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.4

发布时间:2023-05-10 浏览次数:
字号: + - 14

PubMed

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

Filters applied: from 2023/4/1 - 2023/4/30.

 

1. Nature. 2023 Apr;616(7957):553-562. doi: 10.1038/s41586-023-05776-4. Epub 2023

Apr 13.

 

Tracking early lung cancer metastatic dissemination in TRACERx using ctDNA.

 

Abbosh C(#)(1), Frankell AM(#)(2)(3), Harrison T(#)(4), Kisistok J(#)(5)(6)(7),

Garnett A(#)(4), …

 

Author information:

(1)Cancer Research UK Lung Cancer Centre of Excellence, University College

London Cancer Institute, London, UK. c.abbosh@ucl.ac.uk.

(2)Cancer Research UK Lung Cancer Centre of Excellence, University College

London Cancer Institute, London, UK.

(3)Cancer Evolution and Genome Instability Laboratory, The Francis Crick

Institute, London, UK.

Circulating tumour DNA (ctDNA) can be used to detect and profile residual tumour

cells persisting after curative intent therapy1. The study of large patient

cohorts incorporating longitudinal plasma sampling and extended follow-up is

required to determine the role of ctDNA as a phylogenetic biomarker of relapse

in early-stage non-small-cell lung cancer (NSCLC). Here we developed ctDNA

methods tracking a median of 200 mutations identified in resected NSCLC tissue

across 1,069 plasma samples collected from 197 patients enrolled in the TRACERx

study2. A lack of preoperative ctDNA detection distinguished biologically

indolent lung adenocarcinoma with good clinical outcome. Postoperative plasma

analyses were interpreted within the context of standard-of-care radiological

surveillance and administration of cytotoxic adjuvant therapy. Landmark analyses

of plasma samples collected within 120 days after surgery revealed ctDNA

detection in 25% of patients, including 49% of all patients who experienced

clinical relapse; 3 to 6 monthly ctDNA surveillance identified impending disease

relapse in an additional 20% of landmark-negative patients. We developed a

bioinformatic tool (ECLIPSE) for non-invasive tracking of subclonal architecture

at low ctDNA levels. ECLIPSE identified patients with polyclonal metastatic

dissemination, which was associated with a poor clinical outcome. By measuring

subclone cancer cell fractions in preoperative plasma, we found that subclones

seeding future metastases were significantly more expanded compared with

non-metastatic subclones. Our findings will support (neo)adjuvant trial advances

and provide insights into the process of metastatic dissemination using

low-ctDNA-level liquid biopsy.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41586-023-05776-4

PMID: 37055640 [Indexed for MEDLINE]

 

2. Nature. 2023 Apr;616(7957):525-533. doi: 10.1038/s41586-023-05783-5. Epub 2023

Apr 12.

 

The evolution of lung cancer and impact of subclonal selection in TRACERx.

 

Frankell AM(#)(1)(2), Dietzen M(#)(1)(2)(3), Al Bakir M(#)(1)(2), Lim

EL(#)(1)(2),…

 

Author information:

(1)Cancer Evolution and Genome Instability Laboratory, The Francis Crick

Institute, London, UK.

(2)Cancer Research UK Lung Cancer Centre of Excellence, University College

London Cancer Institute, London, UK.

(3)Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre

of Excellence, University College London Cancer Institute, London, UK.

Lung cancer is the leading cause of cancer-associated mortality worldwide1. Here

we analysed 1,644 tumour regions sampled at surgery or during follow-up from the

first 421 patients with non-small cell lung cancer prospectively enrolled into

the TRACERx study. This project aims to decipher lung cancer evolution and

address the primary study endpoint: determining the relationship between

intratumour heterogeneity and clinical outcome. In lung adenocarcinoma,

mutations in 22 out of 40 common cancer genes were under significant subclonal

selection, including classical tumour initiators such as TP53 and KRAS. We

defined evolutionary dependencies between drivers, mutational processes and

whole genome doubling (WGD) events. Despite patients having a history of

smoking, 8% of lung adenocarcinomas lacked evidence of tobacco-induced

mutagenesis. These tumours also had similar detection rates for EGFR mutations

and for RET, ROS1, ALK and MET oncogenic isoforms compared with tumours

in never-smokers, which suggests that they have a similar aetiology and

pathogenesis. Large subclonal expansions were associated with positive subclonal

selection. Patients with tumours harbouring recent subclonal expansions, on the

terminus of a phylogenetic branch, had significantly shorter disease-free

survival. Subclonal WGD was detected in 19% of tumours, and 10% of tumours

harboured multiple subclonal WGDs in parallel. Subclonal, but not truncal, WGD

was associated with shorter disease-free survival. Copy number heterogeneity was

associated with extrathoracic relapse within 1 year after surgery. These data

demonstrate the importance of clonal expansion, WGD and copy number instability

in determining the timing and patterns of relapse in non-small cell lung cancer

and provide a comprehensive clinical cancer evolutionary data resource.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41586-023-05783-5

PMCID: PMC10115649

PMID: 37046096 [Indexed for MEDLINE]

 

3. Nature. 2023 Apr;616(7957):534-542. doi: 10.1038/s41586-023-05729-x. Epub 2023

Apr 12.

 

The evolution of non-small cell lung cancer metastases in TRACERx.

 

Al Bakir M(#)(1)(2), Huebner A(#)(1)(2)(3), Martínez-Ruiz C(#)(1)(3),

Grigoriadis K(#)(1)(2)(3), Watkins TBK(#)(2), Pich O(#)(2),…

 

Author information:

(1)Cancer Research UK Lung Cancer Centre of Excellence, University College

London Cancer Institute, London, UK.

(2)Cancer Evolution and Genome Instability Laboratory, The Francis Crick

Institute, London, UK.

(3)Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre

of Excellence, University College London Cancer Institute, London, UK.

Metastatic disease is responsible for the majority of cancer-related deaths1. We

report the longitudinal evolutionary analysis of 126 non-small cell lung cancer

(NSCLC) tumours from 421 prospectively recruited patients in TRACERx who

developed metastatic disease, compared with a control cohort of 144

non-metastatic tumours. In 25% of cases, metastases diverged early, before the

last clonal sweep in the primary tumour, and early divergence was enriched for

patients who were smokers at the time of initial diagnosis. Simulations

suggested that early metastatic divergence more frequently occurred at smaller

tumour diameters (less than 8 mm). Single-region primary tumour sampling

resulted in 83% of late divergence cases being misclassified as early,

highlighting the importance of extensive primary tumour sampling. Polyclonal

dissemination, which was associated with extrathoracic disease recurrence, was

found in 32% of cases. Primary lymph node disease contributed to metastatic

relapse in less than 20% of cases, representing a hallmark of metastatic

potential rather than a route to subsequent recurrences/disease progression.

Metastasis-seeding subclones exhibited subclonal expansions within primary

tumours, probably reflecting positive selection. Our findings highlight the

importance of selection in metastatic clone evolution within untreated primary

tumours, the distinction between monoclonal versus polyclonal seeding in

dictating site of recurrence, the limitations of current radiological screening

approaches for early diverging tumours and the need to develop strategies to

target metastasis-seeding subclones before relapse.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41586-023-05729-x

PMCID: PMC10115651

PMID: 37046095 [Indexed for MEDLINE]

 

4. Nature. 2023 Apr;616(7957):563-573. doi: 10.1038/s41586-023-05771-9. Epub 2023

Apr 12.

 

Antibodies against endogenous retroviruses promote lung cancer immunotherapy.

 

Ng KW(#)(1), Boumelha J(#)(2), Enfield KSS(#)(3), Almagro J(4), Cha H(5)(6),

Pich O(3),...

 

Author information:

(1)Retroviral Immunology Laboratory, The Francis Crick Institute, London, UK.

(2)Oncogene Biology Laboratory, The Francis Crick Institute, London, UK.

(3)Cancer Evolution and Genome Instability Laboratory, The Francis Crick

Institute, London, UK.

B cells are frequently found in the margins of solid tumours as organized

follicles in ectopic lymphoid organs called tertiary lymphoid structures

(TLS)1,2. Although TLS have been found to correlate with improved patient

survival and response to immune checkpoint blockade (ICB), the underlying

mechanisms of this association remain elusive1,2. Here we investigate

lung-resident B cell responses in patients from the TRACERx 421 (Tracking

Non-Small-Cell Lung Cancer Evolution Through Therapy) and other lung cancer

cohorts, and in a recently established immunogenic mouse model for lung

adenocarcinoma3. We find that both human and mouse lung adenocarcinomas elicit

local germinal centre responses and tumour-binding antibodies, and further

identify endogenous retrovirus (ERV) envelope glycoproteins as a dominant

anti-tumour antibody target. ERV-targeting B cell responses are amplified by ICB

in both humans and mice, and by targeted inhibition of KRAS(G12C) in the mouse

model. ERV-reactive antibodies exert anti-tumour activity that extends survival

in the mouse model, and ERV expression predicts the outcome of ICB in human lung

adenocarcinoma. Finally, we find that effective immunotherapy in the mouse model

requires CXCL13-dependent TLS formation. Conversely, therapeutic CXCL13

treatment potentiates anti-tumour immunity and synergizes with ICB. Our findings

provide a possible mechanistic basis for the association of TLS with

immunotherapy response.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41586-023-05771-9

PMCID: PMC10115647

PMID: 37046094 [Indexed for MEDLINE]

 

5. Nature. 2023 Apr;616(7957):543-552. doi: 10.1038/s41586-023-05706-4. Epub 2023

Apr 12.

 

Genomic-transcriptomic evolution in lung cancer and metastasis.

 

Martínez-Ruiz C(#)(1)(2), Black JRM(#)(1)(2), Puttick C(#)(1)(2)(3), Hill

MS(#)(3), …

 

Author information:

(1)Cancer Research UK Lung Cancer Centre of Excellence, University College

London Cancer Institute, London, UK.

