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 >>2022年

高分SCI文摘

2022年

No.8

发布时间:2022-10-12 浏览次数:
字号: + - 14

PubMed  Filters applied: from 2022/8/1 - 2022/8/31

1. Nat Genet. 2022 Aug;54(8):1167-1177. doi: 10.1038/s41588-022-01115-x. Epub 2022 Aug 1.

 

Cross-ancestry genome-wide meta-analysis of 61,047 cases and 947,237 controls

identifies new susceptibility loci contributing to lung cancer.

 

Byun J(#)(1)(2), Han Y(#)(1)(2), Li Y(#)(1)(2)(3), Xia J(#)(1)(4), Long E(#)(5),

Choi J(5), Xiao X(1), Zhu M(6), Zhou W(1), Sun R(7), Bossé Y(8), Song Z(1)(4),

Schwartz A(9)(10), Lusk C(9)(10), Rafnar T(11), Stefansson K(11), Zhang T(5),

Author information:

(1)Institute for Clinical and Translational Research, Baylor College of

Medicine, Houston, TX, USA.

(2)Section of Epidemiology and Population Sciences, Department of Medicine,

Baylor College of Medicine, Houston, TX, USA.

(3)Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine,

Houston, TX, USA.

To identify new susceptibility loci to lung cancer among diverse populations, we

performed cross-ancestry genome-wide association studies in European, East Asian

and African populations and discovered five loci that have not been previously

reported. We replicated 26 signals and identified 10 new lead associations from

previously reported loci. Rare-variant associations tended to be specific to

populations, but even common-variant associations influencing smoking behavior,

such as those with CHRNA5 and CYP2A6, showed population specificity.

Fine-mapping and expression quantitative trait locus colocalization nominated

several candidate variants and susceptibility genes such as IRF4 and FUBP1. DNA

damage assays of prioritized genes in lung fibroblasts indicated that a subset

of these genes, including the pleiotropic gene IRF4, potentially exert effects

by promoting endogenous DNA damage.

 

© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.

 

DOI: 10.1038/s41588-022-01115-x

PMCID: PMC9373844

PMID: 35915169 [Indexed for MEDLINE]

 

2. Cell. 2022 Aug 18;185(17):3214-3231.e23. doi: 10.1016/j.cell.2022.06.038. Epub

2022 Jul 30.

 

Mitochondrial ROS promotes susceptibility to infection via gasdermin D-mediated

necroptosis.

 

Weindel CG(1), Martinez EL(1), Zhao X(2), Mabry CJ(1), Bell SL(3), Vail KJ(4),

Coleman AK(1), VanPortfliet JJ(1), Zhao B(5), Wagner AR(1), Azam S(1), Scott

HM(1), Li P(5), West AP(1), Karpac J(2), Patrick KL(6), Watson RO(7).

 

Author information:

(1)Department of Microbial Pathogenesis and Immunology, Texas A&M Health,

College of Medicine, Bryan, TX 77807, USA.

(2)Department of Molecular and Cellular Medicine, Texas A&M Health, College of

Medicine, Bryan, TX 77807, USA.

(3)Department of Microbial Pathogenesis and Immunology, Texas A&M Health,

College of Medicine, Bryan, TX 77807, USA; Department of Microbiology,

Biochemistry & Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ

07103, USA.

Comment in

    Curr Biol. 2022 Aug 22;32(16):R891-R894.

 

Although mutations in mitochondrial-associated genes are linked to inflammation

and susceptibility to infection, their mechanistic contributions to immune

outcomes remain ill-defined. We discovered that the disease-associated

gain-of-function allele Lrrk2G2019S (leucine-rich repeat kinase 2) perturbs

mitochondrial homeostasis and reprograms cell death pathways in macrophages.

When the inflammasome is activated in Lrrk2G2019S macrophages, elevated

mitochondrial ROS (mtROS) directs association of the pore-forming protein

gasdermin D (GSDMD) to mitochondrial membranes. Mitochondrial GSDMD pore

formation then releases mtROS, promoting a switch to RIPK1/RIPK3/MLKL-dependent

necroptosis. Consistent with enhanced necroptosis, infection of Lrrk2G2019S mice

with Mycobacterium tuberculosis elicits hyperinflammation and severe

immunopathology. Our findings suggest a pivotal role for GSDMD as an executer of

multiple cell death pathways and demonstrate that mitochondrial dysfunction can

direct immune outcomes via cell death modality switching. This work provides

insights into how LRRK2 mutations manifest or exacerbate human diseases and

identifies GSDMD-dependent necroptosis as a potential target to limit

Lrrk2G2019S-mediated immunopathology.

 

Copyright © 2022 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.cell.2022.06.038

PMCID: PMC9531054

PMID: 35907404 [Indexed for MEDLINE]

 

3. Cancer Cell. 2022 Aug 30:S1535-6108(22)00377-4. doi:

10.1016/j.ccell.2022.08.013. Online ahead of print.

 

Analyses of rare predisposing variants of lung cancer in 6,004 whole genomes in

Chinese.

 

Wang C(1), Dai J(2), Qin N(2), Fan J(2), Ma H(3), Chen C(2), An M(2), Zhang

J(2), Yan C(2), Gu Y(4), Xie Y(5), He Y(4), Jiang Y(6), Zhu M(2), Song C(2),

Jiang T(7), Liu J(8), Zhou J(2), Wang N(2), Hua T(2), Liang S(2), Wang L(8), Xu

J(9), Yin R(10), Chen L(9), Xu L(10), Jin G(6), Lin D(11), Hu Z(12), Shen H(13).

 

Author information:

(1)Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative

Innovation Center for Cancer Personalized Medicine, Nanjing Medical University,

Nanjing 211166, Jiangsu, China; State Key Laboratory of Reproductive Medicine,

Nanjing Medical University, Nanjing 211166, Jiangsu, China; Department of

Epidemiology, Center for Global Health, School of Public Health, Nanjing Medical

University, Nanjing 211166, Jiangsu, China; Department of Bioinformatics, School

of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing

211166, Jiangsu, China.

We present the largest whole-genome sequencing (WGS) study of non-small cell

lung cancer (NSCLC) to date among 6,004 individuals of Chinese ancestry, coupled

with 23,049 individuals genotyped by SNP array. We construct a high-quality

haplotype reference panel for imputation and identify 20 common and

low-frequency loci (minor allele frequency [MAF] ≥ 0.5%), including five loci

that have never been reported before. For rare loss-of-function (LoF) variants

(MAF < 0.5%), we identify BRCA2 and 18 other cancer predisposition genes that

affect 5.29% of individuals with NSCLC, and 98.91% (181 of 183) of LoF variants

have not been linked previously to NSCLC risk. Promoter variants of BRCA2 also

have a substantial effect on NSCLC risk, and their prevalence is comparable with

BRCA2 LoF variants. The associations are validated in an independent

case-control study including 4,410 individuals and a prospective cohort study

including 23,826 individuals. Our findings not only provide a high-quality

reference panel for future array-based association studies but depict the whole

picture of rare pathogenic variants for NSCLC.

 

Copyright © 2022 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2022.08.013

PMID: 36113475

 

4. Cancer Cell. 2022 Sep 12;40(9):1010-1026.e11. doi: 10.1016/j.ccell.2022.08.003.

Epub 2022 Aug 25.

 

Personalized neoantigen vaccine NEO-PV-01 with chemotherapy and anti-PD-1 as

first-line treatment for non-squamous non-small cell lung cancer.

 

Awad MM(1), Govindan R(2), Balogh KN(3), Spigel DR(4), Garon EB(5), Bushway

ME(3), Poran A(3), Sheen JH(3), Kohler V(3), Esaulova E(3), Srouji J(3), Ramesh

S(3), Vyasamneni R(3), Karki B(5), Sciuto TE(3), Sethi H(6), Dong JZ(3), Moles

MA(3), Manson K(3), Rooney MS(3), Khondker ZS(3), DeMario M(3), Gaynor RB(7),

Srinivasan L(8).

 

Author information:

(1)Dana Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical

School, Boston, MA, USA.

(2)Washington University School of Medicine, St. Louis, MO, USA.

(3)BioNTech US, Cambridge, MA, USA.

Comment in

    Cancer Cell. 2022 Sep 12;40(9):903-905.

 

Neoantigens arising from mutations in tumor DNA provide targets for immune-based

therapy. Here, we report the clinical and immune data from a Phase Ib clinical

trial of a personalized neoantigen-vaccine NEO-PV-01 in combination with

pemetrexed, carboplatin, and pembrolizumab as first-line therapy for advanced

non-squamous non-small cell lung cancer (NSCLC). This analysis of 38 patients

treated with the regimen demonstrated no treatment-related serious adverse

events. Multiple parameters including baseline tumor immune infiltration and

on-treatment circulating tumor DNA levels were highly correlated with clinical

response. De novo neoantigen-specific CD4+ and CD8+ T cell responses were

observed post-vaccination. Epitope spread to non-vaccinating neoantigens,

including responses to KRAS G12C and G12V mutations, were detected

post-vaccination. Neoantigen-specific CD4+ T cells generated post-vaccination

revealed effector and cytotoxic phenotypes with increased CD4+ T cell

infiltration in the post-vaccine tumor biopsy. Collectively, these data support

the safety and immunogenicity of this regimen in advanced non-squamous NSCLC.

