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.11

发布时间:2022-01-14 浏览次数:
字号: + - 14

Filters applied: from 2021/9/1 - 2021/9/30

 

1. Nat Genet. 2021 Sep;53(9):1348-1359. doi: 10.1038/s41588-021-00920-0. Epub 2021 Sep 6.

 

Genomic and evolutionary classification of lung cancer in never smokers.

 

Zhang T(1), Joubert P(2), Ansari-Pour N(3), Zhao W(1), Hoang PH(1), Lokanga

R(4), …

Author information:

(1)Division of Cancer Epidemiology and Genetics, National Cancer Institute,

Bethesda, MD, USA.

(2)Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval

University, Quebec City, Quebec, Canada.

(3)Big Data Institute, Nuffield Department of Medicine, University of Oxford,

Oxford, UK.

Comment in

    Lancet Oncol. 2021 Oct;22(10):1363.

 

Lung cancer in never smokers (LCINS) is a common cause of cancer mortality but

its genomic landscape is poorly characterized. Here high-coverage whole-genome

sequencing of 232 LCINS showed 3 subtypes defined by copy number aberrations.

The dominant subtype (piano), which is rare in lung cancer in smokers, features

somatic UBA1 mutations, germline AR variants and stem cell-like properties,

including low mutational burden, high intratumor heterogeneity, long telomeres,

frequent KRAS mutations and slow growth, as suggested by the occurrence of

cancer drivers' progenitor cells many years before tumor diagnosis. The other

subtypes are characterized by specific amplifications and EGFR mutations

(mezzo-forte) and whole-genome doubling (forte). No strong tobacco smoking

signatures were detected, even in cases with exposure to secondhand tobacco

smoke. Genes within the receptor tyrosine kinase-Ras pathway had distinct

impacts on survival; five genomic alterations independently doubled mortality.

These findings create avenues for personalized treatment in LCINS.

 

© 2021. This is a U.S. government work and not under copyright protection in the

U.S.; foreign copyright protection may apply.

 

DOI: 10.1038/s41588-021-00920-0

PMCID: PMC8432745

PMID: 34493867 [Indexed for MEDLINE]

 

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

Sep 15.

 

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

 

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

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

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

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

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

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

 

Author information:

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

Center, Houston, TX, USA.

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

USA.

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

FL, USA.

 

Comment in

    Cancer Cell. 2021 Nov 8;39(11):1455-1457.

 

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

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

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

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

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

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

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

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

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

subgroups on the basis of sensitivity and structural changes that

retrospectively predict patient outcomes following treatment with EGFR

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

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

mutations that can effectively guide treatment and clinical trial choices for

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

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

oncogenes with diverse mutations.

 

© 2021. The Author(s).

 

DOI: 10.1038/s41586-021-03898-1

PMCID: PMC8481125

PMID: 34526717

 

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

 

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

 

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

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

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

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

Investigators.

 

Author information:

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

 

Comment in

    Nat Rev Clin Oncol. 2021 Dec;18(12):748.

 

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

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

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

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

investigated extensively.

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

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

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

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

central review. Secondary outcomes included the duration of response,

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

alterations were assessed.

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

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

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

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

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

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

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

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

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

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

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

detectable HER2 expression or HER2 amplification.

CONCLUSIONS: Trastuzumab deruxtecan showed durable anticancer activity in

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

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

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

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

NCT03505710.).

 

Copyright © 2021 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2112431

PMID: 34534430

 

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

Epub 2021 Sep 20.

 

Adjuvant atezolizumab after adjuvant chemotherapy in resected stage IB-IIIA

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

phase 3 trial.

 

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

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

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

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

Investigators.

 

Author information:

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

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

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

Hospital, New York, NY, USA.

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

Hospital, Shanghai, China.

Erratum in

    Lancet. 2021 Sep 23;:

 

Comment in

    Lancet. 2021 Oct 9;398(10308):1281-1283.

 

BACKGROUND: Novel adjuvant strategies are needed to optimise outcomes after

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

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

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

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

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

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

Union Internationale Contre le Cancer and American Joint Committee on Cancer

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

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

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

(observation and regular scans for disease recurrence) after adjuvant

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

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

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

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

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

evaluated in all patients who were randomly assigned and received atezolizumab

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

NCT02486718 (active, not recruiting).

