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.8

发布时间:2021-09-13 浏览次数:
字号: + - 14

Medical Abstracts

Filters applied: from 2021/7/1 - 2021/7/31

Key words:   tuberculosis ;  lung cancer


1. Cell. 2021 Aug 19;184(17):4579-4592.e24. doi: 10.1016/j.cell.2021.06.033. Epub

2021 Jul 22.

 

Genome-wide gene expression tuning reveals diverse vulnerabilities of

M.tuberculosis.

 

Bosch B(1), DeJesus MA(1), Poulton NC(1), Zhang W(2), Engelhart CA(3), Zaveri

A(3), Lavalette S(3), Ruecker N(3), Trujillo C(3), Wallach JB(3), Li S(1), Ehrt

S(3), Chait BT(2), Schnappinger D(4), Rock JM(5).

 

Author information:

(1)Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY

10065, USA.

(2)Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller

University, New York, NY 10065, USA.

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

NY 10065, USA.

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

NY 10065, USA. Electronic address: dis2003@med.cornell.edu.

(5)Laboratory of Host-Pathogen Biology, The Rockefeller University, New York, NY

10065, USA. Electronic address: rock@rockefeller.edu.

 

Antibacterial agents target the products of essential genes but rarely achieve

complete target inhibition. Thus, the all-or-none definition of essentiality

afforded by traditional genetic approaches fails to discern the most attractive

bacterial targets: those whose incomplete inhibition results in major fitness

costs. In contrast, gene "vulnerability" is a continuous, quantifiable trait

that relates the magnitude of gene inhibition to the effect on bacterial

fitness. We developed a CRISPR interference-based functional genomics method to

systematically titrate gene expression in Mycobacterium tuberculosis (Mtb) and

monitor fitness outcomes. We identified highly vulnerable genes in various

processes, including novel targets unexplored for drug discovery. Equally

important, we identified invulnerable essential genes, potentially explaining

failed drug discovery efforts. Comparison of vulnerability between the reference

and a hypervirulent Mtb isolate revealed incomplete conservation of

vulnerability and that differential vulnerability can predict differential

antibacterial susceptibility. Our results quantitatively redefine essential

bacterial processes and identify high-value targets for drug development.

 

Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.cell.2021.06.033

PMCID: PMC8382161

PMID: 34297925

 

2. Nature. 2021 Jul;595(7868):578-584. doi: 10.1038/s41586-021-03651-8. Epub 2021

Jun 16.

 

Tissue-resident macrophages provide a pro-tumorigenic niche to early NSCLC

cells.

 

Casanova-Acebes M(1)(2)(3)(4), Dalla E(#)(5)(6)(7)(8), Leader AM(#)(9)(10)(11),

LeBerichel J(#)(9)(10)(11), Nikolic J(12), Morales BM(13), Brown M(13), Chang

C(9)(10)(11), Troncoso L(9)(10)(11), Chen ST(9)(10)(11), Sastre-Perona

A(14)(15), Park MD(9)(10)(11), Tabachnikova A(9)(10)(11), Dhainaut

M(10)(11)(16), Hamon P(9)(10)(11), Maier B(9)(10)(11)(17), Sawai CM(18),

Agulló-Pascual E(19), Schober M(14), Brown BD(10)(11)(16)(20), Reizis B(21),

Marron T(9)(10)(11)(5)(22), Kenigsberg E(10)(20), Moussion C(13), Benaroch

P(12), Aguirre-Ghiso JA(9)(10)(11)(5)(6)(7)(8), Merad M(23)(24)(25)(26).

 

Author information:

(1)Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai,

New York, NY, USA. mcasanova@cnio.es.

(2)Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New

York, NY, USA. mcasanova@cnio.es.

(3)Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York,

NY, USA. mcasanova@cnio.es.

Comment in

    Nat Immunol. 2021 Sep;22(9):1078-1079.

 

Macrophages have a key role in shaping the tumour microenvironment (TME), tumour

immunity and response to immunotherapy, which makes them an important target for

cancer treatment1,2. However, modulating macrophages has proved extremely

difficult, as we still lack a complete understanding of the molecular and

functional diversity of the tumour macrophage compartment. Macrophages arise

from two distinct lineages. Tissue-resident macrophages self-renew locally,

independent of adult haematopoiesis3-5, whereas short-lived monocyte-derived

macrophages arise from adult haematopoietic stem cells, and accumulate mostly in

inflamed lesions1. How these macrophage lineages contribute to the TME and

cancer progression remains unclear. To explore the diversity of the macrophage

compartment in human non-small cell lung carcinoma (NSCLC) lesions, here we

performed single-cell RNA sequencing of tumour-associated leukocytes. We

identified distinct populations of macrophages that were enriched in human and

mouse lung tumours. Using lineage tracing, we discovered that these macrophage

populations differ in origin and have a distinct temporal and spatial

distribution in the TME. Tissue-resident macrophages accumulate close to tumour

cells early during tumour formation to promote epithelial-mesenchymal transition

and invasiveness in tumour cells, and they also induce a potent regulatory T

cell response that protects tumour cells from adaptive immunity. Depletion of

tissue-resident macrophages reduced the numbers and altered the phenotype of

regulatory T cells, promoted the accumulation of CD8+ T cells and reduced tumour

invasiveness and growth. During tumour growth, tissue-resident macrophages

became redistributed at the periphery of the TME, which becomes dominated by

monocyte-derived macrophages in both mouse and human NSCLC. This study

identifies the contribution of tissue-resident macrophages to early lung cancer

and establishes them as a target for the prevention and treatment of early lung

cancer lesions.

 

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

 

DOI: 10.1038/s41586-021-03651-8

PMID: 34135508

 

3. CA Cancer J Clin. 2021 Jul;71(4):299-314. doi: 10.3322/caac.21671. Epub 2021 May

20.

 

Racial and socioeconomic disparities in lung cancer screening in the United

States: A systematic review.

 

Sosa E(1), D'Souza G(2), Akhtar A(2), Sur M(1), Love K(3), Duffels J(3), Raz

DJ(2), Kim JY(2), Sun V(1)(2), Erhunmwunsee L(1)(2).

 

Author information:

(1)Department of Populations Sciences, City of Hope National Medical Center,

Duarte, California.

(2)Department of Surgery, City of Hope Comprehensive Cancer Center, Duarte,

California.