(2)Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre

of Excellence, University College London Cancer Institute, London, UK.

(3)Cancer Evolution and Genome Instability Laboratory, The Francis Crick

Institute and University College London Cancer Institute, London, UK.

Intratumour heterogeneity (ITH) fuels lung cancer evolution, which leads to

immune evasion and resistance to therapy1. Here, using paired whole-exome and

RNA sequencing data, we investigate intratumour transcriptomic diversity in 354

non-small cell lung cancer tumours from 347 out of the first 421 patients

prospectively recruited into the TRACERx study2,3. Analyses of 947 tumour

regions, representing both primary and metastatic disease, alongside 96

tumour-adjacent normal tissue samples implicate the transcriptome as a major

source of phenotypic variation. Gene expression levels and ITH relate to

patterns of positive and negative selection during tumour evolution. We observe

frequent copy number-independent allele-specific expression that is linked to

epigenomic dysfunction. Allele-specific expression can also result in

genomic-transcriptomic parallel evolution, which converges on cancer gene

disruption. We extract signatures of RNA single-base substitutions and link

their aetiology to the activity of the RNA-editing enzymes ADAR and APOBEC3A,

thereby revealing otherwise undetected ongoing APOBEC activity in tumours.

Characterizing the transcriptomes of primary-metastatic tumour pairs, we combine

multiple machine-learning approaches that leverage genomic and transcriptomic

variables to link metastasis-seeding potential to the evolutionary context of

mutations and increased proliferation within primary tumour regions. These

results highlight the interplay between the genome and transcriptome in

influencing ITH, lung cancer evolution and metastasis.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41586-023-05706-4

PMCID: PMC10115639

PMID: 37046093 [Indexed for MEDLINE]

 

6. Nat Genet. 2023 Apr 6. doi: 10.1038/s41588-023-01355-5. Online ahead of print.

 

Genomic and transcriptomic analysis of checkpoint blockade response in advanced

non-small cell lung cancer.

 

Ravi A(#)(1)(2), Hellmann MD(#)(3), Arniella MB(#)(1), Holton M(1), Freeman

SS(1), …

 

Author information:

(1)Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard,

Cambridge, MA, USA.

(2)Lank Center for Genitourinary Oncology, Dana-Farber Cancer Institute, Boston,

MA, USA.

(3)AstraZeneca, Oncology R&D, New York, NY, USA.

Anti-PD-1/PD-L1 agents have transformed the treatment landscape of advanced

non-small cell lung cancer (NSCLC). To expand our understanding of the molecular

features underlying response to checkpoint inhibitors in NSCLC, we describe here

the first joint analysis of the Stand Up To Cancer-Mark Foundation cohort, a

resource of whole exome and/or RNA sequencing from 393 patients with NSCLC

treated with anti-PD-(L)1 therapy, along with matched clinical response

annotation. We identify a number of associations between molecular features and

outcome, including (1) favorable (for example, ATM altered) and unfavorable (for

example, TERT amplified) genomic subgroups, (2) a prominent association between

expression of inducible components of the immunoproteasome and response and (3)

a dedifferentiated tumor-intrinsic subtype with enhanced response to checkpoint

blockade. Taken together, results from this cohort demonstrate the complexity of

biological determinants underlying immunotherapy outcomes and reinforce the

discovery potential of integrative analysis within large, well-curated,

cancer-specific cohorts.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41588-023-01355-5

PMID: 37024582

 

7. Mol Cell. 2023 May 4;83(9):1474-1488.e8. doi: 10.1016/j.molcel.2023.04.007. Epub 2023 Apr 27.

 

Structural and functional basis of the universal transcription factor NusG

pro-pausing activity in Mycobacterium tuberculosis.

 

Delbeau M(1), Omollo EO(2), Froom R(3), Koh S(1), Mooney RA(2), Lilic M(1),

Brewer JJ(1), Rock J(4), Darst SA(5), Campbell EA(6), Landick R(7).

 

Author information:

(1)Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY

10065, USA.

(2)Department of Biochemistry, University of Wisconsin-Madison, Madison, WI

53706, USA.

(3)Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY

10065, USA; Laboratory of Host-Pathogen Biology, The Rockefeller University, New

York, NY 10065, USA.

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

10065, USA.

(5)Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY

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

(6)Laboratory of Molecular Biophysics, The Rockefeller University, New York, NY

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

(7)Department of Biochemistry, University of Wisconsin-Madison, Madison, WI

53706, USA; Department of Bacteriology, University of Wisconsin-Madison,

Madison, WI 53706, USA. Electronic address: rlandick@wisc.edu.

 

Transcriptional pauses mediate regulation of RNA biogenesis. DNA-encoded pause

signals trigger pausing by stabilizing RNA polymerase (RNAP) swiveling and

inhibiting DNA translocation. The N-terminal domain (NGN) of the only universal

transcription factor, NusG/Spt5, modulates pausing through contacts to RNAP and

DNA. Pro-pausing NusGs enhance pauses, whereas anti-pausing NusGs suppress

pauses. Little is known about pausing and NusG in the human pathogen

Mycobacterium tuberculosis (Mtb). We report that MtbNusG is pro-pausing. MtbNusG

captures paused, swiveled RNAP by contacts to the RNAP protrusion and

nontemplate-DNA wedged between the NGN and RNAP gate loop. In contrast,

anti-pausing Escherichia coli (Eco) NGN contacts the MtbRNAP gate loop,

inhibiting swiveling and pausing. Using CRISPR-mediated genetics, we show that

pro-pausing NGN is required for mycobacterial fitness. Our results define an

essential function of mycobacterial NusG and the structural basis of pro- versus

anti-pausing NusG activity, with broad implications for the function of all NusG

orthologs.

 

Copyright © 2023 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.molcel.2023.04.007

PMID: 37116494 [Indexed for MEDLINE]

 

8. J Clin Oncol. 2023 Apr 25:JCO2202524. doi: 10.1200/JCO.22.02524. Online ahead of print.

 

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

 

9. 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.

 

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]

 

10. Nat Commun. 2023 Apr 20;14(1):2280. doi: 10.1038/s41467-023-38045-z.

 

Structural insights into RNase J that plays an essential role in Mycobacterium

tuberculosis RNA metabolism.

 

Bao L(#)(1), Hu J(#)(1), Zhan B(#)(1), Chi M(1), Li Z(1), Wang S(1), Shan C(2),

Zhao Z(2), Guo Y(1), Ding X(2), Ji C(1), Tao S(3), Ni T(2), Zhang X(4), Zhao

G(5)(6), Li J(7)(8).

 

Author information:

(1)State Key Laboratory of Genetic Engineering, School of Life Sciences and

Huashan Hospital, Shanghai Engineering Research Center of Industrial

Microorganisms, Engineering Research Center of Gene Technology of MOE, Fudan

University, 200438, Shanghai, China.

(2)State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan

University, 200438, Shanghai, China.

(3)Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai

Center for Systems Biomedicine, Shanghai Jiao Tong University, 200240, Shanghai,

China.

Ribonucleases (RNases) are responsible for RNA metabolism. RNase J, the core

enzyme of the RNA degradosome, plays an essential role in global mRNA decay.

Emerging evidence showed that the RNase J of Mycobacterium tuberculosis

(Mtb-RNase J) could be an excellent target for treating Mtb infection. Here,

crystal structures of Mtb-RNase J in apo-state and complex with the

single-strand RNA reveal the conformational change upon RNA binding and

hydrolysis. Mtb-RNase J forms an active homodimer through the interactions

between the β-CASP and the β-lactamase domain. Knockout of RNase J slows the

growth rate and changes the colony morphologies and cell length in Mycobacterium

smegmatis, which is restored by RNase J complementation. Finally, RNA-seq

analysis shows that the knockout strain significantly changes the expression

levels of 49 genes in metabolic pathways. Thus, our current study explores the

structural basis of Mtb-RNase J and might provide a promising candidate in

pharmacological treatment for tuberculosis.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41467-023-38045-z

PMCID: PMC10119312

PMID: 37080992 [Indexed for MEDLINE]

 

11. J Clin Invest. 2023 Apr 17;133(8):e159941. doi: 10.1172/JCI159941.

 

Mycobacterium tuberculosis hijacks host TRIM21- and NCOA4-dependent

ferritinophagy to enhance intracellular growth.

 

Dai Y(1)(2), Zhu C(3), Xiao W(1)(4), Huang K(5), Wang X(6), Shi C(1), Lin D(1),

Zhang H(1)(2), Liu X(7)(8), Peng B(9), Gao Y(10), Liu CH(11), Ge B(12), Kaufmann

SH(13)(14)(15), Feng CG(16), Chen X(1)(4), Cai Y(1).

 

Author information:

(1)Guangdong Provincial Key Laboratory of Regional Immunity and Diseases,

Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen,

China.

(2)Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou,

China.

(3)Laboratory of Molecular Biology, Beijing Key Laboratory for Drug Resistance

Tuberculosis Research, Beijing Chest Hospital, Capital Medical University,

Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, China.

Ferritin, a key regulator of iron homeostasis in macrophages, has been reported

to confer host defenses against Mycobacterium tuberculosis (Mtb) infection.

Nuclear receptor coactivator 4 (NCOA4) was recently identified as a cargo

receptor in ferritin degradation. Here, we show that Mtb infection enhanced

NCOA4-mediated ferritin degradation in macrophages, which in turn increased the

bioavailability of iron to intracellular Mtb and therefore promoted bacterial

growth. Of clinical relevance, the upregulation of FTH1 in macrophages was

associated with tuberculosis (TB) disease progression in humans.

Mechanistically, Mtb infection enhanced NCOA4-mediated ferritin degradation

through p38/AKT1- and TRIM21-mediated proteasomal degradation of HERC2, an E3

ligase of NCOA4. Finally, we confirmed that NCOA4 deficiency in myeloid cells

expedites the clearance of Mtb infection in a murine model. Together, our

findings revealed a strategy by which Mtb hijacks host ferritin metabolism for

its own intracellular survival. Therefore, this represents a potential target

for host-directed therapy against tuberculosis.

