboptimal sensitivity, which restricts its use in clinical practice. The novel

Fujifilm SILVAMP TB LAM (FujiLAM) assay has been developed to improve the

sensitivity of AlereLAM. We assessed the diagnostic accuracy of the FujiLAM assay

for the detection of tuberculosis in hospital inpatients with HIV compared with

the AlereLAM assay.

METHODS: For this diagnostic accuracy study, we assessed biobanked urine samples

obtained from the FIND Specimen Bank and the University of Cape Town Biobank,

which had been collected from hospital inpatients (aged ≥18 years) with HIV

during three independent prospective cohort studies done at two South African

hospitals. Urine samples were tested using FujiLAM and AlereLAM assays. The

conduct and reporting of each test was done blind to other test results. The

primary objective was to assess the diagnostic accuracy of FujiLAM compared with

AlereLAM, against microbiological and composite reference standards (including

clinical diagnoses).

FINDINGS: Between April 18, 2018, and May 3, 2018, urine samples from 968

hospital inpatients with HIV were evaluated. The prevalence of

microbiologically-confirmed tuberculosis was 62% and the median CD4 count was 86

cells per μL. Using the microbiological reference standard, the estimated

sensitivity of FujiLAM was 70·4% (95% CI 53·0 to 83·1) compared with 42·3% (31·7

to 51·8) for AlereLAM (difference 28·1%) and the estimated specificity of FujiLAM

was 90·8% (86·0 to 94·4) and 95·0% (87·7-98·8) for AlereLAM (difference -4·2%).Against the composite reference standard, the specificity of both assays was

higher (95·7% [92·0 to 98·0] for FujiLAM vs 98·2% [95·7 to 99·6] for AlereLAM;

difference -2·5%), but the sensitivity of both assays was lower (64·9% [50·1 to

76·7] for FujiLAM vs 38·2% [28·1 to 47·3] for AlereLAM; difference 26·7%).

INTERPRETATION: In comparison to AlereLAM, FujiLAM offers superior diagnostic

sensitivity, while maintaining specificity, and could transform rapid

point-of-care tuberculosis diagnosis for hospital inpatients with HIV. The

applicability of FujiLAM for settings of intended use requires prospective

assessment.

FUNDING: Global Health Innovative Technology Fund, UK Department for

International Development, Dutch Ministry of Foreign Affairs, Bill & Melinda

Gates Foundation, German Federal Ministry of Education and Research, Australian

Department of Foreign Affairs and Trade, Wellcome Trust, Department of Science

and Technology and National Research Foundation of South Africa, and South

African Medical Research Council.

Copyright © 2019 Author(s). Published by Elsevier Ltd. This is an Open Access

article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights

reserved.

DOI: 10.1016/S1473-3099(19)30001-5

PMID: 31155318

4. Nat Commun. 2019 May 27;10(1):2329. doi: 10.1038/s41467-019-10065-8.

Heterogeneous GM-CSF signaling in macrophages is associated with control of

Mycobacterium tuberculosis.

Bryson BD(1)(2), Rosebrock TR(1)(2), Tafesse FG(3), Itoh CY(1)(2), Nibasumba

A(1)(2), Babunovic GH(1)(2), Corleis B(2), Martin C(1)(2), Keegan C(4), Andrade

P(4), Realegeno S(4), Kwon D(2), Modlin RL(4), Fortune SM(5)(6).

Author information:

(1)Harvard T. H. Chan School of Public Health, 655 Huntington Avenue Boston,

Boston, MA, 02115, USA.

(2)Ragon Institute of MGH, MIT, and Harvard, 400 Technology Square Cambridge,

Cambridge, MA, 02139, USA.

(3)Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, Portland,

OR, 97239, USA.

Variability in bacterial sterilization is a key feature of Mycobacterium

tuberculosis (Mtb) disease. In a population of human macrophages, there are

macrophages that restrict Mtb growth and those that do not. However, the sources

of heterogeneity in macrophage state during Mtb infection are poorly understood.