(3)Division of Library Services, City of Hope National Medical Center, Duarte,

California.

 

Nonsmall cell lung cancer (NSCLC) is the leading cause of cancer deaths. Lung

cancer screening (LCS) reduces NSCLC mortality; however, a lack of diversity in

LCS studies may limit the generalizability of the results to marginalized groups

who face higher risk for and worse outcomes from NSCLC. Identifying sources of

inequity in the LCS pipeline is essential to reduce disparities in NSCLC

outcomes. The authors searched 3 major databases for studies published from

January 1, 2010 to February 27, 2020 that met the following criteria: 1)

included screenees between ages 45 and 80 years who were current or former

smokers, 2) written in English, 3) conducted in the United States, and 4)

discussed socioeconomic and race-based LCS outcomes. Eligible studies were

assessed for risk of bias. Of 3721 studies screened, 21 were eligible. Eligible

studies were evaluated, and their findings were categorized into 3 themes

related to LCS disparities faced by Black and socioeconomically disadvantaged

individuals: 1) eligibility; 2) utilization, perception, and utility; and 3)

postscreening behavior and care. Disparities in LCS exist along racial and

socioeconomic lines. There are several steps along the LCS pipeline in which

Black and socioeconomically disadvantaged individuals miss the potential

benefits of LCS, resulting in increased mortality. This study identified

potential sources of inequity that require further investigation. The authors

recommend the implementation of prospective trials that evaluate eligibility

criteria for underserved groups and the creation of interventions focused on

improving utilization and follow-up care to decrease LCS disparities.

 

© 2021 The Authors. CA: A Cancer Journal for Clinicians published by Wiley

Periodicals LLC on behalf of American Cancer Society.

 

DOI: 10.3322/caac.21671

PMCID: PMC8266751

PMID: 34015860

 

4. Cancer Cell. 2021 Jul 20:S1535-6108(21)00382-2. doi:

10.1016/j.ccell.2021.07.005. Online ahead of print.

 

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

 

5. Cancer Cell. 2021 Jul 27:S1535-6108(21)00383-4. doi:

10.1016/j.ccell.2021.07.006. Online ahead of print.

 

Targeting Aurora B kinase prevents and overcomes resistance to EGFR inhibitors

in lung cancer by enhancing BIM- and PUMA-mediated apoptosis.

 

Tanaka K(1), Yu HA(2), Yang S(1), Han S(1), Selcuklu SD(1), Kim K(3), Ramani

S(1), Ganesan YT(1), Moyer A(4), Sinha S(1), Xie Y(5), Ishizawa K(1),

Osmanbeyoglu HU(6), Lyu Y(7), Roper N(8), Guha U(9), Rudin CM(10), Kris MG(2),

Hsieh JJ(7), Cheng EH(11).

 

Author information:

(1)Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer

Center, New York, NY 10065, USA.

(2)Thoracic Oncology Service, Department of Medicine, Memorial Sloan Kettering

Cancer Center, Department of Medicine, Weill Cornell Medical College, New York,

NY 10065, USA.

(3)Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY

10065, USA.

The clinical success of EGFR inhibitors in EGFR-mutant lung cancer is limited by

the eventual development of acquired resistance. We hypothesize that enhancing

apoptosis through combination therapies can eradicate cancer cells and reduce

the emergence of drug-tolerant persisters. Through high-throughput screening of

a custom library of 1,000 compounds, we discover Aurora B kinase inhibitors as

potent enhancers of osimertinib-induced apoptosis. Mechanistically, Aurora B

inhibition stabilizes BIM through reduced Ser87 phosphorylation, and

transactivates PUMA through FOXO1/3. Importantly, osimertinib resistance caused

by epithelial-mesenchymal transition (EMT) activates the ATR-CHK1-Aurora B

signaling cascade and thereby engenders hypersensitivity to respective kinase

inhibitors by activating BIM-mediated mitotic catastrophe. Combined inhibition

of EGFR and Aurora B not only efficiently eliminates cancer cells but also

overcomes resistance beyond EMT.

 

Copyright © 2021 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2021.07.006

PMID: 34388376

 

6. Lancet Oncol. 2021 Jul;22(7):959-969. doi: 10.1016/S1470-2045(21)00247-3. Epub

2021 Jun 9.

 

Pralsetinib for RET fusion-positive non-small-cell lung cancer (ARROW): a

multi-cohort, open-label, phase 1/2 study.

 

Gainor JF(1), Curigliano G(2), Kim DW(3), Lee DH(4), Besse B(5), Baik CS(6),

Doebele RC(7), Cassier PA(8), Lopes G(9), Tan DSW(10), Garralda E(11), Paz-Ares

LG(12), Cho BC(13), Gadgeel SM(14), Thomas M(15), Liu SV(16), Taylor MH(17),

Mansfield AS(18), Zhu VW(19), Clifford C(20), Zhang H(21), Palmer M(22), Green

J(23), Turner CD(23), Subbiah V(24).

 

Author information:

(1)Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.

Electronic address: jgainor@partners.org.

(2)Department of Oncology and Hemato-Oncology, University of Milan and European

Institute of Oncology, IRCCS, Milan, Italy.

(3)Department of Internal Medicine, Seoul National University College of

Medicine and Seoul National University Hospital, Seoul, South Korea.

Erratum in

    Lancet Oncol. 2021 Aug;22(8):e347.

 

BACKGROUND: Oncogenic alterations in RET have been identified in multiple tumour

types, including 1-2% of non-small-cell lung cancers (NSCLCs). We aimed to

assess the safety, tolerability, and antitumour activity of pralsetinib, a

highly potent, oral, selective RET inhibitor, in patients with RET

fusion-positive NSCLC.

METHODS: ARROW is a multi-cohort, open-label, phase 1/2 study done at 71 sites

(community and academic cancer centres) in 13 countries (Belgium, China, France,

Germany, Hong Kong, Italy, Netherlands, Singapore, South Korea, Spain, Taiwan,

the UK, and the USA). Patients aged 18 years or older with locally advanced or

metastatic solid tumours, including RET fusion-positive NSCLC, and an Eastern

Cooperative Oncology Group performance status of 0-2 (later limited to 0-1 in a

protocol amendment) were enrolled. In phase 2, patients received 400 mg

once-daily oral pralsetinib, and could continue treatment until disease

progression, intolerance, withdrawal of consent, or investigator decision. Phase

2 primary endpoints were overall response rate (according to Response Evaluation

Criteria in Solid Tumours version 1·1 and assessed by blinded independent

central review) and safety. Tumour response was assessed in patients with RET

fusion-positive NSCLC and centrally adjudicated baseline measurable disease who

had received platinum-based chemotherapy or were treatment-naive because they

were ineligible for standard therapy. This ongoing study is registered with

ClinicalTrials.gov, NCT03037385, and enrolment of patients with treatment-naive

RET fusion-positive NSCLC was ongoing at the time of this interim analysis.

