您好!欢迎来到首都医科大学附属北京胸科医院官方网站!

高分SCI文摘

您所在的位置: 首页 >>胸科图书馆 >>信息推送 >>高分SCI文摘 >>2021年

高分SCI文摘

2021年

No. 5

发布时间:2021-07-26 浏览次数:
字号: + - 14

  Medical Abstracts

  Filters applied: from 2021/4/1 - 2021/4/30

  1. Science. 2021 Apr 30;372(6541):eabb8699. doi: 10.1126/science.abb8699.

  Stepwise pathogenic evolution of Mycobacterium abscessus.

  Bryant JM(1)(2), Brown KP(1)(3), Burbaud S(1), Everall I(1)(4), Belardinelli

  JM(5), Rodriguez-Rincon D(1), Grogono DM(1)(3), Peterson CM(5), Verma D(5),

  Evans IE(1)(3), Ruis C(1)(2), Weimann A(1)(2), Arora D(1), Malhotra S(6)(7),

  Bannerman B(1)(2), Passemar C(1), Templeton K(8), MacGregor G(8), Jiwa K(9),

  Fisher AJ(9), Blundell TL(6), Ordway DJ(5), Jackson M(5), Parkhill J(10)(11),

  Floto RA(12)(2)(3).

  Author information:

  (1)Molecular Immunity Unit, University of Cambridge Department of Medicine, MRC

  Laboratory of Molecular Biology, Cambridge, UK.

  (2)University of Cambridge Centre for AI in Medicine, Cambridge, UK.

  (3)Cambridge Centre for Lung Infection, Royal Papworth Hospital, Cambridge, UK.

  …

  Comment in

  Science. 2021 Apr 30;372(6541):465-466.

  Although almost all mycobacterial species are saprophytic environmental

  organisms, a few, such as Mycobacterium tuberculosis, have evolved to cause

  transmissible human infection. By analyzing the recent emergence and spread of

  the environmental organism M. abscessus through the global cystic fibrosis

  population, we have defined key, generalizable steps involved in the pathogenic

  evolution of mycobacteria. We show that epigenetic modifiers, acquired through

  horizontal gene transfer, cause saltational increases in the pathogenic

  potential of specific environmental clones. Allopatric parallel evolution during

  chronic lung infection then promotes rapid increases in virulence through

  mutations in a discrete gene network; these mutations enhance growth within

  macrophages but impair fomite survival. As a consequence, we observe constrained

  pathogenic evolution while person-to-person transmission remains indirect, but

  postulate accelerated pathogenic adaptation once direct transmission is

  possible, as observed for M. tuberculosis Our findings indicate how key

  interventions, such as early treatment and cross-infection control, might

  restrict the spread of existing mycobacterial pathogens and prevent new,

  emergent ones.

  Copyright © 2021 The Authors, some rights reserved; exclusive licensee American

  Association for the Advancement of Science. No claim to original U.S. Government

  Works.

  DOI: 10.1126/science.abb8699

  PMCID: PMC7611193

  PMID: 33926925 [Indexed for MEDLINE]

  2. Nature. 2021 Apr;592(7855):629-633. doi: 10.1038/s41586-021-03430-5. Epub 2021

  Apr 7.

  Evaluating eligibility criteria of oncology trials using real-world data and AI.

  Liu R(1), Rizzo S(2), Whipple S(2), Pal N(2), Pineda AL(2), Lu M(2), Arnieri

  B(2), Lu Y(3), Capra W(2), Copping R(4), Zou J(5)(6)(7)(8).

  Author information:

  (1)Department of Electrical Engineering, Stanford University, Stanford, CA, USA.

  (2)Genentech, South San Francisco, CA, USA.

  (3)Department of Biomedical Data Science, Stanford University, Stanford, CA,

  USA.

  (4)Genentech, South San Francisco, CA, USA. copping.ryan@gene.com.

  (5)Department of Electrical Engineering, Stanford University, Stanford, CA, USA.

  jamesz@stanford.edu.

  (6)Department of Biomedical Data Science, Stanford University, Stanford, CA,

  USA. jamesz@stanford.edu.

  (7)Department of Computer Science, Stanford University, Stanford, CA, USA.

  jamesz@stanford.edu.

  (8)Chan Zuckerberg Biohub, San Francisco, CA, USA. jamesz@stanford.edu.

  Comment in

  Nature. 2021 Apr;592(7855):512-513.

  There is a growing focus on making clinical trials more inclusive but the design

  of trial eligibility criteria remains challenging1-3. Here we systematically

  evaluate the effect of different eligibility criteria on cancer trial

  populations and outcomes with real-world data using the computational framework

  of Trial Pathfinder. We apply Trial Pathfinder to emulate completed trials of

  advanced non-small-cell lung cancer using data from a nationwide database of

  electronic health records comprising 61,094 patients with advanced

  non-small-cell lung cancer. Our analyses reveal that many common criteria,

  including exclusions based on several laboratory values, had a minimal effect on

  the trial hazard ratios. When we used a data-driven approach to broaden

  restrictive criteria, the pool of eligible patients more than doubled on average

  and the hazard ratio of the overall survival decreased by an average of 0.05.

  This suggests that many patients who were not eligible under the original trial

  criteria could potentially benefit from the treatments. We further support our

  findings through analyses of other types of cancer and patient-safety data from

  diverse clinical trials. Our data-driven methodology for evaluating eligibility

  criteria can facilitate the design of more-inclusive trials while maintaining

  safeguards for patient safety.

  DOI: 10.1038/s41586-021-03430-5

  PMID: 33828294

  3. Cell. 2021 Apr 1;184(7):1757-1774.e14. doi: 10.1016/j.cell.2021.02.046. Epub

  2021 Mar 23.

  A non-canonical type 2 immune response coordinates tuberculous granuloma

  formation and epithelialization.

  Cronan MR(1), Hughes EJ(2), Brewer WJ(3), Viswanathan G(3), Hunt EG(3), Singh

  B(4), Mehra S(5), Oehlers SH(6), Gregory SG(7), Kaushal D(4), Tobin DM(8).

  Author information:

  (1)Department of Molecular Genetics and Microbiology, Duke University School of

  Medicine, Durham, NC 27710, USA. Electronic address: cronan@mpiib-berlin.mpg.de.

  (2)Department of Molecular Genetics and Microbiology, Duke University School of

  Medicine, Durham, NC 27710, USA; University Program in Genetics and Genomics,

  Duke University School of Medicine, Durham, NC 27710, USA.

  (3)Department of Molecular Genetics and Microbiology, Duke University School of

  Medicine, Durham, NC 27710, USA.

  …

  The central pathogen-immune interface in tuberculosis is the granuloma, a

  complex host immune structure that dictates infection trajectory and physiology.