Here, we perform RNAseq on restrictive and permissive macrophages and reveal that

the expression of genes involved in GM-CSF signaling discriminates between the

two subpopulations. We demonstrate that blocking GM-CSF makes macrophages more

permissive of Mtb growth while addition of GM-CSF increases bacterial control. In

parallel, we find that the loss of bacterial control that occurs in HIV-Mtb

coinfected macrophages correlates with reduced GM-CSF secretion. Treatment of

coinfected cells with GM-CSF restores bacterial control. Thus, we leverage the

natural variation in macrophage control of Mtb to identify a critical cytokine

response for regulating Mtb survival and identify components of the antimicrobial

response induced by GM-CSF.

DOI: 10.1038/s41467-019-10065-8

PMCID: PMC6536549

PMID: 31133636 [Indexed for MEDLINE]

5. Nat Commun. 2019 May 13;10(1):2128. doi: 10.1038/s41467-019-10110-6.

GWAS for quantitative resistance phenotypes in Mycobacterium tuberculosis reveals

resistance genes and regulatory regions.

Farhat MR(1)(2), Freschi L(3), Calderon R(4), Ioerger T(5), Snyder M(6), Meehan

CJ(7), de Jong B(7), Rigouts L(7), Sloutsky A(8), Kaur D(9), Sunyaev S(3)(10),

van Soolingen D(11), Shendure J(6)(12)(13), Sacchettini J(5), Murray M(14).

Author information:

(1)Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.

Maha_Farhat@hms.harvard.edu.

(2)Division of Pulmonary and Critical Care, Massachusetts General Hospital,

Boston, MA, USA. Maha_Farhat@hms.harvard.edu.

(3)Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.

Drug resistance diagnostics that rely on the detection of resistance-related

mutations could expedite patient care and TB eradication. We perform minimum

inhibitory concentration testing for 12 anti-TB drugs together with Illumina

whole-genome sequencing on 1452 clinical Mycobacterium tuberculosis (MTB)

isolates. We evaluate genome-wide associations between mutations in MTB genes or

non-coding regions and resistance, followed by validation in an independent data

set of 792 patient isolates. We confirm associations at 13 non-canonical loci,

with two involving non-coding regions. Promoter mutations are measured to have

smaller average effects on resistance than gene body mutations. We estimate the

heritability of the resistance phenotype to 11 anti-TB drugs and identify a lower

than expected contribution from known resistance genes. This study highlights the

complexity of the genomic mechanisms associated with the MTB resistance

phenotype, including the relatively large number of potentially causal loci, and

emphasizes the contribution of the non-coding portion of the genome.

DOI: 10.1038/s41467-019-10110-6

PMCID: PMC6513847

PMID: 31086182 [Indexed for MEDLINE]

6. PLoS Biol. 2019 May 13;17(5):e3000265. doi: 10.1371/journal.pbio.3000265.

eCollection 2019 May.

Transition bias influences the evolution of antibiotic resistance in

Mycobacterium tuberculosis.

Payne JL(1)(2), Menardo F(3)(4), Trauner A(3)(4), Borrell S(3)(4), Gygli

SM(3)(4), Loiseau C(3)(4), Gagneux S(3)(4), Hall AR(1).

Author information:

(1)Institute of Integrative Biology, ETH Zurich, Switzerland.

(2)Swiss Institute of Bioinformatics, Lausanne, Switzerland.

(3)Swiss Tropical and Public Health Institute, Basel, Switzerland.

(4)University of Basel, Basel, Switzerland.

Transition bias, an overabundance of transitions relative to transversions, has

been widely reported among studies of the rates and spectra of spontaneous

mutations. However, demonstrating the role of transition bias in adaptive

evolution remains challenging. In particular, it is unclear whether such biases

direct the evolution of bacterial pathogens adapting to treatment. We addressed

this challenge by analyzing adaptive antibiotic-resistance mutations in the major

human pathogen Mycobacterium tuberculosis (MTB). We found strong evidence for

transition bias in two independently curated data sets comprising 152 and 208

antibiotic-resistance mutations. This was true at the level of mutational paths

(distinct adaptive DNA sequence changes) and events (individual instances of the

adaptive DNA sequence changes) and across different genes and gene promoters

conferring resistance to a diversity of antibiotics. It was also true for

mutations that do not code for amino acid changes (in gene promoters and the 16S

ribosomal RNA gene rrs) and for mutations that are synonymous to each other and

are therefore likely to have similar fitness effects, suggesting that transition

bias can be caused by a bias in mutation supply. These results point to a central

role for transition bias in determining which mutations drive adaptive antibiotic

resistance evolution in a key pathogen.

