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 >>2023年

高分SCI文摘

2023年

No.8

发布时间:2023-09-14 浏览次数:
字号: + - 14

PubMed

(tuberculosis[Title/Abstract]) OR (lung cancer[Title/Abstract])

Filters applied: from 2023/8/1 - 2023/8/31.

 

1. Lancet. 2023 Aug 19;402(10402):627-640. doi: 10.1016/S0140-6736(23)01231-X. Epub 2023 Aug 8.

 

Nutritional supplementation to prevent tuberculosis incidence in household

contacts of patients with pulmonary tuberculosis in India (RATIONS): a

field-based, open-label, cluster-randomised, controlled trial.

 

Bhargava A(1), Bhargava M(2), Meher A(3), Benedetti A(4), Velayutham B(3), Sai

Teja G(5), Watson B(3), Barik G(3), Pathak RR(6), Prasad R(7), Dayal R(8),

Madhukeshwar AK(9), Chadha V(10), Pai M(11), Joshi R(12), Menzies D(13),

Swaminathan S(14).

 

Author information:

(1)Department of Medicine, Yenepoya Medical College, Mangalore, India; Center

for Nutrition Studies, Yenepoya (Deemed to be University), Mangalore, India;

Department of Medicine, McGill University, Montreal, QC, Canada. Electronic

address: anuragb17@gmail.com.

(2)Department of Community Medicine, Yenepoya Medical College, Mangalore, India;

Center for Nutrition Studies, Yenepoya (Deemed to be University), Mangalore,

India.

(3)Indian Council of Medical Research-National Institute for Research in

Tuberculosis, Chennai, India.

 

Comment in

    Lancet. 2023 Aug 19;402(10402):588-590.

 

BACKGROUND: In India, tuberculosis and undernutrition are syndemics with a high

burden of tuberculosis coexisting with a high burden of undernutrition in

patients and in the population. The aim of this study was to determine the

effect of nutritional supplementation on tuberculosis incidence in household

contacts of adults with microbiologically confirmed pulmonary tuberculosis.

METHODS: In this field-based, open-label, cluster-randomised controlled trial,

we enrolled household contacts of 2800 patients with microbiologically confirmed

pulmonary tuberculosis across 28 tuberculosis units of the National Tuberculosis

Elimination Programme in four districts of Jharkhand, India. The tuberculosis

units were randomly allocated 1:1 by block randomisation to the control group or

the intervention group, by a statistician using computer-generated random

numbers. Although microbiologically confirmed pulmonary tuberculosis patients in

both groups received food rations (1200 kcal, 52 grams of protein per day with

micronutrients) for 6 months, only household contacts in the intervention group

received monthly food rations and micronutrients (750 kcal, 23 grams of protein

per day with micronutrients). After screening all household contacts for

co-prevalent tuberculosis at baseline, all participants were followed up

actively until July 31, 2022, for the primary outcome of incident tuberculosis

(all forms). The ascertainment of the outcome was by independent medical staff

in health services. We used Cox proportional hazards model and Poisson

regression via the generalised estimating equation approach to estimate

unadjusted hazard ratios, adjusted hazard ratios (aHRs), and incidence rate

ratios (IRRs). This study is registered with CTRI-India, CTRI/2019/08/020490.

FINDINGS: Between Aug 16, 2019, and Jan 31, 2021, there were 10345 household

contacts, of whom 5328 (94·8%) of 5621 household contacts in the intervention

group and 4283 (90·7%) of 4724 household contacts in the control group completed

the primary outcome assessment. Almost two-thirds of the population belonged to

Indigenous communities (eg, Santhals, Ho, Munda, Oraon, and Bhumij) and 34%

(3543 of 10345) had undernutrition. We detected 31 (0·3%) of 10345 household

contact patients with co-prevalent tuberculosis disease in both groups at

baseline and 218 (2·1%) people were diagnosed with incident tuberculosis (all

forms) over 21869 person-years of follow-up, with 122 of 218 incident cases in

the control group (2·6% [122 of 4712 contacts at risk], 95% CI 2·2-3·1;

incidence rate 1·27 per 100 person-years) and 96 incident cases in the

intervention group (1·7% [96 of 5602], 1·4-2·1; 0·78 per 100 person-years), 

of whom 152 (69·7%) of 218 were patients with microbiologically confirmed pulmonary

tuberculosis. Tuberculosis incidence (all forms) in the intervention group had an 

adjusted IRR of 0·61 (95% CI 0·43-0·85; aHR 0·59 [0·42-0·83]), with an even 

greater decline in incidence of microbiologically confirmed pulmonary

tuberculosis (0·52 [0·35-0·79]; 0·51 [0·34-0·78]). This translates into a

relative reduction of tuberculosis incidence of 39% (all forms) to 48%

(microbiologically confirmed pulmonary tuberculosis) in the intervention group.

An estimated 30 households (111 household contacts) would need to be provided

nutritional supplementation to prevent one incident tuberculosis.

INTERPRETATION: To our knowledge, this is the first randomised trial looking at

the effect of nutritional support on tuberculosis incidence in household

contacts, whereby the nutritional intervention was associated with substantial

(39-48%) reduction in tuberculosis incidence in the household during 2 years of

follow-up. This biosocial intervention can accelerate reduction in tuberculosis

incidence in countries or communities with a tuberculosis and undernutrition

syndemic.

FUNDING: Indian Council of Medical Research-India TB Research Consortium.

 

Copyright © 2023 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S0140-6736(23)01231-X

PMID: 37567200 [Indexed for MEDLINE]

 

2. Nat Genet. 2023 Aug;55(8):1301-1310. doi: 10.1038/s41588-023-01446-3. Epub 2023 Jul 27.

 

Single-molecule genome-wide mutation profiles of cell-free DNA for non-invasive

detection of cancer.

 

Bruhm DC(1), Mathios D(1), Foda ZH(1), Annapragada AV(1), Medina JE(1), Adleff

V(1), Chiao EJ(1), Ferreira L(1), Cristiano S(1), White JR(1), Mazzilli SA(2),

Billatos E(2)(3), Spira A(2)(3), Zaidi AH(4), Mueller J(4), Kim AK(1),

Anagnostou V(1), Phallen J(1), Scharpf RB(5), Velculescu VE(6).

 

Author information:

(1)The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University

School of Medicine, Baltimore, MD, USA.

(2)Division of Computational Biomedicine, Department of Medicine, Boston

University, Boston, MA, USA.

(3)Section of Pulmonary and Critical Care Medicine, Department of Medicine,

Boston University, Boston, MA, USA.

 

Comment in

    Nat Genet. 2023 Aug;55(8):1261-1262.

