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 >>2018年

高分SCI文摘

2018年

No.22

来源:tushuguan 发布时间:2019-01-14 浏览次数:
字号: + - 14

Medical Abstracts

Keyword: lung cancer

1. N Engl J Med. 2018 Nov 22;379(21):2027-2039. doi: 10.1056/NEJMoa1810171. Epub

2018 Sep 25.

Brigatinib versus Crizotinib in ALK-Positive Non-Small-Cell Lung Cancer.

Camidge DR(1), Kim HR(1), Ahn MJ(1), Yang JC(1), Han JY(1), Lee JS(1), Hochmair

MJ(1), Li JY(1), Chang GC(1), Lee KH(1), Gridelli C(1), Delmonte A(1), Garcia

Campelo R(1), Kim DW(1), Bearz A(1), Griesinger F(1), Morabito A(1), Felip E(1),

Califano R(1), Ghosh S(1), Spira A(1), Gettinger SN(1), Tiseo M(1), Gupta N(1),

Haney J(1), Kerstein D(1), Popat S(1).

Author information:

(1)From the University of Colorado Cancer Center, Aurora (D.R.C.); Division of

Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei

University College of Medicine (H.R.K.), Samsung Medical Center (M.-J.A.), and

Seoul National University Hospital (D.-W.K.), Seoul, National Cancer Center,

Goyang (J.-Y.H.), Seoul National University Bundang Hospital, Seongnam (J.-S.L.),

and Chungbuk National University Hospital, Chungbuk National University College

of Medicine, Cheongju (K.H.L.) - all in South Korea; National Taiwan University

Hospital (J.C.-H.Y.) and the Faculty of Medicine, School of Medicine, National

Yang-Ming University (G.-C.C.), …

BACKGROUND: Brigatinib, a next-generation anaplastic lymphoma kinase (ALK)

inhibitor, has robust efficacy in patients with ALK-positive non-small-cell lung

cancer (NSCLC) that is refractory to crizotinib. The efficacy of brigatinib, as

compared with crizotinib, in patients with advanced ALK-positive NSCLC who have

not previously received an ALK inhibitor is unclear.

METHODS: In an open-label, phase 3 trial, we randomly assigned, in a 1:1 ratio,

patients with advanced ALK-positive NSCLC who had not previously received ALK

inhibitors to receive brigatinib at a dose of 180 mg once daily (with a 7-day

lead-in period at 90 mg) or crizotinib at a dose of 250 mg twice daily. The

primary end point was progression-free survival as assessed by blinded

independent central review. Secondary end points included the objective response

rate and intracranial response. The first interim analysis was planned when

approximately 50% of 198 expected events of disease progression or death had

occurred.

RESULTS: A total of 275 patients underwent randomization; 137 were assigned to

brigatinib and 138 to crizotinib. At the first interim analysis (99 events), the

median follow-up was 11.0 months in the brigatinib group and 9.3 months in the

crizotinib group. The rate of progression-free survival was higher with

brigatinib than with crizotinib (estimated 12-month progression-free survival,

67% [95% confidence interval {CI}, 56 to 75] vs. 43% [95% CI, 32 to 53]; hazard

ratio for disease progression or death, 0.49 [95% CI, 0.33 to 0.74]; P<0.001 by

the log-rank test). The confirmed objective response rate was 71% (95% CI, 62 to

78) with brigatinib and 60% (95% CI, 51 to 68) with crizotinib; the confirmed

rate of intracranial response among patients with measurable lesions was 78% (95%

CI, 52 to 94) and 29% (95% CI, 11 to 52), respectively. No new safety concerns

were noted.

CONCLUSIONS: Among patients with ALK-positive NSCLC who had not previously

received an ALK inhibitor, progression-free survival was significantly longer

among patients who received brigatinib than among those who received crizotinib.

(Funded by Ariad Pharmaceuticals; ALTA-1L ClinicalTrials.gov number, NCT02737501

.).

DOI: 10.1056/NEJMoa1810171

PMID: 30280657 [Indexed for MEDLINE]

2. N Engl J Med. 2018 Nov 22;379(21):2040-2051. doi: 10.1056/NEJMoa1810865. Epub

2018 Sep 25.

Pembrolizumab plus Chemotherapy for Squamous Non-Small-Cell Lung Cancer.

Paz-Ares L(1), Luft A(1), Vicente D(1), Tafreshi A(1), Gümü? M(1), Mazières J(1),

Hermes B(1), Çay ?enler F(1), Cs?szi T(1), Fülöp A(1), Rodríguez-Cid J(1), Wilson

J(1), Sugawara S(1), Kato T(1), Lee KH(1), Cheng Y(1), Novello S(1), Halmos B(1),

Li X(1), Lubiniecki GM(1), Piperdi B(1), Kowalski DM(1); KEYNOTE-407

Investigators.

Author information:

(1)From Hospital Universitario 12 de Octubre, Spanish National Cancer Research

Center, Universidad Complutense and Ciberonc, Madrid (L.P.-A.), and Hospital

Universitario Virgen Macarena, Seville (D.V.) - both in Spain; Leningrad Regional

Clinical Hospital, St. Petersburg, Russia (A.L.); Wollongong Oncology and

Wollongong Private Hospital, Wollongong, NSW, Australia (A.T.); Istanbul

Medeniyet University Hospital, Istanbul (M.G.), and Ankara University, Ankara

(F.Ç.?.) - both in Turkey; Centre Hospitalier Universitaire de Toulouse,

Université Paul Sabatier, Toulouse, France (J.M.); …

BACKGROUND: Standard first-line therapy for metastatic, squamous non-small-cell

lung cancer (NSCLC) is platinum-based chemotherapy or pembrolizumab (for patients

with programmed death ligand 1 [PD-L1] expression on ≥50% of tumor cells). More

recently, pembrolizumab plus chemotherapy was shown to significantly prolong

overall survival among patients with nonsquamous NSCLC.