  Granuloma macrophages undergo a dramatic transition in which entire epithelial

  modules are induced and define granuloma architecture. In tuberculosis,

  relatively little is known about the host signals that trigger this transition.

  Using the zebrafish-Mycobacterium marinum model, we identify the basis of

  granuloma macrophage transformation. Single-cell RNA-sequencing analysis of

  zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected

  macaques reveal that, even in the presence of robust type 1 immune responses,

  countervailing type 2 signals associate with macrophage epithelialization. We

  find that type 2 immune signaling, mediated via stat6, is absolutely required

  for epithelialization and granuloma formation. In mixed chimeras, stat6 acts

  cell autonomously within macrophages, where it is required for epithelioid

  transformation and incorporation into necrotic granulomas. These findings

  establish the signaling pathway that produces the hallmark structure of

  mycobacterial infection.

  Copyright © 2021 Elsevier Inc. All rights reserved.

  DOI: 10.1016/j.cell.2021.02.046

  PMCID: PMC8055144

  PMID: 33761328

  4. Annu Rev Immunol. 2021 Apr 26;39:611-637. doi:

  10.1146/annurev-immunol-093019-010426. Epub 2021 Feb 26.

  The Innate Immune Response to Mycobacterium tuberculosis Infection.

  Ravesloot-Chávez MM(1), Van Dis E(2), Stanley SA(2)(3).

  Author information:

  (1)Department of Plant and Microbial Biology, University of California,

  Berkeley, California 94720, USA; email: mchavez@berkeley.edu.

  (2)Division of Immunology and Pathogenesis, Department of Molecular and Cell

  Biology, University of California, Berkeley, California 94720, USA; email:

  sastanley@berkeley.edu, vandise@berkeley.edu.

  (3)Division of Infectious Diseases and Vaccinology, School of Public Health,

  University of California, Berkeley, California 94720, USA.

  Infection with Mycobacterium tuberculosis causes >1.5 million deaths worldwide

  annually. Innate immune cells are the first to encounter M. tuberculosis, and

  their response dictates the course of infection. Dendritic cells (DCs) activate

  the adaptive response and determine its characteristics. Macrophages are

  responsible both for exerting cell-intrinsic antimicrobial control and for

  initiating and maintaining inflammation. The inflammatory response to M.

  tuberculosis infection is a double-edged sword. While cytokines such as TNF-α

  and IL-1 are important for protection, either excessive or insufficient cytokine

  production results in progressive disease. Furthermore, neutrophils-cells

  normally associated with control of bacterial infection-are emerging as key

  drivers of a hyperinflammatory response that results in host mortality. The

  roles of other innate cells, including natural killer cells and innate-like T

  cells, remain enigmatic. Understanding the nuances of both cell-intrinsic

  control of infection and regulation of inflammation will be crucial for the

  successful development of host-targeted therapeutics and vaccines.

  DOI: 10.1146/annurev-immunol-093019-010426

  PMID: 33637017

  5. Nat Rev Cancer. 2021 Apr;21(4):217-238. doi: 10.1038/s41568-020-00329-7. Epub

  2021 Feb 15.

  The matrix in cancer.

  Cox TR(1)(2).

  Author information:

  (1)The Kinghorn Cancer Centre, The Garvan Institute of Medical Research, Sydney,

  New South Wales, Australia. t.cox@garvan.org.au.

  (2)St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New

  South Wales, Australia. t.cox@garvan.org.au.

  The extracellular matrix is a fundamental, core component of all tissues and

  organs, and is essential for the existence of multicellular organisms. From the

  earliest stages of organism development until death, it regulates and fine-tunes

  every cellular process in the body. In cancer, the extracellular matrix is

  altered at the biochemical, biomechanical, architectural and topographical

  levels, and recent years have seen an exponential increase in the study and

  recognition of the importance of the matrix in solid tumours. Coupled with the

  advancement of new technologies to study various elements of the matrix and

  cell-matrix interactions, we are also beginning to see the deployment of

  matrix-centric, stromal targeting cancer therapies. This Review touches on many

  of the facets of matrix biology in solid cancers, including breast, pancreatic

  and lung cancer, with the aim of highlighting some of the emerging interactions

  of the matrix and influences that the matrix has on tumour onset, progression

  and metastatic dissemination, before summarizing the ongoing work in the field

  aimed at developing therapies to co-target the matrix in cancer and cancer

  metastasis.

  DOI: 10.1038/s41568-020-00329-7

  PMID: 33589810 [Indexed for MEDLINE]

  6. Acc Chem Res. 2021 May 18;54(10):2361-2376. doi: 10.1021/acs.accounts.0c00878.

  Epub 2021 Apr 22.

  Strategies to Combat Multi-Drug Resistance in Tuberculosis.

  Singh V(1)(2), Chibale K(1)(2).

  Author information:

  (1)Drug Discovery and Development Centre (H3D), University of Cape Town,

  Rondebosch 7701, South Africa.

  (2)South African Medical Research Council Drug Discovery and Development

  Research Unit, Department of Chemistry and Institute of Infectious Disease and

  Molecular Medicine, University of Cape Town, Rondebosch 7701, South Africa.

  "Drug resistance is an unavoidable consequence of the use of drugs; however, the

  emergence of multi-drug resistance can be managed by accurate diagnosis and

  tailor-made regimens."Antimicrobial resistance (AMR), is one of the most

  paramount health perils that has emerged in the 21st century. The global

  increase in drug-resistant strains of various bacterial pathogens prompted the

  World Health Organization (WHO) to develop a priority list of AMR pathogens.

  Mycobacterium tuberculosis (Mtb), an acid-fast bacillus that causes tuberculosis

  (TB), merits being one of the highest priority pathogens on this list since

  drug-resistant TB (DR-TB) accounts for ∼29% of deaths attributable to AMR. In

  recent years, funded collaborative efforts of researchers from academia,

  not-for-profit virtual R&D organizations and industry have resulted in the

  continuous growth of the TB drug discovery and development pipeline. This has so

  far led to the accelerated regulatory approval of bedaquiline and delamanid for

  the treatment of DR-TB. However, despite the availability of drug regimes, the

  current cure rate for multi-drug-resistant TB (MDR-TB) and extensively

  drug-resistant TB (XDR-TB) treatment regimens is 50% and 30%, respectively. It

  is to be noted that these regimens are administered over a long duration and

  have a serious side effect profile. Coupled with poor patient adherence, this

  has led to further acquisition of drug resistance and treatment failure. There

  is therefore an urgent need to develop new TB drugs with novel mechanism of

  actions (MoAs) and associated regimens.This Account recapitulates drug

  resistance in TB, existing challenges in addressing DR-TB, new drugs and

  regimens in development, and potential ways to treat DR-TB. We highlight our

  research aimed at identifying novel small molecule leads and associated targets

  against TB toward contributing to the global TB drug discovery and development

  pipeline. Our work mainly involves screening of various small molecule chemical

  libraries in phenotypic whole-cell based assays to identify hits for medicinal

  chemistry optimization, with attendant deconvolution of the MoA. We discuss the

  identification of small molecule chemotypes active against Mtb and subsequent

  structure-activity relationships (SAR) and MoA deconvolution studies. This is

  followed by a discussion on a chemical series identified by whole-cell

  cross-screening against Mtb, for which MoA deconvolution studies revealed a

  pathway that explained the lack of in vivo efficacy in a mouse model of TB and

  reiterated the importance of selecting an appropriate growth medium during

  phenotypic screening. We also discuss our efforts on drug repositioning toward

  addressing DR-TB. In the concluding section, we preview some promising future

  directions and the challenges inherent in advancing the drug pipeline to address

  DR-TB.