 

Copyright © 2022 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2022.08.003

PMID: 36027916 [Indexed for MEDLINE]

 

5. Nat Med. 2022 Aug 25. doi: 10.1038/s41591-022-01977-y. Online ahead of print.

 

Cemiplimab plus chemotherapy versus chemotherapy alone in non-small cell lung

cancer: a randomized, controlled, double-blind phase 3 trial.

 

Gogishvili M(1), Melkadze T(2), Makharadze T(3), Giorgadze D(4), Dvorkin M(5),

Penkov K(6), Laktionov K(7), Nemsadze G(8), Nechaeva M(9), Rozhkova I(10),

Kalinka E(11), Gessner C(12)(13), Moreno-Jaime B(14), Passalacqua R(15), Li

S(16), McGuire K(16), Kaul M(16), Paccaly A(16), Quek RGW(16), Gao B(16),

Seebach F(16), Weinreich DM(16), Yancopoulos GD(16), Lowy I(16), Gullo G(16),

Rietschel P(16).

 

Author information:

(1)High Technology Medical Centre, University Clinic Ltd., Tbilisi, Georgia.

mirandagogishvili@yahoo.com.

(2)Acad. F. Todua Medical Center, Tbilisi, Georgia.

(3)LTD High Technology Hospital Med Center, Batumi, Georgia.

First-line cemiplimab (anti-programmed cell death-1 (PD-1)) monotherapy has

previously shown significant improvement in overall survival (OS) and

progression-free survival (PFS) versus chemotherapy in patients with advanced

non-small cell lung cancer (aNSCLC) and PD-ligand 1 (PD-L1) expression ≥50%.

EMPOWER-Lung 3 ( NCT03409614 ), a double-blind, placebo-controlled, phase 3

study, examined cemiplimab plus platinum-doublet chemotherapy as first-line

treatment for aNSCLC, irrespective of PD-L1 expression or histology. In this

study, 466 patients with stage III/IV aNSCLC without EGFR, ALK or ROS1 genomic

tumor aberrations were randomized (2:1) to receive cemiplimab 350 mg (n = 312)

or placebo (n = 154) every 3 weeks for up to 108 weeks in combination with four

cycles of platinum-doublet chemotherapy (followed by pemetrexed maintenance as

indicated). In total, 57.1% (266/466 patients) had non-squamous NSCLC, and 85.2%

(397/466 patients) had stage IV disease. The primary endpoint was OS. The trial

was stopped early per recommendation of the independent data monitoring

committee, based on meeting preset OS efficacy criteria: median OS was

21.9 months (95% confidence interval (CI), 15.5-not evaluable) with cemiplimab

plus chemotherapy versus 13.0 months (95% CI, 11.9-16.1) with placebo plus

chemotherapy (hazard ratio (HR) = 0.71; 95% CI, 0.53-0.93; P = 0.014). Grade ≥3

adverse events occurred with cemiplimab plus chemotherapy (43.6%, 136/312

patients) and placebo plus chemotherapy (31.4%, 48/153 patients). Cemiplimab is

only the second anti-PD-1/PD-L1 agent to show efficacy in aNSCLC as both

monotherapy and in combination with chemotherapy for both squamous and

non-squamous histologies.

 

© 2022. The Author(s).

 

DOI: 10.1038/s41591-022-01977-y

PMID: 36008722

 

6. Lancet Oncol. 2022 Sep;23(9):1180-1188. doi: 10.1016/S1470-2045(22)00451-X. Epub 2022 Aug 11.

 

Chest CT scan plus x-ray versus chest x-ray for the follow-up of completely

resected non-small-cell lung cancer (IFCT-0302): a multicentre, open-label,

randomised, phase 3 trial.

 

Westeel V(1), Foucher P(2), Scherpereel A(3), Domas J(4), Girard P(5), Trédaniel

J(6), Wislez M(7), Dumont P(8), Quoix E(9), Raffy O(10), Braun D(11), Derollez

M(12), Goupil F(13), Hermann J(14), Devin E(15), Barbieux H(16), Pichon E(17),

Debieuvre D(18), Ozenne G(19), Muir JF(19), Dehette S(20), Virally J(21),

Grivaux M(22), Lebargy F(23), Souquet PJ(24), Freijat FA(25), Girard N(26),

Courau E(27), Azarian R(28), Farny M(29), Duhamel JP(30), Langlais A(31), Morin

F(31), Milleron B(7), Zalcman G(32), Barlesi F(33).

 

Author information:

(1)Department of Pneumology, University Hospital of Besançon, INSERM UMR1098,

Université de Bourgogne-Franche-Comté, Besançon, France. Electronic address:

virginie.westeel@univ-fcomte.fr.

(2)Department of Pneumology, University Hospital Bocage, Dijon, France.

(3)University of Lille, CHU Lille, Thoracic Oncology Department, CNRS, Inserm,

Institut Pasteur de Lille, UMR9020-UMR-S 1277-Canther, Lille, France.

Comment in

    Lancet Oncol. 2022 Sep;23(9):1115-1116.

 

BACKGROUND: Even after resection of early-stage non-small-cell lung cancer

(NSCLC), patients have a high risk of developing recurrence and second primary

lung cancer. We aimed to assess efficacy of a follow-up approach including

clinic visits, chest x-rays, chest CT scans, and fibre-optic bronchoscopy versus

clinical visits and chest x-rays after surgery for resectable NSCLC.

METHODS: In this multicentre, open-label, randomised, phase 3 trial (IFCT-0302),

patients aged 18 years or older and after complete resection of pathological

stage I-IIIA NSCLC according to the sixth edition of the TNM classification were

enrolled within 8 weeks of resection from 122 hospitals and tertiary centres in

France. Patients were randomly assigned (1:1) to CT-based follow-up (clinic

visits, chest x-rays, thoraco-abdominal CT scans, and fibre-optic bronchoscopy

for non-adenocarcinoma histology) or minimal follow-up (visits and chest x-rays)

after surgery for NSCLC, by means of a computer-generated sequence using the

minimisation method. Procedures were repeated every 6 months for the first 2

years and yearly until 5 years. The primary endpoint was overall survival

analysed in the intention-to-treat population. Secondary endpoints, also

analysed in the intention-to-treat population, included disease-free survival.

This trial is registered with ClinicalTrials.gov, NCT00198341, and is active,

but not enrolling.

FINDINGS: Between Jan 3, 2005, and Nov 30, 2012, 1775 patients were enrolled and

randomly assigned to a follow-up group (888 patients to the minimal follow-up

group; 887 patients to the CT-based follow-up group). Median overall survival

was not significantly different between follow-up groups (8·5 years [95% CI

7·4-9·6] in the minimal follow-up group vs 10·3 years [8·1-not reached] in the CT-based follow-up group; adjusted hazard ratio [HR] 0·95, 95% CI 0·83-1·10; log-rank p=0·49). Disease-free survival was not significantly different between follow-up groups (median not reached [95% CI not estimable-not estimable] in the minimal follow-up group vs 4·9 [4·3-not reached] in the CT-based follow-up group; adjusted HR 1·14, 95% CI 0·99-1·30; log-rank p=0·063). Recurrence was detected in 246 (27·7%) of 888 patients in the minimal follow-up group and in 289 (32·6%) patients of 887 in the CT-based follow-up group. Second primary lung cancer was diagnosed in 27 (3·0%) patients in the minimal follow-up group and 40 patients (4·5%) in the CT-based follow-up group. No serious adverse events

related to the trial procedures were reported.

INTERPRETATION: The addition of thoracic CT scans during follow-up, which

included clinic visits and chest x-rays after surgery, did not result in longer

survival among patients with NSCLC. However, it did enable the detection of more

cases of early recurrence and second primary lung cancer, which are more

amenable to curative-intent treatment, supporting the use of CT-based follow-up,

especially in countries where lung cancer screening is already implemented,

alongside with other supportive measures.

FUNDING: French Health Ministry, French National Cancer Institute,

Weisbrem-Benenson Foundation, La Ligue Nationale Contre Le Cancer, and Lilly

Oncology.

TRANSLATION: For the French translation of the abstract see Supplementary

Materials section.

 

Copyright © 2022 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1470-2045(22)00451-X

PMID: 35964621 [Indexed for MEDLINE]

 

7. Genes Dev. 2022 Aug 1;36(15-16):936-949. doi: 10.1101/gad.349659.122. Epub 2022 Sep 29.

 

Modeling diverse genetic subtypes of lung adenocarcinoma with a next-generation

alveolar type 2 organoid platform.

