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.6

发布时间:2023-07-17 浏览次数:
字号: + - 14

PubMed

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

Filters applied: from 2023/6/1 - 2023/6/30.

 

1. N Engl J Med. 2023 Jun 28. doi: 10.1056/NEJMoa2215530. Online ahead of print.

 

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, …

 

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

 

2. CA Cancer J Clin. 2023 Jun 17. doi: 10.3322/caac.21785. Online ahead of print.

 

Clinical insights into small cell lung cancer: Tumor heterogeneity, diagnosis,

therapy, and future directions.

 

Megyesfalvi Z(1)(2)(3), Gay CM(4), Popper H(5), Pirker R(6), Ostoros G(3), Heeke

S(4), Lang C(1)(7), Hoetzenecker K(1), Schwendenwein A(1), Boettiger K(1), Bunn

PA Jr(8), Renyi-Vamos F(2)(3), Schelch K(1)(9), Prosch H(10), Byers LA(4),

Hirsch FR(11)(12), Dome B(1)(2)(3)(13).

 

Author information:

(1)Department of Thoracic Surgery, Comprehensive Cancer Center, Medical

University of Vienna, Vienna, Austria.

(2)Department of Thoracic Surgery, Semmelweis University and National Institute

of Oncology, Budapest, Hungary.

(3)National Koranyi Institute of Pulmonology, Budapest, Hungary.

Small cell lung cancer (SCLC) is characterized by rapid growth and high

metastatic capacity. It has strong epidemiologic and biologic links to tobacco

carcinogens. Although the majority of SCLCs exhibit neuroendocrine features, an

important subset of tumors lacks these properties. Genomic profiling of SCLC

reveals genetic instability, almost universal inactivation of the tumor

suppressor genes TP53 and RB1, and a high mutation burden. Because of early

metastasis, only a small fraction of patients are amenable to curative-intent

lung resection, and these individuals require adjuvant platinum-etoposide

chemotherapy. Therefore, the vast majority of patients are currently being

treated with chemoradiation with or without immunotherapy. In patients with

disease confined to the chest, standard therapy includes thoracic radiotherapy

and concurrent platinum-etoposide chemotherapy. Patients with metastatic

(extensive-stage) disease are treated with a combination of platinum-etoposide

chemotherapy plus immunotherapy with an anti-programmed death-ligand 1

monoclonal antibody. Although SCLC is initially very responsive to

platinum-based chemotherapy, these responses are transient because of the

development of drug resistance. In recent years, the authors have witnessed an

accelerating pace of biologic insights into the disease, leading to the

redefinition of the SCLC classification scheme. This emerging knowledge of SCLC

molecular subtypes has the potential to define unique therapeutic

vulnerabilities. Synthesizing these new discoveries with the current knowledge

of SCLC biology and clinical management may lead to unprecedented advances in

SCLC patient care. Here, the authors present an overview of multimodal clinical

approaches in SCLC, with a special focus on illuminating how recent advancements

in SCLC research could accelerate clinical development.

 

© 2023 The Authors. CA: A Cancer Journal for Clinicians published by Wiley

Periodicals LLC on behalf of American Cancer Society.

 

DOI: 10.3322/caac.21785

PMID: 37329269

 

3. N Engl J Med. 2023 Jul 13;389(2):137-147. doi: 10.1056/NEJMoa2304594. Epub 2023 Jun 4.

 

Overall Survival with Osimertinib in Resected EGFR-Mutated NSCLC.

 

Tsuboi M(1), Herbst RS(1), John T(1), Kato T(1), Majem M(1), Grohé C(1), Wang

J(1), Goldman JW(1), Lu S(1), Su WC(1), de Marinis F(1), Shepherd FA(1), Lee

KH(1), Le NT(1), Dechaphunkul A(1), Kowalski D(1), Poole L(1), Bolanos A(1),

Rukazenkov Y(1), Wu YL(1); ADAURA Investigators.

 

Author information:

(1)From the Department of Thoracic Surgery and Oncology, National Cancer Center

Hospital East, Kashiwa (M.T.), the Department of Thoracic Oncology, Kanagawa

Cancer Center, Yokohama (T.K.) - both in Japan; the Section of Medical Oncology,

Yale School of Medicine and Yale Cancer Center, New Haven, CT (R.S.H.);…

 

BACKGROUND: Among patients with resected, epidermal growth factor receptor

(EGFR)-mutated, stage IB to IIIA non-small-cell lung cancer (NSCLC), adjuvant

osimertinib therapy, with or without previous adjuvant chemotherapy, resulted in

significantly longer disease-free survival than placebo in the ADAURA trial. We

report the results of the planned final analysis of overall survival.

METHODS: In this phase 3, double-blind trial, we randomly assigned eligible

patients in a 1:1 ratio to receive osimertinib (80 mg once daily) or placebo

until disease recurrence was observed, the trial regimen was completed (3

years), or a discontinuation criterion was met. The primary end point was

investigator-assessed disease-free survival among patients with stage II to IIIA

disease. Secondary end points included disease-free survival among patients with

stage IB to IIIA disease, overall survival, and safety.

RESULTS: Of 682 patients who underwent randomization, 339 received osimertinib

and 343 received placebo. Among patients with stage II to IIIA disease, the

5-year overall survival was 85% in the osimertinib group and 73% in the placebo

group (overall hazard ratio for death, 0.49; 95.03% confidence interval [CI],

0.33 to 0.73; P<0.001). In the overall population (patients with stage IB to

IIIA disease), the 5-year overall survival was 88% in the osimertinib group and

78% in the placebo group (overall hazard ratio for death, 0.49; 95.03% CI, 0.34

to 0.70; P<0.001). One new serious adverse event, pneumonia related to

coronavirus disease 2019, was reported after the previously published

data-cutoff date (the event was not considered by the investigator to be related

to the trial regimen, and the patient fully recovered). Adjuvant osimertinib had

a safety profile consistent with that in the primary analysis.

CONCLUSIONS: Adjuvant osimertinib provided a significant overall survival

benefit among patients with completely resected, EGFR-mutated, stage IB to IIIA

NSCLC. (Funded by AstraZeneca; ADAURA ClinicalTrials.gov number, NCT02511106.).

 

Copyright © 2023 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2304594

PMID: 37272535

 

4. N Engl J Med. 2023 Jun 3. doi: 10.1056/NEJMoa2302983. Online ahead of print.

 

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;…

 

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

 

5. Nat Commun. 2023 Jun 28;14(1):3828. doi: 10.1038/s41467-023-39300-z.

 

DprE2 is a molecular target of the anti-tubercular nitroimidazole compounds

pretomanid and delamanid.

 

Abrahams KA(#)(1), Batt SM(#)(1), Gurcha SS(1), Veerapen N(1), Bashiri G(2),

Besra GS(3).

 

Author information:

(1)Institute of Microbiology and Infection, School of Biosciences, University of

Birmingham, Edgbaston, Birmingham, B15 2TT, UK.

(2)Laboratory of Molecular and Microbial Biochemistry, School of Biological

Sciences, University of Auckland, 3A Symonds Street, Auckland, 1010, New

Zealand.

(3)Institute of Microbiology and Infection, School of Biosciences, University of

Birmingham, Edgbaston, Birmingham, B15 2TT, UK. g.besra@bham.ac.uk.