METHODS: In this double-blind, phase 3 trial, we randomly assigned, in a 1:1

ratio, 559 patients with untreated metastatic, squamous NSCLC to receive 200 mg

of pembrolizumab or saline placebo for up to 35 cycles; all the patients also

received carboplatin and either paclitaxel or nanoparticle albumin-bound

[nab]-paclitaxel for the first 4 cycles. Primary end points were overall survival

and progression-free survival.

RESULTS: After a median follow-up of 7.8 months, the median overall survival was

15.9 months (95% confidence interval [CI], 13.2 to not reached) in the

pembrolizumab-combination group and 11.3 months (95% CI, 9.5 to 14.8) in the

placebo-combination group (hazard ratio for death, 0.64; 95% CI, 0.49 to 0.85;

P<0.001). The overall survival benefit was consistent regardless of the level of

PD-L1 expression. The median progression-free survival was 6.4 months (95% CI,

6.2 to 8.3) in the pembrolizumab-combination group and 4.8 months (95% CI, 4.3 to

5.7) in the placebo-combination group (hazard ratio for disease progression or

death, 0.56; 95% CI, 0.45 to 0.70; P<0.001). Adverse events of grade 3 or higher

occurred in 69.8% of the patients in the pembrolizumab-combination group and in

68.2% of the patients in the placebo-combination group. Discontinuation of

treatment because of adverse events was more frequent in the

pembrolizumab-combination group than in the placebo-combination group (13.3% vs.

6.4%).

CONCLUSIONS: In patients with previously untreated metastatic, squamous NSCLC,

the addition of pembrolizumab to chemotherapy with carboplatin plus paclitaxel or

nab-paclitaxel resulted in significantly longer overall survival and

progression-free survival than chemotherapy alone. (Funded by Merck Sharp &

Dohme; KEYNOTE-407 ClinicalTrials.gov number, NCT02775435 .).

DOI: 10.1056/NEJMoa1810865

PMID: 30280635 [Indexed for MEDLINE]

3. Nat Med. 2019 Jan;25(1):111-118. doi: 10.1038/s41591-018-0264-7. Epub 2018 Nov

26.

Aurora kinase A drives the evolution of resistance to third-generation EGFR

inhibitors in lung cancer.

Shah KN(1)(2), Bhatt R(1)(2), Rotow J(2)(3), Rohrberg J(2)(3), Olivas V(2)(3),

Wang VE(2), Hemmati G(2)(3), Martins MM(2)(3), Maynard A(2)(3), Kuhn J(4), Galeas

J(2), Donnella HJ(1)(2), Kaushik S(1)(2), Ku A(1)(2), Dumont S(4), Krings G(5),

Haringsma HJ(6), Robillard L(6), Simmons AD(6), Harding TC(6), McCormick F(2),

Goga A(2)(4), Blakely CM(2)(3), Bivona TG(2)(3), Bandyopadhyay S(7)(8).

Author information:

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

California, San Francisco, San Francisco, CA, USA.

(2)Helen Diller Family Comprehensive Cancer Center, University of California, San

Francisco, San Francisco, CA, USA.

(3)Department of Medicine, University of California, San Francisco, San

Francisco, CA, USA.

(4)Department of Cell and Tissue Biology, University of California, San

Francisco, San Francisco, CA, USA.

Although targeted therapies often elicit profound initial patient responses,

these effects are transient due to residual disease leading to acquired

resistance. How tumors transition between drug responsiveness, tolerance and

resistance, especially in the absence of preexisting subclones, remains unclear.

In epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma cells, we

demonstrate that residual disease and acquired resistance in response to EGFR

inhibitors requires Aurora kinase A (AURKA) activity. Nongenetic resistance

through the activation of AURKA by its coactivator TPX2 emerges in response to

chronic EGFR inhibition where it mitigates drug-induced apoptosis. Aurora kinase

inhibitors suppress this adaptive survival program, increasing the magnitude and

duration of EGFR inhibitor response in preclinical models. Treatment-induced

activation of AURKA is associated with resistance to EGFR inhibitors in vitro, in

vivo and in most individuals with EGFR-mutant lung adenocarcinoma. These findings

delineate a molecular path whereby drug resistance emerges from drug-tolerant

cells and unveils a synthetic lethal strategy for enhancing responses to EGFR

inhibitors by suppressing AURKA-driven residual disease and acquired resistance.

DOI: 10.1038/s41591-018-0264-7

PMCID: PMC6324945 [Available on 2019-05-26]

PMID: 30478424

4. Lancet Oncol. 2018 Dec;19(12):1654-1667. doi: 10.1016/S1470-2045(18)30649-1. Epub 2018 Nov 6.

Lorlatinib in patients with ALK-positive non-small-cell lung cancer: results from

a global phase 2 study.

Solomon BJ(1), Besse B(2), Bauer TM(3), Felip E(4), Soo RA(5), Camidge DR(6),

Chiari R(7), Bearz A(8), Lin CC(9), Gadgeel SM(10), Riely GJ(11), Tan EH(12),

Seto T(13), James LP(14), Clancy JS(15), Abbattista A(16), Martini JF(17), Chen

J(14), Peltz G(18), Thurm H(17), Ou SI(19), Shaw AT(20).

Author information:

(1)Peter MacCallum Cancer Centre, Melbourne, VIC, Australia. Electronic address:

ben.solomon@petermac.org.

(2)Gustave Roussy Cancer Campus, Villejuif, France; Department of Cancer

Medicine, Paris-Sud University, Orsay, France.

(3)Sarah Cannon Cancer Research Institute/Tennessee Oncology, PLLC, Nashville,

TN, USA.

Erratum in

Lancet Oncol. 2019 Jan;20(1):e10.

BACKGROUND: Lorlatinib is a potent, brain-penetrant, third-generation inhibitor

of ALK and ROS1 tyrosine kinases with broad coverage of ALK mutations. In a phase

1 study, activity was seen in patients with ALK-positive non-small-cell lung

cancer, most of whom had CNS metastases and progression after ALK-directed

therapy. We aimed to analyse the overall and intracranial antitumour activity of

lorlatinib in patients with ALK-positive, advanced non-small-cell lung cancer.

