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 >>2021年

高分SCI文摘

2021年

No.10

发布时间:2022-01-14 浏览次数:
字号: + - 14

Medical Abstracts

Filters applied: from 2021/8/1 - 2021/8/31

Key words:   tuberculosis ;  lung cancer

 

1. Lancet. 2021 Oct 9;398(10308):1344-1357. doi: 10.1016/S0140-6736(21)02098-5.

Epub 2021 Sep 20.

 

Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA

non-small-cell lung cancer (IMpower010): a randomised, multicentre, open-label,

phase 3 trial.

 

Felip E(1), Altorki N(2), Zhou C(3), Csőszi T(4), Vynnychenko I(5), Goloborodko

O(6), Luft A(7), Akopov A(8), Martinez-Marti A(9), Kenmotsu H(10), Chen YM(11),

Chella A(12), Sugawara S(13), Voong D(14), Wu F(15), Yi J(14), Deng Y(14),

McCleland M(14), Bennett E(14), Gitlitz B(14), Wakelee H(16); IMpower010

Investigators.

 

Author information:

(1)Vall d'Hebron Institute of Oncology, Vall d'Hebron University Hospital,

Barcelona, Spain. Electronic address: efelip@vhio.net.

(2)Division of Thoracic Surgery, Weill Cornell Medicine, New York-Presbyterian

Hospital, New York, NY, USA.

(3)Department of Oncology, Tongji University Affiliated Shanghai Pulmonary

Hospital, Shanghai, China.

 

Erratum in

    Lancet. 2021 Sep 23;:

 

BACKGROUND: Novel adjuvant strategies are needed to optimise outcomes after

complete surgical resection in patients with early-stage non-small-cell lung

cancer (NSCLC). We aimed to evaluate adjuvant atezolizumab versus best

supportive care after adjuvant platinum-based chemotherapy in these patients.

METHODS: IMpower010 was a randomised, multicentre, open-label, phase 3 study

done at 227 sites in 22 countries and regions. Eligible patients were 18 years

or older with completely resected stage IB (tumours ≥4 cm) to IIIA NSCLC per the

Union Internationale Contre le Cancer and American Joint Committee on Cancer

staging system (7th edition). Patients were randomly assigned (1:1) by a

permuted-block method (block size of four) to receive adjuvant atezolizumab

(1200 mg every 21 days; for 16 cycles or 1 year) or best supportive care

(observation and regular scans for disease recurrence) after adjuvant

platinum-based chemotherapy (one to four cycles). The primary endpoint,

investigator-assessed disease-free survival, was tested hierarchically first in

the stage II-IIIA population subgroup whose tumours expressed PD-L1 on 1% or

more of tumour cells (SP263), then all patients in the stage II-IIIA population,

and finally the intention-to-treat (ITT) population (stage IB-IIIA). Safety was

evaluated in all patients who were randomly assigned and received atezolizumab

or best supportive care. IMpower010 is registered with ClinicalTrials.gov,

NCT02486718 (active, not recruiting).

FINDINGS: Between Oct 7, 2015, and Sept 19, 2018, 1280 patients were enrolled

after complete resection. 1269 received adjuvant chemotherapy, of whom 1005

patients were eligible for randomisation to atezolizumab (n=507) or best

supportive care (n=498); 495 in each group received treatment. After a median

follow-up of 32·2 months (IQR 27·4-38·3) in the stage II-IIIA population,

atezolizumab treatment improved disease-free survival compared with best

supportive care in patients in the stage II-IIIA population whose tumours

expressed PD-L1 on 1% or more of tumour cells (HR 0·66; 95% CI 0·50-0·88;

p=0·0039) and in all patients in the stage II-IIIA population (0·79; 0·64-0·96; p=0·020). In the ITT population, HR for disease-free survival was 0·81

(0·67-0·99; p=0·040). Atezolizumab-related grade 3 and 4 adverse events occurred in 53 (11%) of 495 patients and grade 5 events in four patients (1%).

INTERPRETATION: IMpower010 showed a disease-free survival benefit with

atezolizumab versus best supportive care after adjuvant chemotherapy in patients

with resected stage II-IIIA NSCLC, with pronounced benefit in the subgroup whose

tumours expressed PD-L1 on 1% or more of tumour cells, and no new safety

signals. Atezolizumab after adjuvant chemotherapy offers a promising treatment

option for patients with resected early-stage NSCLC.

FUNDING: F Hoffmann-La Roche and Genentech.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S0140-6736(21)02098-5

PMID: 34555333

 

2. N Engl J Med. 2021 Sep 18. doi: 10.1056/NEJMoa2112431. Online ahead of print.

 

Trastuzumab Deruxtecan in HER2-Mutant Non-Small-Cell Lung Cancer.

 

Li BT(1), Smit EF(1), Goto Y(1), Nakagawa K(1), Udagawa H(1), Mazières J(1),

Nagasaka M(1), Bazhenova L(1), Saltos AN(1), Felip E(1), Pacheco JM(1), Pérol

M(1), Paz-Ares L(1), Saxena K(1), Shiga R(1), Cheng Y(1), Acharyya S(1), Vitazka

P(1), Shahidi J(1), Planchard D(1), Jänne PA(1); DESTINY-Lung01 Trial

Investigators.

 

Author information:

(1)From Memorial Sloan Kettering Cancer Center and Weill Cornell Medicine, New

York (B.T.L.); the Netherlands Cancer Institute, Amsterdam (E.F.S); the National

Cancer Center Hospital, Tokyo (Y.G.), Kindai University Hospital, Osaka (K.N.),

and the National Cancer Center East, Kashiwa (H.U.) - all in Japan; Centre

Hospitalier Universitaire, Toulouse (J.M.), Centre Léon Bérard, Lyon (M.P.), and

the Department of Medical Oncology, Thoracic Group, Gustave Roussy, Villejuif

(D.P.) - all in France; Karmanos Cancer Institute, Detroit (M.N.); the

University of California, San Diego, Moores Cancer Center, San Diego (L.B.);

Moffitt Cancer Center, Tampa, FL (A.N.S.); Vall d'Hebron University Hospital and

Vall d'Hebron Institute of Oncology, Barcelona (E.F.); University of Colorado,

Aurora (J.M.P.); Hospital Universitario 12 de Octubre, H12O-Centro Nacional de

Investigaciones Oncológicas (CNIO) Lung Cancer Clinical Research Unit, and

Complutense University, Madrid (L.P.-A.); Daiichi Sankyo, Basking Ridge, NJ

(K.S., R.S., Y.C., S.A., P.V., J.S.); and Dana-Farber Cancer Institute and the

Belfer Center for Applied Cancer Science, Boston (P.A.J.).

 

BACKGROUND: Human epidermal growth factor receptor 2 (HER2)-targeted therapies

have not been approved for patients with non-small-cell lung cancer (NSCLC). The

efficacy and safety of trastuzumab deruxtecan (formerly DS-8201), a HER2

antibody-drug conjugate, in patients with HER2-mutant NSCLC have not been

investigated extensively.

METHODS: We conducted a multicenter, international, phase 2 study in which

trastuzumab deruxtecan (6.4 mg per kilogram of body weight) was administered to

patients who had metastatic HER2-mutant NSCLC that was refractory to standard

treatment. The primary outcome was objective response as assessed by independent

central review. Secondary outcomes included the duration of response,

progression-free survival, overall survival, and safety. Biomarkers of HER2

alterations were assessed.

RESULTS: A total of 91 patients were enrolled. The median duration of follow-up

was 13.1 months (range, 0.7 to 29.1). Centrally confirmed objective response

occurred in 55% of the patients (95% confidence interval [CI], 44 to 65). The

median duration of response was 9.3 months (95% CI, 5.7 to 14.7). Median

progression-free survival was 8.2 months (95% CI, 6.0 to 11.9), and median

overall survival was 17.8 months (95% CI, 13.8 to 22.1). The safety profile was

generally consistent with those from previous studies; grade 3 or higher

drug-related adverse events occurred in 46% of patients, the most common event

being neutropenia (in 19%). Adjudicated drug-related interstitial lung disease

occurred in 26% of patients and resulted in death in 2 patients. Responses were

observed across different HER2 mutation subtypes, as well as in patients with no

detectable HER2 expression or HER2 amplification.

CONCLUSIONS: Trastuzumab deruxtecan showed durable anticancer activity in

patients with previously treated HER2-mutant NSCLC. The safety profile included

interstitial lung disease that was fatal in two cases. Observed toxic effects

were generally consistent with those in previously reported studies. (Funded by

Daiichi Sankyo and AstraZeneca; DESTINY-Lung01 ClinicalTrials.gov number,

NCT03505710.).

 

Copyright © 2021 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2112431

PMID: 34534430

 

3. Nature. 2021 Sep;597(7878):732-737. doi: 10.1038/s41586-021-03898-1. Epub 2021

Sep 15.

 

Structure-based classification predicts drug response in EGFR-mutant NSCLC.