 

Somatic mutations are a hallmark of tumorigenesis and may be useful for

non-invasive diagnosis of cancer. We analyzed whole-genome sequencing data from

2,511 individuals in the Pan-Cancer Analysis of Whole Genomes (PCAWG) study as

well as 489 individuals from four prospective cohorts and found distinct

regional mutation type-specific frequencies in tissue and cell-free DNA from

patients with cancer that were associated with replication timing and other

chromatin features. A machine-learning model using genome-wide mutational

profiles combined with other features and followed by CT imaging detected >90%

of patients with lung cancer, including those with stage I and II disease. The

fixed model was validated in an independent cohort, detected patients with

cancer earlier than standard approaches and could be used to monitor response to

therapy. This approach lays the groundwork for non-invasive cancer detection

using genome-wide mutation features that may facilitate cancer screening and

monitoring.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41588-023-01446-3

PMCID: PMC10412448

PMID: 37500728 [Indexed for MEDLINE]

 

3. Nature. 2023 Aug;620(7973):445-452. doi: 10.1038/s41586-023-06366-0. Epub 2023

Jul 26.

 

Structure of an endogenous mycobacterial MCE lipid transporter.

 

Chen J(1), Fruhauf A(1), Fan C(1), Ponce J(2), Ueberheide B(2)(3)(4), Bhabha

G(5), Ekiert DC(6)(7).

 

Author information:

(1)Department of Cell Biology, New York University School of Medicine, New York,

NY, USA.

(2)Proteomics Laboratory, Division of Advanced Research Technologies, New York

University School of Medicine, New York, NY, USA.

(3)Department of Biochemistry and Molecular Pharmacology, New York University

School of Medicine, New York, NY, USA.

 

Update of

    Res Sq. 2023 Jan 10;:

 

To replicate inside macrophages and cause tuberculosis, Mycobacterium

tuberculosis must scavenge a variety of nutrients from the host1,2. The

mammalian cell entry (MCE) proteins are important virulence factors in M.

tuberculosis1,3, where they are encoded by large gene clusters and have been

implicated in the transport of fatty acids4-7 and cholesterol1,4,8 across the

impermeable mycobacterial cell envelope. Very little is known about how cargos

are transported across this barrier, and it remains unclear how the

approximately ten proteins encoded by a mycobacterial mce gene cluster assemble

to transport cargo across the cell envelope. Here we report the cryo-electron

microscopy (cryo-EM) structure of the endogenous Mce1 lipid-import machine of

Mycobacterium smegmatis-a non-pathogenic relative of M. tuberculosis. The

structure reveals how the proteins of the Mce1 system assemble to form an

elongated ABC transporter complex that is long enough to span the cell envelope.

The Mce1 complex is dominated by a curved, needle-like domain that appears to be

unrelated to previously described protein structures, and creates a protected

hydrophobic pathway for lipid transport across the periplasm. Our structural

data revealed the presence of a subunit of the Mce1 complex, which we identified

using a combination of cryo-EM and AlphaFold2, and name LucB. Our data lead to a

structural model for Mce1-mediated lipid import across the mycobacterial cell

envelope.

 

© 2023. The Author(s), under exclusive licence to Springer Nature Limited.

 

DOI: 10.1038/s41586-023-06366-0

PMID: 37495693 [Indexed for MEDLINE]

 

4. Lancet. 2023 Aug 5;402(10400):451-463. doi: 10.1016/S0140-6736(23)00774-2. Epub 2023 Jul 6.

 

First-line atezolizumab monotherapy versus single-agent chemotherapy in patients

with non-small-cell lung cancer ineligible for treatment with a

platinum-containing regimen (IPSOS): a phase 3, global, multicentre, open-label,

randomised controlled study.

 

Lee SM(1), Schulz C(2), Prabhash K(3), Kowalski D(4), Szczesna A(5), Han B(6),

Rittmeyer A(7), Talbot T(8), Vicente D(9), Califano R(10), …

 

Author information:

(1)Department of Oncology, University College London Hospitals NHS Foundation

Trust, CRUK Lung Cancer Centre of Excellence and UCL Cancer Institute, London,

UK. Electronic address: sm.lee@nhs.net.

(2)Bereich Pneumologie Klinik und Poliklinik für Innere Medizin II, University

Hospital Regensburg, Regensburg, Germany.

(3)Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India.

Erratum in

    Lancet. 2023 Aug 5;402(10400):450.

 

Comment in

    Lancet. 2023 Aug 5;402(10400):426-427.

 

BACKGROUND: Despite immunotherapy advancements for patients with advanced or

metastatic non-small-cell lung cancer (NSCLC), pivotal first-line trials were

limited to patients with an Eastern Cooperative Oncology Group performance

status (ECOG PS) 0-1 and a median age of 65 years or younger. We aimed to

compare the efficacy and safety of first-line atezolizumab monotherapy with

single-agent chemotherapy in patients ineligible for platinum-based

chemotherapy.

METHODS: This trial was a phase 3, open-label, randomised controlled study

conducted at 91 sites in 23 countries across Asia, Europe, North America, and

South America. Eligible patients had stage IIIB or IV NSCLC in whom

platinum-doublet chemotherapy was deemed unsuitable by the investigator due to

an ECOG PS 2 or 3, or alternatively, being 70 years or older with an ECOG PS 0-1

with substantial comorbidities or contraindications for platinum-doublet

chemotherapy. Patients were randomised 2:1 by permuted-block randomisation

(block size of six) to receive 1200 mg of atezolizumab given intravenously every

3 weeks or single-agent chemotherapy (vinorelbine [oral or intravenous] or

gemcitabine [intravenous]; dosing per local label) at 3-weekly or 4-weekly

cycles. The primary endpoint was overall survival assessed in the

intention-to-treat population. Safety analyses were conducted in the

safety-evaluable population, which included all randomised patients who received

any amount of atezolizumab or chemotherapy. This trial is registered with

ClinicalTrials.gov, NCT03191786.

FINDINGS: Between Sept 11, 2017, and Sept 23, 2019, 453 patients were enrolled

and randomised to receive atezolizumab (n=302) or chemotherapy (n=151).

Atezolizumab improved overall survival compared with chemotherapy (median

overall survival 10·3 months [95% CI 9·4-11·9] vs 9·2 months [5·9-11·2];

stratified hazard ratio 0·78 [0·63-0·97], p=0·028), with a 2-year survival 

rate of 24% (95% CI 19·3-29·4) with atezolizumab compared with 12% (6·7-18·0) 

with chemotherapy. Compared with chemotherapy, atezolizumab was associated with

stabilisation or improvement of patient-reported health-related quality-of-life

functioning scales and symptoms and fewer grade 3-4 treatment-related adverse

events (49 [16%] of 300 vs 49 [33%] of 147) and treatment-related deaths (three

[1%] vs four [3%]).

INTERPRETATION: First-line treatment with atezolizumab monotherapy was

associated with improved overall survival, a doubling of the 2-year survival

rate, maintenance of quality of life, and a favourable safety profile compared

with single-agent chemotherapy. These data support atezolizumab monotherapy as a

potential first-line treatment option for patients with advanced NSCLC who are

ineligible for platinum-based chemotherapy.

FUNDING: F Hoffmann-La Roche and Genentech Inc, a member of the Roche group.

 

Copyright © 2023 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S0140-6736(23)00774-2

PMID: 37423228 [Indexed for MEDLINE]

 

5. N Engl J Med. 2023 Aug 10;389(6):504-513. doi: 10.1056/NEJMoa2215530. Epub 2023 Jun 28.

 

Perioperative Nivolumab and Chemotherapy in Stage III Non-Small-Cell Lung

Cancer.