METHODS: In this phase 2 study, patients with histologically or cytologically

ALK-positive or ROS1-positive, advanced, non-small-cell lung cancer, with or

without CNS metastases, with an Eastern Cooperative Oncology Group performance

status of 0, 1, or 2, and adequate end-organ function were eligible. Patients

were enrolled into six different expansion cohorts (EXP1-6) on the basis of ALK

and ROS1 status and previous therapy, and were given lorlatinib 100 mg orally

once daily continuously in 21-day cycles. The primary endpoint was overall and

intracranial tumour response by independent central review, assessed in pooled

subgroups of ALK-positive patients. Analyses of activity and safety were based on

the safety analysis set (ie, all patients who received at least one dose of

lorlatinib) as assessed by independent central review. Patients with measurable

CNS metastases at baseline by independent central review were included in the

intracranial activity analyses. In this report, we present lorlatinib activity

data for the ALK-positive patients (EXP1-5 only), and safety data for all treated

patients (EXP1-6). This study is ongoing and is registered with

ClinicalTrials.gov, number NCT01970865.

FINDINGS: Between Sept 15, 2015, and Oct 3, 2016, 276 patients were enrolled: 30

who were ALK positive and treatment naive (EXP1); 59 who were ALK positive and

received previous crizotinib without (n=27; EXP2) or with (n=32; EXP3A) previous

chemotherapy; 28 who were ALK positive and received one previous non-crizotinib

ALK tyrosine kinase inhibitor, with or without chemotherapy (EXP3B); 112 who were

ALK positive with two (n=66; EXP4) or three (n=46; EXP5) previous ALK tyrosine

kinase inhibitors with or without chemotherapy; and 47 who were ROS1 positive

with any previous treatment (EXP6). One patient in EXP4 died before receiving

lorlatinib and was excluded from the safety analysis set. In treatment-naive

patients (EXP1), an objective response was achieved in 27 (90·0%; 95% CI

73·5-97·9) of 30 patients. Three patients in EXP1 had measurable baseline CNS

lesions per independent central review, and objective intracranial responses were

observed in two (66·7%; 95% CI 9·4-99·2). In ALK-positive patients with at least

one previous ALK tyrosine kinase inhibitor (EXP2-5), objective responses were

achieved in 93 (47·0%; 39·9-54·2) of 198 patients and objective intracranial

response in those with measurable baseline CNS lesions in 51 (63·0%; 51·5-73·4)

of 81 patients. Objective response was achieved in 41 (69·5%; 95% CI 56·1-80·8)

of 59 patients who had only received previous crizotinib (EXP2-3A), nine (32·1%;

15·9-52·4) of 28 patients with one previous non-crizotinib ALK tyrosine kinase

inhibitor (EXP3B), and 43 (38·7%; 29·6-48·5) of 111 patients with two or more

previous ALK tyrosine kinase inhibitors (EXP4-5). Objective intracranial response

was achieved in 20 (87·0%; 95% CI 66·4-97·2) of 23 patients with measurable

baseline CNS lesions in EXP2-3A, five (55·6%; 21·2-86·3) of nine patients in

EXP3B, and 26 (53·1%; 38·3-67·5) of 49 patients in EXP4-5. The most common

treatment-related adverse events across all patients were hypercholesterolaemia

(224 [81%] of 275 patients overall and 43 [16%] grade 3-4) and

hypertriglyceridaemia (166 [60%] overall and 43 [16%] grade 3-4). Serious

treatment-related adverse events occurred in 19 (7%) of 275 patients and seven

patients (3%) permanently discontinued treatment because of treatment-related

adverse events. No treatment-related deaths were reported.

INTERPRETATION: Consistent with its broad ALK mutational coverage and CNS

penetration, lorlatinib showed substantial overall and intracranial activity both

in treatment-naive patients with ALK-positive non-small-cell lung cancer, and in

those who had progressed on crizotinib, second-generation ALK tyrosine kinase

inhibitors, or after up to three previous ALK tyrosine kinase inhibitors. Thus,

lorlatinib could represent an effective treatment option for patients with

ALK-positive non-small-cell lung cancer in first-line or subsequent therapy.

FUNDING: Pfizer.

Copyright © 2018 Elsevier Ltd. All rights reserved.

DOI: 10.1016/S1470-2045(18)30649-1

PMID: 30413378

5. Nat Med. 2018 Dec;24(12):1845-1851. doi: 10.1038/s41591-018-0232-2. Epub 2018 Nov 5.

Radiotherapy induces responses of lung cancer to CTLA-4 blockade.

Formenti SC(1), Rudqvist NP(2), Golden E(2)(3), Cooper B(4), Wennerberg E(2),

Lhuillier C(2), Vanpouille-Box C(2), Friedman K(5), Ferrari de Andrade L(6)(7),

Wucherpfennig KW(6)(7), Heguy A(8)(9), Imai N(10), Gnjatic S(10), Emerson RO(11),

Zhou XK(12), Zhang T(13), Chachoua A(14), Demaria S(15)(16).

Author information:

(1)Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA.

formenti@med.cornell.edu.

(2)Department of Radiation Oncology, Weill Cornell Medicine, New York, NY, USA.

(3)Department of Radiation Oncology, University of California, San Francisco, CA,

USA.

Focal radiation therapy enhances systemic responses to anti-CTLA-4 antibodies in

preclinical studies and in some patients with melanoma1-3, but its efficacy in

inducing systemic responses (abscopal responses) against tumors unresponsive to

CTLA-4 blockade remained uncertain. Radiation therapy promotes the activation of

anti-tumor T cells, an effect dependent on type I interferon induction in the

irradiated tumor4-6. The latter is essential for achieving abscopal responses in

murine cancers6. The mechanisms underlying abscopal responses in patients treated

with radiation therapy and CTLA-4 blockade remain unclear. Here we report that

radiation therapy and CTLA-4 blockade induced systemic anti-tumor T cells in

chemo-refractory metastatic non-small-cell lung cancer (NSCLC), where anti-CTLA-4

antibodies had failed to demonstrate significant efficacy alone or in combination

with chemotherapy7,8. Objective responses were observed in 18% of enrolled

patients, and 31% had disease control. Increased serum interferon-β after

radiation and early dynamic changes of blood T cell clones were the strongest

response predictors, confirming preclinical mechanistic data. Functional analysis

in one responding patient showed the rapid in vivo expansion of CD8 T cells

recognizing a neoantigen encoded in a gene upregulated by radiation, supporting

the hypothesis that one explanation for the abscopal response is

radiation-induced exposure of immunogenic mutations to the immune system.