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

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

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

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

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

atezolizumab treatment improved disease-free survival compared with best

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

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

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

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

INTERPRETATION: IMpower010 showed a disease-free survival benefit with

atezolizumab versus best supportive care after adjuvant chemotherapy in patients

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

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

signals. Atezolizumab after adjuvant chemotherapy offers a promising treatment

option for patients with resected early-stage NSCLC.

FUNDING: F Hoffmann-La Roche and Genentech.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

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

PMID: 34555333 [Indexed for MEDLINE]

 

5. Annu Rev Pharmacol Toxicol. 2022 Jan 6;62:197-210. doi:

10.1146/annurev-pharmtox-041921-074800. Epub 2021 Sep 30.

 

Emerging Therapeutics, Technologies, and Drug Development Strategies to Address

Patient Nonadherence and Improve Tuberculosis Treatment.

 

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

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

 

Author information:

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

California, San Francisco, California 94158, USA; email: rada.savic@ucsf.edu.

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

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

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

South Africa.

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

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

 

Imperfect medication adherence remains the biggest predictor of treatment

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

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

individual patient phenotypes, population pharmacokinetics, resistance

development, drug distribution to tuberculosis lesions, and pharmacodynamics at

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

patient outcomes. To decrease the impact of expected variabilityin drug intake

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

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

summarize emerging technologies to improve medication adherence in clinical

practice and provide suggestions on how digital adherence technologies can be

incorporated in clinical trials and practice and the drug development pipeline

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

tuberculosis.

 

DOI: 10.1146/annurev-pharmtox-041921-074800

PMID: 34591605

 

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

Epub 2021 Sep 13.

 

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

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

with prespecified comparison to surgery.

 

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

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

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

 

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

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

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

 

Author information:

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

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

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

Anderson Cancer Center, Houston, TX, USA.

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

Center, Houston, TX, USA.

Comment in

    Lancet Oncol. 2021 Dec;22(12):e534.

    Lancet Oncol. 2021 Dec;22(12):e535.

    Lancet Oncol. 2021 Dec;22(12):e536.

    Lancet Oncol. 2021 Dec;22(12):e537-e538.

 

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

higher survival after stereotactic ablative radiotherapy (SABR) than with

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

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

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

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

prospectively registered, contemporary institutional cohort of patients who

underwent video-assisted thoracoscopic surgical lobectomy with mediastinal lymph

node dissection (VATS L-MLND).

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

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

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

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

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

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

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

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

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

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

Anderson Department of Thoracic and Cardiovascular Surgery's prospectively

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

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

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

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

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

Propensity matching consisted of determining a propensity score using a

multivariable logistic regression model including several covariates (age,

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

analysis.

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

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

multidisciplinary management is strongly recommended.

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

Institutes of Health).

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

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

PMCID: PMC8521627

PMID: 34529930 [Indexed for MEDLINE]

 

7. Lancet Infect Dis. 2022 Jan;22(1):e2-e12. doi: 10.1016/S1473-3099(21)00403-5.

Epub 2021 Sep 7.

 

100 years of Mycobacterium bovis bacille Calmette-Guérin.

 

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

Mandalakas AM(7).

 

Author information:

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

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

Tuberculosis Unit, Borstel, Germany; Respiratory Medicine and International

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

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

clange@fz-borstel.de.

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

Project, Southern Danish University, Copenhagen, Denmark.

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

Montreal, QC, Canada.

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

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

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

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

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

BCG provides long-lasting strong protection against miliary and meningeal

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

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

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

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

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

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

infections in immunocompromised hosts, which can also occur following

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

of BCG includes fundamental discoveries about tuberculosis-specific and

non-specific immunity and the demonstration that tuberculosis is a

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

tuberculosis elimination.

 

Copyright © 2022 Elsevier Ltd. All rights reserved.

 

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

PMID: 34506734 [Indexed for MEDLINE]

 

8. Autophagy. 2021 Sep;17(9):2629-2638. doi: 10.1080/15548627.2020.1825273. Epub

2020 Oct 4.

 

Neutrophil autophagy during human active tuberculosis is modulated by SLAMF1.

 

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

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

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

 

Author information:

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

UBA, Buenos Aires, Argentina.

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

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

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

Argentina.

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

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

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

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

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

 

Neutrophils infected with Mycobacterium tuberculosis (Mtb) predominate in

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

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

crucial mechanism for several neutrophil functions, can be modulated by

immunological mediators. The costimulatory molecule SLAMF1 can act as a

microbial sensor in macrophages being also able to interact with

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

tuberculosis; TLR: toll like receptor.