 

Provencio M(1), Nadal E(1), González-Larriba JL(1), Martínez-Martí A(1), Bernabé

R(1), Bosch-Barrera J(1), Casal-Rubio J(1), Calvo V(1), Insa A(1),…

 

Author information:

(1)From Hospital Universitario Puerta de Hierro-Majadahonda (M.P., V.C.,

R.S.-B., J.M.-L., A.C.-B., A.R.), Hospital Universitario Clínico San Carlos

(J.L.G.-L., C.A.R., F.H.-T.), Hospital Universitario 12 de Octubre (S.P.),

Hospital Universitario La Paz (J.C.), and the Genetic and Molecular Epidemiology

Group, Spanish National Cancer Research Center (R. Benítez), Madrid, Institut

Català d'Oncologia, L'Hospitalet de Llobregat (E.N., R.P.), Vall d'Hebron

Institute of Oncology, Hospital Universitari Vall d'Hebrón (A.M.-M.), …

 

BACKGROUND: Approximately 20% of patients with non-small-cell lung cancer

(NSCLC) receive a diagnosis of stage III disease. There is no current consensus

regarding the most appropriate treatment for these patients.

METHODS: In this open-label, phase 2 trial, we randomly assigned patients with

resectable stage IIIA or IIIB NSCLC to receive neoadjuvant nivolumab plus

platinum-based chemotherapy (experimental group) or chemotherapy alone (control

group), followed by surgery. Patients in the experimental group who had R0

resections received adjuvant treatment with nivolumab for 6 months. The primary

end point was a pathological complete response (0% viable tumor in resected lung

and lymph nodes). Secondary end points included progression-free survival and

overall survival at 24 months and safety.

RESULTS: A total of 86 patients underwent randomization; 57 were assigned to the

experimental group and 29 were assigned to the control group. A pathological

complete response occurred in 37% of the patients in the experimental group and

in 7% in the control group (relative risk, 5.34; 95% confidence interval [CI],

1.34 to 21.23; P=0.02). Surgery was performed in 93% of the patients in the

experimental group and in 69% in the control group (relative risk, 1.35; 95% CI,

1.05 to 1.74). Kaplan-Meier estimates of progression-free survival at 24 months

were 67.2% in the experimental group and 40.9% in the control group (hazard

ratio for disease progression, disease recurrence, or death, 0.47; 95% CI, 0.25

to 0.88). Kaplan-Meier estimates of overall survival at 24 months were 85.0% in

the experimental group and 63.6% in the control group (hazard ratio for death,

0.43; 95% CI, 0.19 to 0.98). Grade 3 or 4 adverse events occurred in 11 patients

in the experimental group (19%; some patients had events of both grades) and 3

patients in the control group (10%).

CONCLUSIONS: In patients with resectable stage IIIA or IIIB NSCLC, perioperative

treatment with nivolumab plus chemotherapy resulted in a higher percentage of

patients with a pathological complete response and longer survival than

chemotherapy alone. (Funded by Bristol Myers Squibb and others; NADIM II

ClinicalTrials.gov number, NCT03838159; EudraCT number, 2018-004515-45.).

 

Copyright © 2023 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2215530

PMID: 37379158 [Indexed for MEDLINE]

 

6. N Engl J Med. 2023 Aug 10;389(6):491-503. doi: 10.1056/NEJMoa2302983. Epub 2023 Jun 3.

 

Perioperative Pembrolizumab for Early-Stage Non-Small-Cell Lung Cancer.

 

Wakelee H(1), Liberman M(1), Kato T(1), Tsuboi M(1), Lee SH(1), Gao S(1), Chen

KN(1), Dooms C(1), Majem M(1), Eigendorff E(1), Martinengo GL(1), Bylicki O(1),

Rodríguez-Abreu D(1), Chaft JE(1), Novello S(1), Yang J(1), Keller SM(1),

Samkari A(1), Spicer JD(1); KEYNOTE-671 Investigators.

 

Author information:

(1)From Stanford University School of Medicine, Stanford Cancer Institute,

Stanford, CA (H.W.); Centre Hospitalier de l'Université de Montréal (M.L.) and

McGill University Health Centre (J.D.S.) - both in Montreal; Kanagawa Cancer

Center, Yokohama (T.K.), …

 

BACKGROUND: Among patients with resectable early-stage non-small-cell lung

cancer (NSCLC), a perioperative approach that includes both neoadjuvant and

adjuvant immune checkpoint inhibition may provide benefit beyond either approach

alone.

METHODS: We conducted a randomized, double-blind, phase 3 trial to evaluate

perioperative pembrolizumab in patients with early-stage NSCLC. Participants

with resectable stage II, IIIA, or IIIB (N2 stage) NSCLC were assigned in a 1:1

ratio to receive neoadjuvant pembrolizumab (200 mg) or placebo once every 3

weeks, each of which was given with cisplatin-based chemotherapy for 4 cycles,

followed by surgery and adjuvant pembrolizumab (200 mg) or placebo once every 3

weeks for up to 13 cycles. The dual primary end points were event-free survival

(the time from randomization to the first occurrence of local progression that

precluded the planned surgery, unresectable tumor, progression or recurrence, or

death) and overall survival. Secondary end points included major pathological

response, pathological complete response, and safety.

RESULTS: A total of 397 participants were assigned to the pembrolizumab group,

and 400 to the placebo group. At the prespecified first interim analysis, the

median follow-up was 25.2 months. Event-free survival at 24 months was 62.4% in

the pembrolizumab group and 40.6% in the placebo group (hazard ratio for

progression, recurrence, or death, 0.58; 95% confidence interval [CI], 0.46 to

0.72; P<0.001). The estimated 24-month overall survival was 80.9% in the

pembrolizumab group and 77.6% in the placebo group (P=0.02, which did not meet

the significance criterion). A major pathological response occurred in 30.2% of

the participants in the pembrolizumab group and in 11.0% of those in the placebo

group (difference, 19.2 percentage points; 95% CI, 13.9 to 24.7; P<0.0001;

threshold, P=0.0001), and a pathological complete response occurred in 18.1%

and 4.0%, respectively (difference, 14.2 percentage points; 95% CI, 10.1 to

18.7; P<0.0001; threshold, P=0.0001). Across all treatment phases, 44.9% of

the participants in the pembrolizumab group and 37.3% of those in the placebo

group had treatment-related adverse events of grade 3 or higher, including 1.0%

and 0.8%, respectively, who had grade 5 events.

CONCLUSIONS: Among patients with resectable, early-stage NSCLC, neoadjuvant

pembrolizumab plus chemotherapy followed by resection and adjuvant pembrolizumab

significantly improved event-free survival, major pathological response, and

pathological complete response as compared with neoadjuvant chemotherapy alone

followed by surgery. Overall survival did not differ significantly between the

groups in this analysis. (Funded by Merck Sharp and Dohme; KEYNOTE-671

ClinicalTrials.gov number, NCT03425643.).