 

Naranjo S(#)(1)(2), Cabana CM(#)(1)(2), LaFave LM(1), Romero R(1)(2), Shanahan

SL(1)(2), Bhutkar A(1), Westcott PMK(1), Schenkel JM(1)(3)(4), Ghosh A(1), Liao

LZ(2), Del Priore I(1), Yang D(1)(5), Jacks T(1)(2).

 

Author information:

(1)David H. Koch Institute for Integrative Cancer Research, Massachusetts

Institute of Technology, Cambridge, Massachusetts 02142, USA.

(2)Department of Biology, Massachusetts Institute of Technology, Cambridge,

Massachusetts 02142, USA.

(3)Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts

02115, USA.

(4)Harvard Medical School, Boston, Massachusetts 02115, USA.

(5)Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142,

USA.

(#)Contributed equally

 

Lung cancer is the leading cause of cancer-related death worldwide. Lung

adenocarcinoma (LUAD), the most common histological subtype, accounts for 40% of

all cases. While existing genetically engineered mouse models (GEMMs)

recapitulate the histological progression and transcriptional evolution of human

LUAD, they are time-consuming and technically demanding. In contrast, cell line

transplant models are fast and flexible, but these models fail to capture the

full spectrum of disease progression. Organoid technologies provide a means to

create next-generation cancer models that integrate the most advantageous

features of autochthonous and transplant-based systems. However, robust and

faithful LUAD organoid platforms are currently lacking. Here, we describe

optimized conditions to continuously expand murine alveolar type 2 (AT2) cells,

a prominent cell of origin for LUAD, in organoid culture. These organoids

display canonical features of AT2 cells, including marker gene expression, the

presence of lamellar bodies, and an ability to differentiate into the AT1

lineage. We used this system to develop flexible and versatile immunocompetent

organoid-based models of KRAS, BRAF, and ALK mutant LUAD. Notably,

organoid-based tumors display extensive burden and complete penetrance and are

histopathologically indistinguishable from their autochthonous counterparts.

Altogether, this organoid platform is a powerful, versatile new model system to

study LUAD.

 

© 2022 Naranjo et al.; Published by Cold Spring Harbor Laboratory Press.

 

DOI: 10.1101/gad.349659.122

PMID: 36175034 [Indexed for MEDLINE]

 

8. Nat Commun. 2022 Aug 30;13(1):5105. doi: 10.1038/s41467-022-32455-1.

 

Transcontinental spread and evolution of Mycobacterium tuberculosis W148

European/Russian clade toward extensively drug resistant tuberculosis.

 

Merker M(#)(1)(2)(3), Rasigade JP(#)(4)(5)(6), Barbier M(#)(4)(5), Cox H(7),

Feuerriegel S(1)(2), Kohl TA(1)(2), Shitikov E(8), Klaos K(9), Gaudin C(10),

Antoine R(11), Diel R(12)(13), Borrell S(14)(15), Gagneux S(14)(15),

Nikolayevskyy V(16), Andres S(17), Crudu V(18), Supply P(19), Niemann S(20)(21),

Wirth T(22)(23).

 

Author information:

(1)Molecular and Experimental Mycobacteriology, Research Center Borstel,

Borstel, Germany.

(2)German Center for Infection Research, Partner site

Hamburg-Lübeck-Borstel-Riems, Borstel, Germany.

(3)Evolution of the Resistome, Research Center Borstel, Borstel, Germany.

Transmission-driven multi-/extensively drug resistant (M/XDR) tuberculosis (TB)

is the largest single contributor to human mortality due to antimicrobial

resistance. A few major clades of the Mycobacterium tuberculosis complex

belonging to lineage 2, responsible for high prevalence of MDR-TB in Eurasia,

show outstanding transnational distributions. Here, we determined factors

underlying the emergence and epidemic spread of the W148 clade by genome

sequencing and Bayesian demogenetic analyses of 720 isolates from 23 countries.

We dated a common ancestor around 1963 and identified two successive epidemic

expansions in the late 1980s and late 1990s, coinciding with major

socio-economic changes in the post-Soviet Era. These population expansions

favored accumulation of resistance mutations to up to 11 anti-TB drugs, with MDR

evolving toward additional resistances to fluoroquinolones and second-line

injectable drugs within 20 years on average. Timescaled haplotypic density

analysis revealed that widespread acquisition of compensatory mutations was

associated with transmission success of XDR strains. Virtually all W148 strains

harbored a hypervirulence-associated ppe38 gene locus, and incipient recurrent

emergence of prpR mutation-mediated drug tolerance was detected. The outstanding

genetic arsenal of this geographically widespread M/XDR strain clade represents

a "perfect storm" that jeopardizes the successful introduction of new

anti-M/XDR-TB antibiotic regimens.

 

© 2022. The Author(s).

 

DOI: 10.1038/s41467-022-32455-1

PMCID: PMC9426364

PMID: 36042200 [Indexed for MEDLINE]

 

9. J Clin Oncol. 2022 Aug 26:JCO2200428. doi: 10.1200/JCO.22.00428. Online ahead of print.

 

Updated Overall Survival and Exploratory Analysis From Randomized, Phase II EVAN

Study of Erlotinib Versus Vinorelbine Plus Cisplatin Adjuvant Therapy in Stage

IIIA Epidermal Growth Factor Receptor+ Non-Small-Cell Lung Cancer.

 

Yue D(1), Xu S(2), Wang Q(3), Li X(4), Shen Y(5), Zhao H(6), Chen C(7), Mao

W(8), Liu W(9), Liu J(10), Zhang L(11), Ma H(12), Li Q(13), Yang Y(14), Liu

Y(15), Chen H(16), Zhang Z(1), Zhang B(1), Wang C(1).

 

Author information:

(1)Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.

(2)Harbin Medical University Cancer Hospital, Harbin, China.

(3)Zhongshan Hospital, Fudan University, Shanghai, China.

Clinical trials frequently include multiple end points that mature at different

times. The initial report, typically based on the primary end point, may be

published when key planned co-primary or secondary analyses are not yet

available. Clinical Trial Updates provide an opportunity to disseminate

additional results from studies, published in JCO or elsewhere, for which the

primary end point has already been reported.The randomized, open-label, phase II

EVAN study investigated the efficacy (disease-free survival [DFS] and 5-year

overall survival [OS]) and safety of erlotinib versus vinorelbine/cisplatin as

adjuvant chemotherapy after complete resection (R0) for stage III epidermal

growth factor receptor (EGFR) mutation+ non-small-cell lung cancer. We describe

the updated results at the 43-month follow-up. In EVAN, patients were randomly

assigned (1:1) to erlotinib (n = 51) or vinorelbine/cisplatin (n = 51). The

median follow-up was 54.8 and 63.9 months in the erlotinib and chemotherapy

arms, respectively. With erlotinib, the respective 5-year DFS by Kaplan-Meier

analysis was 48.2% (95% CI, 29.4 to 64.7) and 46.2% (95% CI, 27.6 to 62.9) in

the intention-to-treat and per-protocol populations. The median OS was 84.2

months with erlotinib versus 61.1 months with chemotherapy (hazard ratio, 0.318;

95% CI, 0.151 to 0.670). The 5-year survival rates were 84.8% and 51.1% with

erlotinib and chemotherapy, respectively. In whole-exome sequencing analysis,

frequent genes with variants co-occurring at baseline were TP53, MUC16, FAM104B,

KMT5A, and DNAH9. With erlotinib, a single-nucleotide polymorphism mutation in

UBXN11 was associated with significantly worse DFS (P = .01). To our knowledge,

this study is the first to demonstrate clinically meaningful OS improvement with

adjuvant erlotinib compared with chemotherapy in R0 stage III EGFR+

non-small-cell lung cancer.

 

DOI: 10.1200/JCO.22.00428

PMID: 36027483

 

10. J Clin Invest. 2022 Aug 25:e157917. doi: 10.1172/JCI157917. Online ahead of

print.

 

IL20RB mediates tumoral response to osteoclastic niches and promotes bone

metastasis of lung cancer.

 

He Y(1), Luo W(1), Liu Y(1), Wang Y(1), Ma C(1), Wu Q(1), Tian P(1), He D(1),

Jia Z(1), Lv X(1), Ma YS(2), Yang H(3), Xu K(3), Zhang X(1), Xiao Y(1), Zhang

P(1), Liang Y(1), Fu D(2), Yao F(3), Hu G(1).

 

Author information:

(1)CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute

of Nutrition and Health, Shanghai, China.

(2)Department of Nuclear Medicine, Shanghai Tenth People's Hospital, Shanghai,

China.

(3)Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai, China.