 

DOI: 10.1080/15548627.2020.1825273

PMCID: PMC8496709

PMID: 32954947

 

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

 

Barriers and enablers to implementing tuberculosis control strategies in EU and

European Economic Area countries: a systematic review.

 

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

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

 

Author information:

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

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

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

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

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

Sweden.

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

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

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

 

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

levels requires the implementation of country-specific tuberculosis control

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

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

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

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

accessibility of tuberculosis services to recipients of care. Databases were

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

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

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

common barrier to tuberculosis control strategies described the divergence of

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

perceived usefulness of the guidelines by clinicians. The most commonly

identified enabler to tuberculosis control strategies was the documented

positive attitudes of health-care workers towards tuberculosis programmes.

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

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

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

tuberculosis control strategies analysed.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

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

PMID: 34450080 [Indexed for MEDLINE]

 

10. Nat Rev Clin Oncol. 2021 Sep;18(9):547-557. doi: 10.1038/s41571-021-00501-4.

Epub 2021 Apr 28.

 

Evolution of systemic therapy for stages I-III non-metastatic non-small-cell

lung cancer.

 

Chaft JE(#)(1), Rimner A(#)(2), Weder W(3), Azzoli CG(4), Kris MG(5), Cascone

T(6).

 

Author information:

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

Cancer Center, New York, NY and Weill Cornell Medical College, New York, NY,

USA. chaftj@mskcc.org.

(2)Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New

York, NY, USA.

(3)Thoracic Surgery, Klinik Bethanien Zurich, Zurich, Switzerland.

The treatment goal for patients with early-stage lung cancer is cure.

Multidisciplinary discussions of surgical resectability and medical operability

determine the modality of definitive local treatment (surgery or radiotherapy)

and the associated systemic therapies to further improve the likelihood of cure.

Trial evidence supports cisplatin-based adjuvant therapy either after surgical

resection or concurrently with radiotherapy. Consensus guidelines support

neoadjuvant chemotherapy in lieu of adjuvant chemotherapy and carboplatin-based

regimens for patients who are ineligible for cisplatin. The incorporation of

newer agents, now standard for patients with stage IV lung cancer, into the

curative therapy paradigm has lagged owing to inefficient trial designs, the

lengthy follow-up needed to assess survival end points and a developmental focus

on the advanced-stage disease setting. Surrogate end points, such as

pathological response, are being studied and might shorten trial durations. In

2018, the anti-PD-L1 antibody durvalumab was approved for patients with stage

III lung cancer after concurrent chemoradiotherapy. Since then, the study of

targeted therapies and immunotherapies in patients with early-stage lung cancer

has rapidly expanded. In this Review, we present the current considerations in

the treatment of patients with early-stage lung cancer and explore the current

and future state of clinical research to develop systemic therapies for

non-metastatic lung cancer.

 

© 2021. Springer Nature Limited.

 

DOI: 10.1038/s41571-021-00501-4

PMID: 33911215 [Indexed for MEDLINE]

 

11. Am J Respir Crit Care Med. 2021 Sep 1;204(5):583-595. doi:

10.1164/rccm.202101-0032OC.

 

Micro-Computed Tomography Analysis of the Human Tuberculous Lung Reveals

Remarkable Heterogeneity in Three-dimensional Granuloma Morphology.

 

Wells G(1), Glasgow JN(2), Nargan K(1), Lumamba K(1), Madansein R(3), Maharaj

K(3), Hunter RL(4), Naidoo T(5), Coetzer L(6), le Roux S(6), du Plessis A(6),

Steyn AJC(1)(2)(7).

 

Author information:

(1)Africa Health Research Institute, University of KwaZulu-Natal, Durban, South

Africa.

(2)Department of Microbiology and.

(3)Department of Cardiothoracic Surgery, Nelson Mandela School of Medicine,

University of KwaZulu-Natal, Durban, South Africa.

(4)Department of Pathology and Laboratory Medicine, University of Texas Health

Sciences Center at Houston, Houston, Texas.

(5)Department of Anatomical Pathology, National Health Laboratory Service,

Inkosi Albert Luthuli Central Hospital, Durban, South Africa; and.

(6)Computed Tomography Scanner Facility, Central Analytical Facilities,

Stellenbosch University, Stellenbosch, South Africa.