 

Copyright © 2023 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2302983

PMID: 37272513 [Indexed for MEDLINE]

 

7. Cancer Cell. 2023 Aug 9:S1535-6108(23)00252-0. doi: 10.1016/j.ccell.2023.07.012. Online ahead of print.

 

CD44(+) lung cancer stem cell-derived pericyte-like cells cause brain metastases

through GPR124-enhanced trans-endothelial migration.

 

Huang Q(1), Liu L(2), Xiao D(2), Huang Z(1), Wang W(2), Zhai K(1), Fang X(1),

Kim J(3), Liu J(3), Liang W(2), He J(4), Bao S(5).

 

Author information:

(1)Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic,

Cleveland, OH 44195, USA.

(2)Department of Thoracic Surgery, the First Affiliated Hospital of Guangzhou

Medical University, the State Key Laboratory of Respiratory Disease, and the

National Clinical Research Centre for Respiratory Disease, Guangzhou 510120,

China.

(3)Department of Neuro-Oncology, H. Lee Moffitt Cancer Center and Research

Institute, Tampa, FL 33612, USA.

 

Brain metastasis of lung cancer causes high mortality, but the exact mechanisms

underlying the metastasis remain unclear. Here we report that vascular pericytes

derived from CD44+ lung cancer stem cells (CSCs) in lung adenocarcinoma (ADC)

potently cause brain metastases through the G-protein-coupled receptor 124

(GPR124)-enhanced trans-endothelial migration (TEM). CD44+ CSCs in perivascular

niches generate the majority of vascular pericytes in lung ADC. CSC-derived

pericyte-like cells (Cd-pericytes) exhibit remarkable TEM capacity to

effectively intravasate into the vessel lumina, survive in the circulation,

extravasate into the brain parenchyma, and then de-differentiate into

tumorigenic CSCs to form metastases. Cd-pericytes uniquely express GPR124 that

activates Wnt7-β-catenin signaling to enhance TEM capacity of Cd-pericytes for

intravasation and extravasation, two critical steps during tumor metastasis.

Furthermore, selective disruption of Cd-pericytes, GPR124, or the Wnt7-β-catenin

signaling markedly reduces brain and liver metastases of lung ADC. Our findings

uncover an unappreciated cellular and molecular paradigm driving tumor

metastasis.

 

Copyright © 2023 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2023.07.012

PMID: 37595587

 

8. Nat Cell Biol. 2023 Sep;25(9):1346-1358. doi: 10.1038/s41556-023-01210-z. Epub

2023 Aug 17.

 

KDM6A epigenetically regulates subtype plasticity in small cell lung cancer.

 

Duplaquet L(#)(1), Li Y(#)(1), Booker MA(2), Xie Y(1)(3), Olsen SN(4), Patel

RA(5), Hong D(1), Hatton C(4), Denize T(6), Walton E(6), Laimon YN(6), Li

R(1)(3), Jiang Y(1)(3), Bronson RT(7), Southard J(8), Li S(8), Signoretti

S(6)(9)(10), Qiu X(1)(3), Cejas P(1)(3), Armstrong SA(4), Long HW(1)(3),

Tolstorukov MY(2), Haffner MC(5)(11)(12), Oser MG(13)(14).

 

Author information:

(1)Department of Medical Oncology, Dana-Farber Cancer Institute and Brigham and

Women's Hospital, Harvard Medical School, Boston, MA, USA.

(2)Department of Informatics and Analytics, Dana-Farber Cancer Institute,

Boston, MA, USA.

(3)Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute,

Boston, MA, USA.

 

Small cell lung cancer (SCLC) exists broadly in four molecular subtypes: ASCL1,

NEUROD1, POU2F3 and Inflammatory. Initially, SCLC subtypes were thought to be

mutually exclusive, but recent evidence shows intra-tumoural subtype

heterogeneity and plasticity between subtypes. Here, using a CRISPR-based

autochthonous SCLC genetically engineered mouse model to study the consequences

of KDM6A/UTX inactivation, we show that KDM6A inactivation induced plasticity

from ASCL1 to NEUROD1 resulting in SCLC tumours that express both ASCL1 and

NEUROD1. Mechanistically, KDM6A normally maintains an active chromatin state

that favours the ASCL1 subtype with its loss decreasing H3K4me1 and increasing

H3K27me3 at enhancers of neuroendocrine genes leading to a cell state that is

primed for ASCL1-to-NEUROD1 subtype switching. This work identifies KDM6A as an

epigenetic regulator that controls ASCL1 to NEUROD1 subtype plasticity and

provides an autochthonous SCLC genetically engineered mouse model to model ASCL1

and NEUROD1 subtype heterogeneity and plasticity, which is found in 35-40% of

human SCLCs.

 

© 2023. The Author(s), under exclusive licence to Springer Nature Limited.

 

DOI: 10.1038/s41556-023-01210-z

PMID: 37591951

 

9. Lancet Oncol. 2023 Sep;24(9):989-1001. doi: 10.1016/S1470-2045(23)00329-7. Epub 2023 Aug 14.

 

First-line cemiplimab monotherapy and continued cemiplimab beyond progression

plus chemotherapy for advanced non-small-cell lung cancer with PD-L1 50% or more

(EMPOWER-Lung 1): 35-month follow-up from a mutlicentre, open-label, randomised,

phase 3 trial.

 

Özgüroğlu M(1), Kilickap S(2), Sezer A(3), Gümüş M(4), Bondarenko I(5),

Gogishvili M(6), Nechaeva M(7), Schenker M(8), Cicin I(9), Ho GF(10), Kulyaba

Y(11), Zyuhal K(12), Scheusan RI(13), Garassino MC(14), He X(15), Kaul M(15),

Okoye E(15), Li Y(15), Li S(15), Pouliot JF(15), Seebach F(15), Lowy I(15),

Gullo G(15), Rietschel P(15).

 

Author information:

(1)Cerrahpaşa Faculty of Medicine, Division of Medical Oncology, Istanbul

University Cerrahpaşa, Istanbul, Türkiye. Electronic address:

ozguroglu@gmail.com.

(2)Faculty of Medicine, Department of Internal Medicine and Medical Oncology,

Istinye University Istanbul, Türkiye.

(3)Department of Medical Oncology, Başkent University, Adana, Türkiye.

 

BACKGROUND: Cemiplimab provided significant survival benefit to patients with

advanced non-small-cell lung cancer with PD-L1 tumour expression of at least 50%

and no actionable biomarkers at 1-year follow-up. In this exploratory analysis,

we provide outcomes after 35 months' follow-up and the effect of adding

chemotherapy to cemiplimab at the time of disease progression.

METHODS: EMPOWER-Lung 1 was a multicentre, open-label, randomised, phase 3

trial. We enrolled patients (aged ≥18 years) with histologically confirmed

squamous or non-squamous advanced non-small-cell lung cancer with PD-L1 tumour

expression of 50% or more. We randomly assigned (1:1) patients to intravenous

cemiplimab 350 mg every 3 weeks for up to 108 weeks, or until disease

progression, or investigator's choice of chemotherapy. Central randomisation

scheme generated by an interactive web response system governed the

randomisation process that was stratified by histology and geographical region.