FINDINGS: Of 233 patients with RET fusion-positive NSCLC enrolled between March

17, 2017, and May 22, 2020 (data cutoff), 92 with previous platinum-based

chemotherapy and 29 who were treatment-naive received pralsetinib before July

11, 2019 (efficacy enrolment cutoff); 87 previously treated patients and 27

treatment-naive patients had centrally adjudicated baseline measurable disease.

Overall responses were recorded in 53 (61%; 95% CI 50-71) of 87 patients with

previous platinum-based chemotherapy, including five (6%) patients with a

complete response; and 19 (70%; 50-86) of 27 treatment-naive patients, including

three (11%) with a complete response. In 233 patients with RET fusion-positive

NSCLC, common grade 3 or worse treatment-related adverse events were neutropenia

(43 patients [18%]), hypertension (26 [11%]), and anaemia (24 [10%]); there were

no treatment-related deaths in this population.

INTERPRETATION: Pralsetinib is a new, well-tolerated, promising, once-daily oral

treatment option for patients with RET fusion-positive NSCLC.

FUNDING: Blueprint Medicines.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1470-2045(21)00247-3

PMID: 34118197 [Indexed for MEDLINE]

 

7. Nat Med. 2021 Jul;27(7):1171-1177. doi: 10.1038/s41591-021-01358-x. Epub 2021

May 24.

 

Prisons as ecological drivers of fitness-compensated multidrug-resistant

Mycobacterium tuberculosis.

 

Gygli SM(#)(1)(2), Loiseau C(#)(1)(2), Jugheli L(1)(2)(3), Adamia N(3), Trauner

A(1)(2), Reinhard M(1)(2), Ross A(1)(2), Borrell S(1)(2), Aspindzelashvili R(3),

Maghradze N(1)(2)(3), Reither K(1)(2), Beisel C(4), Tukvadze N(1)(2)(3),

Avaliani Z(3), Gagneux S(5)(6).

 

Author information:

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

(2)University of Basel, Basel, Switzerland.

(3)National Center for Tuberculosis and Lung Diseases (NCTLD), Tbilisi, Georgia.

(4)Department of Biosystems Science and Engineering, ETH Zürich, Basel,

Switzerland.

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

sebastien.gagneux@swisstph.ch.

(6)University of Basel, Basel, Switzerland. sebastien.gagneux@swisstph.ch.

(#)Contributed equally

 

Erratum in

    Nat Med. 2021 Jun 1;:

 

Multidrug-resistant tuberculosis (MDR-TB) accounts for one third of the annual

deaths due to antimicrobial resistance1. Drug resistance-conferring mutations

frequently cause fitness costs in bacteria2-5. Experimental work indicates that

these drug resistance-related fitness costs might be mitigated by compensatory

mutations6-10. However, the clinical relevance of compensatory evolution remains

poorly understood. Here we show that, in the country of Georgia, during a 6-year

nationwide study, 63% of MDR-TB was due to patient-to-patient transmission.

Compensatory mutations and patient incarceration were independently associated

with transmission. Furthermore, compensatory mutations were overrepresented

among isolates from incarcerated individuals that also frequently spilled over

into the non-incarcerated population. As a result, up to 31% of MDR-TB in

Georgia was directly or indirectly linked to prisons. We conclude that prisons

fuel the epidemic of MDR-TB in Georgia by acting as ecological drivers of

fitness-compensated strains with high transmission potential.

 

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

 

DOI: 10.1038/s41591-021-01358-x

PMID: 34031604

 

8. Mol Aspects Med. 2021 Jul 31:101002. doi: 10.1016/j.mam.2021.101002. Online

ahead of print.

 

Protein synthesis in Mycobacterium tuberculosis as a potential target for

therapeutic interventions.

 

Kumar N(1), Sharma S(1), Kaushal PS(2).

 

Author information:

(1)Structural Biology & Translation Regulation Laboratory, Regional Centre for

Biotechnology, NCR Biotech Science Cluster, Faridabad, 121 001, India.

(2)Structural Biology & Translation Regulation Laboratory, Regional Centre for

Biotechnology, NCR Biotech Science Cluster, Faridabad, 121 001, India.

Electronic address: prem.kaushal@rcb.res.in.

 

Mycobacterium tuberculosis (Mtb) causes one of humankind's deadliest diseases,

tuberculosis. Mtb protein synthesis machinery possesses several unique

species-specific features, including its ribosome that carries two mycobacterial

specific ribosomal proteins, bL37 and bS22, and ribosomal RNA segments. Since

the protein synthesis is a vital cellular process that occurs on the ribosome, a

detailed knowledge of the structure and function of mycobacterial ribosomes is

essential to understand the cell's proteome by translation regulation. Like in

many bacterial species such as Bacillus subtilis and Streptomyces coelicolor,

two distinct populations of ribosomes have been identified in Mtb. Under

low-zinc conditions, Mtb ribosomal proteins S14, S18, L28, and L33 are replaced

with their non-zinc binding paralogues. Depending upon the nature of

physiological stress, species-specific modulation of translation by stress

factors and toxins that interact with the ribosome have been reported. In

addition, about one-fourth of messenger RNAs in mycobacteria have been reported

to be leaderless, i.e., without 5' UTR regions. However, the mechanism by which

they are recruited to the Mtb ribosome is not understood. In this review, we

highlight the mycobacteria-specific features of the translation apparatus and

propose exploiting these features to improve the efficacy and specificity of

existing antibiotics used to treat tuberculosis.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/j.mam.2021.101002

PMID: 34344520

 

9. 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.

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

 

10. Nat Commun. 2021 Jul 16;12(1):4360. doi: 10.1038/s41467-021-24537-3.

 

Glucocorticoid receptor triggers a reversible drug-tolerant dormancy state with

acquired therapeutic vulnerabilities in lung cancer.