  DOI: 10.1021/acs.accounts.0c00878

  PMCID: PMC8154215

  PMID: 33886255

  7. Acc Chem Res. 2021 May 4;54(9):2065-2075. doi: 10.1021/acs.accounts.1c00048.

  Epub 2021 Apr 20.

  The Enzymes of the Rifamycin Antibiotic Resistome.

  Surette MD(1), Spanogiannopoulos P(1), Wright GD(1).

  Author information:

  (1)M.G. DeGroote Institute for Infectious Disease Research, David Braley Center

  for Antibiotic Discovery, Department of Biochemistry and Biomedical Sciences,

  McMaster University, Hamilton, Ontario L8S 3Z5, Canada.

  Rifamycin antibiotics include the WHO essential medicines rifampin, rifabutin,

  and rifapentine. These are semisynthetic derivatives of the natural product

  rifamycins, originally isolated from the soil bacterium Amycolatopsis

  rifamycinica. These antibiotics are primarily used to treat mycobacterial

  infections, including tuberculosis. Rifamycins act by binding to the β-subunit

  of bacterial RNA polymerase, inhibiting transcription, which results in cell

  death. These antibiotics consist of a naphthalene core spanned by a polyketide

  ansa bridge. This structure presents a unique 3D configuration that engages RNA

  polymerase through a series of hydrogen bonds between hydroxyl groups linked to

  the naphthalene core and C21 and C23 of the ansa bridge. This binding occurs not

  in the enzyme active site where template-directed RNA synthesis occurs but

  instead in the RNA exit tunnel, thereby blocking productive formation of

  full-length RNA. In their clinical use to treat tuberculosis, resistance to

  rifamycin antibiotics arises principally from point mutations in RNA polymerase

  that decrease the antibiotic's affinity for the binding site in the RNA exit

  tunnel. In contrast, the rifamycin resistome of environmental mycobacteria and

  actinomycetes is much richer and diverse. In these organisms, rifamycin

  resistance includes many different enzymatic mechanisms that modify and alter

  the antibiotic directly, thereby inactivating it. These enzymes include ADP

  ribosyltransferases, glycosyltransferases, phosphotransferases, and

  monooxygenases.ADP ribosyltransferases catalyze group transfer of ADP ribose

  from the cofactor NAD+, which is more commonly deployed for metabolic redox

  reactions. ADP ribose is transferred to the hydroxyl linked to C23 of the

  antibiotic, thereby sterically blocking productive interaction with RNA

  polymerase. Like ADP ribosyltransferases, rifamycin glycosyl transferases also

  modify the hydroxyl of position C23 of rifamycins, transferring a glucose moiety

  from the donor molecule UDP-glucose. Unlike other antibiotic resistance kinases

  that transfer the γ-phosphate of ATP to inactivate antibiotics such as

  aminoglycosides or macrolides, rifamycin phosphotransferases are ATP-dependent

  dikinases. These enzymes transfer the β-phosphate of ATP to the C21 hydroxyl of

  the rifamycin ansa bridge. The result is modification of a critical RNA

  polymerase binding group that blocks productive complex formation. On the other

  hand, rifamycin monooxygenases are FAD-dependent enzymes that hydroxylate the

  naphthoquinone core. The result of this modification is untethering of the ansa

  chain from the naphthyl moiety, disrupting the essential 3D shape necessary for

  productive RNA polymerase binding and inhibition that leads to cell death.All of

  these enzymes have homologues in bacterial metabolism that either are their

  direct precursors or share common ancestors to the resistance enzyme. The

  diversity of these resistance mechanisms, often redundant in individual

  bacterial isolates, speaks to the importance of protecting RNA polymerase from

  these compounds and validates this enzyme as a critical antibiotic target.

  DOI: 10.1021/acs.accounts.1c00048

  PMID: 33877820

  8. Cancer Cell. 2021 Apr 12;39(4):566-579.e7. doi: 10.1016/j.ccell.2021.02.014.

  Therapeutic targeting of ATR yields durable regressions in small cell lung

  cancers with high replication stress.

  Thomas A(1), Takahashi N(2), Rajapakse VN(2), Zhang X(3), Sun Y(2), Ceribelli

  M(3), Wilson KM(3), Zhang Y(2), Beck E(3), Sciuto L(2), Nichols S(2), Elenbaas

  B(4), Puc J(4), Dahmen H(5), Zimmermann A(5), Varonin J(6), Schultz CW(2), Kim

  S(2), Shimellis H(2), Desai P(2), Klumpp-Thomas C(3), Chen L(3), Travers J(3),

  McKnight C(3), Michael S(3), Itkin Z(3), Lee S(2), Yuno A(2), Lee MJ(2), Redon

  CE(2), Kindrick JD(7), Peer CJ(7), Wei JS(8), Aladjem MI(2), Figg WD(7),

  Steinberg SM(9), Trepel JB(2), Zenke FT(5), Pommier Y(2), Khan J(8), Thomas

  CJ(10).

  Author information:

  (1)Developmental Therapeutics Branch, Center for Cancer Research, National

  Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.

  Electronic address: anish.thomas@nih.gov.

  (2)Developmental Therapeutics Branch, Center for Cancer Research, National

  Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.

  (3)Division of Preclinical Innovation, National Center for Advancing

  Translational Sciences, National Institute of Health, Rockville, MD 20850, USA.

  …

  Small cell neuroendocrine cancers (SCNCs) are recalcitrant cancers arising from

  diverse primary sites that lack effective treatments. Using chemical genetic

  screens, we identified inhibition of ataxia telangiectasia and rad3 related

  (ATR), the primary activator of the replication stress response, and

  topoisomerase I (TOP1), nuclear enzyme that suppresses genomic instability, as

  synergistically cytotoxic in small cell lung cancer (SCLC). In a

  proof-of-concept study, we combined M6620 (berzosertib), first-in-class ATR

  inhibitor, and TOP1 inhibitor topotecan in patients with relapsed SCNCs.