DOI: 10.1371/journal.pbio.3000265

PMCID: PMC6532934

PMID: 31083647

Conflict of interest statement: The authors have declared that no competing

interests exist.

7. Microbiol Mol Biol Rev. 2019 Mar 27;83(2). pii: e00062-18. doi:

10.1128/MMBR.00062-18. Print 2019 May 15.

Deciphering Within-Host Microevolution of Mycobacterium tuberculosis through

Whole-Genome Sequencing: the Phenotypic Impact and Way Forward.

Ley SD(1), de Vos M(1), Van Rie A(#)(2), Warren RM(#)(3).

Author information:

(1)DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South

African Medical Research Council Centre for Tuberculosis Research; Division of

Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences,

Stellenbosch University, Cape Town, South Africa.

(2)Department of Epidemiology and Social Medicine, Faculty of Medicine and Health

Sciences, University of Antwerp, Antwerp, Belgium.

(3)DST-NRF Centre of Excellence for Biomedical Tuberculosis Research; South

African Medical Research Council Centre for Tuberculosis Research; Division of

Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences,

Stellenbosch University, Cape Town, South Africa rw1@sun.ac.za.

(#)Contributed equally

The Mycobacterium tuberculosis genome is more heterogenous and less genetically

stable within the host than previously thought. Currently, only limited data

exist on the within-host microevolution, diversity, and genetic stability of M.

tuberculosis As a direct consequence, our ability to infer M. tuberculosis

transmission chains and to understand the full complexity of drug resistance

profiles in individual patients is limited. Furthermore, apart from the

acquisition of certain drug resistance-conferring mutations, our knowledge on the

function of genetic variants that emerge within a host and their phenotypic

impact remains scarce. We performed a systematic literature review of

whole-genome sequencing studies of serial and parallel isolates to summarize the

knowledge on genetic diversity and within-host microevolution of M. tuberculosis

We identified genomic loci of within-host emerged variants found across multiple

studies and determined their functional relevance. We discuss important remaining

knowledge gaps and finally make suggestions on the way forward.

Copyright © 2019 American Society for Microbiology.

DOI: 10.1128/MMBR.00062-18

PMID: 30918049 [Indexed for MEDLINE]

8. Lancet Infect Dis. 2019 May;19(5):519-528. doi: 10.1016/S1473-3099(18)30753-9.

Epub 2019 Mar 22.

Active and passive case-finding in tuberculosis-affected households in Peru: a

10-year prospective cohort study.

Saunders MJ(1), Tovar MA(2), Collier D(3), Baldwin MR(4), Montoya R(5), Valencia

TR(6), Gilman RH(7), Evans CA(2).

Author information:

(1)Infectious Diseases and Immunity, Imperial College London, and Wellcome Trust

Imperial College Centre for Global Health Research, London, UK; Innovation for

Health and Development (IFHAD), Laboratory of Research and Development,

Universidad Peruana Cayetano Heredia, Lima, Peru; Innovación Por la Salud Y

Desarrollo (IPSYD), Asociación Benéfica PRISMA, Lima, Peru. Electronic address:

matthew.saunders@ifhad.org.

(2)Infectious Diseases and Immunity, Imperial College London, and Wellcome Trust

Imperial College Centre for Global Health Research, London, UK; Innovation for

Health and Development (IFHAD), Laboratory of Research and Development,

Universidad Peruana Cayetano Heredia, Lima, Peru; Innovación Por la Salud Y

Desarrollo (IPSYD), Asociación Benéfica PRISMA, Lima, Peru.

(3)Innovation for Health and Development (IFHAD), Laboratory of Research and

Development, Universidad Peruana Cayetano Heredia, Lima, Peru; Innovación Por la

Salud Y Desarrollo (IPSYD), Asociación Benéfica PRISMA, Lima, Peru.

BACKGROUND: Active case-finding among contacts of patients with tuberculosis is a

global health priority, but the effects of active versus passive case-finding are

poorly characterised. We assessed the contribution of active versus passive

case-finding to tuberculosis detection among contacts and compared sex and

disease characteristics between contacts diagnosed through these strategies.