 

DOI: 10.1172/JCI159941

PMCID: PMC10104892

PMID: 37066876 [Indexed for MEDLINE]

 

12. Cancer Discov. 2023 Apr 16:CD-22-0620. doi: 10.1158/2159-8290.CD-22-0620. Online ahead of print.

 

Integrative analysis of a large real-world cohort of small cell lung cancer

identifies distinct genetic subtypes and insights into histological

transformation.

 

Sivakumar S(1), Moore JA(2), Montesion M(3), Sharaf R(3), Lin DI(1), Colon

CI(4), Fleischmann Z(1), Ebot EM(1), Newberg JY(5), Mills JM(6), Hegde PS(3),

Pan Q(7), Dowlati A(8), Frampton GM(2), Sage J(9), Lovly CM(10).

 

Author information:

(1)Foundation Medicine, Cambridge, MA, United States.

(2)Foundation Medicine Inc., Cambridge, MA, United States.

(3)Foundation Medicine, Inc., Cambridge, MA, United States.

Small cell lung cancer (SCLC) is a recalcitrant neuroendocrine carcinoma with

dismal survival outcomes. A major barrier in the field has been the relative

paucity of human tumors studied. Here we provide an integrated analysis of 3,600

"real-world" SCLC cases. This large cohort allowed us to identify new recurrent

alterations and genetic subtypes, including STK11-mutant tumors (1.7%) and

TP53/RB1 wild-type tumors (5.5%), as well as rare cases that were human

papillomavirus-positive. In our cohort, gene amplifications on 4q12 are

associated with increased overall survival while CCNE1 amplification is

associated with decreased overall survival. We also identify more frequent

alterations in the PTEN pathway in brain metastases. Finally, profiling cases of

SCLC containing oncogenic drivers typically associated with NSCLC demonstrates

that SCLC transformation may occur across multiple distinct molecular cohorts of

NSCLC. These novel and unsuspected genetic features of SCLC may help personalize

treatment approaches for this fatal form of cancer.

 

DOI: 10.1158/2159-8290.CD-22-0620

PMID: 37062002

 

13. Nat Commun. 2023 Apr 8;14(1):1988. doi: 10.1038/s41467-023-37719-y.

 

The relative transmission fitness of multidrug-resistant Mycobacterium

tuberculosis in a drug resistance hotspot.

 

Loiseau C(#)(1)(2), Windels EM(#)(3)(4), Gygli SM(1)(2), Jugheli L(1)(2)(5),

Maghradze N(1)(2)(5), Brites D(1)(2), Ross A(1)(2), Goig G(1)(2), Reinhard

M(1)(2), Borrell S(1)(2), Trauner A(1)(2), Dötsch A(1)(2), Aspindzelashvili

R(5), Denes R(6), Reither K(1)(2), Beisel C(6), Tukvadze N(1)(2)(5), Avaliani

Z(5), Stadler T(6)(7), Gagneux S(8)(9).

 

Author information:

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

(2)University of Basel, Basel, Switzerland.

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

Switzerland. etthel.windels@bsse.ethz.ch.

Multidrug-resistant tuberculosis (MDR-TB) is among the most frequent causes of

death due to antimicrobial resistance. Although only 3% of global TB cases are

MDR, geographical hotspots with up to 40% of MDR-TB have been observed in

countries of the former Soviet Union. While the quality of TB control and

patient-related factors are known contributors to such hotspots, the role of the

pathogen remains unclear. Here we show that in the country of Georgia, a known

hotspot of MDR-TB, MDR Mycobacterium tuberculosis strains of lineage 4 (L4)

transmit less than their drug-susceptible counterparts, whereas most MDR strains

of L2 suffer no such defect. Our findings further indicate that the high

transmission fitness of these L2 strains results from epistatic interactions

between the rifampicin resistance-conferring mutation RpoB S450L, compensatory

mutations in the RNA polymerase, and other pre-existing genetic features of

L2/Beijing clones that circulate in Georgia. We conclude that the transmission

fitness of MDR M. tuberculosis strains is heterogeneous, but can be as high as

drug-susceptible forms, and that such highly drug-resistant and transmissible

strains contribute to the emergence and maintenance of hotspots of MDR-TB. As

these strains successfully overcome the metabolic burden of drug resistance, and

given the ongoing rollout of new treatment regimens against MDR-TB, proper

surveillance should be implemented to prevent these strains from acquiring

resistance to the additional drugs.

 

© 2023. The Author(s).

 

DOI: 10.1038/s41467-023-37719-y

PMCID: PMC10082831

PMID: 37031225 [Indexed for MEDLINE]

 

14. J Clin Oncol. 2023 Apr 6:JCO2300282. doi: 10.1200/JCO.23.00282. Online ahead of print.

 

Therapy for Stage IV Non-Small-Cell Lung Cancer Without Driver Alterations: ASCO

Living Guideline, Version 2023.1.

 

Singh N(1), Jaiyesimi IA(2), Ismaila N(3), Leighl NB(4), Mamdani H(5), Phillips

T(6), Owen DH(7).

 

Author information:

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

(2)Corewell Health William Beaumont University Hospital Royal Oak and Oakland

University William Beaumont School of Medicine, Rochester, MI.

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

(4)Princess Margaret Cancer Center, University Health Network, Toronto, Ontario,

Canada.

(5)Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State

University, Detroit, MI.

(6)City of Hope, Duarte, CA.

(7)Ohio State University, Columbus, OH.

 

Update of

    J Clin Oncol. 2022 Oct 1;40(28):3323-3343.

 

Living guidelines are developed for selected topic areas with rapidly evolving

evidence that drives frequent change in recommended 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-non-da-living-guideline.

 

DOI: 10.1200/JCO.23.00282

PMID: 37023387

 

15. J Clin Oncol. 2023 Apr 6:JCO2300281. doi: 10.1200/JCO.23.00281. Online ahead of print.

 

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

Living Guideline, Version 2023.1.

 

Singh N(1), Jaiyesimi IA(2), Ismaila N(3), Leighl NB(4), Mamdani H(5), Phillips

T(6), Owen DH(7).

 

Author information:

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

(2)Corewell Health William Beaumont University Hospital Royal Oak and Oakland

University William Beaumont School of Medicine, Rochester, MI.

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

(4)Princess Margaret Cancer Center, University Health Network, Toronto, Ontario,

Canada.

(5)Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State

University, Detroit, MI.

(6)City of Hope, Duarte, CA.

(7)Ohio State University, Columbus, OH.

 

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 recommended 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, online only). Updates are

published regularly and can be found at

https://ascopubs.org/nsclc-da-living-guideline.

 

DOI: 10.1200/JCO.23.00281

PMID: 37023367

 

16. J Clin Oncol. 2023 Apr 5:JCO2201728. doi: 10.1200/JCO.22.01728. Online ahead of print.

 

Endosonography With or Without Confirmatory Mediastinoscopy for Resectable Lung

Cancer: A Randomized Clinical Trial.

 

Bousema JE(1), Dijkgraaf MGW(2), van der Heijden EHFM(3), Verhagen AFTM(4),

Annema JT(5), van den Broek FJC(1); MEDIASTrial study group.

 

Author information:

(1)Department of Surgery, Máxima MC, Veldhoven, the Netherlands.

(2)Amsterdam UMC Location University of Amsterdam, Epidemiology and Data

Science, Amsterdam Public Health, Methodology, Amsterdam, the Netherlands.

(3)Department of Pulmonary Medicine, Radboud University Medical Center,

Nijmegen, the Netherlands.

PURPOSE: Resectable non-small-cell lung cancer (NSCLC) with a high probability

of mediastinal nodal involvement requires mediastinal staging by endosonography

and, in the absence of nodal metastases, confirmatory mediastinoscopy according

to current guidelines. However, randomized data regarding immediate lung tumor

resection after systematic endosonography versus additional confirmatory

mediastinoscopy before resection are lacking.

METHODS: Patients with (suspected) resectable NSCLC and an indication for

mediastinal staging after negative systematic endosonography were randomly

assigned to immediate lung tumor resection or confirmatory mediastinoscopy

followed by tumor resection. The primary outcome in this noninferiority trial

(noninferiority margin of 8% that previously showed to not compromise survival,

Pnoninferior < .0250) was the presence of unforeseen N2 disease after tumor

resection with lymph node dissection. Secondary outcomes were 30-day major

morbidity and mortality.

RESULTS: Between July 17, 2017, and October 5, 2020, 360 patients were randomly

assigned, 178 to immediate lung tumor resection (seven dropouts) and 182 to

confirmatory mediastinoscopy first (seven dropouts before and six after

mediastinoscopy). Mediastinoscopy detected metastases in 8.0% (14/175; 95% CI,

4.8 to 13.0) of patients. Unforeseen N2 rate after immediate resection (8.8%)

was noninferior compared with mediastinoscopy first (7.7%) in both

intention-to-treat (Δ, 1.03%; UL 95% CIΔ, 7.2%; Pnoninferior = .0144) and

per-protocol analyses (Δ, 0.83%; UL 95% CIΔ, 7.3%; Pnoninferior = .0157). Major

morbidity and 30-day mortality was 12.9% after immediate resection versus 15.4%

after mediastinoscopy first (P = .4940).

CONCLUSION: On the basis of our chosen noninferiority margin in the rate of

unforeseen N2, confirmatory mediastinoscopy after negative systematic

endosonography can be omitted in patients with resectable NSCLC and an

indication for mediastinal staging.

 

DOI: 10.1200/JCO.22.01728

PMID: 37018653

 

17. ACS Nano. 2023 Apr 11;17(7):6998-7006. doi: 10.1021/acsnano.3c01374. Epub 2023 Apr 3.

 

Sensitive Urine Immunoassay for Visualization of Lipoarabinomannan for

Noninvasive Tuberculosis Diagnosis.