 

Bone is a common site of metastasis in lung cancer but the regulatory mechanism

remains incompletely understood. Osteoclasts are known to play crucial roles in

osteolytic bone metastasis by digesting bone matrix and indirectly enhancing

tumor colonization. In this study, we found that IL20RB mediated a direct

tumoral response to osteoclasts. Tumoral expression of IL20RB was associated

with bone metastasis of lung cancer, and functionally IL20RB promoted metastatic

growth of lung cancer cells in bone. Mechanistically, tumor cells induced

osteoclasts to secrete the IL20RB ligand IL19, and IL19 stimulated

IL20RB-expressing tumor cells to activate the downstream JAK1-STAT3 signaling

and enhanced proliferation of tumor cells in bone. Importantly, blocking IL20RB

with a neutralizing antibody significantly suppressed bone metastasis of lung

cancer. Overall, our data revealed a direct pro-tumor role of osteoclastic niche

in bone metastasis and supported IL20RB-targeting approaches for metastasis

treatment.

 

DOI: 10.1172/JCI157917

PMID: 36006737

 

11. Nat Chem Biol. 2022 Aug 22. doi: 10.1038/s41589-022-01102-7. Online ahead of

print.

 

Evybactin is a DNA gyrase inhibitor that selectively kills Mycobacterium

tuberculosis.

 

Imai Y(#)(1)(2), Hauk G(#)(3), Quigley J(1), Liang L(1), Son S(1), Ghiglieri

M(1), Gates MF(1), Morrissette M(1), Shahsavari N(1), Niles S(1), Baldisseri

D(4), Honrao C(5), Ma X(5), Guo JJ(5)(6), Berger JM(7), Lewis K(8).

 

Author information:

(1)Antimicrobial Discovery Center, Department of Biology, Northeastern

University, Boston, MA, USA.

(2)Department of Biomolecular Innovation, Institute for Biomedical Sciences,

Shinshu University, Nagano, Japan.

(3)Department of Biophysics and Biophysical Chemistry, Johns Hopkins University

School of Medicine, Baltimore, MD, USA.

The antimicrobial resistance crisis requires the introduction of novel

antibiotics. The use of conventional broad-spectrum compounds selects for

resistance in off-target pathogens and harms the microbiome. This is especially

true for Mycobacterium tuberculosis, where treatment requires a 6-month course

of antibiotics. Here we show that a novel antimicrobial from Photorhabdus

noenieputensis, which we named evybactin, is a potent and selective antibiotic

acting against M. tuberculosis. Evybactin targets DNA gyrase and binds to a site

overlapping with synthetic thiophene poisons. Given the conserved nature of DNA

gyrase, the observed selectivity against M. tuberculosis is puzzling. We found

that evybactin is smuggled into the cell by a promiscuous transporter of

hydrophilic compounds, BacA. Evybactin is the first, but likely not the only,

antimicrobial compound found to employ this unusual mechanism of selectivity.

 

© 2022. The Author(s), under exclusive licence to Springer Nature America, Inc.

 

DOI: 10.1038/s41589-022-01102-7

PMID: 35996001

 

12. J Natl Cancer Inst. 2022 Aug 22:djac154. doi: 10.1093/jnci/djac154. Online ahead of print.

 

Facilitators and Barriers to Implementation of Lung Cancer Screening: A

Framework Driven Systematic Review.

 

Sedani AE(1), Davis OC(2), Clifton SC(3), Campbell JE(1), Chou AF(4).

 

Author information:

(1)Department of Biostatistics and Epidemiology, Hudson College of Public

Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.

(2)College of Medicine, University of Oklahoma Health Sciences Center, Oklahoma

City, OK, USA.

(3)Robert M. Bird Health Sciences Library, University of Oklahoma Health

Sciences Center, Oklahoma City, OK, USA.

(4)Department of Family and Preventive Medicine, College of Medicine, University

of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.

 

BACKGROUND: The purpose of this study is to undertake a comprehensive systematic

review to describe multi-level factors (barriers and facilitators) that may

influence the implementation of low-dose chest computed tomography (LDCT) for

lung cancer screening in the United States.

METHODS: Systematic literature searches were performed using six online

databases and citation indexes for peer-reviewed studies, for articles published

from 2013-2021. Studies were classified into three perspectives, based on the

study's unit of analysis: system, healthcare provider, and patient. Barriers and

facilitators identified for each study included in our final review were then

coded and categorized using the Consolidate Framework for Implementation

Research (CFIR) domains.

RESULTS: At the system level, the two most common constructs were external

policy and incentives, and executing the implementation process. At the provider

level, the most common constructs were evidence strength and quality of the

intervention characteristics, patient needs and resources, implementation

climate, and individual's knowledge and beliefs about the intervention. At the

patient-level, the most common constructs were patient needs and resources,

individual's knowledge and beliefs about the intervention, and engaging in the

implementation process. These constructs can act both as facilitators or

barriers to lung cancer screening implementation.

CONCLUSIONS: Applying the CFIR domains and constructs to understand and specify

factors facilitating uptake of lung cancer screening, as well as cataloguing the

lessons learned from previous efforts, help to inform the development and

implementation processes of lung cancer screening programs in the community

setting.

REGISTRATION: PROSPERO, CRD42021247677.

 

© The Author(s) 2022. Published by Oxford University Press.

 

DOI: 10.1093/jnci/djac154

PMID: 35993616

 

13. Lancet Infect Dis. 2022 Aug 10:S1473-3099(22)00500-X. doi:

10.1016/S1473-3099(22)00500-X. Online ahead of print.

 

Ending tuberculosis in a post-COVID-19 world: a person-centred, equity-oriented

approach.

 

Ryckman T(1), Robsky K(2), Cilloni L(3), Zawedde-Muyanja S(4), Ananthakrishnan

R(5), Kendall EA(6), Shrestha S(3), Turyahabwe S(7), Katamba A(8), Dowdy DW(9).

 

Author information:

(1)Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health,

Baltimore, MD, USA. Electronic address: tryckma1@jh.edu.

(2)Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health,

Baltimore, MD, USA; Uganda Tuberculosis Implementation Research Consortium,

Kampala, Uganda.

(3)Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health,

Baltimore, MD, USA.

The COVID-19 pandemic has disrupted systems of care for infectious

diseases-including tuberculosis-and has exposed pervasive inequities that have

long marred efforts to combat these diseases. The resulting health disparities

often intersect at the individual and community levels in ways that heighten

vulnerability to tuberculosis. Effective responses to tuberculosis (and other

infectious diseases) must respond to these realities. Unfortunately, current

tuberculosis programmes are generally not designed from the perspectives of

affected individuals and fail to address structural determinants of health

disparities. We describe a person-centred, equity-oriented response that would

identify and focus on communities affected by disparities, tailor interventions

to the mechanisms by which disparities worsen tuberculosis, and address upstream

determinants of those disparities. We detail four key elements of the approach

(data collection, programme design, implementation, and sustainability). We then

illustrate how organisations at multiple levels might partner and adapt current

practices to incorporate these elements. Such an approach could generate more

substantial, sustainable, and equitable reductions in tuberculosis burden at the

community level, highlighting the urgency of restructuring post-COVID-19 health

systems in a more person-centred, equity-oriented way.

 

Copyright © 2022 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(22)00500-X

PMCID: PMC9365311

PMID: 35963272

 

14. Nat Commun. 2022 Aug 12;13(1):4731. doi: 10.1038/s41467-022-32085-7.

 

Mycobacterial resistance to zinc poisoning requires assembly of

P-ATPase-containing membrane metal efflux platforms.

 

Boudehen YM(#)(1)(2), Faucher M(#)(1), Maréchal X(3)(4), Miras R(3), Rech J(5),

Rombouts Y(1), Sénèque O(3), Wallat M(1)(6), Demange P(1), Bouet JY(5), Saurel

O(1), Catty P(3), Gutierrez C(1), Neyrolles O(7).

 

Author information:

(1)Institut de Pharmacologie et de Biologie Structurale, IPBS, Université de

Toulouse, CNRS, UPS, Toulouse, France.

(2)Centre National de la Recherche Scientifique UMR 9004, Institut de Recherche

en Infectiologie de Montpellier (IRIM), Université de Montpellier, Montpellier,

France.

(3)University Grenoble Alpes, CNRS, CEA, IRIG, Laboratoire de Chimie et Biologie

des Métaux, 38054, Grenoble, France.

The human pathogen Mycobacterium tuberculosis requires a P1B-ATPase metal

exporter, CtpC (Rv3270), for resistance to zinc poisoning. Here, we show that

zinc resistance also depends on a chaperone-like protein, PacL1 (Rv3269). PacL1

contains a transmembrane domain, a cytoplasmic region with glutamine/alanine

repeats and a C-terminal metal-binding motif (MBM). PacL1 binds Zn2+, but the

MBM is required only at high zinc concentrations. PacL1 co-localizes with CtpC

in dynamic foci in the mycobacterial plasma membrane, and the two proteins form

high molecular weight complexes. Foci formation does not require flotillin nor

the PacL1 MBM. However, deletion of the PacL1 Glu/Ala repeats leads to loss of

CtpC and sensitivity to zinc. Genes pacL1 and ctpC appear to be in the same

operon, and homologous gene pairs are found in the genomes of other bacteria.