METHODS: In shanty towns in Callao, Peru, we identified index patients with

tuberculosis and followed up contacts aged 15 years or older for tuberculosis.

All patients and contacts were offered free programmatic active case-finding

entailing sputum smear microscopy and clinical assessment. Additionally, all

contacts were offered intensified active case-finding with sputum smear and

culture testing monthly for 6 months and then once every 4 years. Passive

case-finding at local health facilities was ongoing throughout follow-up.

FINDINGS: Between Oct 23, 2002, and May 26, 2006, we identified 2666 contacts,

who were followed up until March 1, 2016. Median follow-up was 10·0 years (IQR

7·5-11·0). 232 (9%) of 2666 contacts were diagnosed with tuberculosis. The 2-year

cumulative risk of tuberculosis was 4·6% (95% CI 3·5-5·5), and overall incidence

was 0·98 cases (95% CI 0·86-1·10) per 100 person-years. 53 (23%) of 232 contacts

with tuberculosis were diagnosed through active case-finding and 179 (77%) were

identified through passive case-finding. During the first 6 months of the study,

23 (45%) of 51 contacts were diagnosed through active case-finding and 28 (55%)

were identified through passive case-finding. Contacts diagnosed through active

versus passive case-finding were more frequently female (36 [68%] of 53 vs 85

[47%] of 179; p=0·009), had a symptom duration of less than 15 days (nine [25%]

of 36 vs ten [8%] of 127; p=0·03), and were more likely to be sputum

smear-negative (33 [62%] of 53 vs 62 [35%] of 179; p=0·0003).

INTERPRETATION: Although active case-finding made an important contribution to

tuberculosis detection among contacts, passive case-finding detected most of the

tuberculosis burden. Compared with passive case-finding, active case-finding was

equitable, helped to diagnose tuberculosis earlier and usually before a positive

result on sputum smear microscopy, and showed a high burden of undetected

tuberculosis among women.

FUNDING: Wellcome Trust, Department for International Development Civil Society

Challenge Fund, Joint Global Health Trials consortium, Bill & Melinda Gates

Foundation, Imperial College National Institutes of Health Research Biomedical

Research Centre, Foundation for Innovative New Diagnostics, Sir Halley Stewart

Trust, WHO, TB REACH, and IFHAD: Innovation for Health and Development.

Copyright © 2019 The Author(s). Published by Elsevier Ltd. This is an Open Access

article under the CC BY 4.0 license. Published by Elsevier Ltd.. All rights

reserved.

DOI: 10.1016/S1473-3099(18)30753-9

PMCID: PMC6483977

PMID: 30910427

9. J Antimicrob Chemother. 2019 May 22. pii: dkz215. doi: 10.1093/jac/dkz215. [Epub

ahead of print]

WGS more accurately predicts susceptibility of Mycobacterium tuberculosis to

first-line drugs than phenotypic testing.

Jajou R(1), van der Laan T(1), de Zwaan R(1), Kamst M(1), Mulder A(1), de Neeling

A(1), Anthony R(1), van Soolingen D(1).

Author information:

(1)National Tuberculosis Reference Laboratory, Centre for Infectious Disease

Control, National Institute for Public Health and the Environment (RIVM),

Bilthoven, The Netherlands.

BACKGROUND: Drug-susceptibility testing (DST) of Mycobacterium tuberculosis

complex (MTBC) isolates by the Mycobacteria Growth Indicator Tube (MGIT) approach

is the most widely applied reference standard. However, the use of WGS is

increasing in many developed countries to detect resistance and predict

susceptibility. We investigated the reliability of WGS in predicting drug

susceptibility, and analysed the discrepancies between WGS and MGIT against the

first-line drugs rifampicin, isoniazid, ethambutol and pyrazinamide.

METHODS: DST by MGIT and WGS was performed on MTBC isolates received in

2016/2017. Nine genes and/or their promotor regions were investigated for

resistance-associated mutations: rpoB, katG, fabG1, ahpC, inhA, embA, embB, pncA

and rpsA. Isolates that were discrepant in their MGIT/WGS results and a control

group with concordant results were retested in the MGIT, at the critical

concentration and a lower concentration, and incubated for up to 45days after

the control tube became positive in the MGIT.