 

Chen P(1), Meng Y(1), Liu T(1), Peng W(1), Gao Y(1), He Y(1), Qu R(1), Zhang

C(1), Hu W(1), Ying B(1).

 

Author information:

(1)Department of Laboratory Medicine, Med+X Center for Manufacturing, Department

of Radiology, National Clinical Research Center for Geriatrics, West China

Hospital, Sichuan University, Chengdu, Sichuan 610041, China.

 

Lipoarabinomannan (LAM) is a prospective noninvasive biomarker for tuberculosis

(TB) diagnosis. Here, we report a visual immunoassay of high sensitivity for

detecting LAM in urine samples toward TB diagnosis. This method uses a

DNA-linked immunosorbent of LAM, followed by a transduction cascade into

amplified visual signals using quantum dots (QDs) and calcein reaction with Cu2+

and copper nanoparticles (Cu NPs). The limit of detection (LOD) for LAM in the

urine reaches 2.5 fg/mL and 25 fg/mL using a fluorometer and length readouts on

strips, respectively, demonstrating an ultrahigh sensitivity. The clinical

validation of the proposed assay was performed with 147 HIV-negative clinical

urine specimens. The results show the sensitivity of test is 94.1% (16/17) for

confirmed TB (culture-positive) and 85% (51/60) for unconfirmed TB (clinical

diagnosis without positive culture results), respectively, when the test cutoff

value is 40 fg/mL for TB. Its specificity is 89.2% (25/28) in non-TB and

nontuberculous mycobacterial patients. The area under the curve (AUC) was 0.86

when controls were non-TB and LTBI patients, while the AUC was 0.92 when

controls were only non-TB patients. This highly sensitive visual immunoassay of

LAM has shown potential for noninvasive diagnosis of TB using urine samples.

 

DOI: 10.1021/acsnano.3c01374

PMID: 37010068 [Indexed for MEDLINE]

 

18. Lancet Infect Dis. 2023 Apr;23(4):e138-e150. doi: 10.1016/S1473-3099(23)00004-X.

 

Post-tuberculosis sequelae in children and adolescents: a systematic review.

 

Igbokwe V(1), Ruby LC(2), Sultanli A(3), Bélard S(4).

 

Author information:

(1)Department of Pediatric Respiratory Medicine, Immunology and Critical Care

Medicine, Humboldt-Universität zu Berlin, Berlin, Germany.

(2)Department of Infectious Diseases, and Respiratory Medicine,

Charité-Universitätsmedizin Berlin, Freie Universität Berlin and

Humboldt-Universität zu Berlin, Berlin, Germany.

(3)Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany;

German Center for Infection Research, Tübingen, Germany.

(4)Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany;

German Center for Infection Research, Tübingen, Germany. Electronic address:

sabine.belard@med.uni-tuebingen.de.

 

In 2020, an estimated total of 155 million people had survived tuberculosis.

Among this number, a sizable proportion have considerable post-tuberculosis

morbidity, as shown for the adult population. This systematic review aims to

identify the spectrum and prevalence of post-tuberculosis sequelae in children

and adolescents. Four databases were systematically searched from database

inception to Feb 7, 2022, for literature on post-treatment outcomes of

tuberculosis acquired during childhood. Of the 4613 identified publications, 71

studies were included in this systematic review. Studies on cohorts with

comparably rare (most of which were extrapulmonary) tuberculosis presentations,

such as spinal tuberculosis and tuberculous meningitis were over-represented;

however, no study assessed long-term sequelae in a cohort with an average

childhood tuberculosis spectrum. The descriptive analysis includes long-term

outcomes of 3529 paediatric patients 1 month to 36 years after confirmed (47%)

or clinical (53%) tuberculosis. In a considerable proportion of children, a

broad spectrum of post-tuberculosis sequelae were identified, ranging from

radiological residua after pulmonary tuberculosis, to disabling deformities

after musculoskeletal and cutaneous tuberculosis, to somatic and psychosocial

impairment after tuberculous meningitis. A better understanding and

comprehensive assessment of post-tuberculosis sequelae in children are needed to

improve tuberculosis care beyond antituberculous treatment.

 

Copyright © 2023 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(23)00004-X

PMID: 36963920 [Indexed for MEDLINE]

 

19. Nano Lett. 2023 Apr 12;23(7):2964-2973. doi: 10.1021/acs.nanolett.3c00354. Epub 2023 Mar 22.

 

Alveolar Macrophages-Mediated Translocation of Intratracheally Delivered

Perfluorocarbon Nanoparticles to Achieve Lung Cancer (19)F-MR Imaging.

 

Wang H(1)(2), Li X(1)(2), Wang J(1)(2), Wang J(1)(2), Zou H(1)(2), Hu X(1)(2),

Yang L(1)(2), Shen P(1)(2), A R(1)(2), Wang K(1)(2), Li Y(1)(2), Yang J(1)(2),

Wang K(1)(2), Yang L(1)(2), Wu L(1)(2), Sun X(1)(2).

 

Author information:

(1)Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical

University, Harbin 150028, China.

(2)NHC Key Laboratory of Molecular Probe and Targeted Diagnosis and Therapy,

Molecular Imaging Research Center (MIRC), Harbin Medical University, Harbin

150028, China.

 

Recent advances in intratracheal delivery strategies have sparked considerable

biomedical interest in developing this promising approach for lung cancer

diagnosis and treatment. However, there are very few relevant studies on the

behavior and mechanism of imaging nanoparticles (NPs) after intratracheal

delivery. Here, we found that nanosized perfluoro-15-crown-5-ether (PFCE NPs,

200 nm) exhibite significant 19F-MRI signal-to-noise ratio (SNR) enhancement

than perfluorooctyl bromide (PFOB NPs) up to day 7 after intratracheal delivery.

Alveolar macrophages (AMs) engulf PFCE NPs, become PFCE NPs-laden AMs, and then

migrate into the tumor margin, resulting in increased tumor PFCE concentration

and 19F-MRI signals. AMs-mediated translocation of PFCE NPs to lung draning

lymph nodes (dLNs) decreases the background PFCE concentration. Our results shed

light on the dynamic AMs-mediated translocation of intratracheally delivered PFC

NPs for effective lung tumor visualization and reveal a pathway to develop and

promote the clinical translation of an intratracheal delivery-based imaging

strategy.

 

DOI: 10.1021/acs.nanolett.3c00354

PMID: 36947431 [Indexed for MEDLINE]

 

20. J Clin Invest. 2023 Apr 3;133(7):e164413. doi: 10.1172/JCI164413.

 

Kras oncogene ablation prevents resistance in advanced lung adenocarcinomas.

 

Salmón M(1), Álvarez-Díaz R(1), Fustero-Torre C(2), Brehey O(1), Lechuga CG(1),

Sanclemente M(1), Fernández-García F(1), López-García A(1), Martín-Guijarro

MC(3), Rodríguez-Perales S(3), Bousquet-Mur E(1), Morales-Cacho L(1), Mulero

F(4), Al-Shahrour F(2), Martínez L(5), Domínguez O(6), Caleiras E(7), Ortega

S(8), Guerra C(1)(9), Musteanu M(1)(9)(10), Drosten M(1)(9)(11), Barbacid

M(1)(9).

 

Author information:

(1)Experimental Oncology Group, Molecular Oncology Program.

(2)Bioinformatics Unit.

(3)Molecular Cytogenetics and Genome Editing Unit, Human Cancer Genetics

Program.

KRASG12C inhibitors have revolutionized the clinical management of patients with

KRASG12C-mutant lung adenocarcinoma. However, patient exposure to these

inhibitors leads to the rapid onset of resistance. In this study, we have used

genetically engineered mice to compare the therapeutic efficacy and the

emergence of tumor resistance between genetic ablation of mutant Kras expression

and pharmacological inhibition of oncogenic KRAS activity. Whereas Kras ablation

induces massive tumor regression and prevents the appearance of resistant cells

in vivo, treatment of KrasG12C/Trp53-driven lung adenocarcinomas with sotorasib,

a selective KRASG12C inhibitor, caused a limited antitumor response similar to

that observed in the clinic, including the rapid onset of resistance. Unlike in

human tumors, we did not observe mutations in components of the RAS-signaling

pathways. Instead, sotorasib-resistant tumors displayed amplification of the

mutant Kras allele and activation of xenobiotic metabolism pathways, suggesting

that reduction of the on-target activity of KRASG12C inhibitors is the main

mechanism responsible for the onset of resistance. In sum, our results suggest

that resistance to KRAS inhibitors could be prevented by achieving a more robust

inhibition of KRAS signaling mimicking the results obtained upon Kras ablation.

 

DOI: 10.1172/JCI164413

PMCID: PMC10065067

PMID: 36928090 [Indexed for MEDLINE]

 

21. Lancet Infect Dis. 2023 Apr;23(4):e122-e137. doi: 10.1016/S1473-3099(22)00875-1. Epub 2023 Feb 28.

 

Clinical implications of molecular drug resistance testing for Mycobacterium

tuberculosis: a 2023 TBnet/RESIST-TB consensus statement.

 

Domínguez J(1), Boeree MJ(2), Cambau E(3), Chesov D(4), Conradie F(5), Cox V(6),

Dheda K(7), Dudnyk A(8), Farhat MR(9), Gagneux S(10), Grobusch MP(11), Gröschel

MI(12), Guglielmetti L(13), Kontsevaya I(14), Lange B(15), van Leth F(16),

Lienhardt C(17), Mandalakas AM(18), Maurer FP(19), Merker M(20), Miotto P(21),

Molina-Moya B(22), Morel F(13), Niemann S(23), Veziris N(13), Whitelaw A(24),

Horsburgh CR Jr(25), Lange C(18); TBnet and RESIST-TB networks.