 

Robichaux JP(1), Le X(1), Vijayan RSK(2), Hicks JK(3), Heeke S(1), Elamin YY(1),

Lin HY(4), Udagawa H(1), Skoulidis F(1), Tran H(1), Varghese S(1), He J(1),

Zhang F(1), Nilsson MB(1), Hu L(1), Poteete A(1), Rinsurongkawong W(5), Zhang

X(6), Ren C(6), Liu X(1)(7), Hong L(1), Zhang J(1), Diao L(8), Madison R(9),

Schrock AB(9), Saam J(10), Raymond V(10), Fang B(6), Wang J(8), Ha MJ(4), Cross

JB(2), Gray JE(11), Heymach JV(12).

 

Author information:

(1)Department of Thoracic/Head and Neck Medical Oncology, MD Anderson Cancer

Center, Houston, TX, USA.

(2)Institute for Applied Cancer Science, MD Anderson Cancer Center, Houston, TX,

USA.

(3)Department of Individualized Cancer Management, Moffitt Cancer Center, Tampa,

FL, USA.

 

Epidermal growth factor receptor (EGFR) mutations typically occur in exons 18-21

and are established driver mutations in non-small cell lung cancer (NSCLC)1-3.

Targeted therapies are approved for patients with 'classical' mutations and a

small number of other mutations4-6. However, effective therapies have not been

identified for additional EGFR mutations. Furthermore, the frequency and effects

of atypical EGFR mutations on drug sensitivity are unknown1,3,7-10. Here we

characterize the mutational landscape in 16,715 patients with EGFR-mutant NSCLC,

and establish the structure-function relationship of EGFR mutations on drug

sensitivity. We found that EGFR mutations can be separated into four distinct

subgroups on the basis of sensitivity and structural changes that

retrospectively predict patient outcomes following treatment with EGFR

inhibitors better than traditional exon-based groups. Together, these data

delineate a structure-based approach for defining functional groups of EGFR

mutations that can effectively guide treatment and clinical trial choices for

patients with EGFR-mutant NSCLC and suggest that a structure-function-based

approach may improve the prediction of drug sensitivity to targeted therapies in

oncogenes with diverse mutations.

 

© 2021. The Author(s).

 

DOI: 10.1038/s41586-021-03898-1

PMCID: PMC8481125

PMID: 34526717

 

4. Annu Rev Pharmacol Toxicol. 2021 Sep 30. doi:

10.1146/annurev-pharmtox-041921-074800. Online ahead of print.

 

Emerging Therapeutics, Technologies, and Drug Development Strategies to Address

Patient Nonadherence and Improve Tuberculosis Treatment.

 

Garcia-Cremades M(1), Solans BP(1), Strydom N(1), Vrijens B(2)(3), Pillai

GC(4)(5), Shaffer C(1), Thomas B(6), Savic RM(1).

 

Author information:

(1)Department of Bioengineering and Therapeutic Sciences, University of

California San Francisco, San Francisco, California 94158, USA; email:

rada.savic@ucsf.edu.

(2)AARDEX Group, B-4102 Liège Science Park, Belgium.

(3)Department of Public Health, University of Liège, B-4000 Liège, Belgium.

(4)Division of Clinical Pharmacology, University of Cape Town, Observatory 7925,

South Africa.

(5)CP+ Associates GmbH, Basel 4102, Switzerland.

(6)The Arcady Group, Richmond, Virginia 23226, USA.

 

Imperfect medication adherence remains the biggest predictor of treatment

failure for patients with tuberculosis. Missed doses during treatment lead to

relapse, tuberculosis resistance, and further spread of disease. Understanding

individual patient phenotypes, population pharmacokinetics, resistance

development, drug distribution to tuberculosis lesions, and pharmacodynamics at

the site of infection is necessary to fully measure the impact of adherence on

patient outcomes. To decrease the impact of expected variability in drug intake

on tuberculosis outcomes, an improvement in patient adherence and new forgiving

regimens that protect against missed doses are needed. In this review, we

summarize emerging technologies to improve medication adherence in clinical

practice and provide suggestions on how digital adherence technologies can be

incorporated in clinical trials and practice and the drug development pipeline

that will lead to more forgiving regimens and benefit patients suffering from

tuberculosis. Expected final online publication date for the Annual Review of

Pharmacology and Toxicology, Volume 62 is January 2022. Please see

http://www.annualreviews.org/page/journal/pubdates for revised estimates.

 

DOI: 10.1146/annurev-pharmtox-041921-074800

PMID: 34591605

 

5. Lancet Oncol. 2021 Oct;22(10):1448-1457. doi: 10.1016/S1470-2045(21)00401-0.

Epub 2021 Sep 13.

 

Stereotactic ablative radiotherapy for operable stage I non-small-cell lung

cancer (revised STARS): long-term results of a single-arm, prospective trial

with prespecified comparison to surgery.

 

Chang JY(1), Mehran RJ(2), Feng L(3), Verma V(4), Liao Z(4), Welsh JW(4), Lin

SH(4), O'Reilly MS(4), Jeter MD(4), Balter PA(5), McRae SE(6), Berry D(3),

Heymach JV(7), Roth JA(2); STARS Lung Cancer Trials Group.

 

Collaborators: Antonoff M, Hofstetter W, Rajaram R, Rice D, Sepesi B, Swisher S,

Vaporciyan A, Walsh G, DeGraaf C, Correa A, Chen A, Gandhi S, Komaki R, Lee P,

Nguyen QN, Ning M, Gao S, Pollard-Larkin J, Nitsch P, Sadagopan R, Wang X.

 

Author information:

(1)Department of Radiation Oncology, The University of Texas MD Anderson Cancer

Center, Houston, TX, USA. Electronic address: jychang@mdanderson.org.

(2)Department of Thoracic and Cardiovascular Surgery, The University of Texas MD

Anderson Cancer Center, Houston, TX, USA.

(3)Department of Biostatistics, The University of Texas MD Anderson Cancer

Center, Houston, TX, USA.

 

BACKGROUND: A previous pooled analysis of the STARS and ROSEL trials showed

higher survival after stereotactic ablative radiotherapy (SABR) than with

surgery for operable early-stage non-small-cell lung cancer (NSCLC), but that

analysis had notable limitations. This study reports long-term results of the

revised STARS trial, in which the SABR group was re-accrued with a larger sample

size, along with a protocol-specified propensity-matched comparison with a

prospectively registered, contemporary institutional cohort of patients who

underwent video-assisted thoracoscopic surgical lobectomy with mediastinal lymph

node dissection (VATS L-MLND).

METHODS: This single-arm prospective trial was done at the University of Texas

MD Anderson Cancer Center (Houston, TX, USA) and enrolled patients aged 18 years

or older with a Zubrod performance status of 0-2, newly diagnosed and

histologically confirmed NSCLC with N0M0 disease (squamous cell, adenocarcinoma,

large cell, or NSCLC not otherwise specified), and a tumour diameter of 3 cm or

less. This trial did not include patients from the previous pooled analysis.

SABR dosing was 54 Gy in three fractions (for peripheral lesions) or 50 Gy in

four fractions (for central tumours; simultaneous integrated boost to gross

tumour totalling 60 Gy). The primary endpoint was the 3-year overall survival.

For the propensity-matching analysis, we used a surgical cohort from the MD

Anderson Department of Thoracic and Cardiovascular Surgery's prospectively

registered, institutional review board-approved database of all patients with

clinical stage I NSCLC who underwent VATS L-MLND during the period of enrolment

in this trial. Non-inferiority could be claimed if the 3-year overall survival

rate after SABR was lower than that after VATS L-MLND by 12% or less and the

upper bound of the 95% CI of the hazard ratio (HR) was less than 1·965.

Propensity matching consisted of determining a propensity score using a

multivariable logistic regression model including several covariates (age,

tumour size, histology, performance status, and the interaction of age and sex);

based on the propensity scores, one patient in the SABR group was randomly

matched with one patient in the VATS L-MLND group using a 5:1 digit greedy match

algorithm. This study is registered with ClinicalTrials.gov, NCT02357992.

FINDINGS: Between Sept 1, 2015, and Jan 31, 2017, 80 patients were enrolled and

included in efficacy and safety analyses. Median follow-up time was 5·1 years

(IQR 3·9-5·8). Overall survival was 91% (95% CI 85-98) at 3 years and 87%

(79-95) at 5 years. SABR was tolerated well, with no grade 4-5 toxicity and one

(1%) case each of grade 3 dyspnoea, grade 2 pneumonitis, and grade 2 lung

fibrosis. No serious adverse events were recorded. Overall survival in the

propensity-matched VATS L-MLND cohort was 91% (95% CI 85-98) at 3 years and 84%

(76-93) at 5 years. Non-inferiority was claimed since the 3-year overall

survival after SABR was not lower than that observed in the VATS L-MLND group.

There was no significant difference in overall survival between the two patient

cohorts (hazard ratio 0·86 [95% CI 0·45-1·65], p=0·65) from a multivariable

analysis.

INTERPRETATION: Long-term survival after SABR is non-inferior to VATS L-MLND for

operable stage IA NSCLC. SABR remains promising for such cases but

multidisciplinary management is strongly recommended.

FUNDING: Varian Medical Systems and US National Cancer Institute (National

Institutes of Health).