(7)Centers for AIDS Research and Free Radical Biology, University of Alabama at

Birmingham, Birmingham, Alabama.

 

Comment in

    Am J Respir Crit Care Med. 2021 Sep 1;204(5):505-507.

 

Rationale: Our current understanding of tuberculosis (TB) pathophysiology is

limited by a reliance on animal models, the paucity of human TB lung tissue, and

traditional histopathological analysis, a destructive two-dimensional approach

that provides limited spatial insight. Determining the three-dimensional (3D)

structure of the necrotic granuloma, a characteristic feature of TB, will more

accurately inform preventive TB strategies.Objectives: To ascertain the 3D shape

of the human tuberculous granuloma and its spatial relationship with airways and

vasculature within large lung tissues.Methods: We characterized the 3D

microanatomical environment of human tuberculous lungs by using micro computed

tomography, histopathology, and immunohistochemistry. By using 3D segmentation

software, we accurately reconstructed TB granulomas, vasculature, and airways in

three dimensions and confirmed our findings by using histopathology and

immunohistochemistry.Measurements and Main Results: We observed marked

heterogeneity in the morphology, volume, and number of TB granulomas in human

lung sections. Unlike depictions of granulomas as simple spherical structures,

human necrotic granulomas exhibit complex, cylindrical, branched morphologies

that are connected to the airways and shaped by the bronchi. The use of 3D

imaging of human TB lung sections provides unanticipated insight into the

spatial organization of TB granulomas in relation to the airways and

vasculature.Conclusions: Our findings highlight the likelihood that a single,

structurally complex lesion could be mistakenly viewed as multiple independent

lesions when evaluated in two dimensions. In addition, the lack of

vascularization within obstructed bronchi establishes a paradigm for

antimycobacterial drug tolerance. Lastly, our results suggest that bronchogenic

spread of Mycobacterium tuberculosis reseeds the lung.

 

DOI: 10.1164/rccm.202101-0032OC

PMCID: PMC8491258

PMID: 34015247 [Indexed for MEDLINE]

 

12. J Clin Oncol. 2021 Sep 1;39(25):2791-2802. doi: 10.1200/JCO.20.03307. Epub 2021 Jun 2.

 

Clinicopathologic Features and Response to Therapy of NRG1 Fusion-Driven Lung

Cancers: The eNRGy1 Global Multicenter Registry.

 

Drilon A(1), Duruisseaux M(2)(3)(4), Han JY(5), Ito M(6)(7)(8), Falcon C(1),

Yang SR(1), Murciano-Goroff YR(9), Chen H(10)(11), Okada M(8), Molina MA(12),

Author information:

(1)Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New

York, NY.

(2)Respiratory Department, Louis Pradel Hospital, Hospices Civils de Lyon Cancer

Institute, Lyon, France.

(3)Anticancer Antibodies Laboratory, Cancer Research Center of Lyon, Lyon,

France.

PURPOSE: Although NRG1 fusions are oncogenic drivers across multiple tumor types

including lung cancers, these are difficult to study because of their rarity.

The global eNRGy1 registry was thus established to characterize NRG1

fusion-positive lung cancers in the largest and most diverse series to date.

METHODS: From June 2018 to February 2020, a consortium of 22 centers from nine

countries in Europe, Asia, and the United States contributed data from patients

with pathologically confirmed NRG1 fusion-positive lung cancers. Profiling

included DNA-based and/or RNA-based next-generation sequencing and fluorescence

in situ hybridization. Anonymized clinical, pathologic, molecular, and response

(RECIST v1.1) data were centrally curated and analyzed.

RESULTS: Although the typified never smoking (57%), mucinous adenocarcinoma

(57%), and nonmetastatic (71%) phenotype predominated in 110 patients with NRG1

fusion-positive lung cancer, further diversity, including in smoking history

(43%) and histology (43% nonmucinous and 6% nonadenocarcinoma), was elucidated.