RESULTS: In total, 1136 isolates were included, of which 1121 were routine MTBC

isolates from the Netherlands. The negative predictive value of WGS was ≥99.3%

for all four first-line antibiotics. The majority of discrepancies for isoniazid

and ethambutol were explained by growth at the lower concentrations, and for

rifampicin by prolonged incubation in the MGIT, both indicating low-level

resistance.

CONCLUSIONS: Applying WGS in a country like the Netherlands, with a low TB

incidence and low prevalence of resistance, can reduce the need for phenotypic

DST for 90% of isolates and accurately detect mutations associated with

low-level resistance, often missed in conventional DST.

© The Author(s) 2019. Published by Oxford University Press on behalf of the

British Society for Antimicrobial Chemotherapy. All rights reserved. For

permissions, please email: journals.permissions@oup.com.

DOI: 10.1093/jac/dkz215

PMID: 31119271

10. Eur Respir J. 2019 May 16. pii: 1802242. doi: 10.1183/13993003.02242-2018. [Epub

ahead of print]

IL-4 subverts mycobacterial containment in M. tuberculosis-infected human

macrophages.

Pooran A(1), Davids M(1), Nel A(2), Shoko A(3), Blackburn J(2), Dheda K(4)(5).

Author information:

(1)Division of Pulmonology, Department of Medicine and UCT Lung Institute,

University of Cape Town, Cape Town, South Africa.

(2)Department of Integrative Biomedical Sciences; Institute for Infectious

Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.

(3)Centre for Proteomics and Genomics Research, Cape Town, South Africa.

(4)Division of Pulmonology, Department of Medicine and UCT Lung Institute,

University of Cape Town, Cape Town, South Africa keertan.dheda@uct.ac.za.

(5)Faculty of Infectious and Tropical Diseases, Department of Immunology and

Infection, London School of Hygiene and Tropical Medicine, London, UK.

Protective immunity against Mycobacterium tuberculosis is poorly understood. The

role of interleukin-4 (IL-4), the archetypal T-helper-2 (Th2) cytokine, in the

immunopathogenesis of human tuberculosis remains unclear.Blood and/or

broncho-alveolar lavage fluid (BAL) were obtained from participants with

pulmonary TB (TB; n=23) and presumed latent TB infection (LTBI; n=22). Messenger

RNA expression levels of interferon-gamma (IFN-γ), IL-4, and its splice variant

IL-4δ2 were determined by real-time PCR. The effect of human recombinant IL-4

(hrIL-4) on mycobacterial survival/containment [colony-forming-units (CFU·mL-1]

was evaluated in M. tuberculosis-infected macrophages co-cultured with

mycobacterial antigen-primed effector T-cells. Regulatory T-cell (Treg) and Th1

cytokine levels were evaluated using flow cytometry.In blood, but not BAL, IL-4

mRNA levels (p=0.02) and the IL-4/IFN-γ ratio (p=0.01) was higher in TB versus

LTBI. hrIL-4 reduced mycobacterial containment in infected macrophages (p<0.008)

in a dose-dependent manner and was associated with an increase in Tregs (p<0.001)

but decreased CD4+Th1 cytokine levels (CD4+IFN-γ+: p<0.001; CD4+TNFα+: p=0.01).

Blocking IL-4 significantly neutralised mycobacterial containment (p=0.03),

CD4+IFNγ+ levels (p=0.03) and Treg expression (p=0.03).IL-4 can subvert

mycobacterial containment in human macrophages, likely via perturbations in Treg

and Th1-linked pathways. These data may have implications in the design of

effective TB vaccines and host-directed therapies.

Copyright ©ERS 2019.

DOI: 10.1183/13993003.02242-2018

PMID: 31097521

11. Clin Infect Dis. 2019 May 10. pii: ciz380. doi: 10.1093/cid/ciz380. [Epub ahead

of print]

Sub-therapeutic rifampicin concentration is associated with unfavourable

tuberculosis treatment outcomes.

Ramachandran G(1), Chandrasekaran P(1), Gaikwad S(2), Hemanth Kumar AK(1),

Thiruvengadam K(1), Gupte N(3)(4), Paradkar M(4), Dhanasekaran K(1),

Sivaramakrishnan GN(1), Kagal A(2), Thomas B(1), Pradhan N(4), Kadam D(2), Hanna

LE(1), Balasubramanian U(4), Kulkarni V(4), Murali L(5), Golub J(3)(6), Gupte

A(3), Shivakumar SVBY(7), Swaminathan S(8), Dooley KE(3), Gupta A(3)(4)(6), Mave

V(3)(4); C-TRIUMPh team.