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1470-2045(21)00401-0

PMCID: PMC8521627

PMID: 34529930 [Indexed for MEDLINE]

 

6. Cancer Cell. 2021 Sep 13;39(9):1279-1291.e3. doi: 10.1016/j.ccell.2021.07.005.

Epub 2021 Aug 12.

 

Bevacizumab plus erlotinib in Chinese patients with untreated, EGFR-mutated,

advanced NSCLC (ARTEMIS-CTONG1509): A multicenter phase 3 study.

 

Zhou Q(1), Xu CR(1), Cheng Y(2), Liu YP(3), Chen GY(4), Cui JW(5), Yang N(6),

Song Y(7), Li XL(8), Lu S(9), Zhou JY(10), Ma ZY(11), Yu SY(12), Huang C(13),

Shu YQ(14), Wang Z(1), Yang JJ(1), Tu HY(1), Zhong WZ(1), Wu YL(15).

 

Author information:

(1)Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer,

Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and

Guangdong Academy of Medical Sciences, Guangzhou, China.

(2)Department of Thoracic Oncology, Jilin Provincial Tumor Hospital, Changchun,

China.

(3)Department of Medical Oncology, First Hospital of China Medical University,

Shenyang, China.

 

Dual inhibition of epidermal growth factor receptor (EGFR) and vascular

endothelial growth factor (VEGF) pathways may delay therapeutic resistance in

advanced non-small cell lung cancer (NSCLC). This phase 3 study investigated the

efficacy and safety of an erlotinib plus bevacizumab regimen in untreated

patients with advanced NSCLC. In total, 311 patients received bevacizumab plus

erlotinib (n = 157) or erlotinib only (n = 154). Progression-free survival (PFS)

was 17.9 months (95% confidence interval [CI], 15.2-19.9) for bevacizumab plus

erlotinib and 11.2 months (95% CI, 9.7-13.8) for erlotinib only (hazard ratio

[HR] = 0.55; 95% CI, 0.41-0.73; p < 0.001). A brain metastases subgroup treated

with bevacizumab plus erlotinib also showed improved PFS (HR = 0.48; 95% CI,

0.27-0.84; p = 0.008). Grade ≥3 treatment-related adverse events occurred in 86 (54.8%) and 40 (26.1%) patients, respectively. Bevacizumab plus erlotinib

significantly improved PFS in patients with untreated metastatic EGFR-mutated

NSCLC, including those with brain metastases at baseline.

 

Copyright © 2021 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2021.07.005

PMID: 34388377

 

7. Nat Med. 2021 Sep;27(9):1646-1654. doi: 10.1038/s41591-021-01388-5. Epub 2021

Jun 28.

 

Inherited PD-1 deficiency underlies tuberculosis and autoimmunity in a child.

 

Ogishi M(1)(2), Yang R(3), Aytekin C(4), Langlais D(5), Bourgey M(6), Khan T(7),

Ali FA(7), Rahman M(7), Delmonte OM(8), Chrabieh M(9)(10), Zhang P(3), Gruber

C(11)(12)(13)(14), Pelham SJ(3), Spaan AN(3), Rosain J(9)(10), Lei WT(3),

Drutman S(3), Hellmann MD(15)(16), Callahan MK(15)(16), Adamow M(17)(18), Wong

P(17), Wolchok JD(15)(16)(18)(19), Rao G(20), Ma CS(20)(21), Nakajima Y(22),

Yaguchi T(22), Chamoto K(22), Williams SC(23)(24), Emile JF(25), Rozenberg

F(26), Glickman MS(27), Rapaport F(3), Kerner G(9)(10), Allington G(28)(29)(30),

Tezcan I(31), Cagdas D(31), Hosnut FO(32), Dogu F(33), Ikinciogullari A(33), Rao

VK(34), Kainulainen L(35), Béziat V(3)(9)(10), Bustamante J(3)(9)(10)(36),

Vilarinho S(#)(28)(29)(30), Lifton RP(#)(28)(37), Boisson B(#)(3)(9)(10), Abel

L(#)(3)(9)(10), Bogunovic D(#)(11)(12)(13)(14), Marr N(#)(7)(38), Notarangelo

LD(#)(8), Tangye SG(#)(20)(21), Honjo T(#)(22), Gros P(#)(39)(40),

Boisson-Dupuis S(#)(41)(42)(43), Casanova JL(#)(44)(45)(46)(47).

 

Author information:

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

Branch, Rockefeller University, New York, NY, USA. mogishi@rockefeller.edu.

(2)The David Rockefeller Graduate Program, Rockefeller University, New York, NY,

USA. mogishi@rockefeller.edu.

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

Branch, Rockefeller University, New York, NY, USA.

 

The pathophysiology of adverse events following programmed cell death protein 1

(PD-1) blockade, including tuberculosis (TB) and autoimmunity, remains poorly

characterized. We studied a patient with inherited PD-1 deficiency and TB who

died of pulmonary autoimmunity. The patient's leukocytes did not express PD-1 or

respond to PD-1-mediated suppression. The patient's lymphocytes produced only

small amounts of interferon (IFN)-γ upon mycobacterial stimuli, similarly to

patients with inborn errors of IFN-γ production who are vulnerable to TB. This

phenotype resulted from a combined depletion of Vδ2+ γδ T, mucosal-associated

invariant T and CD56bright natural killer lymphocytes and dysfunction of other T

lymphocyte subsets. Moreover, the patient displayed hepatosplenomegaly and an

expansion of total, activated and RORγT+ CD4-CD8- double-negative αβ T cells,

similar to patients with STAT3 gain-of-function mutations who display

lymphoproliferative autoimmunity. This phenotype resulted from excessive amounts

of STAT3-activating cytokines interleukin (IL)-6 and IL-23 produced by activated

T lymphocytes and monocytes, and the STAT3-dependent expression of RORγT by

activated T lymphocytes. Our work highlights the indispensable role of human

PD-1 in governing both antimycobacterial immunity and self-tolerance, while

identifying potentially actionable molecular targets for the diagnostic and

therapeutic management of TB and autoimmunity in patients on PD-1 blockade.

 

© 2021. The Author(s), under exclusive licence to Springer Nature America, Inc.

 

DOI: 10.1038/s41591-021-01388-5

PMCID: PMC8446316

PMID: 34183838 [Indexed for MEDLINE]

 

8. J Clin Invest. 2021 Sep 1;131(17):e137407. doi: 10.1172/JCI137407.

 

TREM-2 promotes Th1 responses by interacting with the CD3ζ-ZAP70 complex

following Mycobacterium tuberculosis infection.

 

Wu Y(1)(2)(3)(4), Wu M(1), Ming S(1)(2)(5), Zhan X(1), Hu S(1), Li X(1)(2), Yin

H(1)(2), Cao C(1)(2), Liu J(1)(2), Li J(1)(2), Wu Z(6), Zhou J(6), Liu L(5),

Gong S(4), He D(7), Huang X(1)(2)(3)(4)(5).

 

Author information:

(1)Center for Infection and Immunity, The Fifth Affiliated Hospital of Sun

Yat-sen University, Zhuhai, Guangdong Province, China.

(2)Guangdong Provincial Engineering Research Center of Molecular Imaging,

Guangdong Provincial Key Laboratory of Biomedical Imaging, and Department of

Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University,

Zhuhai, China.

(3)Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai,

Guangdong Province, China.

(4)Department of Gastroenterology, Guangzhou Women and Children's Medical

Center, Guangzhou Institute of Pediatrics, Guangzhou Medical University,

Guangzhou, Guangdong Province, China.

(5)National Clinical Research Center for Infectious Disease, Shenzhen Third

People's Hospital, The Second Affiliated Hospital of the Southern University of

Science and Technology, Shenzhen, Guangdong Province, China.

(6)The Fourth People's Hospital of Foshan, Foshan, China.

(7)Shantou No. 3 People's Hospital, Shantou, Guangdong Province, China.

 

Triggering receptor expressed on myeloid cells 2 (TREM-2) is a modulator of

pattern recognition receptors on innate immune cells that regulates the

inflammatory response. However, the role of TREM-2 in in vivo models of

infection and inflammation remains controversial. Here, we demonstrated that

TREM-2 expression on CD4+ T cells was induced by Mycobacterium tuberculosis

infection in both humans and mice and positively associated with T cell

activation and an effector memory phenotype. Activation of TREM-2 in CD4+ T

cells was dependent on interaction with the putative TREM-2 ligand expressed on

DCs. Unlike the observation in myeloid cells that TREM-2 signals through DAP12,

in CD4+ T cells, TREM-2 interacted with the CD3ζ-ZAP70 complex as well as with

the IFN-γ receptor, leading to STAT1/-4 activation and T-bet transcription. In

addition, an infection model using reconstituted Rag2-/- mice (with TREM-2-KO

vs. WT cells or TREM-2+ vs. TREM-2-CD4+ T cells) or CD4+ T cell-specific TREM-2

conditional KO mice demonstrated that TREM-2 promoted a Th1-mediated host

defense against M. tuberculosis infection. Taken together, these findings reveal

a critical role of TREM-2 in evoking proinflammatory Th1 responses that may

provide potential therapeutic targets for infectious and inflammatory diseases.