 

Author information:

(1)Institut d'Investigació Germans Trias i Pujol, Universitat Autònoma de

Barcelona, CIBER Enfermedades Respiratorias, INNOVA4TB Consortium, Barcelona,

Spain. Electronic address: jadominguez@igtp.cat.

(2)Department of Lung Diseases, Radboud Institute for Health Sciences, Radboud

University Medical Center, Nijmegen, Netherlands.

(3)Centre National de Référence des Mycobactéries et de la Résistance des

Mycobactéries aux Antituberculeux, Paris, France, APHP-Hôpital Bichat,

Mycobacteriology Laboratory, INSERM, University Paris Cite, IAME UMR1137, Paris,

France.

 

Erratum in

    Lancet Infect Dis. 2023 Mar 28;:

 

Drug-resistant tuberculosis is a substantial health-care concern worldwide.

Despite culture-based methods being considered the gold standard for drug

susceptibility testing, molecular methods provide rapid information about the

Mycobacterium tuberculosis mutations associated with resistance to

anti-tuberculosis drugs. This consensus document was developed on the basis of a

comprehensive literature search, by the TBnet and RESIST-TB networks, about

reporting standards for the clinical use of molecular drug susceptibility

testing. Review and the search for evidence included hand-searching journals and

searching electronic databases. The panel identified studies that linked

mutations in genomic regions of M tuberculosis with treatment outcome data.

Implementation of molecular testing for the prediction of drug resistance in M

tuberculosis is key. Detection of mutations in clinical isolates has

implications for the clinical management of patients with multidrug-resistant or

rifampicin-resistant tuberculosis, especially in situations when phenotypic drug

susceptibility testing is not available. A multidisciplinary team including

clinicians, microbiologists, and laboratory scientists reached a consensus on

key questions relevant to molecular prediction of drug susceptibility or

resistance to M tuberculosis, and their implications for clinical practice. This

consensus document should help clinicians in the management of patients with

tuberculosis, providing guidance for the design of treatment regimens and

optimising outcomes.

 

Copyright © 2023 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(22)00875-1

PMID: 36868253 [Indexed for MEDLINE]

 

22. J Clin Oncol. 2023 Apr 10;41(11):1992-1998. doi: 10.1200/JCO.22.01989. Epub 2023 Feb 21.

 

Pembrolizumab Plus Pemetrexed and Platinum in Nonsquamous Non-Small-Cell Lung

Cancer: 5-Year Outcomes From the Phase 3 KEYNOTE-189 Study.

 

Garassino MC(1)(2), Gadgeel S(3), Speranza G(4), Felip E(5), Esteban E(6),

Dómine M(7), Hochmair MJ(8), Powell SF(9), Bischoff HG(10), Peled N(11), Grossi

F(12), Jennens RR(13), Reck M(14), Hui R(15), Garon EB(16), Kurata T(17), Gray

JE(18), Schwarzenberger P(19), Jensen E(19), Pietanza MC(19), Rodríguez-Abreu

D(20).

 

Author information:

(1)Knapp Center for Biomedical Discovery, University of Chicago Medicine &

Biological Sciences, Chicago, IL.

(2)Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.

(3)Henry Ford Cancer Institute/Henry Ford Health, Detroit, MI.

Clinical trials frequently include multiple end points that mature at different

times. The initial report, typically on the 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.We present 5-year outcomes from the

phase 3 KEYNOTE-189 study (ClinicalTrials.gov identifier: NCT02578680). Eligible

patients with previously untreated metastatic nonsquamous non-small-cell lung

cancer without EGFR/ALK alterations were randomly assigned 2:1 to pembrolizumab

200 mg or placebo once every 3 weeks for up to 35 cycles with pemetrexed and

investigator's choice of carboplatin/cisplatin for four cycles, followed by

maintenance pemetrexed until disease progression or unacceptable toxicity.

Primary end points were overall survival (OS) and progression-free survival

(PFS). Among 616 randomly assigned patients (n = 410, pembrolizumab plus

pemetrexed-platinum; n = 206, placebo plus pemetrexed-platinum), median time

from random assignment to data cutoff (March 8, 2022) was 64.6 (range,

60.1-72.4) months. Hazard ratio (95% CI) for OS was 0.60 (0.50 to 0.72) and PFS

was 0.50 (0.42 to 0.60) for pembrolizumab plus platinum-pemetrexed versus

placebo plus platinum-pemetrexed. 5-year OS rates were 19.4% versus 11.3%.

Toxicity was manageable. Among 57 patients who completed 35 cycles of

pembrolizumab, objective response rate was 86.0% and 3-year OS rate after

completing 35 cycles (approximately 5 years after random assignment) was 71.9%.

Pembrolizumab plus pemetrexed-platinum maintained OS and PFS benefits versus

placebo plus pemetrexed-platinum, regardless of programmed cell death ligand-1

expression. These data continue to support pembrolizumab plus

pemetrexed-platinum as a standard of care in previously untreated metastatic

non-small-cell lung cancer without EGFR/ALK alterations.

 

DOI: 10.1200/JCO.22.01989

PMCID: PMC10082311

PMID: 36809080 [Indexed for MEDLINE]

 

23. J Clin Oncol. 2023 Apr 10;41(11):e21-e30. doi: 10.1200/JCO.22.02783. Epub 2023 Feb 21.

 

Therapy for Stage IV Non-Small-Cell Lung Cancer Without Driver Alterations: ASCO

Living Guideline, Version 2022.3.

 

Jaiyesimi IA(1)(2), Owen DH(3), Ismaila N(4), Blanchard E(5), Celano P(6),

Florez N(7), Jain D(8), Singh N(9).

 

Author information:

(1)Corewell Health William Beaumont University Hospital Royal Oak, MI.

(2)Oakland University William Beaumont School of Medicine, Rochester, MI.

(3)Ohio State University, Columbus, OH.

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

(5)Southcoast Centers for Cancer Care, New Bedford, MA.

(6)The Cancer Center at GBMC, Towson, MD.

(7)Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA.

(8)Houston Methodist Cancer Center, Houston, TX.

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

 

Update of

    J Clin Oncol. 2022 Oct 1;40(28):3323-3343.

 

Living guidelines are developed for selected topic areas with rapidly evolving

evidence that drives frequent change in recommended 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-non-da-living-guideline.

 

DOI: 10.1200/JCO.22.02783

PMID: 36809066 [Indexed for MEDLINE]

 

24. J Clin Oncol. 2023 Apr 10;41(11):e31-e41. doi: 10.1200/JCO.22.02782. Epub 2023 Feb 21.

 

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

Living Guideline, Version 2022.3.

 

Jaiyesimi IA(1)(2), Owen DH(3), Ismaila N(4), Blanchard E(5), Celano P(6),

Florez N(7), Jain D(8), Singh N(9).

 

Author information:

(1)Corewell Health William Beaumont University Hospital, Royal Oak, MI.

(2)Oakland University William Beaumont School of Medicine, Rochester, MI.

(3)Ohio State University, Columbus, OH.

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.22.02782

PMID: 36802359 [Indexed for MEDLINE]

 

25. Angew Chem Int Ed Engl. 2023 Apr 17;62(17):e202300221. doi:

10.1002/anie.202300221. Epub 2023 Mar 13.

 

Chemical Validation of Mycobacterium tuberculosis Phosphopantetheine

Adenylyltransferase Using Fragment Linking and CRISPR Interference.

 

El Bakali J(1)(2), Blaszczyk M(3)(4), Evans JC(5)(6), Boland JA(1), McCarthy

WJ(1)(7), Fathoni I(8), Dias MVB(3)(9), Johnson EO(6), Coyne AG(1), Mizrahi

V(5), Blundell TL(3), Abell C(1), Spry C(1)(8).

 

Author information:

(1)Yusuf Hamied Department of Chemistry, University of Cambridge, Lensfield

Road, Cambridge, CB2 1EW, UK.

(2)Present address: Univ. Lille, Inserm, CHU Lille, UMR-S 1172-LiNC-Lille

Neuroscience & Cognition, 59000, Lille, France.

(3)Department of Biochemistry, University of Cambridge, 80 Tennis Court Road,

Cambridge, CB2 1GA, UK.

The coenzyme A (CoA) biosynthesis pathway has attracted attention as a potential

target for much-needed novel antimicrobial drugs, including for the treatment of

tuberculosis (TB), the lethal disease caused by Mycobacterium tuberculosis

(Mtb). Seeking to identify inhibitors of Mtb phosphopantetheine

adenylyltransferase (MtbPPAT), the enzyme that catalyses the penultimate step in

CoA biosynthesis, we performed a fragment screen. In doing so, we discovered

three series of fragments that occupy distinct regions of the MtbPPAT active

site, presenting a unique opportunity for fragment linking. Here we show how,

guided by X-ray crystal structures, we could link weakly-binding fragments to

produce an active site binder with a KD<20 μM and on-target anti-Mtb activity,

as demonstrated using CRISPR interference. This study represents a big step

toward validating MtbPPAT as a potential drug target and designing a

MtbPPAT-targeting anti-TB drug.

 

© 2023 The Authors. Angewandte Chemie International Edition published by

Wiley-VCH GmbH.

 

DOI: 10.1002/anie.202300221

PMID: 36757665 [Indexed for MEDLINE]

 

26. Am J Respir Crit Care Med. 2023 Apr 15;207(8):1080-1088. doi:

10.1164/rccm.202208-1543OC.

 

The Long-Term Impact of Early-Life Tuberculosis Disease on Child Health: A

Prospective Birth Cohort Study.

 

Martinez L(1), Gray DM(2)(3), Botha M(2)(3), Nel M(2)(3), Chaya S(2)(3), Jacobs

C(2)(3), Workman L(2)(3), Nicol MP(3)(4)(5), Zar HJ(2)(3).

 

Author information:

(1)Department of Epidemiology, School of Public Health, Boston University,

Boston, Massachusetts.