Author information:

(1)National Institute for Research in Tuberculosis (ICMR), Chennai, Tamil Nadu,

India.

(2)Byramjee Jeejeebhoy Government Medical College, Pune, Maharashtra, India.

(3)Johns Hopkins School of Medicine, Baltimore, Maryland, USA.

BACKGROUND: The relationships between first-line drug concentrations and

clinically-important outcomes among patients with tuberculosis (TB) remain poorly

understood.

METHODS: We enrolled a prospective cohort of patients with new pulmonary TB

receiving thrice-weekly treatment in India. Maximum plasma concentration of each

drug was determined at month 1 and 5 using blood samples drawn 2 hours post-dose.

Sub-therapeutic cut-offs were: rifampicin <8µg/mL; isoniazid <3µg/mL;

pyrazinamide <20µg/mL. Factors associated with lower log-transformed drug

concentrations, unfavourable outcomes (composite of treatment failure, all-cause

mortality, and recurrence) as well as individual outcomes were examined using

Poisson regression models.

RESULTS: Among 404 participants, rifampicin, isoniazid, and pyrazinamide

concentrations were sub-therapeutic in 85%, 29%, and 12% at month 1 (with similar

results for rifampicin and isoniazid at month 5). Rifampicin concentrations were

lower with HIV co-infection (1.6 µg/ml vs 4.6 µg/ml; p = 0.015). Unfavourable

outcome was observed in 19%; a 1 ug/ml decrease in rifampicin concentration was

independently associated with unfavourable outcome (aIRR 1.21, 95% CI: 1.01 -

1.47) and treatment failure (aIRR: 1.16; 95% CI: 1.05 - 1.28). A 1 ug/ml decrease

in pyrazinamide concentration was associated with recurrence (aIRR: 1.05; 95% CI:

1.01-1.11).

CONCLUSIONS: Rifampicin concentrations were sub-therapeutic in most Indian

patients taking a thrice-weekly TB regimen, and low rifampicin and pyrazinamide

concentrations were associated with poor outcomes. Higher or more frequent dosing

is needed to improve TB treatment outcomes in India.

© The Author(s) 2019. Published by Oxford University Press for the Infectious

Diseases Society of America. All rights reserved. For permissions, e-mail:

journals.permissions@oup.com.

DOI: 10.1093/cid/ciz380

PMID: 31075166

12. Proc Natl Acad Sci U S A. 2019 May 21;116(21):10430-10434. doi:

10.1073/pnas.1903561116. Epub 2019 May 8.

Homozygosity for TYK2 P1104A underlies tuberculosis in about 1% of patients in a

cohort of European ancestry.

Kerner G(1)(2), Ramirez-Alejo N(3), Seeleuthner Y(1)(2), Yang R(3), Ogishi M(3),

Cobat A(1)(2), Patin E(4), Quintana-Murci L(4), Boisson-Dupuis S(1)(2)(3),

Casanova JL(5)(2)(3)(6)(7), Abel L(1)(2)(3).

Author information:

(1)Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR

1163, Necker Hospital for Sick Children, 75015 Paris, France.

(2)Imagine Institute, Paris Descartes University, 75015 Paris, France.

(3)St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller

Branch, The Rockefeller University, New York, NY 10065.

(4)Human Evolutionary Genetics Unit, Institut Pasteur, CNRS UMR2000, 75015 Paris,

France.

(5)Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM UMR

1163, Necker Hospital for Sick Children, 75015 Paris, France;

The human genetic basis of tuberculosis (TB) has long remained elusive. We

recently reported a high level of enrichment in homozygosity for the common TYK2

P1104A variant in a heterogeneous cohort of patients with TB from non-European

countries in which TB is endemic. This variant is homozygous in 1/600 Europeans

and 1/5,000 people from other countries outside East Asia and sub-Saharan

Africa. We report a study of this variant in the UK Biobank cohort. The frequency

of P1104A homozygotes was much higher in patients with TB (6/620, 1%) than in

controls (228/114,473, 0.2%), with an odds ratio (OR) adjusted for ancestry of

5.0 [95% confidence interval (CI): 1.96-10.31, P = 2 × 10-3]. Conversely, we did

not observe enrichment for P1104A heterozygosity, or for TYK2 I684S or V362F

homozygosity or heterozygosity. Moreover, it is unlikely that more than 10% of

controls were infected with Mycobacterium tuberculosis, as 97% were of European

genetic ancestry, born between 1939 and 1970, and resided in the United Kingdom.