Furthermore, PacL1 colocalizes and functions redundantly with other PacL

orthologs in M. tuberculosis. Overall, our results indicate that PacL proteins

may act as scaffolds that assemble P-ATPase-containing metal efflux platforms

mediating bacterial resistance to metal poisoning.

 

© 2022. The Author(s).

 

DOI: 10.1038/s41467-022-32085-7

PMCID: PMC9374683

PMID: 35961955 [Indexed for MEDLINE]

 

15. J Clin Oncol. 2022 Aug 12:JCO2102911. doi: 10.1200/JCO.21.02911. Online ahead of print.

 

Updated Analysis of NEJ009: Gefitinib-Alone Versus Gefitinib Plus Chemotherapy

for Non-Small-Cell Lung Cancer With Mutated EGFR.

 

Miyauchi E(1), Morita S(2), Nakamura A(3), Hosomi Y(4), Watanabe K(5), Ikeda

S(6), Seike M(7), Fujita Y(8), Minato K(9), Ko R(10), Harada T(11), Hagiwara

K(12), Kobayashi K(13), Nukiwa T(14), Inoue A(15); North-East Japan Study Group.

 

Author information:

(1)Department of Respiratory Medicine, Tohoku University Hospital, Sendai,

Japan.

(2)Department of Biomedical Statistics and Bioinformatics, Kyoto University

Graduate School of Medicine, Kyoto, Japan.

(3)Department of Respiratory Medicine, Sendai Kosei Hospital, Sendai, Japan.

Clinical trials frequently include multiple end points that mature at different

times. The initial report, typically based on the primary end point, may be

published when key planned coprimary or secondary analyses are not yet

available. Clinical Trial Updates provide an opportunity to disseminate

additional results from studies, published in JCO or elsewhere, for which the

primary end point has already been reported.In a randomized, open-label, phase

III NEJ009 study, gefitinib plus chemotherapy significantly improved

progression-free survival (PFS) and overall survival (OS) compared with

gefitinib-alone in patients with untreated non-small-cell lung cancer harboring

mutations in epidermal growth factor receptor. Herein, we report the updated

survival outcome and long-term tolerability. Patients were randomly assigned to

gefitinib (gefitinib 250 mg orally, once daily) and gefitinib combined with

carboplatin plus pemetrexed (GCP in a 3-week cycle for six cycles followed by

concurrent gefitinib and pemetrexed maintenance) groups. At the data cutoff (May

22, 2020), GCP demonstrated significantly better PFS2 (hazard ratio, 0.77; 95%

CI, 0.62 to 0.97; P = .027) than gefitinib. However, the updated median OS was

38.5 months (95% CI, 31.1 to 47.1) and 49.0 months (95% CI, 41.8 to 56.7) in the

gefitinib and GCP groups, respectively (hazard ratio, 0.82; 95% CI, 0.64 to

1.06; P = .127). The OS in both groups was similar for the overall patient

population. No severe adverse events occurred since the first report. This

updated analysis revealed that the GCP regimen improved PFS and PFS2 with an

acceptable safety profile compared with gefitinib-alone. GCP is more efficient

than gefitinib monotherapy as a first-line treatment for non-small-cell lung

cancer with epidermal growth factor receptor mutations.

 

DOI: 10.1200/JCO.21.02911

PMID: 35960896

 

16. Am J Respir Crit Care Med. 2022 Aug 11. doi: 10.1164/rccm.202201-0144OC. Online ahead of print.

 

Forgiveness Is the Attribute of the Strong: Nonadherence and Regimen-Shortening

in Drug-Sensitive TB.

 

Stagg HR(1), Thompson JA(2), Lipman MC(3), Sloan DJ(4), Flook M(5), Fielding

KL(2); Critical Path to TB Drug Regimens.

 

Author information:

(1)University of Edinburgh, Usher Institute of Population Health Sciences and

Informatics, Edinburgh, United Kingdom of Great Britain and Northern Ireland;

helen.stagg@ed.ac.uk.

(2)London School of Hygiene and Tropical Medicine, London, United Kingdom of

Great Britain and Northern Ireland.

(3)University College London, Respiratory Medicine, London, United Kingdom of

Great Britain and Northern Ireland.

(4)University of St Andrews, Medicine, St Andrews, United Kingdom of Great

Britain and Northern Ireland.

(5)University of Edinburgh, Usher Institute of Population Health Sciences and

Informatics, Edinburgh, United Kingdom of Great Britain and Northern Ireland.

 

RATIONALE: 'Forgiveness' charts the ability of a drug or regimen to withstand

non-adherence without negative clinical consequences.

OBJECTIVES: We aimed to determine the influence of regimen length, regimen drugs

and dosing, and when during treatment non-adherence occurs on the forgiveness of

anti-tuberculosis regimens.

METHODS: Using data from three randomised controlled trials comparing

experimental four-month regimens for drug-sensitive tuberculosis with the

standard six-month regimen, we used generalised linear models to examine how the

risk of a negative composite outcome changed as dose-taking decreased. The

percentage of doses taken and absolute number of doses missed were calculated,

during the intensive and continuation phases of treatment, and overall. A

mediation analysis was undertaken to determine how much of the association

between intensive phase dose-taking and the negative composite outcome was

mediated through continuation phase dose-taking.

MEASUREMENTS AND MAIN RESULTS: Forgiveness of the four-month and six-month

regimens did not differ for any treatment period. Importantly, four-month

regimens were no less forgiving of small numbers of absolute missed doses than

the six-month regimen (e.g. for 3-7 missed doses versus no missed doses

(baseline), six-month regimen adjusted risk ratio 1.65 (95% confidence interval

0.80-3.41) and four-month regimens 1.80 (1.33-2.45)). No four-month regimen was

conclusively more forgiving than another. We found evidence of mediation by

continuation phase dose-taking on the intensive phase dose-taking and negative

composite outcome relationship.

CONCLUSIONS: With the current appetite for, and progress towards, shorter

drug-sensitive tuberculosis regimens worldwide, we offer reassurance that

shorter regimens are not necessarily less forgiving of non-adherence. Given the

importance of continuation phase adherence, patient support during this period

should not be neglected.

 

DOI: 10.1164/rccm.202201-0144OC

PMID: 35952354

 

17. PLoS Biol. 2022 Aug 9;20(8):e3001755. doi: 10.1371/journal.pbio.3001755.

eCollection 2022 Aug.

 

Genome-wide association studies of global Mycobacterium tuberculosis resistance

to 13 antimicrobials in 10,228 genomes identify new resistance mechanisms.

 

The CRyPTIC Consortium.

 

Collaborators: Earle SG, Wilson DJ, Barilar I, Battaglia S, Borroni E, Brandao

AP, Brankin A, Cabibbe AM, Carter J, Chetty D, Cirillo DM, Claxton P, Clifton

DA, …

Comment in

    A data compendium associating the genomes of 12,289 Mycobacterium

tuberculosis isolates with quantitative resistance phenotypes to 13 antibiotics.

 

The emergence of drug-resistant tuberculosis is a major global public health

concern that threatens the ability to control the disease. Whole-genome

sequencing as a tool to rapidly diagnose resistant infections can transform

patient treatment and clinical practice. While resistance mechanisms are well

understood for some drugs, there are likely many mechanisms yet to be uncovered,

particularly for new and repurposed drugs. We sequenced 10,228 Mycobacterium

tuberculosis (MTB) isolates worldwide and determined the minimum inhibitory

concentration (MIC) on a grid of 2-fold concentration dilutions for 13

antimicrobials using quantitative microtiter plate assays. We performed

oligopeptide- and oligonucleotide-based genome-wide association studies using

linear mixed models to discover resistance-conferring mechanisms not currently

catalogued. Use of MIC over binary resistance phenotypes increased sample

heritability for the new and repurposed drugs by 26% to 37%, increasing our

ability to detect novel associations. For all drugs, we discovered uncatalogued

variants associated with MIC, including in the Rv1218c promoter binding site of

the transcriptional repressor Rv1219c (isoniazid), upstream of the vapBC20

operon that cleaves 23S rRNA (linezolid) and in the region encoding an α-helix

lining the active site of Cyp142 (clofazimine, all p < 10-7.7). We observed that

artefactual signals of cross-resistance could be unravelled based on the

relative effect size on MIC. Our study demonstrates the ability of very

large-scale studies to substantially improve our knowledge of genetic variants

associated with antimicrobial resistance in M. tuberculosis.

 

DOI: 10.1371/journal.pbio.3001755

PMCID: PMC9363015

PMID: 35944070 [Indexed for MEDLINE]

 

18. PLoS Biol. 2022 Aug 9;20(8):e3001721. doi: 10.1371/journal.pbio.3001721.

eCollection 2022 Aug.