(#)Contributed equally

 

Mycobacterium tuberculosis is one of the global leading causes of death due to a

single infectious agent. Pretomanid and delamanid are new antitubercular agents

that have progressed through the drug discovery pipeline. These compounds are

bicyclic nitroimidazoles that act as pro-drugs, requiring activation by a

mycobacterial enzyme; however, the precise mechanisms of action of the active

metabolite(s) are unclear. Here, we identify a molecular target of activated

pretomanid and delamanid: the DprE2 subunit of

decaprenylphosphoribose-2'-epimerase, an enzyme required for the synthesis of

cell wall arabinogalactan. We also provide evidence for an NAD-adduct as the

active metabolite of pretomanid. Our results highlight DprE2 as a potential

antimycobacterial target and provide a foundation for future exploration into

the active metabolites and clinical development of pretomanid and delamanid.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41467-023-39300-z

PMCID: PMC10307805

PMID: 37380634 [Indexed for MEDLINE]

 

6. Nat Commun. 2023 Jun 28;14(1):3830. doi: 10.1038/s41467-023-39528-9.

 

A landscape of response to drug combinations in non-small cell lung cancer.

 

Nair NU(1), Greninger P(2), Zhang X(3), Friedman AA(2), Amzallag A(2), Cortez

E(2), Sahu AD(4), Lee JS(5), Dastur A(2), Egan RK(2), Murchie E(2), Ceribelli

M(3), Crowther GS(2), Beck E(3), McClanaghan J(2), Klump-Thomas C(3), Boisvert

JL(2), Damon LJ(2), Wilson KM(3), Ho J(2), Tam A(2), McKnight C(3), Michael

S(3), Itkin Z(3), Garnett MJ(6), Engelman JA(2), Haber DA(2)(3), Thomas

CJ(#)(7)(8), Ruppin E(#)(9), Benes CH(#)(10).

 

Author information:

(1)Cancer Data Science Laboratory, Center for Cancer Research, National Cancer

Institute, National Institutes of Health, Bethesda, MD, USA.

(2)Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.

(3)Howard Hughes Medical Institute, Bethesda, MD, USA.

Combination of anti-cancer drugs is broadly seen as way to overcome the

often-limited efficacy of single agents. The design and testing of combinations

are however very challenging. Here we present a uniquely large dataset screening

over 5000 targeted agent combinations across 81 non-small cell lung cancer cell

lines. Our analysis reveals a profound heterogeneity of response across the

tumor models. Notably, combinations very rarely result in a strong gain in

efficacy over the range of response observable with single agents. Importantly,

gain of activity over single agents is more often seen when co-targeting

functionally proximal genes, offering a strategy for designing more efficient

combinations. Because combinatorial effect is strongly context specific, tumor

specificity should be achievable. The resource provided, together with an

additional validation screen sheds light on major challenges and opportunities

in building efficacious combinations against cancer and provides an opportunity

for training computational models for synergy prediction.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41467-023-39528-9

PMCID: PMC10307832

PMID: 37380628 [Indexed for MEDLINE]

 

7. J Clin Oncol. 2023 Jun 28:101200JCO2300515. doi: 10.1200/JCO.23.00515. Online

ahead of print.

 

Lazertinib Versus Gefitinib as First-line Treatment in Patients With

EGFR-mutated Advanced Non-Small Cell Lung Cancer (NSCLC): Results From LASER301.

 

Cho BC(1), Ahn MJ(2), Kang JH(3), Soo RA(4), Reungwetwattana T(5), Chih-Hsin

Yang J(6), Cicin I(7), Kim DW(8), Wu YL(9), Lu S(10), Lee KH(11), Pang YK(12),

Zimina A(13), Fong CH(14), Poddubskaya E(15), Sezer A(16), How SH(17),

Danchaivijitr P(18), Kim Y(19), Lim Y(19), An T(19), Lee H(19), Byun HM(19),

Zaric B(20).

 

Author information:

(1)Division of Medical Oncology, Yonsei Cancer Center, Yonsei University College

of Medicine, Seoul, Republic of Korea.

(2)Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,

Republic of Korea.

(3)Division of Medical Oncology, Department of Internal Medicine, Seoul St.

Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.

PURPOSE: Lazertinib is a potent, central nervous system (CNS)-penetrant,

third-generation epidermal growth factor receptor (EGFR) tyrosine kinase

inhibitor (TKI). This global, phase 3 study (LASER301) compared lazertinib

versus gefitinib in treatment-naïve patients with EGFR-mutated (exon 19 deletion

[ex19del]/L858R) locally advanced or metastatic non-small cell lung cancer

(NSCLC).

METHODS: Patients were ≥18 years with no prior systemic anti-cancer therapy.

Neurologically stable patients with CNS metastases were allowed. Patients were

randomized 1:1 to lazertinib 240 mg once daily (qd) orally (po) or gefitinib 250

mg qd po, stratified by mutation status and race. The primary endpoint was

investigator-assessed progression-free survival (PFS) by RECIST v1.1.

RESULTS: Overall, 393 patients received double-blind study treatment across 96

sites in 13 countries. Median PFS was significantly longer with lazertinib than

with gefitinib (20.6 vs 9.7 months; hazard ratio [HR], 0.45; 95% confidence

interval [CI], 0.34 to 0.58; P<0.001). The PFS benefit of lazertinib over

gefitinib was consistent across all predefined subgroups. The objective response

rate was 76% in both groups (odds ratio, 0.99; 95% CI, 0.62 to 1.59). Median

duration of response was 19.4 months (95% CI, 16.6 to 24.9) with lazertinib

versus 8.3 months (95% CI, 6.9 to 10.9) with gefitinib. Overall survival data

were immature at the interim analysis (29% maturity). The 18-month survival rate

was 80% with lazertinib and 72% with gefitinib (HR, 0.74; 95% CI, 0.51 to 1.08;

P=0.116). Observed safety of both treatments was consistent with their

previously reported safety profiles.

CONCLUSION: Lazertinib demonstrated significant efficacy improvement compared

with gefitinib in the first-line treatment of EGFR-mutated advanced NSCLC, with

a manageable safety profile.

 

DOI: 10.1200/JCO.23.00515

PMID: 37379502

 

8. Mol Biol Evol. 2023 Jun 1;40(6):msad131. doi: 10.1093/molbev/msad131.

 

Analysis of Genome-Wide Mutational Dependence in Naturally Evolving

Mycobacterium tuberculosis Populations.

 

Green AG(1), Vargas R(1)(2), Marin MG(1), Freschi L(1), Xie J(3), Farhat

MR(1)(4).

 

Author information:

(1)Department of Biomedical Informatics, Harvard Medical School, Boston, MA,

USA.

(2)Center for Computational Biomedicine, Harvard Medical School, Boston, MA,

USA.

(3)Department of Genetics, Johns Hopkins School of Medicine, Baltimore, MD, USA.

(4)Division of Pulmonary and Critical Care Medicine, Massachusetts General

Hospital, Boston, MA, USA.

 

Pathogenic microorganisms are in a perpetual struggle for survival in changing

host environments, where host pressures necessitate changes in pathogen

virulence, antibiotic resistance, or transmissibility. The genetic basis of

phenotypic adaptation by pathogens is difficult to study in vivo. In this work,

we develop a phylogenetic method to detect genetic dependencies that promote

pathogen adaptation using 31,428 in vivo sampled Mycobacterium tuberculosis

genomes, a globally prevalent bacterial pathogen with increasing levels of

antibiotic resistance. We find that dependencies between mutations are enriched

in antigenic and antibiotic resistance functions and discover 23 mutations that

potentiate the development of antibiotic resistance. Between 11% and 92% of

resistant strains harbor a dependent mutation acquired after a

resistance-conferring variant. We demonstrate the pervasiveness of genetic

dependency in adaptation of naturally evolving populations and the utility of

the proposed computational approach.

 

© The Author(s) 2023. Published by Oxford University Press on behalf of Society

for Molecular Biology and Evolution.