 

DOI: 10.1172/JCI137407

PMCID: PMC8409584

PMID: 34623322 [Indexed for MEDLINE]

 

9. Lancet Infect Dis. 2021 Sep 23:S1473-3099(21)00449-7. doi:

10.1016/S1473-3099(21)00449-7. Online ahead of print.

 

Global, regional, and national sex differences in the global burden of

tuberculosis by HIV status, 1990-2019: results from the Global Burden of Disease

Study 2019.

 

GBD 2019 Tuberculosis Collaborators.

 

BACKGROUND: Tuberculosis is a major contributor to the global burden of disease,

causing more than a million deaths annually. Given an emphasis on equity in

access to diagnosis and treatment of tuberculosis in global health targets,

evaluations of differences in tuberculosis burden by sex are crucial. We aimed

to assess the levels and trends of the global burden of tuberculosis, with an

emphasis on investigating differences in sex by HIV status for 204 countries and

territories from 1990 to 2019.

METHODS: We used a Bayesian hierarchical Cause of Death Ensemble model (CODEm)

platform to analyse 21 505 site-years of vital registration data, 705 site-years

of verbal autopsy data, 825 site-years of sample-based vital registration data,

and 680 site-years of mortality surveillance data to estimate mortality due to

tuberculosis among HIV-negative individuals. We used a population attributable

fraction approach to estimate mortality related to HIV and tuberculosis

coinfection. A compartmental meta-regression tool (DisMod-MR 2.1) was then used

to synthesise all available data sources, including prevalence surveys, annual

case notifications, population-based tuberculin surveys, and tuberculosis

cause-specific mortality, to produce estimates of incidence, prevalence, and

mortality that were internally consistent. We further estimated the fraction of

tuberculosis mortality that is attributable to independent effects of risk

factors, including smoking, alcohol use, and diabetes, for HIV-negative

individuals. For individuals with HIV and tuberculosis coinfection, we assessed

mortality attributable to HIV risk factors including unsafe sex, intimate

partner violence (only estimated among females), and injection drug use. We

present 95% uncertainty intervals for all estimates.

FINDINGS: Globally, in 2019, among HIV-negative individuals, there were 1·18

million (95% uncertainty interval 1·08-1·29) deaths due to tuberculosis and 8·50 million (7·45-9·73) incident cases of tuberculosis. Among HIV-positive

individuals, there were 217 000 (153 000-279 000) deaths due to tuberculosis and

1·15 million (1·01-1·32) incident cases in 2019. More deaths and incident cases

occurred in males than in females among HIV-negative individuals globally in

2019, with 342 000 (234 000-425 000) more deaths and 1·01 million (0·82-1·23)

more incident cases in males than in females. Among HIV-positive individuals,

6250 (1820-11 400) more deaths and 81 100 (63 300-100 000) more incident cases

occurred among females than among males in 2019. Age-standardised mortality

rates among HIV-negative males were more than two times greater in 105 countries

and age-standardised incidence rates were more than 1·5 times greater in 74

countries than among HIV-negative females in 2019. The fraction of global

tuberculosis deaths among HIV-negative individuals attributable to alcohol use,

smoking, and diabetes was 4·27 (3·69-5·02), 6·17 (5·48-7·02), and 1·17

(1·07-1·28) times higher, respectively, among males than among females in 2019.

Among individuals with HIV and tuberculosis coinfection, the fraction of

mortality attributable to injection drug use was 2·23 (2·03-2·44) times greater

among males than females, whereas the fraction due to unsafe sex was 1·06

(1·05-1·08) times greater among females than males.

INTERPRETATION: As countries refine national tuberculosis programmes and

strategies to end the tuberculosis epidemic, the excess burden experienced by

males is important. Interventions are needed to actively communicate, especially

to men, the importance of early diagnosis and treatment. These interventions

should occur in parallel with efforts to minimise excess HIV burden among women

in the highest HIV burden countries that are contributing to excess HIV and

tuberculosis coinfection burden for females. Placing a focus on tuberculosis

burden among HIV-negative males and HIV and tuberculosis coinfection among

females might help to diminish the overall burden of tuberculosis. This strategy

will be crucial in reaching both equity and burden targets outlined by global

health milestones.

FUNDING: Bill & Melinda Gates Foundation.

 

Copyright © 2021 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(21)00449-7

PMID: 34563275

 

10. J Clin Oncol. 2021 Sep 22:JCO2101113. doi: 10.1200/JCO.21.01113. Online ahead of print.

 

Poziotinib for Patients With HER2 Exon 20 Mutant Non-Small-Cell Lung Cancer:

Results From a Phase II Trial.

 

Elamin YY(1), Robichaux JP(1), Carter BW(2), Altan M(1), Gibbons DL(1), Fossella

FV(1), Lam VK(1)(3), Patel AB(4), Negrao MV(1), Le X(1), Mott FE(1), Zhang J(1),

Feng L(5), Blumenschein G Jr(1), Tsao AS(1), Heymach JV(1).

 

Author information:

(1)Department of Thoracic/Head and Neck Medical Oncology, The University of

Texas MD Anderson Cancer Center, Houston, TX.

(2)Department of Thoracic Imaging, The University of Texas MD Anderson Cancer

Center, Houston, TX.

(3)Department of Medicine, Johns Hopkins Sidney Kimmel Cancer Center, Baltimore,

MD.

(4)Department of Dermatology, The University of Texas MD Anderson Cancer Center,

Houston, TX.

(5)Department of Biostatistics, The University of Texas MD Anderson Cancer

Center, Houston, TX.

 

PURPOSE: Targeted therapies against non-small-cell lung cancer (NSCLC) harboring

HER2 mutations remain an unmet need. In this study, we assessed the efficacy and

safety of poziotinib in patients with HER2 exon 20 mutant advanced NSCLC in a

single-arm, open-label, phase II study.

PATIENTS AND METHODS: Patients with advanced HER2 exon 20 mutant NSCLC were

enrolled to receive poziotinib at a dose of 16 mg/d for 28-day cycles. The

primary end point was objective response rate per RECIST version 1.1.

Confirmatory scans were performed at least 28 days from initial radiologic

response.

RESULTS: Thirty patients received poziotinib treatment. At baseline, 90% of

patients received prior platinum-based chemotherapy and 53% had two lines or

more prior systemic therapies. As of data cutoff on March 1, 2021, the confirmed

objective response rate was 27% (95% CI, 12 to 46). Responses were observed

across HER2 exon 20 mutation subtypes. The median duration of response was 5.0

months (95% CI, 4.0 to not estimable). The median progression-free survival was

5.5 months (95% CI, 4.0 to 7.0). The median overall survival was 15 months (95%

CI, 9.0 to not estimable). The most common grade 3 treatment-related adverse

events were skin rash (47%) and diarrhea (20%). There was one possible

treatment-related death because of pneumonitis.

CONCLUSION: Poziotinib showed promising antitumor activity in patients with HER2

exon 20 mutant NSCLC including patients who had previously received

platinum-based chemotherapy.

 

DOI: 10.1200/JCO.21.01113

PMID: 34550757

 

11. Nat Commun. 2021 Sep 20;12(1):5548. doi: 10.1038/s41467-021-25867-y.

 

Isonicotinylation is a histone mark induced by the anti-tuberculosis first-line

drug isoniazid.

 

Jiang Y(1), Li Y(1), Liu C(1), Zhang L(1), Lv D(1), Weng Y(2), Cheng Z(2), Chen

X(3), Zhan J(1), Zhang H(4).

 

Author information:

(1)Program for Cancer and Cell Biology, Department of Human Anatomy, Histology

and Embryology, PKU International Cancer Institute, MOE Key Laboratory of

Carcinogenesis and Translational Research and State Key Laboratory of Natural

and Biomimetic Drugs, Peking University Health Science Center, Beijing, PR

China.

(2)Jingjie PTM BioLab Co. Ltd., Hangzhou Economic and Technological Development

Area, Hangzhou, PR China.

(3)Department of Microbiology & Infectious Disease Center, Peking University

Health Science Center, Beijing, PR China.

(4)Program for Cancer and Cell Biology, Department of Human Anatomy, Histology

and Embryology, PKU International Cancer Institute, MOE Key Laboratory of

Carcinogenesis and Translational Research and State Key Laboratory of Natural

and Biomimetic Drugs, Peking University Health Science Center, Beijing, PR

China. Hongquan.Zhang@bjmu.edu.cn.

 

Isoniazid (INH) is a first-line anti-tuberculosis drug used for nearly 70 years.

However, the mechanism underlying the side effects of INH has remained elusive.

Here, we report that INH and its metabolites induce a post-translational

modification (PTM) of histones, lysine isonicotinylation (Kinic), also called

4-picolinylation, in cells and mice. INH promotes the biosynthesis of

isonicotinyl-CoA (Inic-CoA), a co-factor of intracellular isonicotinylation.

Mass spectrometry reveals 26 Kinic sites in histones in HepG2 cells.