Had all of them been infected, the OR for developing TB upon infection would be

higher. These findings suggest that homozygosity for TYK2 P1104A may account for

1% of TB cases in Europeans.

DOI: 10.1073/pnas.1903561116

PMCID: PMC6534977 [Available on 2019-11-08]

PMID: 31068474

13. Proc Natl Acad Sci U S A. 2019 May 21;116(21):10510-10517. doi:

10.1073/pnas.1818009116. Epub 2019 May 6.

Chemical disarming of isoniazid resistance in Mycobacterium tuberculosis.

Flentie K(1), Harrison GA(1), Tükenmez H(2), Livny J(3), Good JAD(4)(5), Sarkar

S(4)(5), Zhu DX(1), Kinsella RL(1), Weiss LA(1), Solomon SD(1), Schene ME(1),

Hansen MR(4)(5), Cairns AG(4)(5), Kulén M(4)(5), Wixe T(4)(5), Lindgren

AEG(4)(5), Chorell E(1)(4)(5), Bengtsson C(4)(5), Krishnan KS(4)(5), Hultgren

SJ(1)(6), Larsson C(2)(5), Almqvist F(7)(5), Stallings CL(8).

Author information:

(1)Department of Molecular Microbiology, Washington University School of

Medicine, St. Louis, MO 63110.

(2)Department of Molecular Biology, Umeå University, SE-90187 Umeå, Sweden.

(3)Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA

02142.

Mycobacterium tuberculosis (Mtb) killed more people in 2017 than any other single

infectious agent. This dangerous pathogen is able to withstand stresses imposed

by the immune system and tolerate exposure to antibiotics, resulting in

persistent infection. The global tuberculosis (TB) epidemic has been exacerbated

by the emergence of mutant strains of Mtb that are resistant to frontline

antibiotics. Thus, both phenotypic drug tolerance and genetic drug resistance are

major obstacles to successful TB therapy. Using a chemical approach to identify

compounds that block stress and drug tolerance, as opposed to traditional screens

for compounds that kill Mtb, we identified a small molecule, C10, that blocks

tolerance to oxidative stress, acid stress, and the frontline antibiotic

isoniazid (INH). In addition, we found that C10 prevents the selection for

INH-resistant mutants and restores INH sensitivity in otherwise INH-resistant Mtb

strains harboring mutations in the katG gene, which encodes the enzyme that

converts the prodrug INH to its active form. Through mechanistic studies, we

discovered that C10 inhibits Mtb respiration, revealing a link between

respiration homeostasis and INH sensitivity. Therefore, by using C10 to dissect

Mtb persistence, we discovered that INH resistance is not absolute and can be

reversed.

DOI: 10.1073/pnas.1818009116

PMCID: PMC6535022 [Available on 2019-11-06]

PMID: 31061116

Conflict of interest statement: Conflict of interest statement: C.L.S., S.J.H.,

and F.A. have ownership interests in Quretech Bio AB, which licenses C10.

14. Clin Pharmacokinet. 2019 May 3. doi: 10.1007/s40262-019-00764-2. [Epub ahead of print]

Clinical Pharmacokinetics and Pharmacodynamics of Rifampicin in Human

Tuberculosis.

Abulfathi AA(1), Decloedt EH(2), Svensson EM(3)(4), Diacon AH(5)(6), Donald P(7),

Reuter H(2).

Author information:

(1)Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine

and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South

Africa. aaabulfathi@sun.ac.za.

(2)Division of Clinical Pharmacology, Department of Medicine, Faculty of Medicine

and Health Sciences, Stellenbosch University, PO Box 241, Cape Town, 8000, South

Africa.

(3)Department of Pharmacy, Radboud Institute for Health Sciences, Radboud

University Medical Center, Nijmegen, The Netherlands.