  Objective response rate among patients with SCLC was 36% (9/25), achieving the

  primary efficacy endpoint. Durable tumor regressions were observed in patients

  with platinum-resistant SCNCs, typically fatal within weeks of recurrence. SCNCs

  with high neuroendocrine differentiation, characterized by enhanced replication

  stress, were more likely to respond. These findings highlight replication stress

  as a potentially transformative vulnerability of SCNCs, paving the way for

  rational patient selection in these cancers, now treated as a single disease.

  Published by Elsevier Inc.

  DOI: 10.1016/j.ccell.2021.02.014

  PMCID: PMC8048383

  PMID: 33848478

  9. Nat Rev Clin Oncol. 2021 Apr 28. doi: 10.1038/s41571-021-00501-4. Online ahead

  of print.

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

  lung cancer.

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

  T(6).

  Author information:

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

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

  USA. chaftj@mskcc.org.

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

  York, NY, USA.

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

  …

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

  Multidisciplinary discussions of surgical resectability and medical operability

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

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

  Trial evidence supports cisplatin-based adjuvant therapy either after surgical

  resection or concurrently with radiotherapy. Consensus guidelines support

  neoadjuvant chemotherapy in lieu of adjuvant chemotherapy and carboplatin-based

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

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

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

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

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

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

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

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

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

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

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

  and future state of clinical research to develop systemic therapies for

  non-metastatic lung cancer.

  DOI: 10.1038/s41571-021-00501-4

  PMID: 33911215

  10. PLoS Med. 2021 Apr 26;18(4):e1003566. doi: 10.1371/journal.pmed.1003566.

  eCollection 2021 Apr.

  Dynamics of sputum conversion during effective tuberculosis treatment: A

  systematic review and meta-analysis.

  Calderwood CJ(1), Wilson JP(1), Fielding KL(1), Harris RC(1), Karat AS(1),

  Mansukhani R(1), Falconer J(2), Bergstrom M(1), Johnson SM(1), McCreesh N(1),

  Monk EJM(1), Odayar J(3), Scott PJ(1), Stokes SA(1), Theodorou H(1), Moore

  DAJ(1).

  Author information:

  (1)TB Centre, London School of Hygiene & Tropical Medicine, London, United

  Kingdom.

  (2)Library & Archives Service, London School of Hygiene & Tropical Medicine,

  London, United Kingdom.

  (3)Division of Epidemiology and Biostatistics, School of Public Health & Family

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

  BACKGROUND: Two weeks' isolation is widely recommended for people commencing

  treatment for pulmonary tuberculosis (TB). The evidence that this corresponds to

  clearance of potentially infectious tuberculous mycobacteria in sputum is not

  well established. This World Health Organization-commissioned review

  investigated sputum sterilisation dynamics during TB treatment.

  METHODS AND FINDINGS: For the main analysis, 2 systematic literature searches of

  OvidSP MEDLINE, Embase, and Global Health, and EBSCO CINAHL Plus were conducted

  to identify studies with data on TB infectiousness (all studies to search date,

  1 December 2017) and all randomised controlled trials (RCTs) for

  drug-susceptible TB (from 1 January 1990 to search date, 20 February 2018).

  Included articles reported on patients receiving effective treatment for

  culture-confirmed drug-susceptible pulmonary TB. The outcome of interest was

  sputum bacteriological conversion: the proportion of patients having converted

  by a defined time point or a summary measure of time to conversion, assessed by

  smear or culture. Any study design with 10 or more particpants was considered.

  Record sifting and data extraction were performed in duplicate. Random effects

  meta-analyses were performed. A narrative summary additionally describes the

  results of a systematic search for data evaluating infectiousness from humans to

  experimental animals (PubMed, all studies to 27 March 2018). Other evidence on

  duration of infectiousness-including studies reporting on cough dynamics, human

  tuberculin skin test conversion, or early bactericidal activity of TB

  treatments-was outside the scope of this review. The literature search was

  repeated on 22 November 2020, at the request of the editors, to identify studies

  published after the previous censor date. Four small studies reporting 3

  different outcome measures were identified, which included no data that would

  alter the findings of the review; they are not included in the meta-analyses. Of

  5,290 identified records, 44 were included. Twenty-seven (61%) were RCTs and 17

  (39%) were cohort studies. Thirteen studies (30%) reported data from Africa, 12

  (27%) from Asia, 6 (14%) from South America, 5 (11%) from North America, and 4

  (9%) from Europe. Four studies reported data from multiple continents. Summary

  estimates suggested smear conversion in 9% of patients at 2 weeks (95% CI

  3%-24%, 1 single study [N = 1]), and 82% of patients at 2 months of treatment

  (95% CI 78%-86%, N = 10). Among baseline smear-positive patients, solid culture

  conversion occurred by 2 weeks in 5% (95% CI 0%-14%, N = 2), increasing to 88%

  at 2 months (95% CI 84%-92%, N = 20). At equivalent time points, liquid culture

  conversion was achieved in 3% (95% CI 1%-16%, N = 1) and 59% (95% CI 47%-70%, N

  = 8). Significant heterogeneity was observed. Further interrogation of the data

  to explain this heterogeneity was limited by the lack of disaggregation of

  results, including by factors such as HIV status, baseline smear status, and the

  presence or absence of lung cavitation.

  CONCLUSIONS: This systematic review found that most patients remained culture

  positive at 2 weeks of TB treatment, challenging the view that individuals are

  not infectious after this interval. Culture positivity is, however, only 1

  component of infectiousness, with reduced cough frequency and aerosol generation

  after TB treatment initiation likely to also be important. Studies that

  integrate our findings with data on cough dynamics could provide a more complete

  perspective on potential transmission of Mycobacterium tuberculosis by

  individuals on treatment.

  TRIAL REGISTRATION: Systematic review registration: PROSPERO 85226.

  DOI: 10.1371/journal.pmed.1003566

  PMCID: PMC8109831

  PMID: 33901173

  11. Lancet Infect Dis. 2021 Apr 20:S1473-3099(20)30728-3. doi:

  10.1016/S1473-3099(20)30728-3. Online ahead of print.

  Quantifying the rates of late reactivation tuberculosis: a systematic review.

  Dale KD(1), Karmakar M(2), Snow KJ(3), Menzies D(4), Trauer JM(5), Denholm

  JT(6).

  Author information:

  (1)Victorian Tuberculosis Program, Royal Melbourne Hospital, Peter Doherty

  Institute for Infection and Immunity, The University of Melbourne, Melbourne,

  VIC, Australia; Department of Microbiology and Immunology, Peter Doherty

  Institute for Infection and Immunity, The University of Melbourne, Melbourne,

  VIC, Australia. Electronic address: katie.dale@mh.org.au.