Acetyltransferases CREB-binding protein (CBP) and P300 catalyse histone Kinic,

while histone deacetylase HDAC3 functions as a deisonicotinylase. Notably, MNase

sensitivity assay and RNA-seq analysis show that histone Kinic relaxes chromatin

structure and promotes gene transcription. INH-mediated histone Kinic

upregulates PIK3R1 gene expression and activates the PI3K/Akt/mTOR signalling

pathway in liver cancer cells, linking INH to tumourigenicity in the liver. We

demonstrate that Kinic is a histone acylation mark with a pyridine ring, which

may have broad biological effects. Therefore, INH-induced isonicotinylation

potentially accounts for the side effects in patients taking INH long-term for

anti-tuberculosis therapy, and this modification may increase the risk of cancer

in humans.

 

© 2021. The Author(s).

 

DOI: 10.1038/s41467-021-25867-y

PMCID: PMC8452692

PMID: 34545082 [Indexed for MEDLINE]

 

12. PLoS Med. 2021 Sep 14;18(9):e1003712. doi: 10.1371/journal.pmed.1003712.

eCollection 2021 Sep.

 

Economic and modeling evidence for tuberculosis preventive therapy among people

living with HIV: A systematic review and meta-analysis.

 

Uppal A(1)(2)(3), Rahman S(1)(2)(3), Campbell JR(1)(2)(3), Oxlade O(3), Menzies

D(1)(2)(3).

 

Author information:

(1)Montréal Chest Institute, Montréal, Québec, Canada.

(2)Respiratory Epidemiology and Clinical Research Unit, Centre for Outcomes

Research and Evaluation, Research Institute of McGill University Health Centre,

Montréal, Québec, Canada.

(3)McGill International Tuberculosis Centre, Montréal, Québec, Canada.

 

Comment in

    Tuberculosis preventive treatment in people living with HIV - is the glass

half empty, or half full?

    The Latent Tuberculosis Cascade-of-Care Among People Living with HIV: A

Systematic Review and Meta-Analysis.

    Tuberculosis Preventive Therapy for People Living with HIV: A Systematic

Review and Network Meta-analysis.

 

BACKGROUND: Human immunodeficiency virus (HIV) is the strongest known risk

factor for tuberculosis (TB) through its impairment of T-cell immunity.

Tuberculosis preventive treatment (TPT) is recommended for people living with

HIV (PLHIV) by the World Health Organization, as it significantly reduces the

risk of developing TB disease. We conducted a systematic review and

meta-analysis of modeling studies to summarize projected costs, risks, benefits,

and impacts of TPT use among PLHIV on TB-related outcomes.

METHODS AND FINDINGS: We searched MEDLINE, Embase, and Web of Science from

inception until December 31, 2020. Two reviewers independently screened titles,

abstracts, and full texts; extracted data; and assessed quality. Extracted data

were summarized using descriptive analysis. We performed quantile regression and

random effects meta-analysis to describe trends in cost, effectiveness, and

cost-effectiveness outcomes across studies and identified key determinants of

these outcomes. Our search identified 6,615 titles; 61 full texts were included

in the final review. Of the 61 included studies, 31 reported both cost and

effectiveness outcomes. A total of 41 were set in low- and middle-income

countries (LMICs), while 12 were set in high-income countries (HICs); 2 were set

in both. Most studies considered isoniazid (INH)-based regimens 6 to 2 months

long (n = 45), or longer than 12 months (n = 11). Model parameters and

assumptions varied widely between studies. Despite this, all studies found that

providing TPT to PLHIV was predicted to be effective at averting TB disease. No

TPT regimen was substantially more effective at averting TB disease than any

other. The cost of providing TPT and subsequent downstream costs (e.g. post-TPT

health systems costs) were estimated to be less than $1,500 (2020 USD) per

person in 85% of studies that reported cost outcomes (n = 36), regardless of

study setting. All cost-effectiveness analyses concluded that providing TPT to

PLHIV was potentially cost-effective compared to not providing TPT. In

quantitative analyses, country income classification, consideration of

antiretroviral therapy (ART) use, and TPT regimen use significantly impacted

cost-effectiveness. Studies evaluating TPT in HICs suggested that TPT may be

more effective at preventing TB disease than studies evaluating TPT in LMICs;

pooled incremental net monetary benefit, given a willingness-to-pay threshold of

country-level per capita gross domestic product (GDP), was $271 in LMICs (95%

confidence interval [CI] -$81 to $622, p = 0.12) and was $2,568 in HICs

(-$32,115 to $37,251, p = 0.52). Similarly, TPT appeared to be more effective at

averting TB disease in HICs; pooled percent reduction in active TB incidence was

20% (13% to 27%, p < 0.001) in LMICs and 37% (-34% to 100%, p = 0.13) in HICs.

Key limitations of this review included the heterogeneity of input parameters

and assumptions from included studies, which limited pooling of effect

estimates, inconsistent reporting of model parameters, which limited sample

sizes of quantitative analyses, and database bias toward English publications.

CONCLUSIONS: The body of literature related to modeling TPT among PLHIV is large

and heterogeneous, making comparisons across studies difficult. Despite this

variability, all studies in all settings concluded that providing TPT to PLHIV

is potentially effective and cost-effective for preventing TB disease.

 

DOI: 10.1371/journal.pmed.1003712

PMCID: PMC8439468

PMID: 34520463

 

13. Am J Respir Crit Care Med. 2021 Sep 14. doi: 10.1164/rccm.202103-0548OC. Online ahead of print.

 

Longitudinal Dynamics of a Blood Transcriptomic Signature of Tuberculosis.

 

Mulenga H(1), Musvosvi M(2), Mendelsohn SC(3), Penn-Nicholson A(4), Kimbung

Mbandi S(4), Gartland AF(5), Tameris M(6), Mabwe S(7), Africa H(8), Bilek N(7),

Kafaar F(4), Khader SA(9), Carstens B(4), Hadley K(4), Hikuam C(4), Erasmus

M(7), Jaxa L(4), Raphela R(4), Nombida O(4), Kaskar M(4), Nicol MP(10), Mbhele

S(10), Van Heerden J(10), Innes C(11), Brumskine W(11), Hiemstra A(12), Malherbe

ST(13), Hassan-Moosa R(14), Walzl G(13), Naidoo K(15), Churchyard G(16),

Hatherill M(17), Scriba TJ(18); CORTIS Study Team.

 

Author information:

(1)University of Cape Town Faculty of Health Sciences, 63726, Pathology,

Observatory, South Africa.

(2)University of Cape Town, Institute of Infectious Diseases and Molecular

Medicine, Observatory, South Africa.

(3)University of Cape Town, 37716, South African Tuberculosis Vaccine

Initiative, Cape Town, South Africa.

 

Objectives We evaluated longitudinal kinetics of an 11-gene blood transcriptomic

tuberculosis (TB) signature, RISK11, and effects of TB preventative therapy

(TPT) and respiratory organisms on RISK11 signature score, in HIV-uninfected and

HIV-infected individuals. Methods RISK11 was measured in a longitudinal study of

RISK11-guided TPT in HIV-uninfected adults, a cross-sectional respiratory

organisms cohort or a longitudinal study in people living with HIV (PLHIV).

HIV-uninfected RISK11+ participants were randomised to TPT or no TPT; RISK11-

participants received no TPT. PLHIV received standard-of-care ART and TPT. In

the cross-sectional respiratory organisms cohort, viruses and bacteria in

nasopharyngeal and oropharyngeal swabs were quantified by RT-qPCR. Measurements

and Main Results RISK11+ status was transient in most of the 128 HIV-negative

participants with longitudinal samples; >70% of RISK11+ participants reverted to

RISK11- by 3 months, irrespective of TPT. By comparison, reversion from a

RISK11-positive state was less common in 645 PLHIV (42.1%). Non-HIV viral and

non-tuberculous bacterial organisms were detected in 7.2% and 38.9% of the 1,000

respiratory organisms cohort participants, respectively, and among those

investigated for TB, 3.8% had prevalent disease. Median RISK11 scores (%) were

higher in participants with viral organisms alone (46.7%), viral and bacterial

organisms (42.8%), or prevalent TB (85.7%), than those with bacterial organisms

other than TB (13.4%), or no organisms (14.2%). RISK11 could not discriminate

between prevalent TB and viral organisms. Conclusions Positive RISK11 signature

status is often transient, possibly due to intercurrent viral infection,

highlighting potentially important challenges for implementation of these

biomarkers as new tools for TB control. This article is open access and

distributed under the terms of the Creative Commons Attribution 4.0

International License (https://creativecommons.org/licenses/by/4.0/).

 

DOI: 10.1164/rccm.202103-0548OC

PMID: 34520313

 

14. Lancet Infect Dis. 2021 Sep 7:S1473-3099(21)00403-5. doi:

10.1016/S1473-3099(21)00403-5. Online ahead of print.

 

100 years of Mycobacterium bovis bacille Calmette-Guérin.

 

Lange C(1), Aaby P(2), Behr MA(3), Donald PR(4), Kaufmann SHE(5), Netea MG(6),

Mandalakas AM(7).

 

Author information:

(1)Division of Clinical Infectious Diseases, Medical Clinic, Research Center

Borstel, Borstel, Germany; German Center for Infection Research (DZIF)

Tuberculosis Unit, Borstel, Germany; Respiratory Medicine and International

Health, University of Lübeck, Lübeck, Germany; Global TB Program, Baylor College

of Medicine and Texas Children's Hospital, Houston, TX, USA. Electronic address:

clange@fz-borstel.de.