 

DOI: 10.1093/molbev/msad131

PMCID: PMC10292908

PMID: 37352142 [Indexed for MEDLINE]

 

9. J Clin Oncol. 2023 Jun 16:JCO2300059. doi: 10.1200/JCO.23.00059. Online ahead of print.

 

First-in-Human, Phase I Dose-Escalation and Dose-Expansion Study of Trophoblast

Cell-Surface Antigen 2-Directed Antibody-Drug Conjugate Datopotamab Deruxtecan

in Non-Small-Cell Lung Cancer: TROPION-PanTumor01.

 

Shimizu T(1)(2), Sands J(3), Yoh K(4), Spira A(5), Garon EB(6), Kitazono S(7),

Johnson ML(8), Meric-Bernstam F(9), Tolcher AW(10), Yamamoto N(1), Greenberg

J(11), Kawasaki Y(12), Zebger-Gong H(11), Kobayashi F(13), Phillips P(12),

Lisberg AE(6), Heist RS(14).

 

Author information:

(1)National Cancer Center Hospital, Tokyo, Japan.

(2)Wakayama Medical University Hospital, Wakayama, Japan.

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

PURPOSE: This first-in-human, dose-escalation and dose-expansion study evaluated

the safety, tolerability, and antitumor activity of datopotamab deruxtecan

(Dato-DXd), a novel trophoblast cell-surface antigen 2 (TROP2)-directed

antibody-drug conjugate in solid tumors, including advanced non-small-cell lung

cancer (NSCLC).

PATIENTS AND METHODS: Adults with locally advanced/metastatic NSCLC received

0.27-10 mg/kg Dato-DXd once every 3 weeks during escalation or 4, 6, or 8 mg/kg

Dato-DXd once every 3 weeks during expansion. Primary end points were safety and

tolerability. Secondary end points included objective response rate (ORR),

survival, and pharmacokinetics.

RESULTS: Two hundred ten patients received Dato-DXd, including 180 in the 4-8

mg/kg dose-expansion cohorts. This population had a median of three prior lines

of therapy. The maximum tolerated dose was 8 mg/kg once every 3 weeks; the

recommended dose for further development was 6 mg/kg once every 3 weeks. In

patients receiving 6 mg/kg (n = 50), median duration on study, including

follow-up, and median exposure were 13.3 and 3.5 months, respectively. The most

frequent any-grade treatment-emergent adverse events (TEAEs) were nausea (64%),

stomatitis (60%), and alopecia (42%). Grade ≥3 TEAEs and treatment-related AEs

occurred in 54% and 26% of patients, respectively. Interstitial lung disease

adjudicated as drug-related (two grade 2 and one grade 4) occurred in three of

50 patients (6%). The ORR was 26% (95% CI, 14.6 to 40.3), and median duration of

response was 10.5 months; median progression-free survival and overall survival

were 6.9 months (95% CI, 2.7 to 8.8 months) and 11.4 months (95% CI, 7.1 to 20.6

months), respectively. Responses occurred regardless of TROP2 expression.

CONCLUSION: Promising antitumor activity and a manageable safety profile were

seen with Dato-DXd in heavily pretreated patients with advanced NSCLC. Further

investigation as first-line combination therapy in advanced NSCLC and as

monotherapy in the second-line setting and beyond is ongoing.

 

DOI: 10.1200/JCO.23.00059

PMID: 37327461

 

10. Cell Host Microbe. 2023 Jun 14;31(6):921-923. doi: 10.1016/j.chom.2023.05.021.

 

BCG: From veins to correlates.

 

Suliman S(1).

 

Author information:

(1)Zuckerberg San Francisco General Hospital, Department of Medicine, Division

of Experimental Medicine, University of California San Francisco, San Francisco,

CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA. Electronic address:

sara.suliman@ucsf.edu.

 

In this issue of Cell Host & Microbe, Darrah et al. define immune responses

against Mycobacterium tuberculosis (Mtb) infection following intravenous

Bacille-Calmette Guérin (BCG) vaccination of nonhuman primates. The results

provide candidate correlates of protection for examination in clinical trials of

TB vaccines against Mtb infection and tuberculosis (TB) disease.

 

Copyright © 2023 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.chom.2023.05.021

PMID: 37321176 [Indexed for MEDLINE]

 

11. PLoS Biol. 2023 Jun 15;21(6):e3002159. doi: 10.1371/journal.pbio.3002159.

eCollection 2023 Jun.

 

Autophagy prevents early proinflammatory responses and neutrophil recruitment

during Mycobacterium tuberculosis infection without affecting pathogen burden in

macrophages.

 

Kinsella RL(1), Kimmey JM(1), Smirnov A(1), Woodson R(1), Gaggioli MR(1), Chavez

SM(1), Kreamalmeyer D(1), Stallings CL(1).

 

Author information:

(1)Department of Molecular Microbiology, Center for Women's Infectious Disease

Research, Washington University School of Medicine, St. Louis, Missouri, United

States of America.

 

The immune response to Mycobacterium tuberculosis infection determines

tuberculosis disease outcomes, yet we have an incomplete understanding of what

immune factors contribute to a protective immune response. Neutrophilic

inflammation has been associated with poor disease prognosis in humans and in

animal models during M. tuberculosis infection and, therefore, must be tightly

regulated. ATG5 is an essential autophagy protein that is required in innate

immune cells to control neutrophil-dominated inflammation and promote survival

during M. tuberculosis infection; however, the mechanistic basis for how ATG5

regulates neutrophil recruitment is unknown. To interrogate what innate immune

cells require ATG5 to control neutrophil recruitment during M. tuberculosis

infection, we used different mouse strains that conditionally delete Atg5 in

specific cell types. We found that ATG5 is required in CD11c+ cells (lung

macrophages and dendritic cells) to control the production of proinflammatory

cytokines and chemokines during M. tuberculosis infection, which would otherwise

promote neutrophil recruitment. This role for ATG5 is autophagy dependent, but

independent of mitophagy, LC3-associated phagocytosis, and inflammasome

activation, which are the most well-characterized ways that autophagy proteins

regulate inflammation. In addition to the increased proinflammatory cytokine

production from macrophages during M. tuberculosis infection, loss of ATG5 in

innate immune cells also results in an early induction of TH17 responses.

Despite prior published in vitro cell culture experiments supporting a role for

autophagy in controlling M. tuberculosis replication in macrophages, the effects

of autophagy on inflammatory responses occur without changes in M. tuberculosis

burden in macrophages. These findings reveal new roles for autophagy proteins in

lung resident macrophages and dendritic cells that are required to suppress

inflammatory responses that are associated with poor control of M. tuberculosis

infection.

 

Copyright: © 2023 Kinsella 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.3002159

PMCID: PMC10306192

PMID: 37319285 [Indexed for MEDLINE]

 

12. Nat Commun. 2023 Jun 12;14(1):3468. doi: 10.1038/s41467-023-39139-4.

 

Phase 1b trial of anti-EGFR antibody JMT101 and Osimertinib in EGFR exon 20

insertion-positive non-small-cell lung cancer.

 

Zhao S(#)(1), Zhuang W(#)(2), Han B(#)(3), Song Z(#)(4), Guo W(#)(5), Luo

F(#)(6), Wu L(7), Hu Y(8), Wang H(9), Dong X(10), Jiang D(11), Wang M(12), Miao

L(13), Wang Q(14), Zhang J(15), Fu Z(16), Huang Y(1), Xu C(17), Hu L(18), Li

L(19), Hu R(19), Yang Y(19), Li M(19), Yang X(20), Zhang L(21), Huang Y(22),

Fang W(23).