 

A data compendium associating the genomes of 12,289 Mycobacterium tuberculosis

isolates with quantitative resistance phenotypes to 13 antibiotics.

 

The CRyPTIC Consortium.

 

Collaborators: Brankin A, Malone KM, Barilar I, Battaglia S, Borroni E, Brandao

AP, Cabibbe AM, Carter J, …

Comment in

    Genome-wide association studies of global Mycobacterium tuberculosis

resistance to thirteen antimicrobials in 10,228 genomes identify new resistance

mechanisms.

 

The Comprehensive Resistance Prediction for Tuberculosis: an International

Consortium (CRyPTIC) presents here a data compendium of 12,289 Mycobacterium

tuberculosis global clinical isolates, all of which have undergone whole-genome

sequencing and have had their minimum inhibitory concentrations to 13

antitubercular drugs measured in a single assay. It is the largest matched

phenotypic and genotypic dataset for M. tuberculosis to date. Here, we provide a

summary detailing the breadth of data collected, along with a description of how

the isolates were selected, collected, and uniformly processed in CRyPTIC

partner laboratories across 23 countries. The compendium contains 6,814 isolates

resistant to at least 1 drug, including 2,129 samples that fully satisfy the

clinical definitions of rifampicin resistant (RR), multidrug resistant (MDR),

pre-extensively drug resistant (pre-XDR), or extensively drug resistant (XDR).

The data are enriched for rare resistance-associated variants, and the current

limits of genotypic prediction of resistance status (sensitive/resistant) are

presented by using a genetic mutation catalogue, along with the presence of

suspected resistance-conferring mutations for isolates resistant to the newly

introduced drugs bedaquiline, clofazimine, delamanid, and linezolid. Finally, a

case study of rifampicin monoresistance demonstrates how this compendium could

be used to advance our genetic understanding of rare resistance phenotypes. The

data compendium is fully open source and it is hoped that it will facilitate and

inspire future research for years to come.

 

DOI: 10.1371/journal.pbio.3001721

PMCID: PMC9363010

PMID: 35944069 [Indexed for MEDLINE]

 

19. Am J Respir Crit Care Med. 2022 Aug 9. doi: 10.1164/rccm.202112-2758OC. Online ahead of print.

 

In Utero and Childhood/Adolescence Exposure to Tobacco Smoke, Genetic Risk and

Lung Cancer Incidence and Mortality in Adulthood.

 

He H(1), He MM(2), Wang H(3), Qiu W(3), Liu L(4), Long L(5), Shen Q(3), Zhang

S(3), Qin S(3), Lu Z(3), Cai Y(3), Zhang M(3), Niu S(3), Li J(6), Shen N(4), Zhu

Y(7), Tian J(7), Chang J(3), Miao X(7), Zhong R(8).

 

Author information:

(1)Huazhong University of Science and Technology, School of Public Health,

Tongji Medical College, Wuhan, Hubei , China.

(2)Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China.

(3)Huazhong University of Science and Technology, Wuhan, Hubei , China.

RATIONALE: The individual effects of early-life tobacco smoke exposure and its

interactions with genetic factors on lung cancer in adulthood remain unclear.

OBJECTIVES: To investigate the associations between early-life tobacco exposures

as well as their interactions with polygenic risk scores (PRSs) and lung cancer

incidence and mortality.

METHODS: A total of 432,831 participants from the UK Biobank study were

included. We estimated the associations of in utero exposure to tobacco smoke,

the age of smoking initiation and their interactions with PRSs with lung cancer

incidence and mortality in adulthood using Cox proportional hazard models.

MEASUREMENTS AND MAIN RESULTS: Lung cancer incidence [hazard ratio (HR): 1.59,

95% confidence interval (CI), 1.44-1.76] increased among participants with in

utero tobacco exposure. Multivariable-adjusted HRs (with 95% CIs) of lung cancer

incidence for smoking initiation in adulthood, adolescence, and childhood (vs.

never smokers) were 6.10 (5.25-7.09), 9.56 (8.31-11.00), and 15.15 (12.90-17.79)

(Ptrend<0.001). Similar findings were observed in lung cancer mortality.

Participants with high PRSs and in utero tobacco exposure (vs. low PRSs

participants without in utero exposure) had HR of 2.35 for lung cancer incidence

(95%CI, 1.97-2.80, Pinteraction=0.089) and 2.43 for mortality (95% CI,

2.05-2.88, Pinteraction=0.032). High PRSs with smoking initiation in childhood

(vs. never smokers with low PRSs) had HR of 18.71 for incidence (95% CI,

14.21-24.63, Pinteraction=0.004) and 19.74 for mortality (95%CI, 14.98-26.01,

Pinteraction=0.033).

CONCLUSIONS: In utero and childhood/adolescence exposure to tobacco smoke and

its interaction with genetic factors may substantially increase the risks of

lung cancer incidence and mortality in adulthood.

 

DOI: 10.1164/rccm.202112-2758OC

PMID: 35943859

 

20. Nat Commun. 2022 Aug 8;13(1):4455. doi: 10.1038/s41467-022-32043-3.

 

A long-acting formulation of rifabutin is effective for prevention and treatment

of Mycobacterium tuberculosis.

 

Kim M(#)(1)(2)(3), Johnson CE(#)(1)(2)(3), Schmalstig AA(4), Annis A(4), Wessel

SE(1)(2)(3), Van Horn B(5), Schauer A(5), Exner AA(6), Stout JE(7), Wahl

A(1)(2)(3), Braunstein M(4), Victor Garcia J(8)(9)(10), Kovarova M(11)(12)(13).

 

Author information:

(1)International Center for the Advancement of Translational Science, University

of North Carolina at Chapel Hill, Chapel Hill, NC, USA.

(2)Division of Infectious Diseases, Department of Medicine, University of North

Carolina at Chapel Hill, Chapel Hill, NC, USA.

(3)Center for AIDS Research, University of North Carolina at Chapel Hill, Chapel

Hill, NC, USA.

Tuberculosis (TB) is a communicable disease caused by Mycobacterium tuberculosis

(Mtb) and is a major cause of morbidity and mortality. Successful treatment

requires strict adherence to drug regimens for prolonged periods of time.

Long-acting (LA) delivery systems have the potential to improve adherence. Here,

we show the development of LA injectable drug formulations of the anti-TB drug

rifabutin made of biodegradable polymers and biocompatible solvents that

solidifies after subcutaneous injection. Addition of amphiphilic compounds

increases drug solubility, allowing to significantly increase formulation drug

load. Solidified implants have organized microstructures that change with

formulation composition. Higher drug load results in smaller pore size that

alters implant erosion and allows sustained drug release. The translational

relevance of these observations in BALB/c mice is demonstrated by (1) delivering

high plasma drug concentrations for 16 weeks, (2) preventing acquisition of Mtb

infection, and (3) clearing acute Mtb infection from the lung and other tissues.

 

© 2022. The Author(s).

 

DOI: 10.1038/s41467-022-32043-3

PMCID: PMC9360445

PMID: 35941109 [Indexed for MEDLINE]

 

21. Lancet Infect Dis. 2022 Aug 4:S1473-3099(22)00425-X. doi:

10.1016/S1473-3099(22)00425-X. Online ahead of print.

 

Nationwide tuberculosis outbreak in the USA linked to a bone graft product: an

outbreak report.

 

Schwartz NG(1), Hernandez-Romieu AC(2), Annambhotla P(3), Filardo TD(4),

Althomsons SP(5), Free RJ(3), Li R(2), Wyatt Wilson W(2), Deutsch-Feldman M(4),

Drees M(6), Hanlin E(7), White K(8), Lehman KA(9), Thacker TC(9), Brubaker

SA(10), Clark B(10), Basavaraju SV(3), Benowitz I(3), Burton Glowicz J(3), Cowan

LS(5), Starks AM(5), Bamrah Morris S(5), LoBue P(5), Stewart RJ(5), Wortham

JM(5), Haddad MB(5); Bone Allograft Tuberculosis Investigators.

 

Author information:

(1)Division of Tuberculosis Elimination, National Center for HIV, Viral

Hepatitis, STD, and TB Prevention, US Centers for Disease Control and

Prevention, Atlanta, GA, USA; Epidemic Intelligence Service, US Centers for

Disease Control and Prevention, Atlanta, GA, USA. Electronic address:

nschwartz@cdc.gov.

(2)Epidemic Intelligence Service, US Centers for Disease Control and Prevention,

Atlanta, GA, USA; Division of Healthcare Quality Promotion, National Center for

Emerging and Zoonotic Infectious Diseases, US Centers for Disease Control and

Prevention, Atlanta, GA, USA.

(3)Division of Healthcare Quality Promotion, National Center for Emerging and

Zoonotic Infectious Diseases, US Centers for Disease Control and Prevention,

Atlanta, GA, USA.