(2)Bandim Health Project, Indepth Network, Bissau, Guinea-Bissau; Bandim Health

Project, Southern Danish University, Copenhagen, Denmark.

(3)McGill International TB Centre and Department of Medicine, McGill University,

Montreal, QC, Canada.

 

Mycobacterium bovis bacille Calmette-Guérin (BCG), an experimental vaccine

designed to protect cattle from bovine tuberculosis, was administered for the

first time to a newborn baby in Paris in 1921. Over the past century, BCG has

saved tens of millions of lives and has been given to more humans than any other

vaccine. It remains the sole tuberculosis vaccine licensed for use in humans.

BCG provides long-lasting strong protection against miliary and meningeal

tuberculosis in children, but it is less effective for the prevention of

pulmonary tuberculosis, especially in adults. Evidence mainly from the past two

decades suggests that BCG has non-specific benefits against non-tuberculous

infections in newborn babies and in older adults, and offers immunotherapeutic

benefit in certain malignancies such as non-muscle invasive bladder cancer.

However, as a live attenuated vaccine, BCG can cause localised or disseminated

infections in immunocompromised hosts, which can also occur following

intravesical installation of BCG for the treatment of bladder cancer. The legacy

of BCG includes fundamental discoveries about tuberculosis-specific and

non-specific immunity and the demonstration that tuberculosis is a

vaccine-preventable disease, providing a foundation for new vaccines to hasten

tuberculosis elimination.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(21)00403-5

PMID: 34506734

 

15. Lancet Infect Dis. 2021 Sep 6:S1473-3099(21)00145-6. doi:

10.1016/S1473-3099(21)00145-6. Online ahead of print.

 

Comparison of three tests for latent tuberculosis infection in high-risk people

in the USA: an observational cohort study.

 

Ho CS(1), Feng PI(2), Narita M(3), Stout JE(4), Chen M(2), Pascopella L(5),

Garfein R(6), Reves R(7), Katz DJ(2); Tuberculosis Epidemiologic Studies

Consortium.

 

Collaborators: Flood J, Pascopella L, Higashi J, Moser K, Moore M, Garfein R,

Benson C, Belknap R, Reves R, Stout JE, Ahmed A, Sterling T, Pettit A, Blumberg

HM, Oladele A, Lauzardo M, Seraphin MN, Brostrom R, Khurana R, Cronin W, Dorman

S, Narita M, Horne D, Miller T.

 

Author information:

(1)Division of Tuberculosis Elimination, US Centers for Disease Control and

Prevention, Atlanta, GA, USA. Electronic address: gtb9@cdc.gov.

(2)Division of Tuberculosis Elimination, US Centers for Disease Control and

Prevention, Atlanta, GA, USA.

(3)TB Control Program, Public Health-Seattle and King County, Seattle, WA, USA.

(4)Division of Infectious Diseases and International Health, Department of

Medicine, Duke University Medical Center, Durham, NC, USA.

(5)Tuberculosis Control Branch, Division of Communicable Disease Control, Center

for Infectious Diseases, California Department of Public Health, Richmond, CA,

USA.

(6)Division of Global Public Health, School of Medicine, University of

California, San Diego, CA, USA.

(7)Denver Health and Hospital Authority, Denver, CO, USA.

 

BACKGROUND: Treatment of latent tuberculosis infection is an important strategy

to prevent tuberculosis disease. In the USA, three tests are used to identify

latent tuberculosis infection: the tuberculin skin test (TST) and two IFN-γ

release assays (T-SPOT.TB and QuantiFERON). To our knowledge, few large studies

have compared all three tests among people at high risk of latent tuberculosis

infection or progression to tuberculosis disease. We aimed to assess test

agreement between IFN-γ release assays and TST to provide guidance on their use

in important risk groups.

METHODS: In this observational cohort study, we enrolled participants at high

risk of latent tuberculosis infection or progression to tuberculosis disease at

ten US sites with 18 affiliated clinics, including close contacts of infectious

tuberculosis cases, people born in countries whose populations in the USA have

high (≥100 cases per 100 000 people) or moderate (10-99 cases per 100 000

people) tuberculosis incidence, and people with HIV. Participants were

interviewed about demographics and medical risk factors, and all three tests

were administered to each participant. The primary endpoints for this study were

the proportions of positive test results by test type stratified by risk group

and test concordance by risk group for participants with valid results for all

three test types. The study is registered at ClinicalTrials.gov, NCT01622140.

FINDINGS: Between July 12, 2012, and May 5, 2017, 26 292 people were approached

and 22 131 (84·2%) were enrolled in the study. Data from 21 846 (98·7%)

participants were available for analysis, including 3790 (17·3%) born in the USA

and 18 023 (82·5%) born outside the USA. Among non-US-born participants overall,

the RR comparing the proportions of TST-positive results (7476 [43·2%] of 17 306 participants) to QuantiFERON-positive results (4732 [26·5%] of 17 882

participants) was 1·6 (95% CI 1·6-1·7). The risk ratio (RR) for the comparison

with the proportion of T-SPOT.TB-positive results (3693 [21·6%] of 17 118

participants) was 2·0 (95% CI 1·9-2·1). US-born participants had less variation

in the proportions of positive results across all tests. The RRs for the

proportion of TST-positive results (391 [10·9%] of 3575 participants) compared

with the proportion of QuantiFERON-positive results (445 [12·0%] of 3693

participants) and T-SPOT.TB-positive results (295 [8·1%] of 3638 participants)

were 0·9 (95% CI 0·8-1·0) and 1·3 (1·2-1·6), respectively. 20 149 (91·0%) of 21 846 participants had results for all three tests, including 16 712 (76%)

non-US-born participants. Discordance between TST and IFN-γ release assay

results varied by age among non-US-born participants and was greatest among the

848 non-US-born children younger than 5 years. 204 (87·2%) of 234 non-US-born

children younger than 5 years with at least one positive test were TST-positive

and IFN-γ release assay-negative. The proportion of non-US-born participants who

were TST-negative but IFN-γ release assay-positive ranged from one (0·5%) of 199 children younger than 2 years to 86 (14·5%) of 594 participants aged 65 years

and older (ptrend<0·0001). Test agreement was higher between the two IFN-γ

release assays than between TST and either IFN-γ release assay, regardless of

birthplace. κ agreement was particularly low between TST and IFN-γ release

assays in non-US-born children younger than 5 years.

INTERPRETATION: Our findings support the preferential use of IFN-γ release

assays for the diagnosis of latent tuberculosis in high-risk populations,

especially in very young and older people born outside the USA.

FUNDING: US Centers for Disease Control and Prevention.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(21)00145-6

PMID: 34499863

 

16. Nat Commun. 2021 Sep 2;12(1):5236. doi: 10.1038/s41467-021-25537-z.

 

The cryo-EM structure of the bd oxidase from M. tuberculosis reveals a unique

structural framework and enables rational drug design to combat TB.

 

Safarian S(1), Opel-Reading HK(2), Wu D(3), Mehdipour AR(4), Hards K(5), Harold

LK(5), Radloff M(3), Stewart I(6), Welsch S(7), Hummer G(4)(8), Cook GM(5),

Krause KL(2), Michel H(9).

 

Author information:

(1)Department of Molecular Membrane Biology, Max Planck Institute of Biophysics,

Frankfurt/Main, Germany. schara.safarian@biophys.mpg.de.

(2)Department of Biochemistry, University of Otago, Dunedin, New Zealand.

(3)Department of Molecular Membrane Biology, Max Planck Institute of Biophysics,

Frankfurt/Main, Germany.

New drugs are urgently needed to combat the global TB epidemic. Targeting

simultaneously multiple respiratory enzyme complexes of Mycobacterium

tuberculosis is regarded as one of the most effective treatment options to

shorten drug administration regimes, and reduce the opportunity for the

emergence of drug resistance. During infection and proliferation, the cytochrome

bd oxidase plays a crucial role for mycobacterial pathophysiology by maintaining

aerobic respiration at limited oxygen concentrations. Here, we present the

cryo-EM structure of the cytochrome bd oxidase from M. tuberculosis at 2.5 Å. In

conjunction with atomistic molecular dynamics (MD) simulation studies we

discovered a previously unknown MK-9-binding site, as well as a unique disulfide

bond within the Q-loop domain that defines an inactive conformation of the

canonical quinol oxidation site in Actinobacteria. Our detailed insights into

the long-sought atomic framework of the cytochrome bd oxidase from M.

tuberculosis will form the basis for the design of highly specific drugs to act

on this enzyme.

 

© 2021. The Author(s).

 

DOI: 10.1038/s41467-021-25537-z

PMCID: PMC8413341

PMID: 34475399 [Indexed for MEDLINE]

 

17. J Clin Invest. 2021 Sep 2:148013. doi: 10.1172/JCI148013. Online ahead of print.

 

Alveolar macrophages from persons living with HIV show impaired epigenetic

response to Mycobacterium tuberculosis.