 

Author information:

(1)Department of Medical Oncology, State Key Laboratory of Oncology in South

China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen

University Cancer Center, Guangzhou, China.

(2)Department of Thoracic Oncology, Fujian Cancer Hospital, Fuzhou, China.

(3)Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai, China.

EGFR exon 20 insertion (20ins)-positive non-small-cell lung cancer (NSCLC) is an

uncommon disease with limited therapeutic options and dismal prognosis. Here we

report the activity, tolerability, potential mechanisms of response and

resistance for dual targeting EGFR 20ins with JMT101 (anti-EGFR monoclonal

antibody) plus osimertinib from preclinical models and an open label,

multi-center phase 1b trial (NCT04448379). Primary endpoint of the trial is

tolerability. Secondary endpoints include objective response rate, duration of

response, disease control rate, progression free survival, overall survival, the

pharmacokinetic profile of JMT101, occurrence of anti-drug antibodies and

correlation between biomarkers and clinical outcomes. A total of 121 patients

are enrolled to receive JMT101 plus osimertinib 160 mg. The most common adverse

events are rash (76.9%) and diarrhea (63.6%). The confirmed objective response

rate is 36.4%. Median progression-free survival is 8.2 months. Median duration

of response is unreached. Subgroup analyses were performed by

clinicopathological features and prior treatments. In patients with

platinum-refractory diseases (n = 53), confirmed objective response rate is

34.0%, median progression-free survival is 9.2 months and median duration of

response is 13.3 months. Responses are observed in distinct 20ins variants and

intracranial lesions. Intracranial disease control rate is 87.5%. Confirmed

intracranial objective response rate is 25%.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41467-023-39139-4

PMCID: PMC10261012

PMID: 37308490 [Indexed for MEDLINE]

 

13. PLoS Med. 2023 Jun 9;20(6):e1004246. doi: 10.1371/journal.pmed.1004246.

eCollection 2023 Jun.

 

Same-day testing with initiation of antiretroviral therapy or tuberculosis

treatment versus standard care for persons presenting with tuberculosis symptoms

at HIV diagnosis: A randomized open-label trial from Haiti.

 

Dorvil N(1), Rivera VR(1), Riviere C(1), Berman R(2), Severe P(1), Bang H(3),

Lavoile K(1), Devieux JG(4), Faustin M(1), Saintyl G(1), Mendicuti MD(5), Pierre

S(1), Apollon A(1), Dumond E(1), Forestal GPL(1), Rouzier V(1)(6), Marcelin

A(1), McNairy ML(6), Walsh KF(6), Dupnik K(6), Reif LK(6), Byrne AL(7),

Bousleiman S(2), Orvis E(2), Joseph P(1), Cremieux PY(2), Pape JW(1)(6), Koenig

SP(5).

 

Author information:

(1)Haitian Group for the Study of Kaposi's Sarcoma and Opportunistic Infections

(GHESKIO), Port-au-Prince, Haiti.

(2)The Analysis Group, Boston, Massachusetts, United States of America.

(3)University of California, Davis School of Medicine, Davis, California, United

States of America.

(4)Florida International University, Miami, Florida, United States of America.

(5)Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts,

United States of America.

(6)Weill Cornell Medicine, New York, New York, United States of America.

(7)St. Vincent's Hospital, Darlinghurst, New South Wales, Australia.

 

BACKGROUND: Same-day HIV testing and antiretroviral therapy (ART) initiation is

being widely implemented. However, the optimal timing of ART among patients with

tuberculosis (TB) symptoms is unknown. We hypothesized that same-day treatment

(TB treatment for those diagnosed with TB; ART for those not diagnosed with TB)

would be superior to standard care in this population.

METHODS AND FINDINGS: We conducted an open-label trial among adults with TB

symptoms at initial HIV diagnosis at GHESKIO in Haiti; participants were

recruited and randomized on the same day. Participants were randomized in a 1:1

ratio to same-day treatment (same-day TB testing with same-day TB treatment if

TB diagnosed; same-day ART if TB not diagnosed) versus standard care (initiating

TB treatment within 7 days and delaying ART to day 7 if TB not diagnosed). In

both groups, ART was initiated 2 weeks after TB treatment. The primary outcome

was retention in care with 48-week HIV-1 RNA<200 copies/mL, with intention to

treat (ITT) analysis. From November 6, 2017 to January 16, 2020, 500

participants were randomized (250/group); the final study visit occurred on

March 1, 2021. Baseline TB was diagnosed in 40 (16.0%) in the standard and 48

(19.2%) in the same-day group; all initiated TB treatment. In the standard

group, 245 (98.0%) initiated ART at median of 9 days; 6 (2.4%) died, 15 (6.0%)

missed the 48-week visit, and 229 (91.6%) attended the 48-week visit. Among all

who were randomized, 220 (88.0%) received 48-week HIV-1 RNA testing; 168 had

<200 copies/mL (among randomized: 67.2%; among tested: 76.4%). In the same-day

group, 249 (99.6%) initiated ART at median of 0 days; 9 (3.6%) died, 23 (9.2%)

missed the 48-week visit, and 218 (87.2%) attended the 48-week visit. Among all

who were randomized, 211 (84.4%) received 48-week HIV-1 RNA; 152 had<200

copies/mL (among randomized: 60.8%; among tested: 72.0%). There was no

difference between groups in the primary outcome (60.8% versus 67.2%; risk

difference: -0.06; 95% CI [-0.15, 0.02]; p = 0.14). Two new grade 3 or 4 events

were reported per group; none were judged to be related to the intervention. The

main limitation of this study is that it was conducted at a single urban clinic,

and the generalizability to other settings is uncertain.

CONCLUSIONS: In patients with TB symptoms at HIV diagnosis, we found that

same-day treatment was not associated with superior retention and viral

suppression. In this study, a short delay in ART initiation did not appear to

compromise outcomes.

TRIAL REGISTRATION: This study is registered with ClinicalTrials.gov

NCT03154320.

 

Copyright: © 2023 Dorvil 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.pmed.1004246

PMCID: PMC10292694

PMID: 37294843 [Indexed for MEDLINE]

 

14. J Clin Oncol. 2023 Jun 4:JCO2300774. doi: 10.1200/JCO.23.00774. Online ahead of print.

 

Phase II, Open-Label Study of Encorafenib Plus Binimetinib in Patients With

BRAF(V600)-Mutant Metastatic Non-Small-Cell Lung Cancer.

 

Riely GJ(1), Smit EF(2), Ahn MJ(3), Felip E(4), Ramalingam SS(5), Tsao A(6),

Johnson M(7), Gelsomino F(8), Esper R(9), Nadal E(10), Offin M(1), Provencio

M(11), Clarke J(12), Hussain M(9), Otterson GA(13), Dagogo-Jack I(14), Goldman

JW(15), Morgensztern D(16), Alcasid A(17), Usari T(18), Wissel P(19), Wilner

K(20), Pathan N(20), Tonkovyd S(21), Johnson BE(22).

 

Author information:

(1)Memorial Sloan Kettering Cancer Center, New York, NY.

(2)Department of Pulmonary Diseases, Leiden University Medical Center, Leiden,

the Netherlands.

(3)Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul,

South Korea.

PURPOSE: The combination of encorafenib (BRAF inhibitor) plus binimetinib (MEK

inhibitor) has demonstrated clinical efficacy with an acceptable safety profile

in patients with BRAFV600E/K-mutant metastatic melanoma. We evaluated the

efficacy and safety of encorafenib plus binimetinib in patients with

BRAFV600E-mutant metastatic non-small-cell lung cancer (NSCLC).