 

Prekovic S(#)(1), Schuurman K(#)(2), Mayayo-Peralta I(2), Manjón AG(3), Buijs

M(2), Yavuz S(2), Wellenstein MD(4), Barrera A(5), Monkhorst K(6), Huber

A(2)(7), Morris B(8), Lieftink C(8), Chalkiadakis T(2), Alkan F(2), Silva J(2),

Győrffy B(9)(10), Hoekman L(11), van den Broek B(12), Teunissen H(13), Debets

DO(14), Severson T(2), Jonkers J(15), Reddy T(5), de Visser KE(4), Faller W(2),

Beijersbergen R(8), Altelaar M(11)(14), de Wit E(13), Medema R(3), Zwart

W(16)(17).

 

Author information:

(1)Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute,

Amsterdam, The Netherlands. s.prekovic@nki.nl.

(2)Division of Oncogenomics, Oncode Institute, The Netherlands Cancer Institute,

Amsterdam, The Netherlands.

(3)Division of Cell Biology, Oncode Institute, The Netherlands Cancer Institute,

Amsterdam, The Netherlands.

The glucocorticoid receptor (GR) regulates gene expression, governing aspects of

homeostasis, but is also involved in cancer. Pharmacological GR activation is

frequently used to alleviate therapy-related side-effects. While prior studies

have shown GR activation might also have anti-proliferative action on tumours,

the underpinnings of glucocorticoid action and its direct effectors in

non-lymphoid solid cancers remain elusive. Here, we study the mechanisms of

glucocorticoid response, focusing on lung cancer. We show that GR activation

induces reversible cancer cell dormancy characterised by anticancer drug

tolerance, and activation of growth factor survival signalling accompanied by

vulnerability to inhibitors. GR-induced dormancy is dependent on a single

GR-target gene, CDKN1C, regulated through chromatin looping of a GR-occupied

upstream distal enhancer in a SWI/SNF-dependent fashion. These insights

illustrate the importance of GR signalling in non-lymphoid solid cancer biology,

particularly in lung cancer, and warrant caution for use of glucocorticoids in

treatment of anticancer therapy related side-effects.

 

© 2021. The Author(s).

 

DOI: 10.1038/s41467-021-24537-3

PMCID: PMC8285479

PMID: 34272384 [Indexed for MEDLINE]

 

11. 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

 

12. PLoS Med. 2021 Jul 14;18(7):e1003717. doi: 10.1371/journal.pmed.1003717.

eCollection 2021 Jul.

 

Evaluating the impact of the nationwide public-private mix (PPM) program for

tuberculosis under National Health Insurance in South Korea: A difference in

differences analysis.

 

Yu S(1)(2)(3), Sohn H(4), Kim HY(2)(3), Kim H(1)(5), Oh KH(1)(6), Kim HJ(1),

Chung H(2)(3), Choi H(1)(7).

 

Author information:

(1)Korean Institute of Tuberculosis, Korean National Tuberculosis Association,

Cheongju, Republic of Korea.

(2)School of Health Policy & Management, College of Health Science, Korea

University, Seoul, Republic of Korea.

(3)BK21 FOUR R&E Center for Learning Health Systems, Korea University, Seoul,

Republic of Korea.

BACKGROUND: Public-private mix (PPM) programs on tuberculosis (TB) have a

critical role in engaging and integrating the private sector into the national

TB control efforts in order to meet the End TB Strategy targets. South Korea's

PPM program can provide important insights on the long-term impact and policy

gaps in the development and expansion of PPM as a nationwide program.

METHODS AND FINDINGS: Healthcare is privatized in South Korea, and a majority

(80.3% in 2009) of TB patients sought care in the private sector. Since 2009,

South Korea has rapidly expanded its PPM program coverage under the National

Health Insurance (NHI) scheme as a formal national program with dedicated PPM

nurses managing TB patients in both the private and public sectors. Using the

difference in differences (DID) analytic framework, we compared relative changes

in TB treatment outcomes-treatment success (TS) and loss to follow-up (LTFU)-in

the private and public sector between the 2009 and 2014 TB patient cohorts.

Propensity score matching (PSM) using the kernel method was done to adjust for

imbalances in the covariates between the 2 population cohorts. The 2009 cohort

included 6,195 (63.0% male, 37.0% female; mean age: 42.1) and 27,396 (56.1%

male, 43.9% female; mean age: 45.7) TB patients in the public and private

sectors, respectively. The 2014 cohort included 2,803 (63.2% male, 36.8% female;

mean age: 50.1) and 29,988 (56.5% male, 43.5% female; mean age: 54.7) patients.

In both the private and public sectors, the proportion of patients with transfer

history decreased (public: 23.8% to 21.7% and private: 20.8% to 17.6%), and

bacteriological confirmed disease increased (public: 48.9% to 62.3% and private:

48.8% to 58.1%) in 2014 compared to 2009. After expanding nationwide PPM,

absolute TS rates improved by 9.10% (87.5% to 93.4%) and by 13.6% (from 70.3% to

83.9%) in the public and private sectors. Relative to the public, the private

saw 4.1% (95% confidence interval [CI] 2.9% to 5.3%, p-value < 0.001) and -8.7%

(95% CI -9.7% to -7.7%, p-value<0.001) higher rates of improvement in TS and

reduction in LTFU. Treatment outcomes did not improve in patients who

experienced at least 1 transfer during their TB treatment. Study limitations

include non-longitudinal nature of our original dataset, inability to assess the

regional disparities, and verify PPM program's impact on TB mortality.

CONCLUSIONS: We found that the nationwide scale-up of the PPM program was

associated with improvements in TB treatment outcomes in the private sector in

South Korea. Centralized financial governance and regulatory mechanisms were

integral in facilitating the integration of highly diverse South Korean private

sector into the national TB control program and scaling up of the PPM

intervention nationwide. However, TB care gaps continued to exist for patients

who transferred at least once during their treatment. These programmatic gaps

may be improved through reducing administrative hurdles and making programmatic

amendments that can help facilitate management TB patients between institutions

and healthcare sectors, as well as across administrative regions.

 

DOI: 10.1371/journal.pmed.1003717

PMCID: PMC8318235

PMID: 34260579

 

13. Immunity. 2021 Aug 10;54(8):1758-1771.e7. doi: 10.1016/j.immuni.2021.06.009.

Epub 2021 Jul 12.