BACKGROUND: Mycobacterium tuberculosis transmission through solid organ

transplantation has been well described, but transmission through transplanted

tissues is rare. We investigated a tuberculosis outbreak in the USA linked to a

bone graft product containing live cells derived from a single deceased donor.

METHODS: In this outbreak report, we describe the management and severity of the

outbreak and identify opportunities to improve tissue transplant safety in the

USA. During early June, 2021, the US Centers for Disease Control and Prevention

(CDC) worked with state and local health departments and health-care facilities

to locate and sequester unused units from the recalled lot and notify, evaluate,

and treat all identified product recipients. Investigators from CDC and the US

Food and Drug Administration (FDA) reviewed donor screening and tissue

processing. Unused product units from the recalled and other donor lots were

tested for the presence of M tuberculosis using real-time PCR (rt PCR) assays

and culture. M tuberculosis isolates from unused product and recipients were

compared using phylogenetic analysis.

FINDINGS: The tissue donor (a man aged 80 years) had unrecognised risk factors,

symptoms, and signs consistent with tuberculosis. Bone was procured from the

deceased donor and processed into 154 units of bone allograft product containing

live cells, which were distributed to 37 hospitals and ambulatory surgical

centres in 20 US states between March 1 and April 2, 2021. From March 3 to June

1, 2021, 136 (88%) units were implanted into 113 recipients aged 24-87 years in

18 states (some individuals received multiple units). The remaining 18 units

(12%) were located and sequestered. 87 (77%) of 113 identified product

recipients had microbiological or imaging evidence of tuberculosis disease.

Eight product recipients died 8-99 days after product implantation (three deaths

were attributed to tuberculosis after recognition of the outbreak). All 105

living recipients started treatment for tuberculosis disease at a median of 69

days (IQR 56-81) after product implantation. M tuberculosis was detected in all

eight sequestered unused units tested from the recalled donor lot, but not in

lots from other donors. M tuberculosis isolates from unused product and

recipients were more than 99·99% genetically identical.

INTERPRETATION: Donor-derived transmission of M tuberculosis via bone allograft

resulted in substantial morbidity and mortality. All prospective tissue and

organ donors should be routinely assessed for tuberculosis risk factors and

clinical findings. When these are present, laboratory testing for M tuberculosis

should be strongly considered.

FUNDING: None.

 

Copyright © 2022 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(22)00425-X

PMID: 35934016

 

22. Immunol Rev. 2022 Aug;309(1):97-122. doi: 10.1111/imr.13116. Epub 2022 Jul 12.

 

Transcriptomics for child and adolescent tuberculosis.

 

Kaforou M(1), Broderick C(1), Vito O(1), Levin M(1), Scriba TJ(2), Seddon

JA(1)(3).

 

Author information:

(1)Department of Infectious Disease, Imperial College London, London, UK.

(2)South African Tuberculosis Vaccine Initiative, Institute of Infectious

Disease and Molecular Medicine and Division of Immunology, Department of

Pathology, University of Cape Town, Cape Town, South Africa.

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

Stellenbosch University, Cape Town, South Africa.

 

Tuberculosis (TB) in humans is caused by Mycobacterium tuberculosis (Mtb). It is

estimated that 70 million children (<15 years) are currently infected with Mtb,

with 1.2 million each year progressing to disease. Of these, a quarter die. The

risk of progression from Mtb infection to disease and from disease to death is

dependent on multiple pathogen and host factors. Age is a central component in

all these transitions. The natural history of TB in children and adolescents is

different to adults, leading to unique challenges in the development of

diagnostics, therapeutics, and vaccines. The quantification of RNA

transcripts in specific cells or in the peripheral blood, using high-throughput

methods, such as microarray analysis or RNA-Sequencing, can shed light into the

host immune response to Mtb during infection and disease, as well as

understanding treatment response, disease severity, and vaccination, in a global

hypothesis-free manner. Additionally, gene expression profiling can be used for

biomarker discovery, to diagnose disease, predict future disease progression and

to monitor response to treatment. Here, we review the role of transcriptomics in

children and adolescents, focused mainly on work done in blood, to understand

disease biology, and to discriminate disease states to assist clinical

decision-making. In recent years, studies with a specific pediatric and

adolescent focus have identified blood gene expression markers with diagnostic

or prognostic potential that meet or exceed the current sensitivity and

specificity targets for diagnostic tools. Diagnostic and prognostic gene

expression signatures identified through high-throughput methods are currently

being translated into diagnostic tests.

 

© 2022 The Authors. Immunological Reviews published by John Wiley & Sons Ltd.

 

DOI: 10.1111/imr.13116

PMID: 35818983 [Indexed for MEDLINE]

 

23. Lancet Infect Dis. 2022 Aug;22(8):1172-1180. doi: 10.1016/S1473-3099(22)00149-9. Epub 2022 May 17.

 

Prevalence of bacteriologically confirmed pulmonary tuberculosis in South

Africa, 2017-19: a multistage, cluster-based, cross-sectional survey.

 

Moyo S(1), Ismail F(2), Van der Walt M(3), Ismail N(4), Mkhondo N(5), Dlamini

S(6), Mthiyane T(3), Chikovore J(7), Oladimeji O(7), Mametja D(8), Maribe P(3),

Seocharan I(3), Ximiya P(6), Law I(4), Tadolini M(9), Zuma K(7), Manda S(3),

Sismanidis C(4), Pillay Y(10), Mvusi L(6).

 

Author information:

(1)Human Sciences Research Council, Cape Town, South Africa; School of Public

Health and Family Medicine, University of Cape Town, Cape Town, South Africa.

Electronic address: smoyo@hsrc.ac.za.

(2)Centre for Tuberculosis, National Institute for Communicable Diseases,

Johannesburg, South Africa; Department of Medical Microbiology, University of

Pretoria, Pretoria, South Africa.

(3)South African Medical Research Council, Cape Town, South Africa.

Erratum in

    Lancet Infect Dis. 2022 Jul;22(7):e177.

 

Comment in

    Lancet Infect Dis. 2022 Aug;22(8):1094-1096.

    Lancet Infect Dis. 2022 Sep;22(9):1273.

 

BACKGROUND: Tuberculosis remains an important clinical and public health issue

in South Africa, which has one of the highest tuberculosis burdens in the world.

We aimed to estimate the burden of bacteriologically confirmed pulmonary

tuberculosis among people aged 15 years or older in South Africa.

METHODS: This multistage, cluster-based, cross-sectional survey included

eligible residents (age ≥15 years, who had slept in a house for ≥10 nights in

the preceding 2 weeks) in 110 clusters nationally (cluster size of 500 people;

selected by probability proportional-to-population size sampling). Participants

completed face-to-face symptom questionnaires (for cough, weight loss, fever,

and night sweats) and manually read digital chest X-ray screening. Screening was

recorded as positive if participants had at least one symptom or an abnormal

chest X-ray suggestive of tuberculosis, or a combination thereof. Sputum samples

from participants who were screen-positive were tested by the Xpert MTB/RIF

Ultra assay (first sample) and Mycobacteria Growth Indicator Tube culture

(second sample), with optional HIV testing. Participants with a positive

Mycobacterium tuberculosis complex culture were considered positive for

bacteriologically confirmed pulmonary tuberculosis; when culture was not

positive, participants with a positive Xpert MTB/RIF Ultra result with an

abnormal chest X-ray suggestive of active tuberculosis and without current or

previous tuberculosis were considered positive for bacteriologically confirmed

pulmonary tuberculosis.

FINDINGS: Between Aug 15, 2017, and July 28, 2019, 68 771 people were enumerated

from 110 clusters, with 53 250 eligible to participate in the survey, of whom

35 191 (66·1%) participated. 9066 (25·8%) of 35 191 participants were

screen-positive and 234 (0·7%) were identified as having bacteriologically

confirmed pulmonary tuberculosis. Overall, the estimated prevalence of

bacteriologically confirmed pulmonary tuberculosis was 852 cases (95% CI

679-1026) per 100 000 population; the prevalence was highest in people aged

35-44 years (1107 cases [95% CI 703-1511] per 100 000 population) and those aged

65 years or older (1104 cases [680-1528] per 100 000 population). The estimated

prevalence was approximately 1·6 times higher in men than in women (1094 cases

[95% CI 835-1352] per 100 000 population vs 675 cases [494-855] per 100 000

population). 135 (57·7%) of 234 participants with tuberculosis screened positive

by chest X-ray only, 16 (6·8%) by symptoms only, and 82 (35·9%) by both. 55

(28·8%) of 191 participants with tuberculosis with known HIV status were

HIV-positive.

INTERPRETATION: Pulmonary tuberculosis prevalence in this survey was high,

especially in men. Despite the ongoing burden of HIV, many participants with

tuberculosis in this survey did not have HIV. As more than half of the

participants with tuberculosis had an abnormal chest X-ray without symptoms,

prioritising chest X-ray screening could substantially increase case finding.