RNA-based testing identified most fusions (74%). Molecularly, six (of 18) novel

5' partners, 20 unique epidermal growth factor domain-inclusive chimeric events,

and heterogeneous 5'/3' breakpoints were found. Platinum-doublet and

taxane-based (post-platinum-doublet) chemotherapy achieved low objective

response rates (ORRs 13% and 14%, respectively) and modest progression-free

survival medians (PFS 5.8 and 4.0 months, respectively). Consistent with a low

programmed death ligand-1 expressing (28%) and low tumor mutational burden

(median: 0.9 mutations/megabase) immunophenotype, the activity of

chemoimmunotherapy and single-agent immunotherapy was poor (ORR 0%/PFS 3.3

months and ORR 20%/PFS 3.6 months, respectively). Afatinib achieved an ORR of

25%, not contingent on fusion type, and a 2.8-month median PFS.

CONCLUSION: NRG1 fusion-positive lung cancers were molecularly, pathologically,

and clinically more heterogeneous than previously recognized. The activity of

cytotoxic, immune, and targeted therapies was disappointing. Further research

examining NRG1-rearranged tumor biology is needed to develop new therapeutic

strategies.

 

DOI: 10.1200/JCO.20.03307

PMCID: PMC8407651

PMID: 34077268 [Indexed for MEDLINE]

 

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

10.1164/rccm.202009-3527OC.

 

Evidence-based Definition for Extensively Drug-Resistant Tuberculosis.

 

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

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

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

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

 

Author information:

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

Geneva, Switzerland.

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

Scientifico, Tradate, Italy.

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

Bern, Bern, Switzerland.

Comment in

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

 

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

the adequacy of this definition for surveillance and treatment choice.

 

DOI: 10.1164/rccm.202009-3527OC

PMID: 34107231 [Indexed for MEDLINE]

 

14. Cancer Discov. 2022 Jan;12(1):74-89. doi: 10.1158/2159-8290.CD-21-0715. Epub

2021 Sep 21.

 

Efficacy and Safety of Patritumab Deruxtecan (HER3-DXd) in EGFR

Inhibitor-Resistant, EGFR-Mutated Non-Small Cell Lung Cancer.

 

Jänne PA(1), Baik C(2), Su WC(3), Johnson ML(4), Hayashi H(5), Nishio M(6), Kim

DW(7), Koczywas M(8), Gold KA(9), Steuer CE(10), Murakami H(11), Yang JC(12),

Kim SW(13), Vigliotti M(14), Shi R(14), Qi Z(14), Qiu Y(14), Zhao L(14),

Sternberg D(14), Yu C(14), Yu HA(15).

 

Author information:

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

Pasi_Janne@dfci.harvard.edu.

(2)Seattle Cancer Care Alliance, Seattle, Washington.

(3)National Cheng Kung University Hospital, Tainan, Taiwan.

Comment in

    Cancer Discov. 2022 Jan;12(1):16-19.

 

Comment on

    Cancer Discov. 2022 Jan;12(1):16-19.

 

Receptor tyrosine-protein kinase ERBB3 (HER3) is expressed in most EGFR-mutated

lung cancers but is not a known mechanism of resistance to EGFR inhibitors.

HER3-DXd is an antibody-drug conjugate consisting of a HER3 antibody attached to

a topoisomerase I inhibitor payload via a tetrapeptide-based cleavable linker.

This phase I, dose escalation/expansion study included patients with locally

advanced or metastatic EGFR-mutated non-small cell lung cancer (NSCLC) with

prior EGFR tyrosine kinase inhibitor (TKI) therapy. Among 57 patients receiving

HER3-DXd 5.6 mg/kg intravenously once every 3 weeks, the confirmed objective

response rate by blinded independent central review (Response Evaluation

Criteria in Solid Tumors v1.1) was 39% [95% confidence interval (CI),

26.0-52.4], and median progression-free survival was 8.2 (95% CI, 4.4-8.3)

months. Responses were observed in patients with known and unknown EGFR TKI

resistance mechanisms. Clinical activity was observed across a broad range of

HER3 membrane expression. The most common grade ≥3 treatment-emergent adverse

events were hematologic toxicities. HER3-DXd has clinical activity in EGFR

TKI-resistant cancers independent of resistance mechanisms, providing an

approach to treat a broad range of drug-resistant cancers. SIGNIFICANCE: In

metastatic EGFR-mutated NSCLC, after disease progression on EGFR TKI therapy,

treatment approaches include genotype-directed therapy targeting a known

resistance mechanism or chemotherapy. HER3-DXd demonstrated clinical activity

spanning known and unknown EGFR TKI resistance mechanisms. HER3-DXd could

present a future treatment option agnostic to the EGFR TKI resistance

mechanism.See related commentary by Lim et al., p. 16.This article is

highlighted in the In This Issue feature, p. 1.