 

Macrophage and neutrophil death programs differentially confer resistance to

tuberculosis.

 

Stutz MD(1), Allison CC(1), Ojaimi S(1), Preston SP(1), Doerflinger M(1),

Arandjelovic P(1), Whitehead L(1), Bader SM(1), Batey D(1), Asselin-Labat ML(1),

Herold MJ(1), Strasser A(1), West NP(2), Pellegrini M(3).

 

Author information:

(1)The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052,

Australia; Department of Medical Biology, The University of Melbourne,

Parkville, VIC 3010, Australia.

(2)School of Chemistry and Molecular Bioscience, The University of Queensland,

St Lucia, QLD 4072, Australia.

(3)The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052,

Australia; Department of Medical Biology, The University of Melbourne,

Parkville, VIC 3010, Australia. Electronic address: pellegrini@wehi.edu.au.

 

Comment in

    Immunity. 2021 Aug 10;54(8):1625-1627.

 

Apoptosis can potently defend against intracellular pathogens by directly

killing microbes and eliminating their replicative niche. However, the reported

ability of Mycobacterium tuberculosis to restrict apoptotic pathways in

macrophages in vitro has led to apoptosis being dismissed as a host-protective

process in tuberculosis despite a lack of in vivo evidence. Here we define

crucial in vivo functions of the death receptor-mediated and BCL-2-regulated

apoptosis pathways in mediating protection against tuberculosis by eliminating

distinct populations of infected macrophages and neutrophils and priming T cell

responses. We further show that apoptotic pathways can be targeted

therapeutically with clinical-stage compounds that antagonize inhibitor of

apoptosis (IAP) proteins to promote clearance of M. tuberculosis in mice. These

findings reveal that any inhibition of apoptosis by M. tuberculosis is

incomplete in vivo, advancing our understanding of host-protective responses to

tuberculosis (TB) and revealing host pathways that may be targetable for

treatment of disease.

 

Copyright © 2021 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.immuni.2021.06.009

PMID: 34256013

 

14. Am J Respir Crit Care Med. 2021 Jul 12. doi: 10.1164/rccm.202011-4063OC. Online

ahead of print.

 

Air Pollution, Genetic Factors and the Risk of Lung Cancer: A Prospective Study

in the UK Biobank.

 

Huang Y(1), Zhu M(1)(2)(3), Ji M(1), Fan J(1), Xie J(1), Wei X(1), Jiang X(1),

Xu J(4), Chen L(4), Yin R(3), Wang Y(1)(3), Dai J(1)(2), Jin G(1)(2), Xu L(3),

Hu Z(1)(2), Ma H(1)(5), Shen H(1)(2)(6).

 

Author information:

(1)Nanjing Medical University School of Public Health, 572407, Department of

Epidemiology, Center for Global Health, Nanjing, China.

(2)Nanjing Medical University, 12461, Jiangsu Key Lab of Cancer Biomarkers,

Prevention and Treatment, Collaborative Innovation Center for Cancer

Personalized Medicine, Nanjing, China.

(3)Jiangsu Institute of Cancer Research, 26481, Department of Thoracic Surgery,

Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Jiangsu

Cancer Hospital, The Affiliated Cancer Hospital of Nanjing Medical University,

Nanjing, China.

 

Rationale: Both genetic and environmental factors contribute to lung cancer, but

the degree to which air pollution modifies the impact of genetic susceptibility

on lung cancer remains unknown. Objectives: To investigate whether air pollution

and genetic factors jointly contribute to incident lung cancer. Methods: We

analyzed data from 455,974 participants (53% women) without previous cancer at

baseline in the UK Biobank. The concentrations of particulate matter (PM2.5,

PMcoarse and PM10), nitrogen dioxide (NO2), and nitrogen oxides (NOx) were

estimated by land-use regression models, and the association between air

pollutants and incident lung cancer was investigated using a Cox proportional

hazard model. Furthermore, we constructed a polygenic risk score and evaluated

whether air pollutants modified the effect of genetic susceptibility on the

development of lung cancer. Measurements and Main Results: The results showed

significant associations between the risk of lung cancer and PM2.5 (hazard ratio

[HR]: 1.63, 95% confidence interval [CI]: 1.33-2.01; per 5 μg/m3), PM10 (1.53,

1.20-1.96; per 10 μg/m3), NO2 (1.10, 1.05-1.15; per 10 μg/m3), and NOx (1.13,

1.07-1.18; per 20 μg/m3). There were additive interactions between air

pollutants and the genetic risk. Compared with participants with low genetic

risk and low air pollution, those with high air pollution and high genetic risk

had the highest risk of lung cancer (PM2.5: HR: 1.71, 95% CI:1.45-2.02; PM10:

1.77, 1.50-2.10; NO2: 1.77, 1.42-2.22; NOx: 1.67, 1.43-1.95). Conclusion:

Long-term exposure to air pollution may increase the risk of lung cancer,

especially in those with high genetic risk.

 

DOI: 10.1164/rccm.202011-4063OC

PMID: 34252012

 

15. 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

 

16. J Clin Invest. 2021 Jul 15;131(14):e140073. doi: 10.1172/JCI140073.

 

Monocyte metabolic transcriptional programs associate with resistance to

tuberculin skin test/interferon-γ release assay conversion.

 

Simmons JD(1), Van PT(2), Stein CM(3)(4), Chihota V(5)(6), Ntshiqa T(6),

Maenetje P(6), Peterson GJ(1), Reynolds A(1), Benchek P(3), Velen K(6), Fielding

KL(5)(7), Grant AD(5)(7)(8), Graustein AD(1), Nguyen FK(1), Seshadri C(1),

Gottardo R(2), Mayanja-Kizza H(9), Wallis RS(6), Churchyard G(6), Boom WH(4),

Hawn TR(1).

 

Author information:

(1)TB Research and Training Center, Department of Medicine, University of

Washington, Seattle, Washington, USA.

(2)Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.

(3)Department of Population & Quantitative Health Sciences and.

 

After extensive exposure to Mycobacterium tuberculosis (Mtb), most individuals

acquire latent Mtb infection (LTBI) defined by a positive tuberculin skin test

(TST) or interferon-γ release assay (IGRA). To identify mechanisms of resistance

to Mtb infection, we compared transcriptional profiles from highly exposed

contacts who resist TST/IGRA conversion (resisters, RSTRs) and controls with

LTBI using RNAseq. Gene sets related to carbon metabolism and free fatty acid

(FFA) transcriptional responses enriched across 2 independent cohorts suggesting

RSTR and LTBI monocytes have distinct activation states. We compared

intracellular Mtb replication in macrophages treated with FFAs and found that

palmitic acid (PA), but not oleic acid (OA), enhanced Mtb intracellular growth.