 

Correa-Macedo W(1), Fava VM(2), Orlova M(2), Cassart P(2), Olivenstein R(3),

Sanz J(4), Xu YZ(2), Dumaine A(5), Sindeaux RH(6), Yotova V(6), Pacis A(7),

Girouard J(8), Kalsdorf B(9), Lange C(9), Routy JP(2), Barreiro LB(5), Schurr

E(2).

 

Author information:

(1)Department of Biochemistry, McGill University, Montréal, Canada.

(2)Program in Infectious Diseases and Global Health, The Research Institute of

the McGill University Health Centre, Montréal, Canada.

(3)Translational Research in Respiratory Diseases Program, The Research

Institute of the McGill University Health Centre, Montréal, Canada.

Persons living with HIV (PLWH) are at increased risk of tuberculosis (TB).

HIV-associated TB is often the result of recent infection with Mycobacterium

tuberculosis (Mtb) followed by rapid progression to disease. Alveolar

macrophages (AM) are the first cells of the innate immune system that engage

Mtb, but how HIV and antiretroviral therapy (ART) impact on the

anti-mycobacterial response of AM is not known. To investigate the impact of HIV

and ART on the transcriptomic and epigenetic response of AM to Mtb, we obtained

AM by bronchoalveolar lavage from 20 PLWH receiving ART, 16 control subjects who

were HIV-free (HC), and 14 subjects who received ART as pre-exposure prophylaxis

(PrEP) to prevent HIV infection. Following in-vitro challenge with Mtb, AM from

each group displayed overlapping but distinct profiles of significantly up- and

down-regulated genes in response to Mtb. Comparatively, AM isolated from both

PLWH and PrEP subjects presented a substantially weaker transcriptional

response. In addition, AM from HC subjects challenged with Mtb responded with

pronounced chromatin accessibility changes while AM obtained from PLWH and PrEP

subjects displayed no significant changes in their chromatin state.

Collectively, these results revealed a stronger adverse effect of ART than HIV

on the epigenetic landscape and transcriptional responsiveness of AM.

 

DOI: 10.1172/JCI148013

PMID: 34473646

 

18. Lancet Infect Dis. 2021 Sep;21(9):e272-e280. doi: 10.1016/S1473-3099(21)00077-3.

 

Barriers and enablers to implementing tuberculosis control strategies in EU and

European Economic Area countries: a systematic review.

 

Conroy O(1), Wurie F(2), Collin SM(2), Edmunds M(2), de Vries G(3), Lönnroth

K(4), Abubakar I(5), Anderson SR(2), Zenner D(6).

 

Author information:

(1)TB Unit, National Infection Service, Public Health England, London, UK.

Electronic address: olivia.conroy@PHE.gov.uk.

(2)TB Unit, National Infection Service, Public Health England, London, UK.

(3)KNCV Tuberculosis Foundation, The Hague, Netherlands.

(4)Department of Public Health Sciences, Karolinska Institute, Stockholm,

Sweden.

(5)Institute for Global Health, University College London, London, UK.

(6)TB Unit, National Infection Service, Public Health England, London, UK;

Institute for Global Health, University College London, London, UK.

 

Meeting the 2035 WHO targets of reducing tuberculosis incidence by 90% from 2015

levels requires the implementation of country-specific tuberculosis control

strategies. This systematic review aims to identify factors that facilitate or

impede the implementation of such strategies in EU and European Economic Area

(EEA) settings. Focusing on providers of care, health system constraints, and

social and political factors, this Review complements available evidence on the

accessibility of tuberculosis services to recipients of care. Databases were

searched for EU and EEA articles published between Jan 1, 1997, and Nov 6, 2020,

that presented empirical data on tuberculosis policies, strategies, guidelines,

or interventions. 2061 articles were screened and 65 were included. The most

common barrier to tuberculosis control strategies described the divergence of

health-care practices from guidelines, often related to inadequate knowledge or

perceived usefulness of the guidelines by clinicians. The most commonly

identified enabler to tuberculosis control strategies was the documented

positive attitudes of health-care workers towards tuberculosis programmes.

Divergence between clinical practice and guidelines was described in most EU and

EEA settings, indicating the need for a focused review of guideline adherence.

Strengths of this study involve its broad inclusion criteria and wide range of

tuberculosis control strategies analysed.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(21)00077-3

PMID: 34450080 [Indexed for MEDLINE]

 

19. J Exp Med. 2021 Sep 6;218(9):e20210615. doi: 10.1084/jem.20210615. Epub 2021 Jul 22.

 

Single cell analysis of M. tuberculosis phenotype and macrophage lineages in the

infected lung.

 

Pisu D(1), Huang L(1)(2), Narang V(3), Theriault M(1), Lê-Bury G(1), Lee B(3),

Lakudzala AE(4), Mzinza DT(4), Mhango DV(4), Mitini-Nkhoma SC(4), Jambo

KC(4)(5), Singhal A(3)(6), Mwandumba HC(4)(5), Russell DG(1).

 

Author information:

(1)Microbiology and Immunology, College of Veterinary Medicine, Cornell

University, Ithaca, NY.

(2)Microbiology and Immunology, University of Arkansas for Medical Sciences,

Little Rock, AR.

(3)Singapore Immunology Network, Agency for Science, Technology and Research,

Singapore.

(4)Malawi Liverpool Wellcome Trust Clinical Research Program, University of

Malawi College of Medicine, Blantyre, Malawi.

(5)Department of Clinical Sciences, Liverpool School of Tropical Medicine,

Liverpool, UK.

(6)A*STAR Infectious Diseases Laboratories, Agency for Science, Technology and

Research, Singapore.

 

In this study, we detail a novel approach that combines bacterial fitness

fluorescent reporter strains with scRNA-seq to simultaneously acquire the host

transcriptome, surface marker expression, and bacterial phenotype for each

infected cell. This approach facilitates the dissection of the functional

heterogeneity of M. tuberculosis-infected alveolar (AMs) and interstitial

macrophages (IMs) in vivo. We identify clusters of pro-inflammatory AMs

associated with stressed bacteria, in addition to three different populations of

IMs with heterogeneous bacterial phenotypes. Finally, we show that the main

macrophage populations in the lung are epigenetically constrained in their

response to infection, while inter-species comparison reveals that most AMs

subsets are conserved between mice and humans. This conceptual approach is

readily transferable to other infectious disease agents with the potential for

an increased understanding of the roles that different host cell populations

play during the course of an infection.

 

© 2021 Pisu et al.

 

DOI: 10.1084/jem.20210615

PMCID: PMC8302446

PMID: 34292313

 

20. J Exp Med. 2021 Sep 6;218(9):e20210332. doi: 10.1084/jem.20210332. Epub 2021 Jul 16.

 

Genetic models of latent tuberculosis in mice reveal differential influence of

adaptive immunity.

 

Su H(1), Lin K(1), Tiwari D(1), Healy C(1), Trujillo C(1), Liu Y(1), Ioerger

TR(2), Schnappinger D(1), Ehrt S(1).

 

Author information:

(1)Department of Microbiology and Immunology, Weill Cornell Medicine, New York,

NY.

(2)Department of Computer Science and Engineering, Texas A&M University, College

Station, TX.

 

Studying latent Mycobacterium tuberculosis (Mtb) infection has been limited by

the lack of a suitable mouse model. We discovered that transient depletion of

biotin protein ligase (BPL) and thioredoxin reductase (TrxB2) results in latent

infections during which Mtb cannot be detected but that relapse in a subset of

mice. The immune requirements for Mtb control during latency, and the frequency

of relapse, were strikingly different depending on how latency was established.

TrxB2 depletion resulted in a latent infection that required adaptive immunity

for control and reactivated with high frequency, whereas latent infection after

BPL depletion was independent of adaptive immunity and rarely reactivated. We

identified immune signatures of T cells indicative of relapse and demonstrated

that BCG vaccination failed to protect mice from TB relapse. These reproducible

genetic latency models allow investigation of the host immunological

determinants that control the latent state and offer opportunities to evaluate

therapeutic strategies in settings that mimic aspects of latency and TB relapse

in humans.

 

© 2021 Su et al.

 

DOI: 10.1084/jem.20210332

PMCID: PMC8289691

PMID: 34269789

 

21. J Clin Oncol. 2021 Sep 10;39(26):2872-2880. doi: 10.1200/JCO.21.00276. Epub 2021 Jul 12.

 

SAKK 16/14: Durvalumab in Addition to Neoadjuvant Chemotherapy in Patients With

Stage IIIA(N2) Non-Small-Cell Lung Cancer-A Multicenter Single-Arm Phase II

Trial.

 

Rothschild SI(1), Zippelius A(1), Eboulet EI(2), Savic Prince S(3), Betticher

D(4), Bettini A(4), Früh M(5)(6), Joerger M(5), Lardinois D(7), Gelpke H(8),

Mauti LA(9), Britschgi C(10), Weder W(11), Peters S(12), Mark M(13), Cathomas

R(13), Ochsenbein AF(6), Janthur WD(14), Waibel C(15), Mach N(16), Froesch

P(17), Buess M(18), Bohanes P(19), Godar G(2), Rusterholz C(2), Gonzalez M(20),

Pless M(9); Swiss Group for Clinical Cancer Research (SAKK).