DOI: 10.1038/s41591-018-0232-2

PMCID: PMC6286242 [Available on 2019-05-05]

PMID: 30397353

6. Lancet Oncol. 2018 Nov;19(11):1468-1479. doi: 10.1016/S1470-2045(18)30673-9. Epub 2018 Sep 24.

Avelumab versus docetaxel in patients with platinum-treated advanced

non-small-cell lung cancer (JAVELIN Lung 200): an open-label, randomised, phase 3

study.

Barlesi F(1), Vansteenkiste J(2), Spigel D(3), Ishii H(4), Garassino M(5), de

Marinis F(6), Özgüro?lu M(7), Szczesna A(8), Polychronis A(9), Uslu R(10),

Krzakowski M(11), Lee JS(12), Calabrò L(13), Arén Frontera O(14), Ellers-Lenz

B(15), Bajars M(16), Ruisi M(16), Park K(17).

Author information:

(1)Aix Marseille University, Assistance Publique Hôpitaux de Marseille,

Marseille, France.

(2)Department of Respiratory Oncology, University Hospital KU Leuven, Leuven,

Belgium.

(3)Sarah Cannon Research Institute, Nashville, TN, USA.

Erratum in

Lancet Oncol. 2018 Nov;19(11):e581.

BACKGROUND: Antibodies targeting the immune checkpoint molecules PD-1 or PD-L1

have demonstrated clinical efficacy in patients with metastatic non-small-cell

lung cancer (NSCLC). In this trial we investigated the efficacy and safety of

avelumab, an anti-PD-L1 antibody, in patients with NSCLC who had already received

platinum-based therapy.

METHODS: JAVELIN Lung 200 was a multicentre, open-label, randomised, phase 3

trial at 173 hospitals and cancer treatment centres in 31 countries. Eligible

patients were aged 18 years or older and had stage IIIB or IV or recurrent NSCLC

and disease progression after treatment with a platinum-containing doublet, an

Eastern Cooperative Oncology Group performance status score of 0 or 1, an

estimated life expectancy of more than 12 weeks, and adequate haematological,

renal, and hepatic function. Participants were randomly assigned (1:1), via an

interactive voice-response system with a stratified permuted block method with

variable block length, to receive either avelumab 10 mg/kg every 2 weeks or

docetaxel 75 mg/m2 every 3 weeks. Randomisation was stratified by PD-L1

expression (≥1% vs <1% of tumour cells), which was measured with the 73-10 assay,

and histology (squamous vs non-squamous). The primary endpoint was overall

survival, analysed when roughly 337 events (deaths) had occurred in the

PD-L1-positive population. Efficacy was analysed in all PD-L1-positive patients

(ie, PD-L1 expression in ≥1% of tumour cells) randomly assigned to study

treatment (the primary analysis population) and then in all randomly assigned

patients through a hierarchical testing procedure. Safety was analysed in all

patients who received at least one dose of study treatment. This trial is

registered with ClinicalTrials.gov, number NCT02395172. Enrolment is complete,

but the trial is ongoing.

FINDINGS: Between March 24, 2015, and Jan 23, 2017, 792 patients were enrolled

and randomly assigned to receive avelumab (n=396) or docetaxel (n=396). 264

participants in the avelumab group and 265 in the docetaxel group had

PD-L1-positive tumours. In patients with PD-L1-positive tumours, median overall

survival did not differ significantly between the avelumab and docetaxel groups

(11·4 months [95% CI 9·4-13·9] vs 10·3 months [8·5-13·0]; hazard ratio 0·90 [96%

CI 0·72-1·12]; one-sided p=0·16). Treatment-related adverse events occurred in

251 (64%) of 393 avelumab-treated patients and 313 (86%) of 365 docetaxel-treated

patients, including grade 3-5 events in 39 (10%) and 180 (49%) patients,

respectively. The most common grade 3-5 treatment-related adverse events were

infusion-related reaction (six patients [2%]) and increased lipase (four [1%]) in

the avelumab group and neutropenia (51 [14%]), febrile neutropenia (37 [10%]),

and decreased neutrophil counts (36 [10%]) in the docetaxel group. Serious

treatment-related adverse events occurred in 34 (9%) patients in the avelumab

group and 75 (21%) in the docetaxel group. Treatment-related deaths occurred in

four (1%) participants in the avelumab group, two due to interstitial lung

disease, one due to acute kidney injury, and one due to a combination of

autoimmune myocarditis, acute cardiac failure, and respiratory failure.

Treatment-related deaths occurred in 14 (4%) patients in the docetaxel group,

three due to pneumonia, and one each due to febrile neutropenia, septic shock,

febrile neutropenia with septic shock, acute respiratory failure, cardiovascular

insufficiency, renal impairment, leucopenia with mucosal inflammation and

pyrexia, infection, neutropenic infection, dehydration, and unknown causes.

INTERPRETATION: Compared with docetaxel, avelumab did not improve overall

survival in patients with platinum-treated PD-L1-positive NSCLC, but had a

favourable safety profile.

FUNDING: Merck and Pfizer.

Copyright © 2018 Elsevier Ltd. All rights reserved.

DOI: 10.1016/S1470-2045(18)30673-9

PMID: 30262187

7. J Clin Oncol. 2019 Jan 10;37(2):97-104. doi: 10.1200/JCO.18.00131. Epub 2018 Nov

16.