  (2)Department of Microbiology and Immunology, Peter Doherty Institute for

  Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia;

  Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.

  (3)Centre for International Child Health, Department of Paediatrics, Royal

  Children's Hospital, University of Melbourne, Parkville, VIC, Australia;

  Australia Department of Paediatrics, University of Melbourne, Parkville, VIC,

  Australia.

  …

  The risk of tuberculosis is greatest soon after infection, but Mycobacterium

  tuberculosis can remain in the body latently, and individuals can develop

  disease in the future, sometimes years later. However, there is uncertainty

  about how often reactivation of latent tuberculosis infection (LTBI) occurs. We

  searched eight databases (inception to June 25, 2019) to identify studies that

  quantified tuberculosis reactivation rates occurring more than 2 years after

  infection (late reactivation), with a focus on identifying untreated study

  cohorts with defined timing of LTBI acquisition (PROSPERO registered:

  CRD42017070594). We included 110 studies, divided into four methodological

  groups. Group 1 included studies that documented late reactivation rates from

  conversion (n=14) and group 2 documented late reactivation rates in LTBI cohorts

  from exposure (n=11). Group 3 included 86 studies in LTBI cohorts with an

  unknown exposure history, and group 4 included seven ecological studies. Since

  antibiotics have been used to treat tuberculosis, only 11 studies have

  documented late reactivation rates in infected, untreated cohorts from either

  conversion (group 1) or exposure (group 2); six of these studies lasted at least

  4 years and none lasted longer than 10 years. These studies found that

  tuberculosis rates declined over time, reaching approximately 200 cases per

  100 000 person-years or less by the fifth year, and possibly declining further

  after 5 years but interpretation was limited by decreasing or unspecified cohort

  sizes. In cohorts with latent tuberculosis and an unknown exposure history

  (group 3), tuberculosis rates were generally lower than those seen in groups 1

  and 2, and beyond 10 years after screening, rates had declined to less than 100

  per 100 000 person-years. Reinfection risks limit interpretation in all studies

  and the effect of age is unclear. Late reactivation rates are commonly estimated

  or modelled to prioritise tuberculosis control strategies towards tubuculosis

  elimination, but significant gaps remain in our understanding that must be

  acknowledged; the relative importance of late reactivation versus early

  progression to the global burden of tuberculosis remains unknown.

  Copyright © 2021 Elsevier Ltd. All rights reserved.

  DOI: 10.1016/S1473-3099(20)30728-3

  PMID: 33891908

  12. J Clin Oncol. 2021 Jul 20;39(21):2339-2349. doi: 10.1200/JCO.21.00174. Epub 2021

  Apr 19.

  Five-Year Outcomes With Pembrolizumab Versus Chemotherapy for Metastatic

  Non-Small-Cell Lung Cancer With PD-L1 Tumor Proportion Score ≥ 50.

  Reck M(1), Rodríguez-Abreu D(2), Robinson AG(3), Hui R(4), Csőszi T(5), Fülöp

  A(6), Gottfried M(7), Peled N(8), Tafreshi A(9), Cuffe S(10), O'Brien M(11), Rao

  S(12), Hotta K(13), Leal TA(14), Riess JW(15), Jensen E(16), Zhao B(16),

  Pietanza MC(16), Brahmer JR(17).

  Author information:

  (1)Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), member of the

  German Center for Lung Research (DZL), Grosshansdorf, Germany.

  (2)Complejo Hospitalario Universitario Insular Materno-Infantil de Gran Canaria,

  Universidad de Las Palmas de Gran Canaria, Las Palmas, Spain.

  (3)Cancer Centre of Southeastern Ontario at Kingston General Hospital, Kingston,

  ON, Canada.

  …

  Comment in

  2321.

  PURPOSE: We report the first 5-year follow-up of any first-line phase III

  immunotherapy trial for non-small-cell lung cancer (NSCLC). KEYNOTE-024

  (ClinicalTrials.gov identifier: NCT02142738) is an open-label, randomized

  controlled trial of pembrolizumab compared with platinum-based chemotherapy in

  patients with previously untreated NSCLC with a programmed death ligand-1

  (PD-L1) tumor proportion score of at least 50% and no sensitizing EGFR or ALK

  alterations. Previous analyses showed pembrolizumab significantly improved

  progression-free survival and overall survival (OS).

  METHODS: Eligible patients were randomly assigned (1:1) to pembrolizumab (200 mg

  once every 3 weeks for up to 35 cycles) or platinum-based chemotherapy. Patients

  in the chemotherapy group with progressive disease could cross over to

  pembrolizumab. The primary end point was progression-free survival; OS was a

  secondary end point.

  RESULTS: Three hundred five patients were randomly assigned: 154 to

  pembrolizumab and 151 to chemotherapy. Median (range) time from randomization to

  data cutoff (June 1, 2020) was 59.9 (55.1-68.4) months. Among patients initially

  assigned to chemotherapy, 99 received subsequent anti-PD-1 or PD-L1 therapy,

  representing a 66.0% effective crossover rate. Median OS was 26.3 months (95%

  CI, 18.3 to 40.4) for pembrolizumab and 13.4 months (9.4-18.3) for chemotherapy

  (hazard ratio, 0.62; 95% CI, 0.48 to 0.81). Kaplan-Meier estimates of the 5-year

  OS rate were 31.9% for the pembrolizumab group and 16.3% for the chemotherapy

  group. Thirty-nine patients received 35 cycles (ie, approximately 2 years) of

  pembrolizumab, 82.1% of whom were still alive at data cutoff (approximately 5

  years). Toxicity did not increase with longer treatment exposure.

  CONCLUSION: Pembrolizumab provides a durable, clinically meaningful long-term OS

  benefit versus chemotherapy as first-line therapy for metastatic NSCLC with

  PD-L1 tumor proportion score of at least 50%.

  DOI: 10.1200/JCO.21.00174

  PMCID: PMC8280089

  PMID: 33872070

  13. Nat Commun. 2021 Apr 15;12(1):2259. doi: 10.1038/s41467-021-22480-x.

  SOD1 regulates ribosome biogenesis in KRAS mutant non-small cell lung cancer.

  Wang X(#)(1)(2)(3), Zhang H(#)(1)(2), Sapio R(4), Yang J(5), Wong J(1), Zhang

  X(1)(2), Guo JY(1)(6), Pine S(1)(2), Van Remmen H(7), Li H(1)(8), White E(1)(9),

  Liu C(1)(10), Kiledjian M(1)(5), Pestov DG(11), Steven Zheng XF(12)(13)(14).

  Author information:

  (1)Rutgers Cancer Institute of New Jersey, Rutgers, The State University of New

  Jersey, New Brunswick, NJ, USA.