The introduction of rifampicin (rifampin) into tuberculosis (TB) treatment five

decades ago was critical for shortening the treatment duration for patients with

pulmonary TB to 6 months when combined with pyrazinamide in the first 2 months.

Resistance or hypersensitivity to rifampicin effectively condemns a patient to

prolonged, less effective, more toxic, and expensive regimens. Because of cost

and fears of toxicity, rifampicin was introduced at an oral daily dose of 600 mg

(8-12 mg/kg body weight). At this dose, clinical trials in 1970s found cure rates

of ≥ 95% and relapse rates of < 5%. However, recent papers report lower cure

rates that might be the consequence of increased emergence of resistance. Several

lines of evidence suggest that higher rifampicin doses, if tolerated and safe,

could shorten treatment duration even further. We conducted a narrative review of

rifampicin pharmacokinetics and pharmacodynamics in adults across a range of

doses and highlight variables that influence its

pharmacokinetics/pharmacodynamics. Rifampicin exposure has considerable inter-

and intra-individual variability that could be reduced by administration during

fasting. Several factors including malnutrition, HIV infection, diabetes

mellitus, dose size, pharmacogenetic polymorphisms, hepatic cirrhosis, and

substandard medicinal products alter rifampicin exposure and/or efficacy. Renal

impairment has no influence on rifampicin pharmacokinetics when dosed at 600 mg.

Rifampicin maximum (peak) concentration (Cmax) > 8.2 μg/mL is an independent

predictor of sterilizing activity and therapeutic drug monitoring at 2, 4, and

6 h post-dose may aid in optimizing dosing to achieve the recommended rifampicin

concentration of ≥ 8 µg/mL. A higher rifampicin Cmax is required for severe forms

TB such as TB meningitis, with Cmax ≥ 22 μg/mL and area under the

concentration-time curve (AUC) from time zero to 6 h (AUC6) ≥ 70 μg·h/mL

associated with reduced mortality. More studies are needed to confirm whether

doses achieving exposures higher than the current standard dosage could translate

into faster sputum conversion, higher cure rates, lower relapse rates, and less

mortality. It is encouraging that daily rifampicin doses up to 35 mg/kg were

found to be safe and well-tolerated over a period of 12 weeks. High-dose

rifampicin should thus be considered in future studies when constructing

potentially shorter regimens. The studies should be adequately powered to

determine treatment outcomes and should include surrogate markers of efficacy

such as Cmax/MIC (minimum inhibitory concentration) and AUC/MIC.

DOI: 10.1007/s40262-019-00764-2

PMID: 31049868

15. Emerg Infect Dis. 2019 May;25(5):936-943. doi: 10.3201/eid2505.181823.

Outcomes of Bedaquiline Treatment in Patients with Multidrug-Resistant

Tuberculosis.

Mbuagbaw L, Guglielmetti L, Hewison C, Bakare N, Bastard M, Caumes E,

Fréchet-Jachym M, Robert J, Veziris N, Khachatryan N, Kotrikadze T, Hayrapetyan

A, Avaliani Z, Schünemann HJ, Lienhardt C.

Bedaquiline is recommended by the World Health Organization for the treatment of

multidrug-resistant (MDR) and extensively drug-resistant (XDR) tuberculosis (TB).

We pooled data from 5 cohorts of patients treated with bedaquiline in France,

Georgia, Armenia, and South Africa and in a multicountry study. The rate of

culture conversion to negative at 6 months (by the end of 6 months of treatment)

was 78% (95% CI 73.5%-81.9%), and the treatment success rate was 65.8% (95% CI

59.9%-71.3%). Death rate was 11.7% (95% CI 7.0%-19.1%). Up to 91.1% (95% CI

82.2%-95.8%) of the patients experienced >1 adverse event, and 11.2% (95% CI

5.0%-23.2%) experienced a serious adverse event. Lung cavitations were

consistently associated with unfavorable outcomes. The use of bedaquiline in MDR

and XDR TB treatment regimens appears to be effective and safe across different

settings, although the certainty of evidence was assessed as very low.

DOI: 10.3201/eid2505.181823

PMCID: PMC6478224

PMID: 31002070

上一篇: No.10

下一篇: No.8