Primary endpoints were overall survival and progression free survival, as

assessed by a blinded independent central review (BICR) per Response Evaluation

Criteria in Solid Tumours version 1.1. Patients with disease progression on

cemiplimab could continue cemiplimab with the addition of up to four cycles of

chemotherapy. We assessed response in these patients by BICR against a new

baseline, defined as the last scan before chemotherapy initiation. The primary

endpoints were assessed in all randomly assigned participants (ie,

intention-to-treat population) and in those with a PD-L1 expression of at least

50%. We assessed adverse events in all patients who received at least one dose

of their assigned treatment. This trial is registered with ClinicalTrials.gov,

NCT03088540.

FINDINGS: Between May 29, 2017, and March 4, 2020, we recruited 712 patients

(607 [85%] were male and 105 [15%] were female). We randomly assigned 357 (50%)

to cemiplimab and 355 (50%) to chemotherapy. 284 (50%) patients assigned to

cemiplimab and 281 (50%) assigned to chemotherapy had verified PD-L1 expression

of at least 50%. At 35 months' follow-up, among those with a verified PD-L1

expression of at least 50% median overall survival in the cemiplimab group was

26·1 months (95% CI 22·1-31·8; 149 [52%] of 284 died) versus 13·3 months

(10·5-16·2; 188 [67%] of 281 died) in the chemotherapy group (hazard ratio [HR]

0·57, 95% CI 0·46-0·71; p<0·0001), median progression-free survival was 8·1

months (95% CI 6·2-8·8; 214 events occurred) in the cemiplimab group versus 5·3 

months (4·3-6·1; 236 events occurred) in the chemotherapy group (HR 0·51, 95% CI 

0·42-0·62; p<0·0001). Continued cemiplimab plus chemotherapy as second-line 

therapy (n=64) resulted in a median progression-free survival of 6·6 months

(6·1-9·3) and overall survival of 15·1 months (11·3-18·7). The most common 

grade 3-4 treatment-emergent adverse events were anaemia (15 [4%] of 356 patients 

in the cemiplimab group vs 60 [17%] of 343 in the control group), neutropenia

(three [1%] vs 35 [10%]), and pneumonia (18 [5%] vs 13 [4%]). Treatment-related

deaths occurred in ten (3%) of 356 patients treated with cemiplimab (due to

autoimmune myocarditis, cardiac failure, cardio-respiratory arrest,

cardiopulmonary failure, septic shock, tumour hyperprogression, nephritis,

respiratory failure, [n=1 each] and general disorders or unknown [n=2]) and in

seven (2%) of 343 patients treated with chemotherapy (due to pneumonia and

pulmonary embolism [n=2 each], and cardiac arrest, lung abscess, and myocardial

infarction [n=1 each]). The safety profile of cemiplimab at 35 months, and of

continued cemiplimab plus chemotherapy, was generally consistent with that

previously observed for these treatments, with no new safety signals

INTERPRETATION: At 35 months' follow-up, the survival benefit of cemiplimab for

patients with advanced non-small-cell lung cancer was at least as pronounced as

at 1 year, affirming its use as first-line monotherapy for this population.

Adding chemotherapy to cemiplimab at progression might provide a new second-line

treatment for patients with advanced non-small-cell lung cancer.

FUNDING: Regeneron Pharmaceuticals and Sanofi.

 

Copyright © 2023 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1470-2045(23)00329-7

PMID: 37591293

 

10. Cancer Cell. 2023 Aug 14;41(8):1516-1534.e9. doi: 10.1016/j.ccell.2023.07.005.

Epub 2023 Aug 3.

 

Mammalian SWI/SNF chromatin remodeling complexes promote tyrosine kinase

inhibitor resistance in EGFR-mutant lung cancer.

 

de Miguel FJ(1), Gentile C(2), Feng WW(3), Silva SJ(4), Sankar A(2), Exposito

F(1), Cai WL(4), Melnick MA(1), Robles-Oteiza C(5), Hinkley MM(2), Tsai JA(3),

Hartley AV(3), Wei J(6), Wurtz A(1), Li F(7), Toki MI(8), Rimm DL(9), Homer

R(8), Wilen CB(6), Xiao AZ(10), Qi J(11), Yan Q(12), Nguyen DX(13), Jänne PA(3),

Kadoch C(14), Politi KA(15).

 

Author information:

(1)Yale Cancer Center, New Haven, CT 06520, USA.

(2)Department of Pediatric Oncology, Dana-Farber Cancer Institute and Harvard

Medical School, Boston, MA 02215, USA; Broad Institute of MIT and Harvard,

Cambridge, MA 02142, USA.

(3)Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard

Medical School, Boston, MA, USA.

 

Acquired resistance to tyrosine kinase inhibitors (TKI), such as osimertinib

used to treat EGFR-mutant lung adenocarcinomas, limits long-term efficacy and is

frequently caused by non-genetic mechanisms. Here, we define the chromatin

accessibility and gene regulatory signatures of osimertinib sensitive and

resistant EGFR-mutant cell and patient-derived models and uncover a role for

mammalian SWI/SNF chromatin remodeling complexes in TKI resistance. By profiling

mSWI/SNF genome-wide localization, we identify both shared and cancer cell

line-specific gene targets underlying the resistant state. Importantly, genetic

and pharmacologic disruption of the SMARCA4/SMARCA2 mSWI/SNF ATPases

re-sensitizes a subset of resistant models to osimertinib via inhibition of

mSWI/SNF-mediated regulation of cellular programs governing cell proliferation,

epithelial-to-mesenchymal transition, epithelial cell differentiation, and NRF2

signaling. These data highlight the role of mSWI/SNF complexes in supporting TKI

resistance and suggest potential utility of mSWI/SNF inhibitors in TKI-resistant

lung cancers.

 

Copyright © 2023 The Authors. Published by Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2023.07.005

PMID: 37541244 [Indexed for MEDLINE]

 

11. Lancet Infect Dis. 2023 Aug 28:S1473-3099(23)00375-4. doi:

10.1016/S1473-3099(23)00375-4. Online ahead of print.

 

Improving measurement of tuberculosis care cascades to enhance people-centred

care.

 

Faust L(1), Naidoo P(2), Caceres-Cardenas G(3), Ugarte-Gil C(4), Muyoyeta M(5),

Kerkhoff AD(6), Nagarajan K(7), Satyanarayana S(8), Rakotosamimanana N(9),

Grandjean Lapierre S(10), Adejumo OA(11), Kuye J(12), Oga-Omenka C(13), Pai

M(1), Subbaraman R(14).

 

Author information:

(1)Department of Epidemiology, Biostatistics and Occupational Health, McGill

University, Montréal, QC, Canada; McGill International TB Centre, Montréal, QC,

Canada.

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

of Medicine and Health Sciences, Stellenbosch University, Cape Town, South

Africa.

(3)Instituto de Medicina Tropical Alexander von Humboldt, Universidad Peruana

Cayetano Heredia, Lima, Peru.