  (2)Department of Pharmacology, Robert Wood Johnson Medical School, Rutgers, The

  State University of New Jersey, Piscataway, NJ, USA.

  (3)Graduate Program in Cellular and Molecular Pharmacology, Rutgers, The State

  University of New Jersey, Piscataway, NJ, USA.

  SOD1 is known as the major cytoplasmic superoxide dismutase and an anticancer

  target. However, the role of SOD1 in cancer is not fully understood. Herein we

  describe the generation of an inducible Sod1 knockout in KRAS-driven NSCLC mouse

  model. Sod1 knockout markedly reduces tumor burden in vivo and blocks growth of

  KRAS mutant NSCLC cells in vitro. Intriguingly, SOD1 is enriched in the nucleus

  and notably in the nucleolus of NSCLC cells. The nuclear and nucleolar, not

  cytoplasmic, form of SOD1 is essential for lung cancer cell proliferation.

  Moreover, SOD1 interacts with PeBoW complex and controls its assembly necessary

  for pre-60S ribosomal subunit maturation. Mechanistically, SOD1 regulates

  co-localization of PeBoW with and processing of pre-rRNA, and maturation of

  cytoplasmic 60S ribosomal subunits in KRAS mutant lung cancer cells.

  Collectively, our study unravels a nuclear SOD1 function essential for ribosome

  biogenesis and proliferation in KRAS-driven lung cancer.

  DOI: 10.1038/s41467-021-22480-x

  PMCID: PMC8050259

  PMID: 33859191 [Indexed for MEDLINE]

  14. Nat Commun. 2021 Apr 12;12(1):2163. doi: 10.1038/s41467-021-22433-4.

  Epigenetic modulation of immune synaptic-cytoskeletal networks potentiates γδ T

  cell-mediated cytotoxicity in lung cancer.

  Weng RR(1), Lu HH(1), Lin CT(2)(3), Fan CC(4), Lin RS(2)(3), Huang TC(1), Lin

  SY(1), Huang YJ(4), Juan YH(1), Wu YC(4), Hung ZC(1), Liu C(5), Lin XH(2)(3),

  Hsieh WC(6)(7), Chiu TY(8), Liao JC(8), Chiu YL(9)(10)(11), Chen SY(6), Yu

  CJ(1)(12), Tsai HC(13)(14)(15).

  Author information:

  (1)Department of Internal Medicine, National Taiwan University Hospital, Taipei,

  Taiwan.

  (2)Tai Cheng Stem Cell Therapy Center, National Taiwan University, Taipei,

  Taiwan.

  (3)Pell Biomedical Technology Ltd, Taipei, Taiwan.

  …

  γδ T cells are a distinct subgroup of T cells that bridge the innate and

  adaptive immune system and can attack cancer cells in an MHC-unrestricted

  manner. Trials of adoptive γδ T cell transfer in solid tumors have had limited

  success. Here, we show that DNA methyltransferase inhibitors (DNMTis) upregulate

  surface molecules on cancer cells related to γδ T cell activation using

  quantitative surface proteomics. DNMTi treatment of human lung cancer

  potentiates tumor lysis by ex vivo-expanded Vδ1-enriched γδ T cells.

  Mechanistically, DNMTi enhances immune synapse formation and mediates

  cytoskeletal reorganization via coordinated alterations of DNA methylation and

  chromatin accessibility. Genetic depletion of adhesion molecules or

  pharmacological inhibition of actin polymerization abolishes the potentiating

  effect of DNMTi. Clinically, the DNMTi-associated cytoskeleton signature

  stratifies lung cancer patients prognostically. These results support a

  combinatorial strategy of DNMTis and γδ T cell-based immunotherapy in lung

  cancer management.

  DOI: 10.1038/s41467-021-22433-4

  PMCID: PMC8042060

  PMID: 33846331 [Indexed for MEDLINE]

  15. Nat Commun. 2021 Apr 6;12(1):2048. doi: 10.1038/s41467-021-22336-4.

  Ferroptosis response segregates small cell lung cancer (SCLC) neuroendocrine

  subtypes.

  Bebber CM(1)(2)(3), Thomas ES(1)(2)(4), Stroh J(1)(2), Chen Z(1)(2),

  Androulidaki A(1)(2), Schmitt A(2)(3), Höhne MN(2)(5), Stüker L(1)(2), de Pádua

  Alves C(1), Khonsari A(1)(6)(7), Dammert MA(1)(6)(7), Parmaksiz F(1)(6)(7),

  Tumbrink HL(1)(6)(7), Beleggia F(3), Sos ML(1)(6)(7), Riemer J(2)(5), George

  J(1), Brodesser S(2), Thomas RK(1)(6)(8), Reinhardt HC(9), von Karstedt

  S(10)(11)(12).

  Author information:

  (1)Department of Translational Genomics, Medical Faculty, University of Cologne,

  Cologne, Germany.

  (2)CECAD Cluster of Excellence, University of Cologne, Cologne, Germany.

  (3)Clinic I for Internal Medicine, Medical Faculty, University Hospital of

  Cologne, Cologne, Germany.

  Loss of TP53 and RB1 in treatment-naïve small cell lung cancer (SCLC) suggests

  selective pressure to inactivate cell death pathways prior to therapy. Yet,

  which of these pathways remain available in treatment-naïve SCLC is unknown.

  Here, through systemic analysis of cell death pathway availability in

  treatment-naïve SCLC, we identify non-neuroendocrine (NE) SCLC to be vulnerable

  to ferroptosis through subtype-specific lipidome remodeling. While NE SCLC is

  ferroptosis resistant, it acquires selective addiction to the TRX anti-oxidant

  pathway. In experimental settings of non-NE/NE intratumoral heterogeneity,

  non-NE or NE populations are selectively depleted by ferroptosis or TRX pathway

  inhibition, respectively. Preventing subtype plasticity observed under single

  pathway targeting, combined treatment kills established non-NE and NE tumors in

  xenografts, genetically engineered mouse models of SCLC and patient-derived

  cells, and identifies a patient subset with drastically improved overall

  survival. These findings reveal cell death pathway mining as a means to identify

  rational combination therapies for SCLC.

  DOI: 10.1038/s41467-021-22336-4

  PMCID: PMC8024350

  PMID: 33824345 [Indexed for MEDLINE]

  16. Am J Respir Crit Care Med. 2021 Apr 6. doi: 10.1164/rccm.202007-2791OC. Online

  ahead of print.

  Machine Learning for Early Lung Cancer Identification Using Routine Clinical and

  Laboratory Data.

  Gould MK(1), Huang BZ(2), Tammemagi MC(3), Kinar Y(4), Shiff R(4).