 

©2021 The Authors; Published by the American Association for Cancer Research.

 

DOI: 10.1158/2159-8290.CD-21-0715

PMID: 34548309

 

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

 

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

drug isoniazid.

 

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

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

 

Author information:

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

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

Carcinogenesis and Translational Research and State Key Laboratory of Natural

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

China.

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

Area, Hangzhou, PR China.

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

Health Science Center, Beijing, PR China.

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

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

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

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

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

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

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

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

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

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

structure and promotes gene transcription. INH-mediated histone Kinic

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

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

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

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

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

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

in humans.

 

© 2021. The Author(s).

 

DOI: 10.1038/s41467-021-25867-y

PMCID: PMC8452692

PMID: 34545082 [Indexed for MEDLINE]

 

16. 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 [Indexed for MEDLINE]

 

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

 

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

infected lung.

 

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

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

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

 

Author information:

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

University, Ithaca, NY.

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

Little Rock, AR.

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

Singapore.

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

Malawi College of Medicine, Blantyre, Malawi.

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

Liverpool, UK.

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

Research, Singapore.

 

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

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

transcriptome, surface marker expression, and bacterial phenotype for each

infected cell. This approach facilitates the dissection of the functional

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

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

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

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

macrophage populations in the lung are epigenetically constrained in their

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

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

readily transferable to other infectious disease agents with the potential for

an increased understanding of the roles that different host cell populations

play during the course of an infection.

 

© 2021 Pisu et al.

 

DOI: 10.1084/jem.20210615

PMCID: PMC8302446

PMID: 34292313 [Indexed for MEDLINE]

 

18. J Exp Med. 2021 Oct 4;218(10):e20210915. doi: 10.1084/jem.20210915. Epub 2021

Sep 7.

 

Blood transcriptomics reveal the evolution and resolution of the immune response

in tuberculosis.

 

Tabone O(#)(1), Verma R(#)(2), Singhania A(1), Chakravarty P(3), Branchett

WJ(1), Graham CM(1), Lee J(2), Trang T(4), Reynier F(4), Leissner P(4), Kaiser

K(5), Rodrigue M(6), Woltmann G(2), Haldar P(#)(2), O'Garra A(#)(1)(7).

 

Author information:

(1)Laboratory of Immunoregulation and Infection, The Francis Crick Institute,

London, UK.

(2)Department of Respiratory Sciences, National Institute for Health Research

Respiratory Biomedical Research Centre, University of Leicester, UK.

(3)Bioinformatics Core, The Francis Crick Institute, London, UK.

Blood transcriptomics have revealed major characteristics of the immune response

in active TB, but the signature early after infection is unknown. In a unique

clinically and temporally well-defined cohort of household contacts of active TB

patients that progressed to TB, we define minimal changes in gene expression in

incipient TB increasing in subclinical and clinical TB. While increasing with

time, changes in gene expression were highest at 30 d before diagnosis, with

heterogeneity in the response in household TB contacts and in a published cohort

of TB progressors as they progressed to TB, at a bulk cohort level and in

individual progressors. Blood signatures from patients before and during anti-TB

treatment robustly monitored the treatment response distinguishing early and

late responders. Blood transcriptomics thus reveal the evolution and resolution

of the immune response in TB, which may help in clinical management of the

disease.

 

© 2021 Tabone et al.

 

DOI: 10.1084/jem.20210915

PMCID: PMC8493863

PMID: 34491266 [Indexed for MEDLINE]

 

19. 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.

Comment in

    J Clin Oncol. 2021 Sep 10;39(26):2855-2858.

 

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 [Indexed for MEDLINE]

 

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

 

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

following Mycobacterium tuberculosis infection.

 

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

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

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

 

Author information:

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

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

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

Guangdong Provincial Key Laboratory of Biomedical Imaging, and Department of

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

Zhuhai, China.

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

Guangdong Province, China.

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

pattern recognition receptors on innate immune cells that regulates the

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

infection and inflammation remains controversial. Here, we demonstrated that

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

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

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

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

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

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

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

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

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

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

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

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

provide potential therapeutic targets for infectious and inflammatory diseases.

 

DOI: 10.1172/JCI137407

PMCID: PMC8409584

PMID: 34623322 [Indexed for MEDLINE]

 

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

 

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

structural framework and enables rational drug design to combat TB.