METHODS: In this ongoing, open-label, single-arm, phase II study, patients with

BRAFV600E-mutant metastatic NSCLC received oral encorafenib 450 mg once daily

plus binimetinib 45 mg twice daily in 28-day cycles. The primary end point was

confirmed objective response rate (ORR) by independent radiology review (IRR).

Secondary end points included duration of response (DOR), disease control rate

(DCR), progression-free survival (PFS), overall survival, time to response, and

safety.

RESULTS: At data cutoff, 98 patients (59 treatment-naïve and 39 previously

treated) with BRAFV600E-mutant metastatic NSCLC received encorafenib plus

binimetinib. Median duration of treatment was 9.2 months with encorafenib and

8.4 months with binimetinib. ORR by IRR was 75% (95% CI, 62 to 85) in

treatment-naïve and 46% (95% CI, 30 to 63) in previously treated patients;

median DOR was not estimable (NE; 95% CI, 23.1 to NE) and 16.7 months (95% CI,

7.4 to NE), respectively. DCR after 24 weeks was 64% in treatment-naïve and 41%

in previously treated patients. Median PFS was NE (95% CI, 15.7 to NE) in

treatment-naïve and 9.3 months (95% CI, 6.2 to NE) in previously treated

patients. The most frequent treatment-related adverse events (TRAEs) were nausea

(50%), diarrhea (43%), and fatigue (32%). TRAEs led to dose reductions in 24

(24%) and permanent discontinuation of encorafenib plus binimetinib in 15 (15%)

patients. One grade 5 TRAE of intracranial hemorrhage was reported. Interactive

visualization of the data presented in this article is available at the PHAROS

dashboard (https://clinical-trials.dimensions.ai/pharos/).

CONCLUSION: For patients with treatment-naïve and previously treated

BRAFV600E-mutant metastatic NSCLC, encorafenib plus binimetinib showed a

meaningful clinical benefit with a safety profile consistent with that observed

in the approved indication in melanoma.

 

DOI: 10.1200/JCO.23.00774

PMID: 37270692

 

15. Cell Host Microbe. 2023 Jun 14;31(6):978-992.e5. doi:

10.1016/j.chom.2023.05.009. Epub 2023 Jun 2.

 

Identification of host regulators of Mycobacterium tuberculosis phenotypes

uncovers a role for the MMGT1-GPR156 lipid droplet axis in persistence.

 

Kalam H(1), Chou CH(1), Kadoki M(2), Graham DB(2), Deguine J(3), Hung DT(4),

Xavier RJ(5).

 

Author information:

(1)Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for

Computational and Integrative Biology, Massachusetts General Hospital, Harvard

Medical School, Boston, MA 02114, USA.

(2)Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for

Computational and Integrative Biology, Massachusetts General Hospital, Harvard

Medical School, Boston, MA 02114, USA; Department of Molecular Biology,

Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.

(3)Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.

 

The ability of Mycobacterium tuberculosis (Mtb) to establish latency affects

disease and response to treatment. The host factors that influence the

establishment of latency remain elusive. We engineered a multi-fluorescent Mtb

strain that reports survival, active replication, and stressed non-replication

states and determined the host transcriptome of the infected macrophages in

these states. Additionally, we conducted a genome-wide CRISPR screen to identify

host factors that modulated the phenotypic state of Mtb. We validated hits in a

phenotype-specific manner and prioritized membrane magnesium transporter 1

(MMGT1) for a detailed mechanistic investigation. Mtb infection of

MMGT1-deficient macrophages promoted a switch to persistence, upregulated lipid

metabolism genes, and accumulated lipid droplets during infection. Targeting

triacylglycerol synthesis reduced both droplet formation and Mtb persistence.

The orphan G protein-coupled receptor GPR156 is a key inducer of droplet

accumulation in ΔMMGT1 cells. Our work uncovers the role of MMGT1-GPR156-lipid

droplets in the induction of Mtb persistence.

 

Copyright © 2023 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.chom.2023.05.009

PMID: 37269834 [Indexed for MEDLINE]

 

16. Cell Host Microbe. 2023 Jun 14;31(6):962-977.e8. doi:

10.1016/j.chom.2023.05.006. Epub 2023 Jun 1.

 

Airway T cells are a correlate of i.v. Bacille Calmette-Guerin-mediated

protection against tuberculosis in rhesus macaques.

 

Darrah PA(1), Zeppa JJ(2), Wang C(3), Irvine EB(4), Bucsan AN(1), Rodgers MA(2),

Pokkali S(1), Hackney JA(1), Kamath M(1), White AG(2), Borish HJ(2), Frye LJ(2),

Tomko J(2), Kracinovsky K(2), Lin PL(5), Klein E(6), Scanga CA(2), Alter G(7),

Fortune SM(4), Lauffenburger DA(8), Flynn JL(2), Seder RA(1), Maiello P(2),

Roederer M(9).

 

Author information:

(1)Vaccine Research Center, National Institute of Allergy and Infectious

Diseases, National Institutes of Health, Bethesda, MD 20892, USA.

(2)Department of Microbiology and Molecular Genetics, University of Pittsburgh

School of Medicine and Center for Vaccine Research, University of Pittsburgh,

Pittsburgh, PA 15261, USA.

(3)Department of Immunology and Microbiology, University of Colorado, Anschuntz

Medical Campus, Aurora, CO 80045, USA; Department of Biological Engineering,

Massachusetts Institute of Technology, Cambridge, MA 02142, USA.

Bacille Calmette-Guerin (BCG), the only approved Mycobacterium tuberculosis

(Mtb) vaccine, provides limited durable protection when administered

intradermally. However, recent work revealed that intravenous (i.v.) BCG

administration yielded greater protection in macaques. Here, we perform a

dose-ranging study of i.v. BCG vaccination in macaques to generate a range of

immune responses and define correlates of protection. Seventeen of 34 macaques

had no detectable infection after Mtb challenge. Multivariate analysis

incorporating longitudinal cellular and humoral immune parameters uncovered an

extensive and highly coordinated immune response from the bronchoalveolar lavage

(BAL). A minimal signature predicting protection contained four BAL immune

features, of which three remained significant after dose correction: frequency

of CD4 T cells producing TNF with interferon γ (IFNγ), frequency of those

producing TNF with IL-17, and the number of NK cells. Blood immune features were

less predictive of protection. We conclude that CD4 T cell immunity and NK cells

in the airway correlate with protection following i.v. BCG.

 

Published by Elsevier Inc.

 

DOI: 10.1016/j.chom.2023.05.006

PMID: 37267955 [Indexed for MEDLINE]

 

17. J Natl Cancer Inst. 2023 Jun 1:djad071. doi: 10.1093/jnci/djad071. Online ahead of print.

 

Lung cancer risk discrimination of prediagnostic proteomics measurements

compared with existing prediction tools.

 

Feng X(1), Wu WY(2), Onwuka JU(1), Haider Z(2), Alcala K(1), Smith-Byrne K(3),

Zahed H(1), Guida F(4), Wang R(5), Bassett JK(6), Stevens V(7), Wang Y(8),

Weinstein S(9), Freedman ND(9), Chen C(10), Tinker L(11), Nøst TH(12), Koh

WP(13)(14), Muller D(15), Colorado-Yohar SM(16)(17)(18), Tumino R(19), Hung

RJ(20)(21), Amos CI(22), Lin X(23)(24)(25), Zhang X(26), Arslan AA(27), Sánchez

MJ(17)(28)(29)(30), Sørgjerd EP(31), Severi G(32), Hveem K(31), Brennan P(1),

Langhammer A(31)(33), Milne RL(6)(34)(35), Yuan JM(5)(36), Melin B(2), Johansson

M(2), Robbins HA(1), Johansson M(1).