  Author information:

  (1)Kaiser Permanente Bernard J Tyson School of Medicine, 547934, Health Systems

  Science, Pasadena, California, United States; michael.k.gould@kp.org.

  (2)Kaiser Permanente, Research and Evaluation, Pasadena, California, United

  States.

  (3)Brock University, 7497, Medical Sciences, Saint Catharines, Ontario, Canada.

  (4)Medial EarlySign, Newton, Massachusetts, United States.

  RATIONALE: Most lung cancers are diagnosed at an advanced stage. Pre-symptomatic

  identification of high-risk individuals can prompt earlier intervention and

  improve long-term outcomes.

  OBJECTIVE: To develop a model to predict a future diagnosis of lung cancer based

  on routine clinical and laboratory data, using machine-learning.

  METHODS: We assembled 6,505 non-small cell lung cancer (NSCLC) cases and 189,597

  contemporaneous controls and compared the accuracy of a novel machine-learning

  model to a modified version of the well-validated PLCOm2012 risk model, using

  the area under the receiver operating characteristic curve (AUC), sensitivity

  and diagnostic odds ratio (OR) as measures of model performance.

  RESULTS: Among ever-smokers in the test set, the a machine-learning model was

  more accurate than the modified PLCOm2012 for identifying NSCLC 9-12 months

  before clinical diagnosis (P<0.00001), with an AUC of 0.86, a diagnostic OR of

  12.8 3 and a sensitivity of 40.31% at a pre-defined specificity of 95%. In

  comparison, the modified PLCOm2012 had an AUC of 0.79, an OR of 7.4 and a

  sensitivity of 27.9% at the same specificity. The machine-learning model was

  more accurate than standard eligibility criteria for lung cancer screening and

  more accurate than the modified PLCOm2012 model when applied to a

  screening-eligible population. Influential model variables included known risk

  factors and novel predictors such as white blood cell and platelet counts.

  CONCLUSIONS: A machine-learning model was more accurate for early diagnosis of

  NSCLC than either standard eligibility criteria for screening or the modified

  PLCOm2012, demonstrating the potential to help prevent lung cancer deaths

  through early detection.

  DOI: 10.1164/rccm.202007-2791OC

  PMID: 33823116

  17. Cell Host Microbe. 2021 Apr 14;29(4):594-606.e6. doi:

  10.1016/j.chom.2021.02.005. Epub 2021 Mar 11.

  TGFβ restricts expansion, survival, and function of T cells within the

  tuberculous granuloma.

  Gern BH(1), Adams KN(2), Plumlee CR(2), Stoltzfus CR(3), Shehata L(3), Moguche

  AO(3), Busman-Sahay K(4), Hansen SG(4), Axthelm MK(4), Picker LJ(4), Estes

  JD(4), Urdahl KB(5), Gerner MY(6).

  Author information:

  (1)Center for Global Infectious Disease Research, Seattle Children's Research

  Institute, Seattle, WA 98109, USA; Department of Pediatrics, University of

  Washington, Seattle, WA 98195, USA.

  (2)Center for Global Infectious Disease Research, Seattle Children's Research

  Institute, Seattle, WA 98109, USA.

  (3)Department of Immunology, University of Washington, Seattle, WA 98109, USA.

  …

  CD4 T cell effector function is required for optimal containment of

  Mycobacterium tuberculosis (Mtb) infection. IFNɣ produced by CD4 T cells is a

  key cytokine that contributes to protection. However, lung-infiltrating CD4

  T cells have a limited ability to produce IFNɣ, and IFNɣ plays a lesser

  protective role within the lung than at sites of Mtb dissemination. In a murine

  infection model, we observed that IFNɣ production by Mtb-specific CD4 T cells is

  rapidly extinguished within the granuloma but not within unaffected lung

  regions, suggesting localized immunosuppression. We identified a signature of

  TGFβ signaling within granuloma-infiltrating T cells in both mice and rhesus

  macaques. Selective blockade of TGFβ signaling in T cells resulted in an

  accumulation of terminally differentiated effector CD4 T cells, improved IFNɣ

  production within granulomas, and reduced bacterial burdens. These findings

  uncover a spatially localized immunosuppressive mechanism associated with Mtb

  infection and provide potential targets for host-directed therapy.

  Copyright © 2021. Published by Elsevier Inc.

  DOI: 10.1016/j.chom.2021.02.005

  PMID: 33711270

  18. J Clin Oncol. 2021 Apr 20;39(12):1349-1359. doi: 10.1200/JCO.20.02212. Epub 2021

  Mar 8.

  Nivolumab and Ipilimumab as Maintenance Therapy in Extensive-Disease Small-Cell

  Lung Cancer: CheckMate 451.

  Owonikoko TK(1), Park K(2), Govindan R(3), Ready N(4), Reck M(5), Peters S(6),

  Dakhil SR(7), Navarro A(8), Rodríguez-Cid J(9), Schenker M(10), Lee JS(11),

  Gutierrez V(12), Percent I(13), Morgensztern D(3), Barrios CH(14), Greillier

  L(15), Baka S(16), Patel M(17), Lin WH(18), Selvaggi G(18), Baudelet C(18),

  Baden J(18), Pandya D(18), Doshi P(18), Kim HR(19).

  Author information:

  (1)Winship Cancer Institute of Emory University, Atlanta, GA.

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

  South Korea.

  (3)Alvin J Siteman Cancer Center at Washington University School of Medicine, St

  Louis, MO.

  …

  PURPOSE: In extensive-disease small-cell lung cancer (ED-SCLC), response rates

  to first-line platinum-based chemotherapy are robust, but responses lack

  durability. CheckMate 451, a double-blind phase III trial, evaluated nivolumab

  plus ipilimumab and nivolumab monotherapy as maintenance therapy following

  first-line chemotherapy for ED-SCLC.

  METHODS: Patients with ED-SCLC, Eastern Cooperative Oncology Group performance

  status 0-1, and no progression after ≤ 4 cycles of first-line chemotherapy were

  randomly assigned (1:1:1) to nivolumab 1 mg/kg plus ipilimumab 3 mg/kg once

  every 3 weeks for 12 weeks followed by nivolumab 240 mg once every 2 weeks,

  nivolumab 240 mg once every 2 weeks, or placebo for ≤ 2 years or until

  progression or unacceptable toxicity. Primary end point was overall survival

  (OS) with nivolumab plus ipilimumab versus placebo. Secondary end points were

  hierarchically tested.