 

Care cascades represent the proportion of people reaching milestones in care for

a disease and are widely used to track progress towards global targets for HIV

and other diseases. Despite recent progress in estimating care cascades for

tuberculosis (TB) disease, they have not been routinely applied at national and

subnational levels, representing a lost opportunity for public health impact. As

researchers who have estimated TB care cascades in high-incidence countries

(India, Madagascar, Nigeria, Peru, South Africa, and Zambia), we describe the

utility of care cascades and identify measurement challenges, including the lack

of population-based disease burden data and electronic data capture, the

under-reporting of people with TB navigating fragmented and privatised health

systems, the heterogeneity of TB tests, and the lack of post-treatment

follow-up. We outline an agenda for rectifying these gaps and argue that

improving care cascade measurement is crucial to enhancing people-centred care

and achieving the End TB goals.

 

Copyright © 2023 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(23)00375-4

PMID: 37652066

 

12. ACS Nano. 2023 Aug 29. doi: 10.1021/acsnano.3c04420. Online ahead of print.

 

Quantitation of Circulating Mycobacterium tuberculosis Antigens by Nanopore

Biosensing in Children Evaluated for Pulmonary Tuberculosis in South Africa.

 

Wang X(1), Wei X(1)(2), van der Zalm MM(3), Zhang Z(2), Subramanian N(2), Demers

AM(3)(4), Ghimenton Walters E(3)(5), Hesseling A(3), Liu C(1)(2).

 

Author information:

(1)Department of Chemical Engineering, University of South Carolina, Columbia,

South Carolina 29208, United States.

(2)Biomedical Engineering Program, University of South Carolina, Columbia, South

Carolina 29208, United States.

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

of Medicine and Health Sciences, Stellenbosch University, Cape Town, 8000, South

Africa.

 

Nanopore sensing of proteomic biomarkers lacks accuracy due to the ultralow

abundance of targets, a wide variety of interferents in clinical samples, and

the mismatch between pore and analyte sizes. By converting antigens to DNA

probes via click chemistry and quantifying their characteristic signals, we show

a nanopore assay with several amplification mechanisms to achieve an attomolar

level limit of detection that enables quantitation of the circulating

Mycobacterium tuberculosis (Mtb) antigen ESAT-6/CFP-10 complex in human serum.

The assay's nonsputum-based feature and low-volume sample requirements make it

particularly well-suited for detecting pediatric tuberculosis (TB) disease,

where establishing an accurate diagnosis is greatly complicated by the

paucibacillary nature of respiratory secretions, nonspecific symptoms, and

challenges with sample collection. In the clinical assessment, the assay was

applied to analyze ESAT-6/CFP-10 levels in serum samples collected during

baseline investigation for TB in 75 children, aged 0-12 years, enrolled in a

diagnostic study conducted in Cape Town, South Africa. This nanopore assay

showed superior sensitivity in children with confirmed TB (94.4%) compared to

clinical "gold standard" diagnostic technologies (Xpert MTB/RIF 44.4% and Mtb

culture 72.2%) and filled the diagnostic gap for children with unconfirmed TB,

where these traditional technologies fell short. We envision that, in

combination with automated sample processing and portable nanopore devices, this

methodology will offer a powerful tool to support the diagnosis of pulmonary TB

in children.

 

DOI: 10.1021/acsnano.3c04420

PMID: 37643288

 

13. J Clin Oncol. 2023 Aug 21:JCO2202561. doi: 10.1200/JCO.22.02561. Online ahead of print.

 

Phase II Trial of Atezolizumab Combined With Carboplatin and Pemetrexed for

Patients With Advanced Nonsquamous Non-Small-Cell Lung Cancer With Untreated

Brain Metastases (Atezo-Brain, GECP17/05).

 

Nadal E(1)(2), Rodríguez-Abreu D(3), Simó M(4), Massutí B(5), Juan O(6),

Huidobro G(7), López R(8), De Castro J(9), Estival A(10), Mosquera J(11),

Sullivan I(12), Felip E(13), Blasco A(14), Guirado M(15), Pereira E(16),

Vilariño N(1)(2)(4), Navarro V(1), Bruna J(4).

 

Author information:

(1)Department of Medical Oncology, Institut Català d'Oncologia (ICO),

L'Hospitalet de Llobregat, Barcelona, Spain.

(2)Preclinical and Experimental Research in Thoracic Tumors (PReTT) Group,

Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL),

L'Hospitalet de Llobregat, Barcelona, Spain.

(3)Department of Medical Oncology, Complejo Hospitalario Universitario

Insular-Materno Infantil de Gran Canaria, Universidad de Las Palmas de Gran

Canaria, Las Palmas de Gran Canaria, Spain.

 

PURPOSE: The Atezo-Brain study evaluated atezolizumab combined with chemotherapy

in patients with advanced non-small-cell lung cancer (NSCLC) with untreated

brain metastases, a population traditionally excluded from trials.

METHODS: This single-arm phase II clinical trial enrolled patients with advanced

nonsquamous NSCLC with untreated brain metastases without neurologic symptoms or

asymptomatic with medical treatment. Dexamethasone was allowed up to 4 mg once

daily. Atezolizumab plus carboplatin and pemetrexed was given for four to six

cycles followed by atezolizumab plus pemetrexed until progression for a maximum

of 2 years. The primary end points were to determine the progression-free

survival (PFS) rate at 12 weeks and the incidence of grade ≥3 adverse events

during the first 9 weeks. Intracranial outcomes were assessed using response

assessment in neuro-oncology brain metastases criteria.

RESULTS: Forty patients were enrolled and 22 (55%) were receiving

corticosteroids at baseline. The overall 12-week PFS rate was 62.2% (95%

credibility interval [CrI], 47.1 to 76.2). The rate of grade 3/4 adverse events

during the first 9 weeks was 27.5%. Most neurologic events were grade 1 and 2

but five patients (12.5%) experienced grade 3-4 neurologic events. With a median

follow-up of 31 months, intracranial median PFS was 6.9 months and response rate

was 42.7% (95% CrI, 28.1 to 57.9). Systemic median PFS was 8.9 months and

response rate was 45% (95% CrI, 28.1 to 57.9). The median overall survival (OS)

was 11.8 months (95% CI, 7.6 to 16.9) and the 2-year OS rate was 27.5% (95% CI,

16.6 to 45.5).

CONCLUSION: Atezolizumab plus carboplatin and pemetrexed demonstrates activity

in patients with advanced nonsquamous NSCLC with untreated brain metastases with

an acceptable safety profile.

 

DOI: 10.1200/JCO.22.02561

PMID: 37603816

 

14. Nat Commun. 2023 Aug 17;14(1):4644. doi: 10.1038/s41467-023-40264-3.

 

MenT nucleotidyltransferase toxins extend tRNA acceptor stems and can be

inhibited by asymmetrical antitoxin binding.

 

Xu X(#)(1), Usher B(#)(2), Gutierrez C(3), Barriot R(1), Arrowsmith TJ(2), Han

X(1), Redder P(1), Neyrolles O(3), Blower TR(4), Genevaux P(5).