(2)Department of Paediatrics and Child Health, Red Cross War Memorial Children's

Hospital.

(3)SA-Medical Research Council Unit on Child and Adolescent Health, and.

(4)Division of Medical Microbiology, University of Cape Town, Cape Town, South

Africa; and.

(5)Marshall Centre for Infection and Immunity, School of Biomedical Sciences,

University of Western Australia, Perth, Western Australia, Australia.

 

Comment in

    Am J Respir Crit Care Med. 2023 Apr 15;207(8):975-977.

 

Rationale: There is growing concern that post-tuberculosis disease (TB) sequelae

and morbidity are substantial, but no studies have controlled for preexisting

factors before disease. Whether children have post-TB morbidity is not well

characterized. Objectives: To assess the effect of a TB diagnosis on wheezing

episodes, lung function, and anthropometric measurements among children enrolled

in a prospective birth cohort study in South Africa. Methods: We prospectively

followed children from birth through 5 years for TB using diagnostic tests

including chest radiography and repeated induced sputum sample testing with

Xpert MTB/RIF and liquid culture. We longitudinally measured health outcomes

including growth, wheezing, and lung function up to 5 years. Mixed-effects

linear regression models were used to assess growth and lung function after TB.

Poisson regression was used to assess risk of subsequent wheezing. Measurements

and Main Results: Among 1,068 participants, 96 TB cases occurred (1,228 cases

per 100,000 person-years [95% confidence interval (CI), 1,006-1,500]) occurred

over 7,815 child-years of follow-up. TB was associated with lower length-for-age

(-0.40 [95% CI, -0.68 to -0.11]), weight-for-age (-0.30 [95% CI, -0.59 to

-0.01]), and body mass index (-0.54 [95% CI, -0.83 to -0.25]) z-scores at 5

years. Children developing TB were consistently more likely to wheeze regardless

of the timing of TB. Children with diagnoses of TB between 0 and 1 year of age

had reduced time to peak tidal expiratory flow over total expiratory time

(-2.35% [95% CI, -4.86% to -0.17%]) and higher fractional exhaled nitric oxide

(2.88 ppb [95% CI, 0.57-5.19 ppb]) at 5 years. Children with diagnoses of TB

between 1 and 4 years of age had impaired Vt (-9.32 ml [95% CI, -14.89 to -3.75

ml]) and time to peak tidal expiratory flow over total expiratory time (-2.73%

[95% CI, -5.45% to -0.01%]) at 5 years. Conclusions: Prevention of TB disease in

the first few years of life may have substantial long-term benefits through

childhood.

 

DOI: 10.1164/rccm.202208-1543OC

PMCID: PMC10112440

PMID: 36746196 [Indexed for MEDLINE]

 

27. J Clin Oncol. 2023 Apr 10;41(11):1999-2006. doi: 10.1200/JCO.22.01990. Epub 2023 Feb 3.

 

Pembrolizumab Plus Chemotherapy in Squamous Non-Small-Cell Lung Cancer: 5-Year

Update of the Phase III KEYNOTE-407 Study.

 

Novello S(1), Kowalski DM(2), Luft A(3), Gümüş M(4), Vicente D(5), Mazières

J(6), Rodríguez-Cid J(7), Tafreshi A(8), Cheng Y(9), Lee KH(10), Golf A(11),

Sugawara S(12), Robinson AG(13), Halmos B(14), Jensen E(15), Schwarzenberger

P(16), Pietanza MC(16), Paz-Ares L(17).

 

Author information:

(1)Department of Oncology, University of Turin, Azienda Ospedaliero

Universitaria San Luigi, Turin, Italy.

(2)Department of Lung Cancer and Thoracic Tumours, Maria Sklodowska-Curie

National Research Institute of Oncology, Warsaw, Poland.

(3)Department of Oncology No. 1 (Thoracic Surgery), Leningrad Regional Clinical

Hospital, Saint Petersburg, Russia.

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.We report 5-year efficacy and safety

outcomes from the phase III KEYNOTE-407 study (ClinicalTrials.gov identifier:

NCT02775435). Eligible patients with previously untreated, metastatic squamous

non-small-cell lung cancer (NSCLC) were randomly assigned 1:1 to pembrolizumab

200 mg or placebo plus carboplatin and paclitaxel/nab-paclitaxel once every 3

weeks for four cycles, followed by pembrolizumab or placebo for up to 35 cycles.

Primary end points were overall survival (OS) and progression-free survival

(PFS) per RECIST version 1.1 by blinded independent central review (BICR). Five

hundred fifty-nine patients were randomly assigned in the intention-to-treat

population (pembrolizumab plus chemotherapy, n = 278; placebo plus chemotherapy,

n = 281). The median time from random assignment to data cutoff was 56.9 (range,

49.9-66.2) months. OS and PFS were improved with pembrolizumab plus chemotherapy

versus placebo plus chemotherapy (hazard ratio [95% CI], 0.71 [0.59 to 0.85] and

0.62 [0.52 to 0.74]), with 5-year OS rates of 18.4% versus 9.7%, respectively.

Toxicity was manageable. Among 55 patients who completed 35 cycles of

pembrolizumab, the objective response rate was 90.9% and the 3-year OS rate

after completion of 35 cycles (approximately 5 years after random assignment)

was 69.5%. Pembrolizumab plus chemotherapy maintained an OS and PFS benefit

versus placebo plus chemotherapy in previously untreated, metastatic squamous

NSCLC and is a standard-of-care first-line treatment option for metastatic

squamous NSCLC regardless of programmed death ligand 1 expression.

 

DOI: 10.1200/JCO.22.01990

PMCID: PMC10082300

PMID: 36735893 [Indexed for MEDLINE]

 

28. J Clin Oncol. 2023 Apr 1;41(10):1830-1840. doi: 10.1200/JCO.22.02186. Epub 2023 Jan 31.

 

Adjuvant Osimertinib for Resected EGFR-Mutated Stage IB-IIIA Non-Small-Cell Lung

Cancer: Updated Results From the Phase III Randomized ADAURA Trial.

 

Herbst RS(1), Wu YL(2), John T(3), Grohe C(4), Majem M(5), Wang J(6), Kato T(7),

Goldman JW(8), Laktionov K(9), Kim SW(10), Yu CJ(11)(12), Vu HV(13), Lu S(14),

Lee KY(15), Mukhametshina G(16), Akewanlop C(17), de Marinis F(18), Bonanno

L(19), Domine M(20), Shepherd FA(21), Urban D(22)(23), Huang X(24), Bolanos

A(25), Stachowiak M(26), Tsuboi M(27).

 

Author information:

(1)Medical Oncology, Yale School of Medicine and Yale Cancer Center, New Haven,

CT.

(2)Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and

Guangdong Academy of Medical Sciences, Guangzhou, China.

(3)Department of Medical Oncology, Austin Health, Melbourne, Australia.

 

Erratum in

    J Clin Oncol. 2023 Apr 27;:JCO2300658.

 

PURPOSE: The phase III ADAURA (ClinicalTrials.gov identifier: NCT02511106)

primary analysis demonstrated a clinically significant disease-free survival

(DFS) benefit with adjuvant osimertinib versus placebo in EGFR-mutated stage

IB-IIIA non-small-cell lung cancer (NSCLC) after complete tumor resection (DFS

hazard ratio [HR], 0.20 [99.12% CI, 0.14 to 0.30]; P < .001). We report an

updated exploratory analysis of final DFS data.

METHODS: Overall, 682 patients with stage IB-IIIA (American Joint Committee on

Cancer/Union for International Cancer Control, seventh edition) EGFR-mutated

(exon 19 deletion/L858R) NSCLC were randomly assigned 1:1 (stratified by stage,

mutational status, and race) to receive osimertinib 80 mg once-daily or placebo

for 3 years. The primary end point was DFS by investigator assessment in stage

II-IIIA disease analyzed by stratified log-rank test; following early reporting

of statistical significance in DFS, no further formal statistical testing was

planned. Secondary end points included DFS in stage IB-IIIA, overall survival,

and safety. Patterns of recurrence and CNS DFS were prespecified exploratory end

points.

RESULTS: At data cutoff (April 11, 2022), in stage II-IIIA disease, median

follow-up was 44.2 months (osimertinib) and 19.6 months (placebo); the DFS HR

was 0.23 (95% CI, 0.18 to 0.30); 4-year DFS rate was 70% (osimertinib) and 29%

(placebo). In the overall population, DFS HR was 0.27 (95% CI, 0.21 to 0.34);

4-year DFS rate was 73% (osimertinib) and 38% (placebo). Fewer patients treated

with osimertinib had local/regional and distant recurrence versus placebo. CNS

DFS HR in stage II-IIIA was 0.24 (95% CI, 0.14 to 0.42). The long-term safety

profile of osimertinib was consistent with the primary analysis.

CONCLUSION: These updated data demonstrate prolonged DFS benefit over placebo,

reduced risk of local and distant recurrence, improved CNS DFS, and a consistent

safety profile, supporting the efficacy of adjuvant osimertinib in resected

EGFR-mutated NSCLC.

 

DOI: 10.1200/JCO.22.02186

PMCID: PMC10082285

PMID: 36720083 [Indexed for MEDLINE]

 

29. Am J Respir Crit Care Med. 2023 Apr 1;207(7):929-935. doi:

10.1164/rccm.202208-1475OC.

 

Assessing Pretomanid for Tuberculosis (APT), a Randomized Phase 2 Trial of

Pretomanid-Containing Regimens for Drug-Sensitive Tuberculosis: 12-Week Results.

 

Dooley KE(1), Hendricks B(2), Gupte N(3), Barnes G(4), Narunsky K(2), Whitelaw

C(2), Smit T(2), Ignatius EH(4), Friedman A(2), Dorman SE(5), Dawson R(2);

Assessing Pretomanid for Tuberculosis (APT) Study Team.

 

Author information:

(1)Vanderbilt University Medical Center, Nashville, Tennessee.