 

Author information:

(1)Department of Medical Oncology and Comprehensive Cancer Center, University

Hospital Basel, Basel, Switzerland.

(2)SAKK Coordinating Center, Bern, Switzerland.

(3)Pathology, Institute of Medical Genetics and Pathology, University Hospital

Basel, Basel, Switzerland.

PURPOSE: For patients with resectable stage IIIA(N2) non-small-cell lung cancer,

neoadjuvant chemotherapy with cisplatin and docetaxel followed by surgery

resulted in a 1-year event-free survival (EFS) rate of 48% in the SAKK 16/00

trial and is an accepted standard of care. We investigated the additional

benefit of perioperative treatment with durvalumab.

METHODS: Neoadjuvant treatment consisted of three cycles of cisplatin 100 mg/m2

and docetaxel 85 mg/m2 once every 3 weeks followed by two doses of durvalumab

750 mg once every 2 weeks. Durvalumab was continued for 1 year after surgery.

The primary end point was 1-year EFS. The hypothesis for statistical

considerations was an improvement of 1-year EFS from 48% to 65%.

RESULTS: Sixty-eight patients were enrolled, 67 were included in the full

analysis set. Radiographic response rate was 43% (95% CI, 31 to 56) after

neoadjuvant chemotherapy and 58% (95% CI, 45 to 71) after sequential neoadjuvant

immunotherapy. Fifty-five patients were resected, of which 34 (62%) achieved a

major pathologic response (MPR; ≤ 10% viable tumor cells) and 10 (18%) among

them a complete pathologic response. Postoperative nodal downstaging (ypN0-1)

was observed in 37 patients (67%). Fifty-one (93%) resected patients had an R0

resection. There was no significant effect of pretreatment PD-L1 expression on

MPR or nodal downstaging. The 1-year EFS rate was 73% (two-sided 90% CI, 63 to

82). Median EFS and overall survival were not reached after 28.6 months of

median follow-up. Fifty-nine (88%) patients had an adverse event grade ≥ 3

including two fatal adverse events that were judged not to be treatment-related.

CONCLUSION: The addition of perioperative durvalumab to neoadjuvant chemotherapy

in patients with stage IIIA(N2) non-small-cell lung cancer is safe and exceeds

historical data of chemotherapy alone with a high MPR and an encouraging 1-year

EFS rate of 73%.

 

DOI: 10.1200/JCO.21.00276

PMID: 34251873

 

22. Am J Respir Crit Care Med. 2021 Sep 15;204(6):713-722. doi:

10.1164/rccm.202009-3527OC.

 

Evidence-based Definition for Extensively Drug-Resistant Tuberculosis.

 

Roelens M(1), Battista Migliori G(2), Rozanova L(1), Estill J(1)(3), Campbell

JR(4)(5), Cegielski JP(6), Tiberi S(7)(8), Palmero D(9), Fox GJ(10),

Guglielmetti L(11)(12), Sotgiu G(13), Brust JCM(14), Bang D(15)(16), Lienhardt

C(17)(18), Lange C(19)(20)(21), Menzies D(4)(5), Keiser O(1), Raviglione M(22).

 

Author information:

(1)Institute of Global Health, Faculty of Medicine, University of Geneva,

Geneva, Switzerland.

(2)Istituti Clinici Scientifici Maugeri, Istituto di Ricovero e Cura a Carattere

Scientifico, Tradate, Italy.

(3)Institute of Mathematical Statistics and Actuarial Science, University of

Bern, Bern, Switzerland.

Comment in

    Am J Respir Crit Care Med. 2021 Sep 15;204(6):629-631.

 

Rationale: Until 2020, extensively drug-resistant tuberculosis (XDR-TB) was

defined as TB with resistance to rifampicin and isoniazid (multidrug-resistant

TB [MDR-TB]), any fluoroquinolone (FQ), and any second-line injectable drug

(SLID). In 2019, the World Health Organization issued new recommendations for

treating patients with drug-resistant TB, substantially limiting the role of

SLIDs in MDR-TB treatment and thus putting the definition of XDR-TB into

question. Objectives: To propose an up-to-date definition for XDR-TB. Methods:

We used a large data set to assess treatment outcomes for patients with MDR-TB

exposed to any type of longer regimen. We included patients with

bacteriologically confirmed MDR-TB and known FQ and SLID resistance results. We

performed logistic regression to estimate the adjusted odds ratios (aORs) for an

unfavorable treatment outcome (failure, relapse, death, loss to follow-up), and

estimates were stratified by the resistance pattern (FQ and/or SLID) and group A

drug use (moxifloxacin/levofloxacin, linezolid, and/or bedaquiline).

Measurements and Main Results: We included 11,666 patients with MDR-TB; 4,653

(39.9%) had an unfavorable treatment outcome. Resistance to FQs increased the

odds of an unfavorable treatment outcome (aOR, 1.91; 95% confidence interval

[CI], 1.63-2.23). Administration of bedaquiline and/or linezolid improved

treatment outcomes regardless of resistance to FQs and/or SLIDs. Among patients

with XDR-TB, compared with persons receiving no group A drug, aORs for an

unfavorable outcome were 0.37 (95% CI, 0.20-0.69) with linezolid only, 0.40 (95%

CI, 0.21-0.77) with bedaquiline only, and 0.21 (95% CI, 0.12-0.38) with both.

Conclusions: Our study supports a new definition of XDR-TB as MDR-TB and

additional resistance to FQ plus bedaquiline and/or linezolid and helps assess

the adequacy of this definition for surveillance and treatment choice.

 

DOI: 10.1164/rccm.202009-3527OC

PMID: 34107231 [Indexed for MEDLINE]

 

23. Autophagy. 2021 Sep;17(9):2629-2638. doi: 10.1080/15548627.2020.1825273. Epub 2020 Oct 4.

 

Neutrophil autophagy during human active tuberculosis is modulated by SLAMF1.

 

Pellegrini JM(1)(2), Sabbione F(3), Morelli MP(1)(2), Tateosian NL(1)(2),

Castello FA(1)(2), Amiano NO(1)(2), Palmero D(4), Levi A(4), Ciallella L(4),

Colombo MI(5), Trevani AS(3), García VE(1)(2).

 

Author information:

(1)Departamento de Química Biológica. Facultad de Ciencias Exactas y Naturales,

UBA, Buenos Aires, Argentina.

(2)Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales

(IQUIBICEN), Facultad de Ciencias Exactas y Naturales, Universidad de Buenos

Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires,

Argentina.

(3)Laboratorio de Inmunidad Innata, Instituto de Medicina Experimental

(IMEX)-CONICET,Academia Nacional de Medicina, Buenos Aires, Argentina.

(4)Hospital F.J. Muñiz, Uspallata 2272, (C1282AEN) Buenos Aires, Argentina.

(5)Instituto de Histología y Embriología de Mendoza, Facultad de Ciencias

Médicas, Universidad Nacional de Cuyo-CONICET, Mendoza, Argentina.

 

Neutrophils infected with Mycobacterium tuberculosis (Mtb) predominate in

tuberculosis patients' lungs. Neutrophils phagocytose the pathogen, but the

mechanism of pathogen elimination is controversial. Macroautophagy/autophagy, a

crucial mechanism for several neutrophil functions, can be modulated by

immunological mediators. The costimulatory molecule SLAMF1 can act as a

microbial sensor in macrophages being also able to interact with

autophagy-related proteins. Here, we demonstrate for the first time that human

neutrophils express SLAMF1 upon Mtb-stimulation. Furthermore, SLAMF1 was found

colocalizing with LC3B+ vesicles, and activation of SLAMF1 increased neutrophil

autophagy induced by Mtb. Finally, tuberculosis patients' neutrophils displayed

reduced levels of SLAMF1 and lower levels of autophagy against Mtb as compared

to healthy controls. Altogether, these results indicate that SLAMF1 participates

in neutrophil autophagy during active tuberculosis.Abbreviations: AFB: acid-fast

bacilli; BafA1: bafilomycin A1; CLL: chronic lymphocytic leukemia; DPI:

diphenyleneiodonium; EVs: extracellular vesicles; FBS: fetal bovine serum; HD:

healthy donors; HR: high responder (tuberculosis patient); IFNG: interferon

gamma; IL1B: interleukin 1 beta; IL17A: interleukin 17A; IL8: interleukin 8; LR:

low responder (tuberculosis patient); mAb: monoclonal antibody; MAP1LC3/LC3:

microtubule associated protein 1 light chain 3; MAPK: mitogen-activated protein

kinase; MAPK1/ERK2: mitogen-activated protein kinase 1; MAPK14/p38:

mitogen-activated protein kinase 14; Mtb: Mycobacterium tuberculosis; Mtb-Ag:

Mycobacterium tuberculosis, Strain H37Rv, whole cell lysate; NETs: neutrophils

extracellular traps; PPD: purified protein derivative; ROS: reactive oxygen

species; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3;

SLAMF1: signaling lymphocytic activation molecule family member 1; TB:

tuberculosis; TLR: toll like receptor.

 

DOI: 10.1080/15548627.2020.1825273

PMCID: PMC8496709

PMID: 32954947

 


上一篇: No.11

下一篇: No.9