 

Author information:

(1)Laboratoire de Microbiologie et Génétique Moléculaires (LMGM), Centre de

Biologie Intégrative (CBI), Université de Toulouse, CNRS, Université Toulouse

III - Paul Sabatier (UT3), Toulouse, France.

(2)Department of Biosciences, Durham University, South Road, Durham, DH1 3LE,

UK.

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

Toulouse, CNRS, Université Toulouse III - Paul Sabatier (UT3), Toulouse, France.

 

Mycobacterium tuberculosis, the bacterium responsible for human tuberculosis,

has a genome encoding a remarkably high number of toxin-antitoxin systems of

largely unknown function. We have recently shown that the M. tuberculosis genome

encodes four of a widespread, MenAT family of nucleotidyltransferase

toxin-antitoxin systems. In this study we characterize MenAT1, using tRNA

sequencing to demonstrate MenT1 tRNA modification activity. MenT1 activity is

blocked by MenA1, a short protein antitoxin unrelated to the MenA3 kinase. X-ray

crystallographic analysis shows blockage of the conserved MenT fold by

asymmetric binding of MenA1 across two MenT1 protomers, forming a heterotrimeric

toxin-antitoxin complex. Finally, we also demonstrate tRNA modification by toxin

MenT4, indicating conserved activity across the MenT family. Our study

highlights variation in tRNA target preferences by MenT toxins, selective use of

nucleotide substrates, and diverse modes of MenA antitoxin activity.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41467-023-40264-3

PMCID: PMC10435456

PMID: 37591829 [Indexed for MEDLINE]

 

15. Lancet Infect Dis. 2023 Aug 14:S1473-3099(23)00371-7. doi:

10.1016/S1473-3099(23)00371-7. Online ahead of print.

 

Incidence and risk factors of tuberculosis among 420 854 household contacts of

patients with tuberculosis in the 100 Million Brazilian Cohort (2004-18): a

cohort study.

 

Pinto PFPS(1), Teixeira CSS(2), Ichihara MY(2), Rasella D(3), Nery JS(4), Sena

SOL(2), Brickley EB(5), Barreto ML(4), Sanchez MN(6), Pescarini JM(7).

 

Author information:

(1)Centro de Integração de Dados e Conhecimentos para Saúde (Cidacs), Fundação

Oswaldo Cruz, Salvador, Brazil. Electronic address: priferscaff@hotmail.com.

(2)Centro de Integração de Dados e Conhecimentos para Saúde (Cidacs), Fundação

Oswaldo Cruz, Salvador, Brazil.

(3)Centro de Integração de Dados e Conhecimentos para Saúde (Cidacs), Fundação

Oswaldo Cruz, Salvador, Brazil; Institute of Global Health (ISGlobal), Hospital

Clínic-Universitat de Barcelona, Barcelona, Spain.

 

BACKGROUND: Although household contacts of patients with tuberculosis are known

to be particularly vulnerable to tuberculosis, the published evidence focused on

this group at high risk within the low-income and middle-income country context

remains sparse. Using nationwide data from Brazil, we aimed to estimate the

incidence and investigate the socioeconomic and clinical determinants of

tuberculosis in a cohort of contacts of tuberculosis patients.

METHODS: In this cohort study, we linked individual socioeconomic and

demographic data from the 100 Million Brazilian Cohort to mortality data and

tuberculosis registries, identified contacts of tuberculosis index patients

diagnosed from Jan 1, 2004 to Dec 31, 2018, and followed up the contacts until

the contact's subsequent tuberculosis diagnosis, the contact's death, or Dec 31,

2018. We investigated factors associated with active tuberculosis using

multilevel Poisson regressions, allowing for municipality-level and

household-level random effects.

FINDINGS: We studied 420 854 household contacts of 137 131 tuberculosis index

patients. During the 15 years of follow-up (median 4·4 years [IQR 1·9-7·6]), we 

detected 8953 contacts with tuberculosis. The tuberculosis incidence among

contacts was 427·8 per 100 000 person-years at risk (95% CI 419·1-436·8),

16-times higher than the incidence in the general population (26·2 [26·1-26·3])

and the risk was prolonged. Tuberculosis incidence was associated with the index

patient being preschool aged (<5 years; adjusted risk ratio 4·15 [95% CI

3·26-5·28]) or having pulmonary tuberculosis (2·84 [2·55-3·17]).

INTERPRETATION: The high and sustained risk of tuberculosis among contacts

reinforces the need to systematically expand and strengthen contact tracing and

preventive treatment policies in Brazil in order to achieve national and

international targets for tuberculosis elimination.

FUNDING: Wellcome Trust and Brazilian Ministry of Health.

 

Copyright © 2023 The Author(s). Published by Elsevier Ltd. This is an Open

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

rights reserved.

 

DOI: 10.1016/S1473-3099(23)00371-7

PMID: 37591301

 

16. PLoS Biol. 2023 Aug 17;21(8):e3002231. doi: 10.1371/journal.pbio.3002231.

eCollection 2023 Aug.

 

NCoR1 controls Mycobacterium tuberculosis growth in myeloid cells by regulating

the AMPK-mTOR-TFEB axis.

 

Biswas VK(1)(2), Sen K(1)(3), Ahad A(1), Ghosh A(1)(2), Verma S(4), Pati R(1),

Prusty S(1)(3), Nayak SP(1), Podder S(1)(2), Kumar D(4), Gupta B(2), Raghav

SK(1)(2).

 

Author information:

(1)Immuno-genomics & Systems Biology Laboratory, Institute of Life Sciences

(ILS), Bhubaneswar, India.

(2)School of Biotechnology, Kalinga Institute of Industrial Technology (KIIT),

Bhubaneswar, India.

(3)Regional Centre for Biotechnology, Faridabad, India.

(4)Molecular Medicine: Cellular Immunology, International Centre for Genetic

Engineering and Biotechnology (ICGEB), New Delhi, India.

 

Mycobacterium tuberculosis (Mtb) defends host-mediated killing by repressing the

autophagolysosome machinery. For the first time, we report NCoR1 co-repressor as

a crucial host factor, controlling Mtb growth in myeloid cells by regulating

both autophagosome maturation and lysosome biogenesis. We found that the dynamic

expression of NCoR1 is compromised in human peripheral blood mononuclear cells

(PBMCs) during active Mtb infection, which is rescued upon prolonged

anti-mycobacterial therapy. In addition, a loss of function in myeloid-specific

NCoR1 considerably exacerbates the growth of M. tuberculosis in vitro in THP1

differentiated macrophages, ex vivo in bone marrow-derived macrophages (BMDMs),

and in vivo in NCoR1MyeKO mice. We showed that NCoR1 depletion controls the

AMPK-mTOR-TFEB signalling axis by fine-tuning cellular adenosine triphosphate

(ATP) homeostasis, which in turn changes the expression of proteins involved in

autophagy and lysosomal biogenesis. Moreover, we also showed that the treatment

of NCoR1 depleted cells by Rapamycin, Antimycin-A, or Metformin rescued the TFEB

activity and LC3 levels, resulting in enhanced Mtb clearance. Similarly,

expressing NCoR1 exogenously rescued the AMPK-mTOR-TFEB signalling axis and Mtb

killing. Overall, our data revealed a central role of NCoR1 in Mtb pathogenesis

in myeloid cells.