SELECT: A Phase II Trial of Adjuvant Erlotinib in Patients With Resected

Epidermal Growth Factor Receptor-Mutant Non-Small-Cell Lung Cancer.

Pennell NA(1), Neal JW(2), Chaft JE(3), Azzoli CG(4), Jänne PA(5), Govindan R(6),

Evans TL(7), Costa DB(8), Wakelee HA(2), Heist RS(4), Shapiro MA(1), Muzikansky

A(4), Murthy S(1), Lanuti M(4), Rusch VW(3), Kris MG(3), Sequist LV(4).

Author information:

(1)1 Cleveland Clinic Taussig Cancer Institute, Cleveland, OH.

(2)2 Stanford Cancer Institute and Stanford School of Medicine, Stanford, CA.

(3)3 Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College,

New York, NY.

(4)4 Massachusetts General Hospital, Boston, MA.

PURPOSE: Given the pivotal role of epidermal growth factor receptor (EGFR)

inhibitors in advanced EGFR-mutant non-small-cell lung cancer (NSCLC), we tested

adjuvant erlotinib in patients with EGFR-mutant early-stage NSCLC.

MATERIALS AND METHODS: In this open-label phase II trial, patients with resected

stage IA to IIIA (7th edition of the American Joint Committee on Cancer staging

system) EGFR-mutant NSCLC were treated with erlotinib 150 mg per day for 2 years

after standard adjuvant chemotherapy with or without radiotherapy. The study was

designed for 100 patients and powered to demonstrate a primary end point of

2-year disease-free survival (DFS) greater than 85%, improving on historic data

of 76%.

RESULTS: Patients (N = 100) were enrolled at seven sites from January 2008 to May

2012; 13% had stage IA disease, 32% had stage IB disease, 11% had stage IIA

disease, 16% had stage IIB disease, and 28% had stage IIIA disease. Toxicities

were typical of erlotinib; there were no grade 4 or 5 adverse events. Forty

percent of patients required erlotinib dose reduction to 100 mg per day and 16%

to 50 mg per day. The intended 2-year course was achieved in 69% of patients. The

median follow-up was 5.2 years, and 2-year DFS was 88% (96% stage I, 78% stage

II, 91% stage III). Median DFS and overall survival have not been reached; 5-year

DFS was 56% (95% CI, 45% to 66%), 5-year overall survival was 86% (95% CI, 77% to

92%). Disease recurred in 40 patients, with only four recurrences during

erlotinib treatment. The median time to recurrence was 25 months after stopping

erlotinib. Of patients with recurrence who underwent rebiopsy (n = 24; 60%), only

one had T790M mutation detected. The majority of patients with recurrence were

retreated with erlotinib (n = 26; 65%) for a median duration of 13 months.

CONCLUSION: Patients with EGFR-mutant NSCLC treated with adjuvant erlotinib had

an improved 2-year DFS compared with historic genotype-matched controls.

Recurrences were rare for patients receiving adjuvant erlotinib, and patients

rechallenged with erlotinib after recurrence experienced durable benefit.

DOI: 10.1200/JCO.18.00131

PMID: 30444685

8. J Natl Cancer Inst. 2018 Nov 13. doi: 10.1093/jnci/djy166. [Epub ahead of print]

Role of INSL4 Signaling in Sustaining the Growth and Viability of

LKB1-Inactivated Lung Cancer.

Yang R(1)(2), Li SW(1)(2)(3), Chen Z(1)(2), Zhou X(1)(2), Ni W(1)(4), Fu DA(5),

Lu J(1)(2)(6), Kaye FJ(2)(3), Wu L(1)(2)(4).

Author information:

(1)Department of Molecular Genetics and Microbiology.

(2)UF Health Cancer Center.

(3)Department of Medicine.

(4)UF Genetics Institute.

(5)Department of Pathology, Immunology and Laboratory Medicine.

(6)Department of Biochemistry and Molecular Biology, College of Medicine,

University of Florida, Gainesville, FL.

Background: The LKB1 tumor suppressor gene is commonly inactivated in non-small

cell lung carcinomas (NSCLC), a major form of lung cancer. Targeted therapies for

LKB1-inactivated lung cancer are currently unavailable. Identification of

critical signaling components downstream of LKB1 inactivation has the potential

to uncover rational therapeutic targets. Here we investigated the role of INSL4,

a member of the insulin/IGF/relaxin superfamily, in LKB1-inactivated NSCLCs.

Methods: INSL4 expression was analyzed using global transcriptome profiling,

quantitative reverse transcription PCR, western blotting, enzyme-linked

immunosorbent assay, and RNA in situ hybridization in human NSCLC cell lines and

tumor specimens. INSL4 gene expression and clinical data from The Cancer Genome

Atlas lung adenocarcinomas (n=515) were analyzed using log-rank and Fisher

exact tests. INSL4 functions were studied using short hairpin RNA (shRNA)

knockdown, overexpression, transcriptome profiling, cell growth, and survival

assays in vitro and in vivo. All statistical tests were two-sided.

Results: INSL4 was identified as a novel downstream target of LKB1 deficiency and

its expression was induced through aberrant CRTC-CREB activation. INSL4 was

highly induced in LKB1-deficient NSCLC cells (up to 543-fold) and 9 of 41 primary

tumors, although undetectable in all normal tissues except the placenta. Lung

adenocarcinomas from The Cancer Genome Atlas with high and low INSL4 expression

(with the top 10th percentile as cutoff) showed statistically significant

differences for advanced tumor stage (P<.001), lymph node metastasis

(P=.001), and tumor size (P=.01). The INSL4-high group showed worse survival

than the INSL4-low group (P<.001). Sustained INSL4 expression was required for

the growth and viability of LKB1-inactivated NSCLC cells in vitro and in a mouse

xenograft model (n=5 mice per group). Expression profiling revealed INSL4 as a

critical regulator of cell cycle, growth, and survival.

Conclusions: LKB1 deficiency induces an autocrine INSL4 signaling that critically

supports the growth and survival of lung cancer cells. Therefore, aberrant INSL4

signaling is a promising therapeutic target for LKB1-deficient lung cancers.