This PA activity correlated with its inhibition of proinflammatory cytokines in

Mtb-infected cells. Mtb growth restriction in PA-treated macrophages was

restored by activation of AMP kinase (AMPK), a central host metabolic regulator

known to be inhibited by PA. Finally, we genotyped AMPK variants and found 7

SNPs in PRKAG2, which encodes the AMPK-γ subunit, that strongly associated with

RSTR status. Taken together, RSTR and LTBI phenotypes are distinguished by FFA

transcriptional programs and by genetic variation in a central metabolic

regulator, which suggests immunometabolic pathways regulate TST/IGRA conversion.

 

DOI: 10.1172/JCI140073

PMCID: PMC8279582

PMID: 34111032

 

17. Autophagy. 2021 Jul 7:1-19. doi: 10.1080/15548627.2021.1938912. Online ahead of print.

 

M. tuberculosis PknG manipulates host autophagy flux to promote pathogen

intracellular survival.

 

Ge P(1)(2), Lei Z(1)(2), Yu Y(1)(2), Lu Z(1)(2), Qiang L(1)(2), Chai Q(1), Zhang

Y(1), Zhao D(1)(2), Li B(1), Pang Y(3), Liu CH(1)(2), Wang J(1).

 

Author information:

(1)CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of

Microbiology, Center for Biosafety Mega-Science, Chinese Academy of Sciences,

Beijing, China.

(2)Savaid Medical School, University of Chinese Academy of Sciences, Beijing,

China.

(3)Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing Chest

Hospital, Capital Medical University, Beijing, China.

 

The eukaryotic-type protein kinase G (PknG), one of the eleven eukaryotic type

serine-threonine protein kinase (STPK) in Mycobacterium tuberculosis (Mtb), is

involved in mycobacterial survival within macrophages, presumably by suppressing

phagosome and autophagosome maturation, which makes PknG an attractive drug

target. However, the exact mechanism by which PknG inhibits pathogen clearance

during mycobacterial infection remains largely unknown. Here, we show that PknG

promotes macroautophagy/autophagy induction but inhibits autophagosome

maturation, causing an overall effect of blocked autophagy flux and enhanced

pathogen intracellular survival. PknG prevents the activation of AKT (AKT

serine/threonine kinase) via competitively binding to its pleckstrin homology

(PH) domain, leading to autophagy induction. Remarkably, PknG could also inhibit

autophagosome maturation to block autophagy flux via targeting host small GTPase

RAB14. Specifically, PknG directly interacts with RAB14 to block RAB14-GTP

hydrolysis. Furthermore, PknG phosphorylates TBC1D4/AS160 (TBC1 domain family

member 4) to suppress its GTPase-activating protein (GAP) activity toward RAB14.

In macrophages and in vivo, PknG promotes Mtb intracellular survival through

blocking autophagy flux, which is dependent on RAB14. Taken together, our data

unveil a dual-functional bacterial effector that tightly regulates host

autophagy flux to benefit pathogen intracellular survival.Abbreviations: AKT:

AKT serine/threonine kinase; ATG5: autophagy related 5; BMDMs: bone

marrow-derived macrophages; DTT: dithiothreitol; FBS: fetal calf serum; GAP:

GTPase-activating protein; MOI: multiplicity of infection; Mtb: Mycobacterium

tuberculosis; MTOR: mechanistic target of rapamycin kinase; OADC: oleic

acid-albumin-dextrose-catalase; PC, phosphatidylcholine; PH: pleckstrin

homology; PI3K: phosphoinositide 3-kinase; PknG: protein kinase G;

PtdIns(3,4,5)P3: phosphatidylinositol(3,4,5)-trisphosphate; SQSTM1: sequestosome

1; STPK: serine-threonine protein kinase; TB: tuberculosis; TBC1D4: TBC1 domain

family member 4; TPR: tetratricopeptide repeat; ULK1: unc-51 like autophagy

activating kinase 1; WT: wild-type.

 

DOI: 10.1080/15548627.2021.1938912

PMID: 34092182

 

18. J Clin Oncol. 2021 Jul 20;39(21):2339-2349. doi: 10.1200/JCO.21.00174. Epub 2021

Apr 19.

 

Five-Year Outcomes With Pembrolizumab Versus Chemotherapy for Metastatic

Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50.

 

Reck M(1), Rodríguez-Abreu D(2), Robinson AG(3), Hui R(4), Csőszi T(5), Fülöp

A(6), Gottfried M(7), Peled N(8), Tafreshi A(9), Cuffe S(10), O'Brien M(11), Rao

S(12), Hotta K(13), Leal TA(14), Riess JW(15), Jensen E(16), Zhao B(16),

Pietanza MC(16), Brahmer JR(17).

 

Author information:

(1)Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), member of the

German Center for Lung Research (DZL), Grosshansdorf, Germany.

(2)Complejo Hospitalario Universitario Insular Materno-Infantil de Gran Canaria,

Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.

(3)Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston,

ON, Canada.

Comment in  2321.

 

PURPOSE: We report the first 5-year follow-up of any first-line phase III

immunotherapy trial for non-small-cell lung cancer (NSCLC). KEYNOTE-024

(ClinicalTrials.gov identifier: NCT02142738) is an open-label, randomized

controlled trial of pembrolizumab compared with platinum-based chemotherapy in

patients with previously untreated NSCLC with a programmed death ligand-1

(PD-L1) tumor proportion score of at least 50% and no sensitizing EGFR or ALK

alterations. Previous analyses showed pembrolizumab significantly improved

progression-free survival and overall survival (OS).

METHODS: Eligible patients were randomly assigned (1:1) to pembrolizumab (200 mg

once every 3 weeks for up to 35 cycles) or platinum-based chemotherapy. Patients

in the chemotherapy group with progressive disease could cross over to

pembrolizumab. The primary end point was progression-free survival; OS was a

secondary end point.