FUNDING: Global Fund, Bill & Melinda Gates Foundation, USAID.

 

World Health Organization; licensee Elsevier. This is an Open Access article

published under the CC BY 3.0 IGO license which permits unrestricted use,

distribution, and reproduction in any medium, provided the original work is

properly cited. In any use of this article, there should be no suggestion that

WHO endorses any specific organisation, products or services. The use of the WHO

logo is not permitted. This notice should be preserved along with the article's

original URL.

 

DOI: 10.1016/S1473-3099(22)00149-9

PMCID: PMC9300471

PMID: 35594897 [Indexed for MEDLINE]

 

24. J Clin Oncol. 2022 Aug 1;40(22):2491-2507. doi: 10.1200/JCO.22.00687. Epub 2022 May 16.

 

Exercise, Diet, and Weight Management During Cancer Treatment: ASCO Guideline.

 

Ligibel JA(1), Bohlke K(2), May AM(3), Clinton SK(4), Demark-Wahnefried W(5),

Gilchrist SC(6), Irwin ML(7), Late M(8), Mansfield S(9), Marshall TF(10),

Meyerhardt JA(1), Thomson CA(11), Wood WA(12), Alfano CM(13).

 

Author information:

(1)Dana-Farber Cancer Institute, Boston, MA.

(2)American Society of Clinical Oncology, Alexandria, VA.

(3)University Medical Center Utrecht, Utrecht University, Utrecht, the

Netherlands.

PURPOSE: To provide guidance on exercise, diet, and weight management during

active cancer treatment in adults.

METHODS: A systematic review of the literature identified systematic reviews and

randomized controlled trials evaluating the impact of aerobic and resistance

exercise, specific diets and foods, and intentional weight loss and avoidance of

weight gain in adults during cancer treatment, on quality of life, treatment

toxicity, and cancer control. PubMed and the Cochrane Library were searched from

January 2000 to May 2021. ASCO convened an Expert Panel to review the evidence

and formulate recommendations.

RESULTS: The evidence base consisted of 52 systematic reviews (42 for exercise,

nine for diet, and one for weight management), and an additional 23 randomized

controlled trials. The most commonly studied types of cancer were breast,

prostate, lung, and colorectal. Exercise during cancer treatment led to

improvements in cardiorespiratory fitness, strength, fatigue, and other

patient-reported outcomes. Preoperative exercise in patients with lung cancer

led to a reduction in postoperative length of hospital stay and complications.

Neutropenic diets did not decrease risk of infection during cancer treatment.

RECOMMENDATIONS: Oncology providers should recommend regular aerobic and

resistance exercise during active treatment with curative intent and may

recommend preoperative exercise for patients undergoing surgery for lung cancer.

Neutropenic diets are not recommended to prevent infection in patients with

cancer during active treatment. Evidence for other dietary and weight loss

interventions during cancer treatment was very limited. The guideline discusses

special considerations, such as exercise in individuals with advanced cancer,

and highlights the critical need for more research in this area, particularly

regarding diet and weight loss interventions during cancer treatment.Additional

information is available at www.asco.org/supportive-care-guidelines.

 

DOI: 10.1200/JCO.22.00687

PMID: 35576506 [Indexed for MEDLINE]

 

25. Nat Rev Clin Oncol. 2022 Aug;19(8):499-514. doi: 10.1038/s41571-022-00639-9.

Epub 2022 May 9.

 

Third-generation EGFR and ALK inhibitors: mechanisms of resistance and

management.

 

Cooper AJ(1), Sequist LV(1), Lin JJ(2).

 

Author information:

(1)Department of Medicine, Massachusetts General Hospital Cancer Center, Boston,

MA, USA.

(2)Department of Medicine, Massachusetts General Hospital Cancer Center, Boston,

MA, USA. jjlin1@partners.org.

 

Erratum in

    Nat Rev Clin Oncol. 2022 Aug 15;:

 

The discoveries of EGFR mutations and ALK rearrangements as actionable oncogenic

drivers in non-small-cell lung cancer (NSCLC) has propelled a biomarker-directed

treatment paradigm for patients with advanced-stage disease. Numerous EGFR and

ALK tyrosine kinase inhibitors (TKIs) with demonstrated efficacy in patients

with EGFR-mutant and ALK-rearranged NSCLCs have been developed, culminating in

the availability of the highly effective third-generation TKIs osimertinib and

lorlatinib, respectively. Despite their marked efficacy, resistance to these

agents remains an unsolved fundamental challenge. Both 'on-target' mechanisms

(largely mediated by acquired resistance mutations in the kinase domains of EGFR

or ALK) and 'off-target' mechanisms of resistance (mediated by non-target kinase

alterations such as bypass signalling activation or phenotypic transformation)

have been identified in patients with disease progression on osimertinib or

lorlatinib. A growing understanding of the biology and spectrum of these

mechanisms of resistance has already begun to inform the development of more

effective therapeutic strategies. In this Review, we discuss the development of

third-generation EGFR and ALK inhibitors, predominant mechanisms of resistance,

and approaches to tackling resistance in the clinic, ranging from novel

fourth-generation TKIs to combination regimens and other investigational

therapies.

 

© 2022. Springer Nature Limited.

 

DOI: 10.1038/s41571-022-00639-9

PMID: 35534623 [Indexed for MEDLINE]

 

26. J Natl Cancer Inst. 2022 Aug 8;114(8):1159-1166. doi: 10.1093/jnci/djac087.

 

Genetic Analysis of Lung Cancer and the Germline Impact on Somatic Mutation

Burden.

 

Gabriel AAG(1), Atkins JR(1), Penha RCC(1), Smith-Byrne K(1)(2), Gaborieau V(1),

Voegele C(1), Abedi-Ardekani B(1), Milojevic M(1), Olaso R(3), Meyer V(3),

Boland A(3), Deleuze JF(3), Zaridze D(4), Mukeriya A(4), Swiatkowska B(5),

Janout V(6), Schejbalová M(7), Mates D(8), Stojšić J(9), Ognjanovic M(10); ILCCO

consortium, Witte JS(11), Rashkin SR(11)(12), Kachuri L(11), Hung RJ(13), Kar

S(14)(15), Brennan P(1), Sertier AS(16), Ferrari A(16), Viari A(16)(17),

Johansson M(1), Amos CI(18), Foll M(1), McKay JD(1).

 

Author information:

(1)Genomic Epidemiology Branch, International Agency for Research on

Cancer/World Health Organization (IARC/WHO), Lyon, France.

(2)Cancer Epidemiology Unit, Nuffield Department of Population Health,

University of Oxford, Oxford, England.

(3)Université Paris-Saclay, The French Alternative Energies and Atomic Energy

Commission (CEA), Centre National de Recherche en Génomique Humaine (CNRGH),

Evry, France.

BACKGROUND: Germline genetic variation contributes to lung cancer (LC)

susceptibility. Previous genome-wide association studies (GWAS) have implicated

susceptibility loci involved in smoking behaviors and DNA repair genes, but

further work is required to identify susceptibility variants.

METHODS: To identify LC susceptibility loci, a family history-based genome-wide

association by proxy (GWAx) of LC (48 843 European proxy LC patients, 195 387

controls) was combined with a previous LC GWAS (29 266 patients, 56 450

controls) by meta-analysis. Colocalization was used to explore candidate genes

and overlap with existing traits at discovered susceptibility loci. Polygenic

risk scores (PRS) were tested within an independent validation cohort (1 666 LC

patients vs 6 664 controls) using variants selected from the LC susceptibility

loci and a novel selection approach using published GWAS summary statistics.

Finally, the effects of the LC PRS on somatic mutational burden were explored in

patients whose tumor resections have been profiled by exome (n = 685) and genome

sequencing (n = 61). Statistical tests were 2-sided.

RESULTS: The GWAx-GWAS meta-analysis identified 8 novel LC loci. Colocalization

implicated DNA repair genes (CHEK1), metabolic genes (CYP1A1), and smoking

propensity genes (CHRNA4 and CHRNB2). PRS analysis demonstrated that these

variants, as well as subgenome-wide significant variants related to expression

quantitative trait loci and/or smoking propensity, assisted in LC genetic risk

prediction (odds ratio = 1.37, 95% confidence interval = 1.29 to 1.45;

P < .001). Patients with higher genetic PRS loads of smoking-related variants

tended to have higher mutation burdens in their lung tumors.

CONCLUSIONS: This study has expanded the number of LC susceptibility loci and

provided insights into the molecular mechanisms by which these susceptibility

variants contribute to LC development.

 

© The Author(s) 2022. Published by Oxford University Press.

 

DOI: 10.1093/jnci/djac087

PMCID: PMC9360465

PMID: 35511172 [Indexed for MEDLINE]

 


上一篇: No.9

下一篇: No.7