 

Copyright: © 2023 Biswas et al. This is an open access article distributed under

the terms of the Creative Commons Attribution License, which permits

unrestricted use, distribution, and reproduction in any medium, provided the

original author and source are credited.

 

DOI: 10.1371/journal.pbio.3002231

PMCID: PMC10465006

PMID: 37590294 [Indexed for MEDLINE]

 

17. Nat Commun. 2023 Aug 3;14(1):4655. doi: 10.1038/s41467-023-40349-z.

 

Neoadjuvant Afatinib for stage III EGFR-mutant non-small cell lung cancer: a

phase II study.

 

Bian D(#)(1), Sun L(#)(1), Hu J(#)(1), Duan L(#)(1), Xia H(#)(1), Zhu X(1), Sun

F(1), Zhang L(2), Yu H(3), Xiong Y(1), Huang Z(1)(4), Zhao D(1), Song N(1), Yang

J(1), Bao X(5), Wu W(6), Huang J(7), He W(8), Zhu Y(9), Jiang G(10), Zhang

P(11)(12)(13).

 

Author information:

(1)Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji

University School of Medicine, Shanghai, 200433, China.

(2)Central Laboratory, Shanghai Pulmonary Hospital, School of Medicine, Tongji

University, Shanghai, 200433, China.

(3)Department of Animal Experimental Center, Shanghai Pulmonary Hospital, Tongji

University School of Medicine, Shanghai, 200433, China.

 

Afatinib, an irreversible ErbB-family blocker, could improve the survival of

advanced epidermal growth factor receptor (EGFR)-mutant non-small cell lung

cancer patients (NSCLCm+). This phase II trial (NCT04201756) aimed to assess the

feasibility of neoadjuvant Afatinib treatment for stage III NSCLCm+. Forty-seven

patients received neoadjuvant Afatinib treatment (40 mg daily). The primary

endpoint was objective response rate (ORR). Secondary endpoints included

pathological complete response (pCR) rate, pathological downstaging rate,

margin-free resection (R0) rate, event-free survival, disease-free survival,

progression-free survival, overall survival, treatment-related adverse events

(TRAEs). The ORR was 70.2% (95% CI: 56.5% to 84.0%), meeting the pre-specified

endpoint. The major pathological response (MPR), pCR, pathological downstaging,

and R0 rates were 9.1%, 3.0%, 57.6%, and 87.9%, respectively. The median

survivals were not reached. The most common TRAEs were diarrhea (78.7%) and rash

(78.7%). Only three patients experienced grade 3/4 TRAEs. Biomarker analysis and

tumor microenvironment dynamics by bulk RNA sequencing were included as

predefined exploratory endpoints. CISH expression was a promising marker for

Afatinib response (AUC = 0.918). In responders, compared to baseline samples,

increasing T-cell- and B-cell-related features were observed in post-treatment

tumor and lymph-node samples, respectively. Neoadjuvant Afatinib is feasible for

stage III NSCLC+ patients and leads to dynamic changes in the tumor

microenvironment.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41467-023-40349-z

PMCID: PMC10400609

PMID: 37537219 [Indexed for MEDLINE]

 

18. J Clin Oncol. 2023 Aug 20;41(24):e63-e72. doi: 10.1200/JCO.23.01055. Epub 2023 Jul 11.

 

Therapy for Stage IV Non-Small-Cell Lung Cancer With Driver Alterations: ASCO

Living Guideline, Version 2023.2.

 

Owen DH(1), Singh N(2), Ismaila N(3), Masters G(4), Riely GJ(5), Robinson AG(6),

Schneider BJ(7), Jaiyesimi IA(8).

 

Author information:

(1)Ohio State University, Columbus OH.

(2)Postgraduate Institute of Medical Education and Research, Chandigarh, India.

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

 

Update of

    J Clin Oncol. 2022 Oct 1;40(28):3310-3322.

 

Living guidelines are developed for selected topic areas with rapidly evolving

evidence that drives frequent change in clinical practice. Living guidelines are

updated on a regular schedule by a standing expert panel that systematically

reviews the health literature on a continuous basis; as described in the ASCO

Guidelines Methodology Manual. ASCO Living Guidelines follow the ASCO Conflict

of Interest Policy Implementation for Clinical Practice Guidelines. Living

Guidelines and updates are not intended to substitute for independent

professional judgment of the treating provider and do not account for individual

variation among patients. See appendix for disclaimers and other important

information (Appendix 1 and Appendix 2). Updates are published regularly and can

be found at https://ascopubs.org/nsclc-da-living-guideline.

 

DOI: 10.1200/JCO.23.01055

PMID: 37433095 [Indexed for MEDLINE]

 

19. J Exp Med. 2023 Aug 7;220(8):e20222090. doi: 10.1084/jem.20222090. Epub 2023 Apr 25.

 

CD30 co-stimulation drives differentiation of protective T cells during

Mycobacterium tuberculosis infection.

 

Foreman TW(1), Nelson CE(1), Sallin MA(1), Kauffman KD(1), Sakai S(1),

Otaizo-Carrasquero F(2), Myers TG(2), Barber DL(1).

 

Author information:

(1)T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National

Institute of Allergy and Infectious Diseases, National Institutes of Health ,

Bethesda, MD, USA.

(2)Genomic Technologies Section, Research Technologies Branch, National

Institute of Allergy and Infectious Diseases, National Institutes of Health ,

Bethesda, MD, USA.

 

Comment in

    J Exp Med. 2023 Aug 7;220(8):

 

Control of Mycobacterium tuberculosis (Mtb) infection requires generation of T

cells that migrate to granulomas, complex immune structures surrounding sites of

bacterial replication. Here we compared the gene expression profiles of T cells

in pulmonary granulomas, bronchoalveolar lavage, and blood of Mtb-infected

rhesus macaques to identify granuloma-enriched T cell genes. TNFRSF8/CD30 was

among the top genes upregulated in both CD4 and CD8 T cells from granulomas. In

mice, CD30 expression on CD4 T cells is required for survival of Mtb infection,

and there is no major role for CD30 in protection by other cell types.

Transcriptomic comparison of WT and CD30-/- CD4 T cells from the lungs of

Mtb-infected mixed bone marrow chimeric mice showed that CD30 directly promotes

CD4 T cell differentiation and the expression of multiple effector molecules.

These results demonstrate that the CD30 co-stimulatory axis is highly

upregulated on granuloma T cells and is critical for protective T cell responses

against Mtb infection.

 

This is a work of the U.S. Government and is not subject to copyright protection

in the United States. Foreign copyrights may apply.

 

DOI: 10.1084/jem.20222090

PMCID: PMC10130742

PMID: 37097292 [Indexed for MEDLINE]

 

 


上一篇: No.9

下一篇: No.7