RESULTS: Three hundred five patients were randomly assigned: 154 to

pembrolizumab and 151 to chemotherapy. Median (range) time from randomization to

data cutoff (June 1, 2020) was 59.9 (55.1-68.4) months. Among patients initially

assigned to chemotherapy, 99 received subsequent anti-PD-1 or PD-L1 therapy,

representing a 66.0% effective crossover rate. Median OS was 26.3 months (95%

CI, 18.3 to 40.4) for pembrolizumab and 13.4 months (9.4-18.3) for chemotherapy

(hazard ratio, 0.62; 95% CI, 0.48 to 0.81). Kaplan-Meier estimates of the 5-year

OS rate were 31.9% for the pembrolizumab group and 16.3% for the chemotherapy

group. Thirty-nine patients received 35 cycles (ie, approximately 2 years) of

pembrolizumab, 82.1% of whom were still alive at data cutoff (approximately 5

years). Toxicity did not increase with longer treatment exposure.

CONCLUSION: Pembrolizumab provides a durable, clinically meaningful long-term OS

benefit versus chemotherapy as first-line therapy for metastatic NSCLC with

PD-L1 tumor proportion score of at least 50%.

 

DOI: 10.1200/JCO.21.00174

PMCID: PMC8280089

PMID: 33872070

 

19. Lancet Infect Dis. 2021 Jul;21(7):984-992. doi: 10.1016/S1473-3099(20)30919-1.

Epub 2021 Feb 25.

 

Quantifying the global number of tuberculosis survivors: a modelling study.

 

Dodd PJ(1), Yuen CM(2), Jayasooriya SM(3), van der Zalm MM(4), Seddon JA(5).

 

Author information:

(1)School of Health and Related Research, University of Sheffield, Sheffield,

UK. Electronic address: p.j.dodd@sheffield.ac.uk.

(2)Harvard Medical School, Harvard University, Boston, MA, USA; Division of

Global Health Equity, Brigham and Women's Hospital, Boston, MA, USA.

(3)Academic Unit of Primary Care, University of Sheffield, Sheffield, UK.

(4)Desmond Tutu TB Centre, Department of Paediatrics and Child Health,

Stellenbosch University, Cape Town, South Africa.

(5)Desmond Tutu TB Centre, Department of Paediatrics and Child Health,

Stellenbosch University, Cape Town, South Africa; Section of Paediatric

Infectious Diseases, Department of Infectious Diseases, Imperial College London,

London, UK.

 

BACKGROUND: People who survive tuberculosis face clinical and societal

consequences after recovery, including increased risks of recurrent

tuberculosis, premature death, reduced lung function, and ongoing stigma. To

describe the size of this issue, we aimed to estimate the number of individuals

who developed first-episode tuberculosis between 1980 and 2019, the number who

survived to 2020, and the number who have been treated within the past 5 years

or 2 years.

METHODS: In this modelling study, we estimated the number of people who survived

treated tuberculosis using country-level WHO data on tuberculosis case

notifications, excluding those who died during treatment. We estimated the

number of individuals surviving untreated tuberculosis using the difference

between WHO country-level incidence estimates and notifications, applying

published age-stratified and HIV-stratified case fatality ratios. To estimate

survival with time, post-tuberculosis life tables were developed for each

country-year by use of UN World Population Prospects 2019 mortality rates and

published post-tuberculosis mortality hazard ratios.

FINDINGS: Between 1980 and 2019, we estimate that 363 million people (95%

uncertainty interval [UI] 287 million-438 million) developed tuberculosis, of

whom 172 million (169 million-174 million) were treated. Individuals who

developed tuberculosis between 1980 and 2019 had lived 3480 million life-years

(95% UI 3040 million-3920 million) after tuberculosis by 2020, with survivors

younger than 15 years at the time of tuberculosis development contributing 12%

(95% UI 7-17) of these life-years. We estimate that 155 million tuberculosis

survivors (95% UI 138 million-171 million) were alive in 2020, the largest

proportion (47% [37-57]) of whom were in the WHO South-East Asia region. Of the

tuberculosis survivors who were alive in 2020, we estimate that 18% (95% UI

16-20) were treated in the past 5 years and 8% (7-9) were treated in the past 2

years.

INTERPRETATION: The number of tuberculosis survivors alive in 2020 is more than

ten times the estimated annual tuberculosis incidence. Interventions to

alleviate respiratory morbidity, screen for and prevent recurrent tuberculosis,

and reduce stigma should be immediately prioritised for recently treated

tuberculosis survivors.

FUNDING: UK Medical Research Council, the UK Department for International

Development, the National Institute for Health Research, and the European and

Developing Countries Clinical Trials Partnership.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(20)30919-1

PMID: 33640076 [Indexed for MEDLINE]

 

20. Lancet Infect Dis. 2021 Jul;21(7):975-983. doi: 10.1016/S1473-3099(20)30770-2.

Epub 2021 Feb 12.

 

QT effects of bedaquiline, delamanid, or both in patients with

rifampicin-resistant tuberculosis: a phase 2, open-label, randomised, controlled

trial.

 

Dooley KE(1), Rosenkranz SL(2), Conradie F(3), Moran L(4), Hafner R(5), von

Groote-Bidlingmaier F(6), Lama JR(7), Shenje J(8), De Los Rios J(7), Comins

K(6), Morganroth J(9), Diacon AH(10), Cramer YS(2), Donahue K(11), Maartens

G(12); AIDS Clinical Trials Group (ACTG) A5343 DELIBERATE Study Team.

 

Collaborators: Alli O, Gottesman J, Guevara M, Hikuam C, Hovind L, Karlsson M,

McClaren J, McIlleron H, Murtaugh W, Rolls B, Shahkolahi A, Stone L, Tegha G,

Tenai J, Upton C, Wimbish C.

 

Author information:

(1)Johns Hopkins University School of Medicine, Baltimore, MD, USA. Electronic

address: kdooley1@jhmi.edu.

(2)Frontier Science Foundation, Brookline, MA, USA.

(3)University of Witwatersrand, Johannesburg, South Africa.

Comment in

    Lancet Infect Dis. 2021 Jul;21(7):894-895.

 

BACKGROUND: Bedaquiline and delamanid are the first drugs of new classes

registered for tuberculosis treatment in 40 years. Each can prolong the QTc

interval, with maximum effects occurring weeks after drug initiation. The

cardiac safety and microbiological activity of these drugs when co-administered

are not well-established. Our aim was to characterise the effects of

bedaquiline, delamanid, or both on the QTc interval, longitudinally over 6

months of multidrug treatment, among patients with multidrug-resistant or

rifampicin-resistant tuberculosis taking multidrug background therapy.