(2)Division of Pulmonology, Department of Medicine and University of Cape Town

Lung Institute, University of Cape Town, Cape Town, South Africa.

(3)Johns Hopkins India Private Limited, Pune, India.

(4)School of Medicine, Johns Hopkins University, Baltimore, Maryland; and.

(5)Medical University of South Carolina, Charleston, South Carolina.

 

Comment in

    Am J Respir Crit Care Med. 2023 Apr 1;207(7):816-818.

 

Rationale: Pretomanid is a new nitroimidazole with proven treatment-shortening

efficacy in drug-resistant tuberculosis. Pretomanid-rifamycin-pyrazinamide

combinations are potent in mice but have not been tested clinically. Rifampicin,

but not rifabutin, reduces pretomanid exposures. Objectives: To evaluate the

safety and efficacy of regimens containing pretomanid-rifamycin-pyrazinamide

among participants with drug-sensitive pulmonary tuberculosis. Methods: A phase

2, 12-week, open-label randomized trial was conducted of isoniazid and

pyrazinamide plus 1) pretomanid and rifampicin (arm 1), 2) pretomanid and

rifabutin (arm 2), or 3) rifampicin and ethambutol (standard of care; arm 3).

Laboratory values of safety and sputum cultures were collected at Weeks 1, 2, 3,

4, 6, 8, 10, and 12. Time to culture conversion on liquid medium was the primary

outcome. Measurements and Main Results: Among 157 participants, 125 (80%) had

cavitary disease. Median time to liquid culture negativity in the modified

intention-to-treat population (n = 150) was 42 (arm 1), 28 (arm 2), and 56 (arm

3) days (P = 0.01) (adjusted hazard ratio for arm 1 vs. arm 3, 1.41 [95%

confidence interval (CI), 0.93-2.12; P = 0.10]; adjusted hazard ratio for arm 2

vs. arm 3, 1.89 [95% CI, 1.24-2.87; P = 0.003]). Eight-week liquid culture

conversion was 79%, 89%, and 69%, respectively. Grade ≥3 adverse events occurred

in 3 of 56 (5%), 5 of 53 (9%), and 2 of 56 (4%) participants. Six participants

were withdrawn because of elevated transaminase concentrations (five in arm 2,

one in arm 1). There were three serious adverse events (arm 2) and no deaths.

Conclusions: Pretomanid enhanced the microbiologic activity of regimens

containing a rifamycin and pyrazinamide. Efficacy and hepatic adverse events

appeared highest with the pretomanid and rifabutin-containing regimen. Whether

this is due to higher pretomanid concentrations merits exploration. Clinical

trial registered with www.clinicaltrials.gov (NCT02256696).

 

DOI: 10.1164/rccm.202208-1475OC

PMID: 36455068 [Indexed for MEDLINE]

 

30. J Clin Oncol. 2023 Apr 10;41(11):1986-1991. doi: 10.1200/JCO.21.02885. Epub 2022 Oct 28.

 

Five-Year Outcomes With Pembrolizumab Versus Chemotherapy as First-Line Therapy

in Patients With Non-Small-Cell Lung Cancer and Programmed Death Ligand-1 Tumor

Proportion Score ≥ 1% in the KEYNOTE-042 Study.

 

de Castro G Jr(1), Kudaba I(2), Wu YL(3), Lopes G(4), Kowalski DM(5), Turna

HZ(6), Caglevic C(7), Zhang L(8), Karaszewska B(9), Laktionov KK(10),

Srimuninnimit V(11), Bondarenko I(12), Kubota K(13), Mukherjee R(14), Lin J(14),

Souza F(14), Mok TSK(15), Cho BC(16).

 

Author information:

(1)Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil.

(2)Latvian Oncology Center, Riga East Clinical University, Riga, Latvia.

(3)Guangdong Lung Cancer Institute, Guangdong Provinicial People's Hospital and

Guangdong Academy of Medical Sciences, Guandong, China.

Clinical trials frequently include multiple end points that mature at different

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

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

available. Clinical Trial Updates provide an opportunity to disseminate

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

primary end point has already been reported.We report 5-year results from the

phase III KEYNOTE-042 study (ClinicalTrials.gov identifier: NCT02220894).

Eligible patients with locally advanced/metastatic non-small-cell lung cancer

(NSCLC) without EGFR/ALK alterations and with programmed death ligand-1 (PD-L1)

tumor proportion score (TPS) ≥ 1% received pembrolizumab 200 mg once every 3

weeks for 35 cycles or chemotherapy (carboplatin + paclitaxel or pemetrexed) for

4-6 cycles with optional maintenance pemetrexed. Primary end points were overall

survival (OS) in PD-L1 TPS ≥ 50%, ≥ 20%, and ≥ 1% groups. Patients who completed

35 cycles of pembrolizumab with ≥ stable disease could begin second-course

pembrolizumab upon progression. One thousand two hundred seventy-four patients

were randomly assigned (pembrolizumab, n = 637; chemotherapy, n = 637). Median

follow-up time was 61.1 (range, 50.0-76.3) months. OS outcomes favored

pembrolizumab (v chemotherapy) regardless of PD-L1 TPS (hazard ratio [95% CI]

for TPS ≥ 50%, 0.68 [0.57 to 0.81]; TPS ≥ 20%, 0.75 [0.64 to 0.87]; TPS ≥ 1%,

0.79 [0.70 to 0.89]), with estimated 5-year OS rates with pembrolizumab of

21.9%, 19.4%, and 16.6%, respectively. No new toxicities were identified.

Objective response rate was 84.3% among 102 patients who completed 35 cycles of

pembrolizumab and 15.2% among 33 patients who received second-course

pembrolizumab. First-line pembrolizumab monotherapy continued to show durable

clinical benefit versus chemotherapy after 5 years of follow-up in

PD-L1-positive, locally advanced/metastatic NSCLC without EGFR/ALK alterations

and remains a standard of care.

 

DOI: 10.1200/JCO.21.02885

PMCID: PMC10082298

PMID: 36306479 [Indexed for MEDLINE]

 

31. Cancer Cell. 2023 May 8;41(5):837-852.e6. doi: 10.1016/j.ccell.2023.03.019. Epub 2023 Apr 21.

 

Early immune pressure initiated by tissue-resident memory T cells sculpts tumor

evolution in non-small cell lung cancer.

 

Weeden CE(1), Gayevskiy V(1), Marceaux C(1), Batey D(2), Tan T(3), Yokote K(2),

Ribera NT(4), Clatch A(5), Christo S(5), Teh CE(6), Mitchell AJ(7), Trussart

M(8), Rankin L(6), Obers A(5), McDonald JA(9), Sutherland KD(9), Sharma VJ(10),

Starkey G(11), D'Costa R(12), Antippa P(13), Leong T(14), Steinfort D(15),

Irving L(15), Swanton C(16), Gordon CL(17), Mackay LK(5), Speed TP(18), Gray

DHD(19), Asselin-Labat ML(20).

 

Author information:

(1)Personalised Oncology Division, Walter and Eliza Hall Institute of Medical

Research, Parkville, VIC, Australia; Department of Medical Biology, the

University of Melbourne, Parkville, VIC, Australia.

(2)Personalised Oncology Division, Walter and Eliza Hall Institute of Medical

Research, Parkville, VIC, Australia.

(3)Immunology Division, Walter and Eliza Hall Institute of Medical Research,

Parkville, VIC, Australia.

Tissue-resident memory T (TRM) cells provide immune defense against local

infection and can inhibit cancer progression. However, it is unclear to what

extent chronic inflammation impacts TRM activation and whether TRM cells

existing in tissues before tumor onset influence cancer evolution in humans. We

performed deep profiling of healthy lungs and lung cancers in never-smokers

(NSs) and ever-smokers (ESs), finding evidence of enhanced immunosurveillance by

cells with a TRM-like phenotype in ES lungs. In preclinical models,

tumor-specific or bystander TRM-like cells present prior to tumor onset boosted

immune cell recruitment, causing tumor immune evasion through loss of MHC class

I protein expression and resistance to immune checkpoint inhibitors. In humans,

only tumors arising in ES patients underwent clonal immune evasion, unrelated to

tobacco-associated mutagenic signatures or oncogenic drivers. These data

demonstrate that enhanced TRM-like activity prior to tumor development shapes

the evolution of tumor immunogenicity and can impact immunotherapy outcomes.

 

Copyright © 2023 The Author(s). Published by Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2023.03.019

PMID: 37086716

 

32. Lancet Oncol. 2023 May;24(5):483-495. doi: 10.1016/S1470-2045(23)00108-0. Epub 2023 Apr 17.

 

Tislelizumab plus chemotherapy versus placebo plus chemotherapy as first-line

treatment for advanced or metastatic oesophageal squamous cell carcinoma

(RATIONALE-306): a global, randomised, placebo-controlled, phase 3 study.

 

Xu J(1), Kato K(2), Raymond E(3), Hubner RA(4), Shu Y(5), Pan Y(6), Park SR(7),

Ping L(8), Jiang Y(9), Zhang J(10), Wu X(11), Yao Y(12), Shen L(13), Kojima

T(14), Gotovkin E(15), Ishihara R(16), Wyrwicz L(17), Van Cutsem E(18),

Jimenez-Fonseca P(19), Lin CY(20), Wang L(21), Shi J(22), Li L(21), Yoon HH(23).

 

Author information:

(1)Fifth Medical Center, Chinese PLA General Hospital, Beijing, China.

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

(3)Centre Hospitalier Paris Saint-Joseph, Paris, France.

BACKGROUND: The options for first-line treatment of advanced oesophageal

squamous cell carcinoma are scarce, and the outcomes remain poor. The anti-PD-1

antibody, tislelizumab, has shown antitumour activity in previously treated

patients with advanced oesophageal squamous cell carcinoma. We report interim

analysis results from the RATIONALE-306 study, which aimed to assess

tislelizumab plus chemotherapy versus placebo plus chemotherapy as first-line

treatment for advanced or metastatic oesophageal squamous cell carcinoma.