  RESULTS: Overall, 834 patients were randomly assigned. The minimum follow-up was

  8.9 months. OS was not significantly prolonged with nivolumab plus ipilimumab

  versus placebo (hazard ratio [HR], 0.92; 95% CI, 0.75 to 1.12; P = .37; median,

  9.2 v 9.6 months). The HR for OS with nivolumab versus placebo was 0.84 (95% CI,

  0.69 to 1.02); the median OS for nivolumab was 10.4 months. Progression-free

  survival HRs versus placebo were 0.72 for nivolumab plus ipilimumab (95% CI,

  0.60 to 0.87) and 0.67 for nivolumab (95% CI, 0.56 to 0.81). A trend toward OS

  benefit with nivolumab plus ipilimumab was observed in patients with tumor

  mutational burden ≥ 13 mutations per megabase. Rates of grade 3-4

  treatment-related adverse events were nivolumab plus ipilimumab (52.2%),

  nivolumab (11.5%), and placebo (8.4%).

  CONCLUSION: Maintenance therapy with nivolumab plus ipilimumab did not prolong

  OS for patients with ED-SCLC who did not progress on first-line chemotherapy.

  There were no new safety signals.

  DOI: 10.1200/JCO.20.02212

  PMCID: PMC8078251

  PMID: 33683919

  19. J Clin Oncol. 2021 Apr 10;39(11):1253-1263. doi: 10.1200/JCO.20.03025. Epub 2021

  Mar 1.

  Updated Integrated Analysis of the Efficacy and Safety of Entrectinib in Locally

  Advanced or Metastatic ROS1 Fusion-Positive Non-Small-Cell Lung Cancer.

  Dziadziuszko R(1), Krebs MG(2), De Braud F(3)(4), Siena S(3)(5), Drilon A(6),

  Doebele RC(7), Patel MR(8), Cho BC(9), Liu SV(10), Ahn MJ(11), Chiu CH(12),

  Farago AF(13), Lin CC(14), Karapetis CS(15), Li YC(16), Day BM(17), Chen D(17),

  Wilson TR(17), Barlesi F(18)(19).

  Author information:

  (1)Medical University of Gdańsk, Gdańsk, Poland.

  (2)Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The

  University of Manchester and The Christie NHS Foundation Trust, Manchester

  Academic Health Science Centre, Manchester, United Kingdom.

  (3)Department of Oncology and Hematology-Oncology, Università degli Studi di

  Milano, Milan, Italy.

  …

  PURPOSE: Genetic rearrangements of the tyrosine receptor kinase ROS

  proto-oncogene 1 (ROS1) are oncogenic drivers in non-small-cell lung cancer

  (NSCLC). We report the results of an updated integrated analysis of three phase

  I or II clinical trials (ALKA-372-001, STARTRK-1, and STARTRK-2) of the ROS1

  tyrosine kinase inhibitor, entrectinib, in ROS1 fusion-positive NSCLC.

  METHODS: The efficacy-evaluable population included adults with locally advanced

  or metastatic ROS1 fusion-positive NSCLC with or without CNS metastases who

  received entrectinib ≥ 600 mg orally once per day. Co-primary end points were

  objective response rate (ORR) assessed by blinded independent central review and

  duration of response (DoR). Secondary end points included progression-free

  survival (PFS), overall survival (OS), intracranial ORR, intracranial DoR,

  intracranial PFS, and safety.

  RESULTS: In total, 161 patients with a follow-up of ≥ 6 months were evaluable.

  The median treatment duration was 10.7 months (IQR, 6.4-17.7). The ORR was 67.1%

  (n = 108, 95% CI, 59.3 to 74.3), and responses were durable (12-month DoR rate,

  63%, median DoR 15.7 months). The 12-month PFS rate was 55% (median PFS 15.7

  months), and the 12-month OS rate was 81% (median OS not estimable). In 24

  patients with measurable baseline CNS metastases by blinded independent central

  review, the intracranial ORR was 79.2% (n = 19; 95% CI, 57.9 to 92.9), the

  median intracranial PFS was 12.0 months (95% CI, 6.2 to 19.3), and the median

  intracranial DoR was 12.9 months (12-month rate, 55%). The safety profile in

  this updated analysis was similar to that reported in the primary analysis, and

  no new safety signals were found.

  CONCLUSION: Entrectinib continued to demonstrate a high level of clinical

  benefit for patients with ROS1 fusion-positive NSCLC, including patients with

  CNS metastases.

  DOI: 10.1200/JCO.20.03025

  PMCID: PMC8078299

  PMID: 33646820

  20. J Clin Oncol. 2021 Apr 20;39(12):1389-1411. doi: 10.1200/JCO.20.03465. Epub 2021

  Feb 22.

  Management of Dyspnea in Advanced Cancer: ASCO Guideline.

  Hui D(1), Bohlke K(2), Bao T(3), Campbell TC(4), Coyne PJ(5), Currow DC(6),

  Gupta A(7), Leiser AL(8), Mori M(9), Nava S(10), Reinke LF(11), Roeland EJ(12),

  Seigel C(13), Walsh D(14), Campbell ML(15).

  Author information:

  (1)MD Anderson Cancer Center, Houston, TX.

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

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

  …

  PURPOSE: To provide guidance on the clinical management of dyspnea in adult

  patients with advanced cancer.

  METHODS: ASCO convened an Expert Panel to review the evidence and formulate

  recommendations. An Agency for Healthcare Research and Quality (AHRQ) systematic

  review provided the evidence base for nonpharmacologic and pharmacologic

  interventions to alleviate dyspnea. The review included randomized controlled

  trials (RCTs) and observational studies with a concurrent comparison group

  published through early May 2020. The ASCO Expert Panel also wished to address

  dyspnea assessment, management of underlying conditions, and palliative care

  referrals, and for these questions, an additional systematic review identified

  RCTs, systematic reviews, and guidelines published through July 2020.

  RESULTS: The AHRQ systematic review included 48 RCTs and two retrospective

  cohort studies. Lung cancer and mesothelioma were the most commonly addressed

  types of cancer. Nonpharmacologic interventions such as fans provided some

  relief from breathlessness. Support for pharmacologic interventions was limited.

  A meta-analysis of specialty breathlessness services reported improvements in

  distress because of dyspnea.

  RECOMMENDATIONS: A hierarchical approach to dyspnea management is recommended,

  beginning with dyspnea assessment, ascertainment and management of potentially

  reversible causes, and referral to an interdisciplinary palliative care team.

  Nonpharmacologic interventions that may be offered to relieve dyspnea include

  airflow interventions (eg, a fan directed at the cheek), standard supplemental

  oxygen for patients with hypoxemia, and other psychoeducational,

  self-management, or complementary approaches. For patients who derive inadequate

  relief from nonpharmacologic interventions, systemic opioids should be offered.

  Other pharmacologic interventions, such as corticosteroids and benzodiazepines,

  are also discussed.Additional information is available at

  www.asco.org/supportive-care-guidelines.

  DOI: 10.1200/JCO.20.03465

  PMID: 33617290

上一篇: No. 6

下一篇: No.4