DOI: 10.1093/jnci/djy166

PMID: 30423141

9. PLoS Med. 2018 Nov 30;15(11):e1002711. doi: 10.1371/journal.pmed.1002711.

eCollection 2018 Nov.

Deep learning for lung cancer prognostication: A retrospective multi-cohort

radiomics study.

Hosny A(1), Parmar C(1), Coroller TP(1), Grossmann P(1), Zeleznik R(1), Kumar

A(1), Bussink J(2), Gillies RJ(3), Mak RH(4), Aerts HJWL(1)(4).

Author information:

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

Women's Hospital, Harvard Medical School, Boston, Massachusetts, United States of

America.

(2)Department of Radiation Oncology, Radboud University Medical Center, Nijmegen,

The Netherlands.

(3)Department of Cancer Physiology, H. Lee Moffitt Cancer Center and Research

Institute, Tampa, Florida, United States of America.

(4)Department of Radiology, Brigham and Women's Hospital, Harvard Medical School,

Boston, Massachusetts, United States of America.

BACKGROUND: Non-small-cell lung cancer (NSCLC) patients often demonstrate varying

clinical courses and outcomes, even within the same tumor stage. This study

explores deep learning applications in medical imaging allowing for the automated

quantification of radiographic characteristics and potentially improving patient

stratification.

METHODS AND FINDINGS: We performed an integrative analysis on 7 independent

datasets across 5 institutions totaling 1,194 NSCLC patients (age median = 68.3

years [range 32.5-93.3], survival median = 1.7 years [range 0.0-11.7]). Using

external validation in computed tomography (CT) data, we identified prognostic

signatures using a 3D convolutional neural network (CNN) for patients treated

with radiotherapy (n = 771, age median = 68.0 years [range 32.5-93.3], survival

median = 1.3 years [range 0.0-11.7]). We then employed a transfer learning

approach to achieve the same for surgery patients (n = 391, age median = 69.1

years [range 37.2-88.0], survival median = 3.1 years [range 0.0-8.8]). We found

that the CNN predictions were significantly associated with 2-year overall

survival from the start of respective treatment for radiotherapy (area under the

receiver operating characteristic curve [AUC] = 0.70 [95% CI 0.63-0.78], p <

0.001) and surgery (AUC = 0.71 [95% CI 0.60-0.82], p < 0.001) patients. The CNN

was also able to significantly stratify patients into low and high mortality risk

groups in both the radiotherapy (p < 0.001) and surgery (p = 0.03) datasets.

Additionally, the CNN was found to significantly outperform random forest models

built on clinical parameters-including age, sex, and tumor node metastasis

stage-as well as demonstrate high robustness against test-retest (intraclass

correlation coefficient = 0.91) and inter-reader (Spearman's rank-order

correlation = 0.88) variations. To gain a better understanding of the

characteristics captured by the CNN, we identified regions with the most

contribution towards predictions and highlighted the importance of

tumor-surrounding tissue in patient stratification. We also present preliminary

findings on the biological basis of the captured phenotypes as being linked to

cell cycle and transcriptional processes. Limitations include the retrospective

nature of this study as well as the opaque black box nature of deep learning

networks.

CONCLUSIONS: Our results provide evidence that deep learning networks may be used

for mortality risk stratification based on standard-of-care CT images from NSCLC

patients. This evidence motivates future research into better deciphering the

clinical and biological basis of deep learning networks as well as validation in

prospective data.

DOI: 10.1371/journal.pmed.1002711

PMCID: PMC6269088

PMID: 30500819

10. Nat Commun. 2018 Nov 19;9(1):4559. doi: 10.1038/s41467-018-07077-1.

H3K9 methyltransferases and demethylases control lung tumor-propagating cells and

lung cancer progression.

Rowbotham SP(1)(2), Li F(3), Dost AFM(1)(2), Louie SM(1)(2), Marsh BP(1)(2),

Pessina P(1)(2), Anbarasu CR(1)(2), Brainson CF(4), Tuminello SJ(5), Lieberman

A(6), Ryeom S(6), Schlaeger TM(1), Aronow BJ(7), Watanabe H(5), Wong KK(3), Kim

CF(8)(9)(10).

Author information:

(1)Stem Cell Program, Division of Hematology/Oncology and Pulmonary and

Respiratory Diseases, Children's Hospital Boston, Boston, MA, 02115, USA.

(2)Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA.

(3)Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical

Center, New York, NY, 10016, USA.

(4)Department of Toxicology and Cancer Biology, University of Kentucky,

Lexington, KY, 40536, USA.

Epigenetic regulators are attractive anticancer targets, but the promise of

therapeutic strategies inhibiting some of these factors has not been proven in

vivo or taken into account tumor cell heterogeneity. Here we show that the

histone methyltransferase G9a, reported to be a therapeutic target in many

cancers, is a suppressor of aggressive lung tumor-propagating cells (TPCs).

Inhibition of G9a drives lung adenocarcinoma cells towards the TPC phenotype by

de-repressing genes which regulate the extracellular matrix. Depletion of G9a

during tumorigenesis enriches tumors in TPCs and accelerates disease progression

metastasis. Depleting histone demethylases represses G9a-regulated genes and TPC

phenotypes. Demethylase inhibition impairs lung adenocarcinoma progression in

vivo. Therefore, inhibition of G9a is dangerous in certain cancer contexts, and

targeting the histone demethylases is a more suitable approach for lung cancer

treatment. Understanding cellular context and specific tumor populations is

critical when targeting epigenetic regulators in cancer for future therapeutic

development.

DOI: 10.1038/s41467-018-07077-1

PMCID: PMC6242814

PMID: 30455465

11. Cell Metab. 2018 Nov 5. pii: S1550-4131(18)30637-5. doi:

10.1016/j.cmet.2018.10.005. [Epub ahead of print]

Genetic Analysis Reveals AMPK Is Required to Support Tumor Growth in Murine

Kras-Dependent Lung Cancer Models.