METHODS: ACTG A5343 is a phase 2, open-label, randomised, controlled trial in

which adults with multidrug-resistant or rifampicin-resistant tuberculosis

receiving multidrug background treatment were randomly assigned 1:1:1 by

centrally, computer-generated randomisation, by means of permuted blocks to

receive bedaquiline, delamanid, or both for 24 weeks. Participants were enrolled

at TASK in Cape Town and the South African Tuberculosis Vaccine Initiative in

Worcester, both in South Africa, and Hospital Maria Auxiliadora in Peru.

Individuals with QTc greater than 450 ms were excluded. HIV-positive

participants received dolutegravir-based antiretroviral therapy. Clofazimine was

disallowed, and levofloxacin replaced moxifloxacin. ECG in triplicate and sputum

cultures were done fortnightly. The primary endpoint was mean QTcF change from

baseline (averaged over weeks 8-24); cumulative culture conversation at week

8-24 was an exploratory endpoint. Analyses included all participants who

initiated study tuberculosis treatment (modified intention-to-treat population).

This trial is registered with ClinicalTrials.gov, NCT02583048 and is ongoing.

FINDINGS: Between Aug 26, 2016 and July 13, 2018, of 174 screened, 84

participants (28 in each treatment group, and 31 in total with HIV) were

enrolled. Two participants did not initiate study treatment (one in the

delamanid group withdrew consent and one in the bedaquiline plus delamanid

group) did not meet the eligibility criterion). Mean change in QTc from baseline

was 12·3 ms (95% CI 7·8-16·7; bedaquiline), 8·6 ms (4·0-13·1; delamanid), and

20·7 ms (16·1-25·3) (bedaquiline plus delamanid). There were no grade 3 or 4

adverse QTc prolongation events and no deaths during study treatment. Cumulative

culture conversion by week 8 was 21 (88%) of 24 (95% CI 71-97; bedaquiline), 20

(83%) of 24 (65-95; delamanid), and 19 (95%) of 20 (79-100; bedaquiline plus

delamanid) and was 92% (77-99) for bedaquiline, 91% (76-99), for delamanid, and

95% (79-100) for bedaquiline plus delamanid at 24 weeks.

INTERPRETATION: Combining bedaquiline and delamanid has a modest, no more than

additive, effect on the QTc interval, and initial microbiology data are

encouraging. This study provides supportive evidence for use of these agents

together in patients with multidrug-resistant or rifampicin-resistant

tuberculosis with normal baseline QTc values.

FUNDING: Division of AIDS, National Institutes of Health.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(20)30770-2

PMCID: PMC8312310

PMID: 33587897 [Indexed for MEDLINE]

 

21. J Clin Oncol. 2021 Jul 20;39(21):2327-2338. doi: 10.1200/JCO.20.03579. Epub 2021

Jan 29.

 

Pembrolizumab Plus Ipilimumab or Placebo for Metastatic Non-Small-Cell Lung

Cancer With PD-L1 Tumor Proportion Score ≥ 50%: Randomized, Double-Blind Phase

III KEYNOTE-598 Study.

 

Boyer M(1), Şendur MAN(2), Rodríguez-Abreu D(3), Park K(4), Lee DH(5), Çiçin

I(6), Yumuk PF(7), Orlandi FJ(8), Leal TA(9), Molinier O(10), Soparattanapaisarn

N(11), Langleben A(12), Califano R(13), Medgyasszay B(14), Hsia TC(15), Otterson

GA(16), Xu L(17), Piperdi B(17), Samkari A(17), Reck M(18); KEYNOTE-598

Investigators.

 

Author information:

(1)Chris O'Brien Lifehouse, Camperdown, NSW, Australia.

(2)Ankara Yıldırım Beyazıt University, Faculty of Medicine and Ankara City

Hospital, Ankara, Turkey.

(3)Complejo Hospitalario Universitario Insular Materno-Infantil de Gran Canaria,

Universidad de Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain.

Comment in

    Nat Rev Clin Oncol. 2021 Apr;18(4):194.

 

PURPOSE: Pembrolizumab monotherapy is standard first-line therapy for metastatic

non-small-cell lung cancer (NSCLC) with programmed death ligand 1 (PD-L1) tumor

proportion score (TPS) ≥ 50% without actionable driver mutations. It is not

known whether adding ipilimumab to pembrolizumab improves efficacy over

pembrolizumab alone in this population.

METHODS: In the randomized, double-blind, phase III KEYNOTE-598 trial

(ClinicalTrials.gov identifier: NCT03302234), eligible patients with previously

untreated metastatic NSCLC with PD-L1 TPS ≥ 50% and no sensitizing EGFR or ALK

aberrations were randomly allocated 1:1 to ipilimumab 1 mg/kg or placebo every 6

weeks for up to 18 doses; all participants received pembrolizumab 200 mg every 3

weeks for up to 35 doses. Primary end points were overall survival and

progression-free survival.

RESULTS: Of the 568 participants, 284 were randomly allocated to each group.

Median overall survival was 21.4 months for pembrolizumab-ipilimumab versus 21.9

months for pembrolizumab-placebo (hazard ratio, 1.08; 95% CI, 0.85 to 1.37; P =

.74). Median progression-free survival was 8.2 months for

pembrolizumab-ipilimumab versus 8.4 months for pembrolizumab-placebo (hazard

ratio, 1.06; 95% CI, 0.86 to 1.30; P = .72). Grade 3-5 adverse events occurred

in 62.4% of pembrolizumab-ipilimumab recipients versus 50.2% of

pembrolizumab-placebo recipients and led to death in 13.1% versus 7.5%. The

external data and safety monitoring committee recommended that the study be

stopped for futility and that participants discontinue ipilimumab and placebo.

CONCLUSION: Adding ipilimumab to pembrolizumab does not improve efficacy and is

associated with greater toxicity than pembrolizumab monotherapy as first-line

treatment for metastatic NSCLC with PD-L1 TPS ≥ 50% and no targetable EGFR or

ALK aberrations. These data do not support use of pembrolizumab-ipilimumab in

place of pembrolizumab monotherapy in this population.

 

DOI: 10.1200/JCO.20.03579

PMID: 33513313

 

上一篇: No.9

下一篇: No.7