 

Author information:

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

Lyon, France.

(2)Department of Radiation Sciences, Oncology, Umea University, Umea, Sweden.

(3)Cancer Epidemiology Unit, University of Oxford, Oxford, UK.

BACKGROUND: We sought to develop a proteomics-based risk model for lung cancer

and evaluate its risk-discriminatory performance in comparison with a

smoking-based risk model (PLCOm2012) and a commercially available autoantibody

biomarker test.

METHODS: We designed a case-control study nested in 6 prospective cohorts,

including 624 lung cancer participants who donated blood samples at most 3 years

prior to lung cancer diagnosis and 624 smoking-matched cancer free participants

who were assayed for 302 proteins. We used 470 case-control pairs from 4 cohorts

to select proteins and train a protein-based risk model. We subsequently used

154 case-control pairs from 2 cohorts to compare the risk-discriminatory

performance of the protein-based model with that of the Early Cancer Detection

Test (EarlyCDT)-Lung and the PLCOm2012 model using receiver operating

characteristics analysis and by estimating models' sensitivity. All tests were

2-sided.

RESULTS: The area under the curve for the protein-based risk model in the

validation sample was 0.75 (95% confidence interval [CI] = 0.70 to 0.81)

compared with 0.64 (95% CI = 0.57 to 0.70) for the PLCOm2012 model (Pdifference

= .001). The EarlyCDT-Lung had a sensitivity of 14% (95% CI = 8.2% to 19%) and a

specificity of 86% (95% CI = 81% to 92%) for incident lung cancer. At the same

specificity of 86%, the sensitivity for the protein-based risk model was

estimated at 49% (95% CI = 41% to 57%) and 30% (95% CI = 23% to 37%) for the

PLCOm2012 model.

CONCLUSION: Circulating proteins showed promise in predicting incident lung

cancer and outperformed a standard risk prediction model and the commercialized

EarlyCDT-Lung.

 

© The Author(s) 2023. Published by Oxford University Press.

 

DOI: 10.1093/jnci/djad071

PMID: 37260165

 

18. J Clin Invest. 2023 Jun 15;133(12):e158630. doi: 10.1172/JCI158630.

 

Poly(ADP-ribose) polymerase 9 mediates early protection against Mycobacterium

tuberculosis infection by regulating type I IFN production.

 

Thirunavukkarasu S(1), Ahmed M(1), Rosa BA(2)(3), Boothby M(4), Cho SH(4),

Rangel-Moreno J(5), Mbandi SK(6), Schreiber V(7), Gupta A(1), Zuniga J(8)(9),

Mitreva M(2)(3), Kaushal D(10), Scriba TJ(6), Khader SA(1).

 

Author information:

(1)Department of Molecular Microbiology.

(2)McDonnell Genome Institute, and.

(3)Division of Infectious Diseases, Department of Internal Medicine, Washington

University in St. Louis, St. Louis, Missouri, USA.

The ADP ribosyltransferases (PARPs 1-17) regulate diverse cellular processes,

including DNA damage repair. PARPs are classified on the basis of their ability

to catalyze poly-ADP-ribosylation (PARylation) or mono-ADP-ribosylation

(MARylation). Although PARP9 mRNA expression is significantly increased in

progressive tuberculosis (TB) in humans, its participation in host immunity to

TB is unknown. Here, we show that PARP9 mRNA encoding the MARylating PARP9

enzyme was upregulated during TB in humans and mice and provide evidence of a

critical modulatory role for PARP9 in DNA damage, cyclic GMP-AMP synthase (cGAS)

expression, and type I IFN production during TB. Thus, Parp9-deficient mice were

susceptible to Mycobacterium tuberculosis infection and exhibited increased TB

disease, cGAS and 2'3'-cyclic GMP-AMP (cGAMP) expression, and type I IFN

production, along with upregulation of complement and coagulation pathways.

Enhanced M. tuberculosis susceptibility is type I IFN dependent, as blockade of

IFN α receptor (IFNAR) signaling reversed the enhanced susceptibility of

Parp9-/- mice. Thus, in sharp contrast to PARP9 enhancement of type I IFN

production in viral infections, this member of the MAR family plays a protective

role by limiting type I IFN responses during TB.

 

DOI: 10.1172/JCI158630

PMCID: PMC10266794

PMID: 37200107 [Indexed for MEDLINE]

 

19. J Clin Oncol. 2023 Jun 10;41(17):3249-3259. doi: 10.1200/JCO.22.02509. Epub 2023 May 4.

 

US Food and Drug Administration Approval Summary: Nivolumab Plus

Platinum-Doublet Chemotherapy for the Neoadjuvant Treatment of Patients With

Resectable Non-Small-Cell Lung Cancer.

 

Akinboro O(1), Drezner N(1), Amatya A(1), Runyan J(1), Fourie-Zirkelbach J(1),

Zhao M(1), Bi Y(1), Korsah K(1), Mixter B(2), Tang S(1), Larkins E(1), Pazdur

R(1)(2), Beaver JA(1)(2), Singh H(1)(2).

 

Author information:

(1)Center for Drug Evaluation and Research (CDER), U.S. Food and Drug

Administration, Silver Spring, MD.

(2)Oncology Center of Excellence, U.S. Food and Drug Administration, Silver

Spring, MD.

 

PURPOSE: On March 4, 2022, the US Food and Drug Administration (FDA) approved

nivolumab plus platinum-doublet chemotherapy for the neoadjuvant treatment of

patients with resectable non-small-cell lung cancer (NSCLC). We discuss the

FDA's review of the key data and regulatory considerations supporting this

approval.

PATIENTS AND METHODS: The approval was based on the results of CheckMate 816, an

international, multiregional, active-controlled trial that randomly assigned 358

patients with resectable NSCLC, stage IB (≥4 cm) to IIIA (N2) per the American

Joint Committee on Cancer seventh staging edition to receive either nivolumab

plus platinum-doublet or platinum-doublet chemotherapy alone for three cycles

before planned surgical resection. The major efficacy end point that supported

this approval was event-free survival (EFS).

RESULTS: At the first planned interim analysis (IA), the hazard ratio (HR) for

EFS was 0.63 (95% CI, 0.45 to 0.87; P = .0052; statistical significance boundary

= .0262) favoring the nivolumab plus chemotherapy arm; the median EFS was 31.6

months (95% CI, 30.2 to not reached) in the nivolumab plus chemotherapy arm

versus 20.8 months (95% CI, 14.0 to 26.7) in the chemotherapy-only arm. At the

time of a prespecified IA for overall survival (OS), 26% of patients had died,

and the HR for OS was 0.57 (95% CI, 0.38 to 0.87; P = .0079; statistical

significance boundary = .0033). Eighty-three percent of patients in the

nivolumab-containing arm versus 75% in the chemotherapy-only arm received

definitive surgery.

CONCLUSION: This approval, the first for any regimen for the neoadjuvant

treatment of NSCLC in the United States, was supported by a statistically

significant and clinically meaningful improvement in EFS with no evidence of

detriment in OS or negative impact on patients' receipt and timing of surgery or

surgical outcomes.

 

DOI: 10.1200/JCO.22.02509

PMCID: PMC10256356

PMID: 37141544 [Indexed for MEDLINE]

 

20. J Clin Oncol. 2023 Jun 20;41(18):3311-3317. doi: 10.1200/JCO.22.02524. Epub 2023 Apr 25.

 

Long-Term Outcomes and Molecular Correlates of Sotorasib Efficacy in Patients

With Pretreated KRAS G12C-Mutated Non-Small-Cell Lung Cancer: 2-Year Analysis of

CodeBreaK 100.