METHODS: This global, randomised, double-blind, parallel-arm,

placebo-controlled, phase 3 study was conducted at 162 medical centres across

Asia, Europe, Oceania, and North America. Patients (aged ≥18 years) with

unresectable, locally advanced, recurrent or metastatic oesophageal squamous

cell carcinoma (regardless of PD-L1 expression), Eastern Cooperative Oncology

Group performance status of 0-1, and measurable or evaluable disease per

Response Evaluation Criteria in Solid Tumours (version 1.1) were recruited.

Patients were randomly assigned (1:1), using permuted block randomisation (block

size of four) and stratified by investigator-chosen chemotherapy, region, and

previous definitive therapy, to tislelizumab 200 mg or placebo intravenously

every 3 weeks on day 1, together with an investigator-chosen chemotherapy

doublet, comprising a platinum agent (cisplatin 60-80 mg/m2 intravenously on day

1 or oxaliplatin 130 mg/m2 intravenously on day 1) plus a fluoropyrimidine

(fluorouracil [750-800 mg/m2 intravenously on days 1-5] or capecitabine [1000

mg/m2 orally twice daily on days 1-14]) or paclitaxel (175 mg/m2 intravenously

on day 1). Treatment was continued until disease progression or unacceptable

toxicity. Investigators, patients, and sponsor staff or designees were masked to

treatment. The primary endpoint was overall survival. The efficacy analysis was

done in the intention-to-treat population (ie, all randomly assigned patients)

and safety was assessed in all patients who received at least one dose of study

treatment. The trial is registered with ClinicalTrials.gov, NCT03783442.

FINDINGS: Between Dec 12, 2018, and Nov 24, 2020, 869 patients were screened, of

whom 649 were randomly assigned to tislelizumab plus chemotherapy (n=326) or

placebo plus chemotherapy (n=323). Median age was 64·0 years (IQR 59·0-69·0),

563 (87%) of 649 participants were male, 86 (13%) were female, 486 (75%) were

Asian, and 155 (24%) were White. 324 (99%) of 326 patients in the tislelizumab

group and 321 (99%) of 323 in the placebo group received at least one dose of

the study drug. As of data cutoff (Feb 28, 2022), median follow-up was 16·3

months (IQR 8·6-21·8) in the tislelizumab group and 9·8 months (IQR 5·8-19·0)

 in the placebo group, and 196 (60%) of 326 patients in the tislelizumab group

versus 226 (70%) of 323 in the placebo group had died. Median overall survival

in the tislelizumab group was 17·2 months (95% CI 15·8-20·1) and in the placebo

group was 10·6 months (9·3-12·1; stratified hazard ratio 0·66 [95% CI

0·54-0·80]; one-sided p<0·0001). 313 (97%) of 324 patients in the tislelizumab

group and 309 (96%) of 321 in the placebo group had treatment-related

treatment-emergent adverse events. The most common grade 3 or 4

treatment-related treatment-emergent adverse events were decreased neutrophil

count (99 [31%] in the tislelizumab group vs 105 [33%] in the placebo group),

decreased white blood cell count (35 [11%] vs 50 [16%]), and anaemia (47 [15%]

vs 41 [13%]). Six deaths in the tislelizumab group (gastrointestinal and upper

gastrointestinal haemorrhage [n=2], myocarditis [n=1], pulmonary tuberculosis

[n=1], electrolyte imbalance [n=1], and respiratory failure [n=1]) and four

deaths in the placebo group (pneumonia [n=1], septic shock [n=1], and

unspecified death [n=2]) were determined to be treatment-related.

INTERPRETATION: Tislelizumab plus chemotherapy as a first-line treatment for

advanced or metastatic oesophageal squamous cell carcinoma provided superior

overall survival with a manageable safety profile versus placebo plus

chemotherapy. Given that the interim analysis met its superiority boundary for

the primary endpoint, as confirmed by the independent data monitoring committee,

this Article represents the primary study analysis.

FUNDING: BeiGene.

 

Copyright © 2023 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1470-2045(23)00108-0

PMID: 37080222 [Indexed for MEDLINE]

 

33. Nat Immunol. 2023 May;24(5):855-868. doi: 10.1038/s41590-023-01476-3. Epub 2023 Apr 3.

 

Antigen-specific B cells direct T follicular-like helper cells into lymphoid

follicles to mediate Mycobacterium tuberculosis control.

 

Swanson RV(#)(1), Gupta A(#)(1)(2), Foreman TW(3)(4), Lu L(1), Choreno-Parra

JA(5), Mbandi SK(6), Rosa BA(7)(8), Akter S(1)(2), Das S(1), Ahmed M(1)(2),

Garcia-Hernandez ML(9), Singh DK(10), Esaulova E(11), Artyomov MN(11), Gommerman

J(12), Mehra S(3)(10), Zuniga J(5)(13), Mitreva M(7)(8), Scriba TJ(6),

Rangel-Moreno J(9), Kaushal D(14), Khader SA(15)(16).

 

Author information:

(1)Department of Molecular Microbiology, Washington University in St. Louis, St.

Louis, MO, USA.

(2)Department of Microbiology, University of Chicago, Chicago, IL, USA.

(3)Divisions of Bacteriology and Parasitology, Tulane National Primate Research

Center, Covington, LA, USA.

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is a global cause

of death. Granuloma-associated lymphoid tissue (GrALT) correlates with

protection during TB, but the mechanisms of protection are not understood.

During TB, the transcription factor IRF4 in T cells but not B cells is required

for the generation of the TH1 and TH17 subsets of helper T cells and follicular

helper T (TFH)-like cellular responses. A population of IRF4+ T cells coexpress

the transcription factor BCL6 during Mtb infection, and deletion of Bcl6

(Bcl6fl/fl) in CD4+ T cells (CD4cre) resulted in reduction of TFH-like cells,

impaired localization within GrALT and increased Mtb burden. In contrast, the

absence of germinal center B cells, MHC class II expression on B cells,

antibody-producing plasma cells or interleukin-10-expressing B cells, did not

increase Mtb susceptibility. Indeed, antigen-specific B cells enhance cytokine

production and strategically localize TFH-like cells within GrALT via

interactions between programmed cell death 1 (PD-1) and its ligand PD-L1 and

mediate Mtb control in both mice and macaques.

 

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

 

DOI: 10.1038/s41590-023-01476-3

PMID: 37012543 [Indexed for MEDLINE]

 

34. Nat Med. 2023 Apr;29(4):1009-1016. doi: 10.1038/s41591-023-02247-1. Epub 2023

Mar 9.

 

Comparison of two diagnostic intervention packages for community-based active

case finding for tuberculosis: an open-label randomized controlled trial.

 

Esmail A(1), Randall P(1), Oelofse S(1), Tomasicchio M(1), Pooran A(1), Meldau

R(1), Makambwa E(1), Mottay L(1), Jaumdally S(1), Calligaro G(1), Meier S(1), de

Kock M(2), Gumbo T(3), Warren RM(2), Dheda K(4)(5)(6).

 

Author information:

(1)Centre for Lung Infection and Immunity, Division of Pulmonology, Department

of Medicine and UCT Lung Institute and South African MRC/UCT Centre for the

Study of Antimicrobial Resistance, University of Cape Town, Cape Town, South

Africa.

(2)DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research/SAMRC

Centre for Tuberculosis Research, Division of Molecular Biology and Human

Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University,

Stellenbosch, South Africa.

(3)Praedicare Laboratories, Dallas, TX, USA.

Two in every five patients with active tuberculosis (TB) remain undiagnosed or

unreported. Therefore community-based, active case-finding strategies require

urgent implementation. However, whether point-of-care (POC), portable

battery-operated, molecular diagnostic tools deployed at a community level,

compared with conventionally used POC smear microscopy, can shorten

time-to-treatment initiation, thus potentially curtailing transmission, remains

unclear. To clarify this issue, we performed an open-label, randomized

controlled trial in periurban informal settlements of Cape Town, South Africa,

where we TB symptom screened 5,274 individuals using a community-based scalable

mobile clinic. Some 584 individuals with HIV infection or symptoms of TB

underwent targeted diagnostic screening and were randomized (1:1) to same-day

smear microscopy (n = 296) or on-site DNA-based molecular diagnosis (n = 288;

GeneXpert). The primary aim was to compare time to TB treatment initiation

between the arms. Secondary aims included feasibility and detection of probably

infectious people. Of participants who underwent targeted screening, 9.9% (58 of

584) had culture-confirmed TB. Time-to-treatment initiation occurred

significantly earlier in the Xpert versus the smear-microscopy arm (8 versus

41 d, P = 0.002). However, overall, Xpert detected only 52% of individuals with

culture-positive TB. Notably, Xpert detected almost all of the probably

infectious patients compared with smear microscopy (94.1% versus 23.5%,

P = <0.001). Xpert was associated with a shorter median time to treatment of

probably infectious patients (7 versus 24 d, P = 0.02) and a greater proportion

of infectious patients were on treatment at 60 d compared with the probably

noninfectious patients (76.5% versus 38.2%, P < 0.01). Overall, a greater

proportion of POC Xpert-positive participants were on treatment at 60 d compared

with all culture-positive participants (100% versus 46.5%, P < 0.01). These

findings challenge the traditional paradigm of a passive case-finding, public

health strategy and argues for the implementation of portable DNA-based

diagnosis with linkage to care as a community-oriented,

transmission-interruption strategy. The study was registered with the South

African National Clinical Trials Registry (application ID 4367;

DOH-27-0317-5367) and ClinicalTrials.gov (NCT03168945).

 

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

 

DOI: 10.1038/s41591-023-02247-1

PMID: 36894651 [Indexed for MEDLINE]

 


上一篇: No.5

下一篇: No.3