 

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

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

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

 

Author information:

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

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

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

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

Frankfurt/Main, Germany.

 

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

simultaneously multiple respiratory enzyme complexes of Mycobacterium

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

shorten drug administration regimes, and reduce the opportunity for the

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

bd oxidase plays a crucial role for mycobacterial pathophysiology by maintaining

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

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

conjunction with atomistic molecular dynamics (MD) simulation studies we

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

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

canonical quinol oxidation site in Actinobacteria. Our detailed insights into

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

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

on this enzyme.

 

© 2021. The Author(s).

 

DOI: 10.1038/s41467-021-25537-z

PMCID: PMC8413341

PMID: 34475399 [Indexed for MEDLINE]

 

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

 

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

Results From a Phase II Trial.

 

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

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

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

 

Author information:

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

Texas MD Anderson Cancer Center, Houston, TX.

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

Center, Houston, TX.

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

MD.

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

Houston, TX.

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

Center, Houston, TX.

 

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

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

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

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

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

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

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

Confirmatory scans were performed at least 28 days from initial radiologic

response.

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

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

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

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

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

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

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

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

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

treatment-related death because of pneumonitis.

CONCLUSION: Poziotinib showed promising antitumor activity in patients with HER2

exon 20 mutant NSCLC including patients who had previously received

platinum-based chemotherapy.

 

DOI: 10.1200/JCO.21.01113

PMID: 34550757

 

23. Nat Commun. 2021 Sep 14;12(1):5431. doi: 10.1038/s41467-021-25787-x.

 

Multi-region exome sequencing reveals the intratumoral heterogeneity of

surgically resected small cell lung cancer.

 

Zhou H(#)(1), Hu Y(#)(2), Luo R(#)(3), Zhao Y(#)(1), Pan H(#)(4), Ji L(#)(5),

Zhou T(1), Zhang L(6), Long H(6), Fu J(6), Wen Z(6), Wang S(6), Wang X(6), Lin

P(6), Yang H(6), Wang J(6), Song M(5), Yi X(5), Yang L(5), Xia X(5), Guan Y(5),

Fang W(1), Yang Y(1), Hong S(1), Huang Y(1), Li P(5), Zhang Y(7), Zhou N(8).

 

Author information:

(1)Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State

Key Laboratory of Oncology in South China, Collaborative Innovation Center for

Cancer Medicine, Guangzhou, China.

(2)Department of Thoracic Surgery, Sun Yat-Sen University Cancer Center, State

Key Laboratory of Oncology in South China, Collaborative Innovation Center for

Cancer Medicine, Guangdong Esophageal Cancer Institute (GECI), Guangzhou, China.

(3)Department of Pathology, Sun Yat-Sen University Cancer Center, State Key

Laboratory of Oncology in South China, Collaborative Innovation Center for

Cancer Medicine, Guangzhou, China.

Small cell lung cancer (SCLC) is a highly malignant tumor which is eventually

refractory to any treatment. Intratumoral heterogeneity (ITH) may contribute to

treatment failure. However, the extent of ITH in SCLC is still largely unknown.

Here, we subject 120 tumor samples from 40 stage I-III SCLC patients to

multi-regional whole-exome sequencing. The most common mutant genes are TP53

(88%) and RB1 (72%). We observe a medium level of mutational heterogeneity

(0.30, range 0.0~0.98) and tumor mutational burden (TMB, 10.2 mutations/Mb,

range 1.1~51.7). Our SCLC samples also exhibit somatic copy number variation

(CNV) across all patients, with an average CNV ITH of 0.49 (range 0.02~0.99). In

terms of mutation distribution, ITH, TMB, mutation clusters, and gene

signatures, patients with combined SCLC behave roughly the same way as patients

with pure SCLC. This condition also exists in smoking patients and patients with

EGFR mutations. A higher TMB per cluster is associated with better disease-free

survival while single-nucleotide variant ITH is linked to worse overall

survival, and therefore these features may be used as prognostic biomarkers for

SCLC. Together, these findings demonstrate the intratumoral genetic

heterogeneity of surgically resected SCLC and provide insights into resistance

to treatment.

 

© 2021. The Author(s).

 

DOI: 10.1038/s41467-021-25787-x

PMCID: PMC8440529

PMID: 34521849 [Indexed for MEDLINE]

 

上一篇: No.12

下一篇: No.10