 

Dy GK(1), Govindan R(2), Velcheti V(3), Falchook GS(4), Italiano A(5), Wolf

J(6), Sacher AG(7), Takahashi T(8), Ramalingam SS(9), Dooms C(10), Kim DW(11),

Addeo A(12), Desai J(13), Schuler M(14), Tomasini P(15), Hong DS(16), Lito

P(17), Tran Q(18), Jones S(18), Anderson A(18), Hindoyan A(18), Snyder W(18),

Skoulidis F(16), Li BT(17).

 

Author information:

(1)Roswell Park Comprehensive Cancer Center, Buffalo, NY.

(2)Siteman Cancer Center, Washington University School of Medicine, St Louis,

MO.

(3)Perlmutter Cancer Center, New York University Langone, New York, NY.

Clinical trials frequently include multiple end points that mature at different

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

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

available. Clinical Trial Updates provide an opportunity to disseminate

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

primary end point has already been reported.In the longest follow-up, to our

knowledge, for a KRASG12C inhibitor, we assessed the long-term efficacy, safety,

and biomarkers of sotorasib in patients with KRAS G12C-mutated advanced

non-small-cell lung cancer (NSCLC) from the CodeBreaK 100 clinical trial

(ClinicalTrials.gov identifier: NCT03600883). This multicenter, single-group,

open-label phase I/phase II trial enrolled 174 patients with KRAS G12C-mutated,

locally advanced or metastatic NSCLC after progression on prior therapies.

Patients (N = 174) received sotorasib 960 mg once daily with the primary end

points for phase I of safety and tolerability and for phase II of objective

response rate (ORR). Sotorasib produced an ORR of 41%, median duration of

response of 12.3 months, progression-free survival (PFS) of 6.3 months, overall

survival (OS) of 12.5 months, and 2-year OS rate of 33%. Long-term clinical

benefit (PFS ≥ 12 months) was observed in 40 (23%) patients across PD-L1

expression levels, in a proportion of patients with somatic STK11 and/or KEAP1

alterations, and was associated with lower baseline circulating tumor DNA

levels. Sotorasib was well tolerated, with few late-onset treatment-related

toxicities, none of which led to treatment discontinuation. These results

demonstrate the long-term benefit of sotorasib, including in subgroups with poor

prognosis.

 

DOI: 10.1200/JCO.22.02524

PMID: 37098232 [Indexed for MEDLINE]

 

21. J Exp Med. 2023 Jun 5;220(6):e20221392. doi: 10.1084/jem.20221392. Epub 2023 Mar 15.

 

Spatial mapping reveals granuloma diversity and histopathological superstructure

in human tuberculosis.

 

Sawyer AJ(1)(2)(3), Patrick E(4)(5)(6), Edwards J(1)(7), Wilmott JS(1)(3)(7),

Fielder T(8), Yang Q(9), Barber DL(10), Ernst JD(11), Britton WJ(2)(12),

Palendira U(1)(2)(3), Chen X(13), Feng CG(1)(2)(3)(14).

 

Author information:

(1)School of Medical Sciences, Faculty of Medicine and Health, The University of

Sydney , Sydney, Australia.

(2)Centenary Institute, The University of Sydney , Sydney, Australia.

(3)Charles Perkins Centre, The University of Sydney , Sydney, Australia.

The hallmark of tuberculosis (TB) is the formation of immune cell-enriched

aggregates called granulomas. While granulomas are pathologically diverse, their

tissue-wide heterogeneity has not been spatially resolved at the single-cell

level in human tissues. By spatially mapping individual immune cells in every

lesion across entire tissue sections, we report that in addition to necrotizing

granulomas, the human TB lung contains abundant non-necrotizing leukocyte

aggregates surrounding areas of necrotizing tissue. These cellular lesions were

more diverse in composition than necrotizing lesions and could be stratified

into four general classes based on cellular composition and spatial distribution

of B cells and macrophages. The cellular composition of non-necrotizing

structures also correlates with their proximity to necrotizing lesions,

indicating these are foci of distinct immune reactions adjacent to necrotizing

granulomas. Together, we show that during TB, diseased lung tissue develops a

histopathological superstructure comprising at least four different types of

non-necrotizing cellular aggregates organized as satellites of necrotizing

granulomas.

 

© 2023 Sawyer et al.

 

DOI: 10.1084/jem.20221392

PMCID: PMC10035589

PMID: 36920308 [Indexed for MEDLINE]

 

22. Am J Respir Crit Care Med. 2023 Jun 1;207(11):1525-1532. doi:

10.1164/rccm.202211-2125OC.

 

Effectiveness of Bedaquiline Use beyond Six Months in Patients with

Multidrug-Resistant Tuberculosis.

 

Trevisi L(1), Hernán MA(2), Mitnick CD(1)(3)(4), Khan U(5), Seung KJ(1)(3)(4),

Rich ML(1)(3)(4), Bastard M(6), Huerga H(6), Melikyan N(6), Atwood SA(3),

Avaliani Z(7), Llanos F(8)(9), Manzur-Ul-Alam M(10), Zarli K(11), Binegdie

AB(12), Adnan S(13), Melikyan A(14), Gelin A(15), Isani AK(16), Vetushko D(17),

Daugarina Z(18), Nkundanyirazo P(19), Putri FA(5), Vilbrun C(20), Khan M(21),

Hewison C(22), Khan PY(23), Franke MF(1).

 

Author information:

(1)Department of Global Health and Social Medicine, Harvard Medical School,

Boston, Massachusetts.

(2)CAUSALab, Departments of Epidemiology, Harvard T.H. Chan School of Public

Health, Harvard University, Boston, Massachusetts.

(3)Division of Global Health Equity, Brigham and Women's Hospital, Boston,

Massachusetts.

Comment in

    Am J Respir Crit Care Med. 2023 Jun 1;207(11):1423-1424.

 

Rationale: Current recommendations for the treatment of rifampicin- and

multidrug-resistant tuberculosis include bedaquiline (BDQ) used for 6 months or

longer. Evidence is needed to inform the optimal duration of BDQ. Objectives: We

emulated a target trial to estimate the effect of three BDQ duration treatment

strategies (6, 7-11, and 12 mo) on the probability of successful treatment

among patients receiving a longer individualized regimen for multidrug-resistant

tuberculosis. Methods: To estimate the probability of successful treatment, we

implemented a three-step approach comprising cloning, censoring, and inverse

probability weighting. Measurements and Main Results: The 1,468 eligible

individuals received a median of 4 (interquartile range, 4-5) likely effective

drugs. In 87.1% and 77.7% of participants, this included linezolid and

clofazimine, respectively. The adjusted probability of successful treatment was

0.85 (95% confidence interval [CI], 0.81-0.88) for 6 months of BDQ, 0.77 (95%

CI, 0.73-0.81) for 7-11 months, and 0.86 (95% CI, 0.83-0.88) for 12 months.

Compared with 6 months of BDQ, the ratio of treatment success was 0.91 (95% CI,

0.85-0.96) for 7-11 months and 1.01 (95% CI, 0.96-1.06) for 12 months. Naive

analyses that did not account for bias revealed a higher probability of

successful treatment with 12 months (ratio, 1.09 [95% CI, 1.05-1.14]).

Conclusions: BDQ use beyond 6 months did not increase the probability of

successful treatment among patients receiving longer regimens that commonly

included new and repurposed drugs. When not properly accounted for, immortal

person-time bias can influence estimates of the effects of treatment duration.

Future analyses should explore the effect of treatment duration of BDQ and other

drugs in subgroups with advanced disease and/or receiving less potent regimens.