Eichner LJ(1), Brun SN(1), Herzig S(1), Young NP(1), Curtis SD(1), Shackelford

DB(1), Shokhirev MN(2), Leblanc M(1), Vera LI(1), Hutchins A(1), Ross DS(1), Shaw

RJ(3), Svensson RU(4).

Author information:

(1)Molecular and Cell Biology Laboratories, The Salk Institute for Biological

Studies, La Jolla, San Diego, CA, USA.

(2)Integrative Genomics and Bioinformatics Core, The Salk Institute for

Biological Studies, La Jolla, San Diego, CA, USA.

(3)Molecular and Cell Biology Laboratories, The Salk Institute for Biological

Studies, La Jolla, San Diego, CA, USA. Electronic address: shaw@salk.edu.

(4)Molecular and Cell Biology Laboratories, The Salk Institute for Biological

Studies, La Jolla, San Diego, CA, USA. Electronic address: rsvensson@salk.edu.

AMPK, a conserved sensor of low cellular energy, can either repress or promote

tumor growth depending on the context. However, no studies have examined AMPK

function in autochthonous genetic mouse models of epithelial cancer. Here, we

examine the role of AMPK in murine KrasG12D-mediated non-small-cell lung cancer

(NSCLC), a cancer type in humans that harbors frequent inactivating mutations in

the LKB1 tumor suppressor-the predominant upstream activating kinase of AMPK and

12 related kinases. Unlike LKB1 deletion, AMPK deletion in KrasG12D lung tumors

did not accelerate lung tumor growth. Moreover, deletion of AMPK in KrasG12D

p53f/f tumors reduced lung tumor burden. We identified a critical role for AMPK

in regulating lysosomal gene expression through the Tfe3 transcription factor,

which was required to support NSCLC growth. Thus, AMPK supports the growth of

KrasG12D-dependent lung cancer through the induction of lysosomes, highlighting

an unrecognized liability of NSCLC.

Copyright © 2018 Elsevier Inc. All rights reserved.

DOI: 10.1016/j.cmet.2018.10.005

PMID: 30415923

12. Cancer Discov. 2018 Nov;8(11):1422-1437. doi: 10.1158/2159-8290.CD-18-0385. Epub 2018 Sep 4.

Crebbp Loss Drives Small Cell Lung Cancer and Increases Sensitivity to HDAC

Inhibition.

Jia D(#)(1), Augert A(#)(1), Kim DW(#)(2), Eastwood E(1), Wu N(1), Ibrahim AH(1),

Kim KB(2), Dunn CT(2), Pillai SPS(3), Gazdar AF(4), Bolouri H(1), Park KS(5),

MacPherson D(6)(7).

Author information:

(1)Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle,

Washington.

(2)Department of Microbiology, Immunology, and Cancer Biology, University of

Virginia School of Medicine, Charlottesville, Virginia.

(3)Division of Comparative Medicine, Fred Hutchinson Cancer Research Center,

Seattle, Washington.

CREBBP, encoding an acetyltransferase, is among the most frequently mutated genes

in small cell lung cancer (SCLC), a deadly neuroendocrine tumor type. We report

acceleration of SCLC upon Crebbp inactivation in an autochthonous mouse model.

Extending these observations beyond the lung, broad Crebbp deletion in mouse

neuroendocrine cells cooperated with Rb1/Trp53 loss to promote neuroendocrine

thyroid and pituitary carcinomas. Gene expression analyses showed that Crebbp

loss results in reduced expression of tight junction and cell adhesion genes,

including Cdh1, across neuroendocrine tumor types, whereas suppression of Cdh1

promoted transformation in SCLC. CDH1 and other adhesion genes exhibited reduced

histone acetylation with Crebbp inactivation. Treatment with the histone

deacetylase (HDAC) inhibitor Pracinostat increased histone acetylation and

restored CDH1 expression. In addition, a subset of Rb1/Trp53/Crebbp-deficient

SCLC exhibited exceptional responses to Pracinostat in vivo Thus, CREBBP acts as

a potent tumor suppressor in SCLC, and inactivation of CREBBP enhances responses

to a targeted therapy.Significance: Our findings demonstrate that CREBBP loss in

SCLC reduces histone acetylation and transcription of cellular adhesion genes,

while driving tumorigenesis. These effects can be partially restored by HDAC

inhibition, which exhibited enhanced effectiveness in Crebbp-deleted tumors.

These data provide a rationale for selectively treating CREBBP-mutant SCLC with

HDAC inhibitors. Cancer Discov; 8(11); 1422-37. ©2018 AACR. This article is

highlighted in the In This Issue feature, p. 1333.

©2018 American Association for Cancer Research.

DOI: 10.1158/2159-8290.CD-18-0385

PMCID: PMC6294438 [Available on 2019-11-01]

PMID: 30181244

13. J Clin Oncol. 2018 Nov 1;36(31):3101-3109. doi: 10.1200/JCO.2018.77.7326. Epub

2018 Aug 29.

Phase Ib/II Study of Capmatinib (INC280) Plus Gefitinib After Failure of

Epidermal Growth Factor Receptor (EGFR) Inhibitor Therapy in Patients With

EGFR-Mutated, MET Factor-Dysregulated Non-Small-Cell Lung Cancer.

Wu YL(1), Zhang L(1), Kim DW(1), Liu X(1), Lee DH(1), Yang JC(1), Ahn MJ(1),

Vansteenkiste JF(1), Su WC(1), Felip E(1), Chia V(1), Glaser S(1), Pultar P(1),

Zhao S(1), Peng B(1), Akimov M(1), Tan DSW(1).