 

DOI: 10.1164/rccm.202211-2125OC

PMCID: PMC10263131

PMID: 36802336 [Indexed for MEDLINE]

 

23. J Clin Oncol. 2023 Jun 1;41(16):2893-2903. doi: 10.1200/JCO.22.02823. Epub 2023 Jan 23.

 

Tarlatamab, a First-in-Class DLL3-Targeted Bispecific T-Cell Engager, in

Recurrent Small-Cell Lung Cancer: An Open-Label, Phase I Study.

 

Paz-Ares L(1), Champiat S(2), Lai WV(3), Izumi H(4), Govindan R(5), Boyer M(6),

Hummel HD(7), Borghaei H(8), Johnson ML(9), Steeghs N(10), Blackhall F(11),

Dowlati A(12), Reguart N(13), Yoshida T(14), He K(15), Gadgeel SM(16), Felip

E(17), Zhang Y(18), Pati A(18), Minocha M(18), Mukherjee S(18), Goldrick A(18),

Nagorsen D(18), Hashemi Sadraei N(18), Owonikoko TK(19).

 

Author information:

(1)Hospital Universitario 12 de Octubre, CNIO-H120 Lung Cancer Unit, Ciberonc

and Universidad Complutense, Madrid, Spain.

(2)Gustave Roussy, DC(c)partement d'Innovation ThC(c)rapeutique et d'Essais

PrC(c)coces (DITEP), Villejuif, France.

(3)Thoracic Oncology Service, Department of Medicine, Division of Solid Tumor

Oncology, Memorial Sloan Kettering Cancer Center, New York, NY.

Comment in

    J Clin Oncol. 2023 Jun 1;41(16):2877-2880.

    Transl Lung Cancer Res. 2023 Jun 30;12(6):1355-1357.

 

PURPOSE: Small-cell lung cancer (SCLC) is an aggressive malignancy with limited

treatments. Delta-like ligand 3 (DLL3) is aberrantly expressed in most SCLC.

Tarlatamab (AMG 757), a bispecific T-cell engager molecule, binds both DLL3 and

CD3 leading to T-cellb-mediated tumor lysis. Herein, we report phase I results

of tarlatamab in patients with SCLC.

PATIENTS AND METHODS: This study evaluated tarlatamab in patients with

relapsed/refractory SCLC. The primary end point was safety. Secondary end points

included antitumor activity by modified RECIST 1.1, overall survival, and

pharmacokinetics.

RESULTS: By July 19, 2022, 107 patients received tarlatamab in dose exploration

(0.003 to 100 mg; n = 73) and expansion (100 mg; n = 34) cohorts. Median prior

lines of anticancer therapy were 2 (range, 1-6); 49.5% received antiprogrammed

death-1/programmed death ligand-1 therapy. Any-grade treatment-related adverse

events occurred in 97 patients (90.7%) and grade b % 3 in 33 patients (30.8%).

One patient (1%) had grade 5 pneumonitis. Cytokine release syndrome was the most

common treatment-related adverse event, occurring in 56 patients (52%) including

grade 3 in one patient (1%). Maximum tolerated dose was not reached. Objective

response rate was 23.4% (95% CI, 15.7 to 32.5) including two complete and 23

partial responses. The median duration of response was 12.3 months (95% CI, 6.6

to 14.9). The disease control rate was 51.4% (95% CI, 41.5 to 61.2). The median

progression-free survival and overall survival were 3.7 months (95% CI, 2.1 to

5.4) and 13.2 months (95% CI, 10.5 to not reached), respectively. Exploratory

analysis suggests that selecting for increased DLL3 expression can result in

increased clinical benefit.

CONCLUSION: In patients with heavily pretreated SCLC, tarlatamab demonstrated

manageable safety with encouraging response durability. Further evaluation of

this promising molecule is ongoing.

 

DOI: 10.1200/JCO.22.02823

PMID: 36689692 [Indexed for MEDLINE]

 

24. J Natl Cancer Inst. 2023 Jun 8;115(6):742-748. doi: 10.1093/jnci/djac015.

 

Randomized Trial of First-Line Tyrosine Kinase Inhibitor With or Without

Radiotherapy for Synchronous Oligometastatic EGFR-Mutated Non-Small Cell Lung

Cancer.

 

Wang XS(1), Bai YF(1), Verma V(2), Yu RL(1), Tian W(1), Ao R(1), Deng Y(1), Zhu

XQ(1), Liu H(1), Pan HX(1), Yang L(1), Bai HS(3), Luo X(3), Guo Y(3), Zhou

MX(3), Sun YM(4), Zhang ZC(4), Li SM(3)(5), Cheng X(3), Tan BX(3), Han LF(6),

Liu YY(7), Zhang K(8), Zeng FX(9), Jia L(10), Hao XB(11), Wang YY(1), Feng G(1),

Xie K(1), Lu Y(12), Zeng M(1).

 

Author information:

(1)Cancer Center, Sichuan Provincial People's Hospital, University of Electronic

Science and Technology of China, Chengdu, China.

(2)Department of Radiation Oncology, University of Nebraska Medical Center,

Omaha, NE, USA.

(3)School of Medicine, University of Electronic Science and Technology of China,

Chengdu, China.

Erratum in

    J Natl Cancer Inst. 2023 May 18;:

 

Expression of concern in

    J Natl Cancer Inst. 2022 Apr 01;:

 

Comment in

    J Natl Cancer Inst. 2023 Jun 8;115(6):605-607.

 

BACKGROUND: Adding radiotherapy (RT) to systemic therapy improves

progression-free survival (PFS) and overall survival (OS) in oligometastatic

non-small cell lung cancer (NSCLC). Whether these findings translate to

epidermal growth factor receptor (EGFR)-mutated NSCLC remains unknown. The

SINDAS trial (NCT02893332) evaluated first-line tyrosine kinase inhibitor (TKI)

therapy for EGFR-mutated synchronous oligometastatic NSCLC and randomized to

upfront RT vs no RT; we now report the prespecified interim analysis at 68%

accrual.

METHODS: Inclusion criteria were biopsy-proven EGFR-mutated adenocarcinoma (per

amplification refractory mutation system or next generation sequencing), with

synchronous (newly diagnosed, treatment naïve) oligometastatic (≤5 metastases;

≤2 lesions in any one organ) NSCLC without brain metastases. All patients

received a first-generation TKI (gefitinib, erlotinib, or icotinib), and

randomization was between no RT vs RT (25-40 Gy in 5 fractions depending on

tumor size and location) to all metastases and the primary tumor/involved

regional lymphatics. The primary endpoint (intention to treat) was PFS.

Secondary endpoints included OS and toxicities. All statistical tests were

2-sided.

RESULTS: A total of 133 patients (n = 65 TKI only, n = 68 TKI with RT) were

enrolled (2016-2019). The median follow-up was 23.6 months. The respective

median PFS was 12.5 months vs 20.2 months (P < .001), and the median OS was

17.4 months vs 25.5 months (P < .001) for TKI only vs TKI with RT. Treatment

yielded no grade 5 events and a 6% rate of symptomatic grade 3-4 pneumonitis in

the TKI with RT arm. Based on the efficacy results of this prespecified interim

analysis, the ethics committee recommended premature cessation of this trial.

CONCLUSIONS: As compared with a first-line TKI alone, addition of upfront local

therapy using RT statistically significantly improved PFS and OS for

EGFR-mutated NSCLC.

 

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

 

DOI: 10.1093/jnci/djac015

PMCID: PMC10248839

PMID: 35094066 [Indexed for MEDLINE]

 


上一篇: No.7

下一篇: No.5