Author information:

(1)Yi-Long Wu, Guangdong General Hospital and Guangdong Academy of Medical

Sciences; Li Zhang, Sun Yat-sen University Cancer Center, Guangdong; Xiaoqing

Liu, Affiliated Hospital of the Chinese Academy of Military Medical Sciences,

Beijing; Sylvia Zhao and Bin Peng, Novartis Institutes for Biomedical Research,

Shanghai, People's Republic of China; Dong-Wan Kim, Seoul National University

Hospital; Dae Ho Lee, University of Ulsan College of Medicine; Myung-Ju Ahn,

Samsung Medical Center, Seoul, Republic of Korea; James Chih-Hsin Yang, National

Taiwan University Hospital, Taipei; Wu-Chou Su, National Cheng Kung University

Hospital, Tainan, Taiwan; Johan F. Vansteenkiste, University Hospital KU Leuven,

Leuven, Belgium; Enriqueta Felip, Vall d'Hebron University Hospital, Barcelona,

Spain; Vincent Chia and Philippe Pultar, Novartis Pharmaceuticals, East Hanover,

NJ; Sabine Glaser and Mikhail Akimov, Novartis Pharma AG, Basel, Switzerland; and

Daniel S.W. Tan, National Cancer Centre Singapore, Singapore.

PURPOSE: MET dysregulation occurs in up to 26% of non-small-cell lung cancers

(NSCLCs) after epidermal growth factor receptor (EGFR)-tyrosine kinase inhibitor

(TKI) treatment. Capmatinib (INC280) is a potent and selective MET inhibitor with

preclinical activity in combination with gefitinib in EGFR-mutant,

MET-amplified/overexpressing models of acquired EGFR-TKI resistance. This phase

Ib/II study investigated the safety and efficacy of capmatinib plus gefitinib in

patients with EGFR-mutated, MET-dysregulated (amplified/overexpressing) NSCLC who

experienced disease progression while receiving EGFR-TKI treatment.

METHODS: Patients in phase Ib received capmatinib 100- to 800-mg capsules once

per day or 200- to 600-mg capsules or tablets twice per day, plus gefitinib 250

mg once per day. Patients in phase II received the recommended phase II dose. The

primary end point was the overall response rate (ORR) per Response Evaluation

Criteria in Solid Tumors (RECIST) version 1.1.

RESULTS: Sixty-one patients were treated in phase Ib, and 100 were treated in

phase II. The recommended phase II dose was capmatinib 400 mg twice per day plus

gefitinib 250 mg once per day. Preliminary clinical activity was observed, with

an ORR across phase Ib/II of 27%. Increased activity was seen in patients with

high MET-amplified tumors, with a phase II ORR of 47% in patients with a MET gene

copy number ≥ 6. Across phases Ib and II, the most common drug-related adverse

events were nausea (28%), peripheral edema (22%), decreased appetite (21%), and

rash (20%); the most common drug-related grade 3/4 adverse events were increased

amylase and lipase levels (both 6%). No significant drug-drug interactions

between capmatinib and gefitinib were evident.

CONCLUSION: This study, focused on a predominant EGFR-TKI resistance mechanism in

patients with EGFR-mutated NSCLC, shows that the combination of capmatinib with

gefitinib is a promising treatment for patients with EGFR-mutated,

MET-dysregulated NSCLC, particularly MET-amplified disease.

DOI: 10.1200/JCO.2018.77.7326

PMID: 30156984

14. Am J Respir Crit Care Med. 2018 Nov 1;198(9):1188-1198. doi:

10.1164/rccm.201710-2118OC.

Airway Microbiota Is Associated with Upregulation of the PI3K Pathway in Lung

Cancer.

Tsay JJ(1), Wu BG(1), Badri MH(2), Clemente JC(3), Shen N(3), Meyn P(4), Li Y(1),

Yie TA(1), Lhakhang T(4), Olsen E(1), Murthy V(1), Michaud G(1), Sulaiman I(1),

Tsirigos A(4), Heguy A(4), Pass H(5), Weiden MD(1), Rom WN(1), Sterman DH(1),

Bonneau R(2)(6), Blaser MJ(7), Segal LN(1).

Author information:

(1)1 Division of Pulmonary and Critical Care Medicine.

(2)2 Flatiron Institute, Center for Computational Biology, Simons Foundation, New

York, New York.

(3)3 Department of Genetics and Genomic Sciences and Immunology Institute, Icahn

School of Medicine at Mount Sinai, New York, New York.

(4)4 New York University Genomic Technology Center, New York, New York; and.

(5)5 Department of Cardiothoracic Surgery, and.

(6)6 New York University Center for Data Science, New York, New York.

(7)7 Department of Medicine, New York University School of Medicine, New York,

New York.

RATIONALE: In lung cancer, upregulation of the PI3K (phosphoinositide 3-kinase)

pathway is an early event that contributes to cell proliferation, survival, and

tissue invasion. Upregulation of this pathway was recently described as

associated with enrichment of the lower airways with bacteria identified as oral

commensals.

OBJECTIVES: We hypothesize that host-microbe interactions in the lower airways of

subjects with lung cancer affect known cancer pathways.

METHODS: Airway brushings were collected prospectively from subjects with lung

nodules at time of diagnostic bronchoscopy, including 39 subjects with final lung

cancer diagnoses and 36 subjects with noncancer diagnoses. In addition, samples

from 10 healthy control subjects were included. 16S ribosomal RNA gene amplicon

sequencing and paired transcriptome sequencing were performed on all airway

samples. In addition, an in vitro model with airway epithelial cells exposed to

bacteria/bacterial products was performed.

MEASUREMENTS AND MAIN RESULTS: The composition of the lower airway transcriptome

in the patients with cancer was significantly different from the control

subjects, which included up-regulation of ERK (extracellular signal-regulated

kinase) and PI3K signaling pathways. The lower airways of patients with lung

cancer were enriched for oral taxa (Streptococcus and Veillonella), which was

associated with up-regulation of the ERK and PI3K signaling pathways. In vitro

exposure of airway epithelial cells to Veillonella, Prevotella, and Streptococcus

led to upregulation of these same signaling pathways.

CONCLUSIONS: The data presented here show that several transcriptomic signatures

previously identified as relevant to lung cancer pathogenesis are associated with

enrichment of the lower airway microbiota with oral commensals.

DOI: 10.1164/rccm.201710-2118OC

PMCID: PMC6221574 [Available on 2019-11-01]

PMID: 29864375

上一篇: No.23

下一篇: No.21