您好!欢迎来到首都医科大学附属北京胸科医院官方网站!

高分SCI文摘

您所在的位置: 首页 >>胸科图书馆 >>信息推送 >>高分SCI文摘 >>2023年

高分SCI文摘

2023年

No.10

发布时间:2023-11-20 浏览次数:
字号: + - 14

PubMed

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

Filters applied: from 2023/10/1 - 2023/10/31.

 

1. N Engl J Med. 2023 Oct 21. doi: 10.1056/NEJMoa2306441. Online ahead of print.

 

Amivantamab plus Chemotherapy in NSCLC with EGFR Exon 20 Insertions.

 

Zhou C(1), Tang KJ(1), Cho BC(1), Liu B(1), Paz-Ares L(1), Cheng S(1), Kitazono

S(1), Thiagarajan M(1), Goldman JW(1), Sabari JK(1), Sanborn RE(1), Mansfield

AS(1), Hung JY(1), Boyer M(1), Popat S(1), Mourão Dias J(1), Felip E(1), Majem

M(1), Gumus M(1), Kim SW(1), Ono A(1), Xie J(1), Bhattacharya A(1), Agrawal

T(1), Shreeve SM(1), Knoblauch RE(1), Park K(1), Girard N(1); PAPILLON

Investigators.

 

Author information:

(1)From Shanghai Pulmonary Hospital, Tongji University School of Medicine,

Shanghai (C.Z.), the Division of Pulmonary and Critical Care Medicine, First

Affiliated Hospital of Sun Yat-sen University, Guangzhou (K.-J.T.), and Harbin

Medical University Cancer Hospital, Harbin (B.L.) - all in China; the Division

of Medical Oncology, Yonsei Cancer Center, Yonsei University College of Medicine

(B.C.C.), Asan Medical Center, University of Ulsan College of Medicine (S. Kim),

and Samsung Medical Center, Sungkyunkwan University School of Medicine (K.P.) -

all in Seoul, South Korea; Hospital Universitario 12 de Octubre, Madrid

(L.P.-A.),…

 

BACKGROUND: Amivantamab has been approved for the treatment of patients with

advanced non-small-cell lung cancer (NSCLC) with epidermal growth factor

receptor (EGFR) exon 20 insertions who have had disease progression during or

after platinum-based chemotherapy. Phase 1 data showed the safety and antitumor

activity of amivantamab plus carboplatin-pemetrexed (chemotherapy). Additional

data on this combination therapy are needed.

METHODS: In this phase 3, international, randomized trial, we assigned in a 1:1

ratio patients with advanced NSCLC with EGFR exon 20 insertions who had not

received previous systemic therapy to receive intravenous amivantamab plus

chemotherapy (amivantamab-chemotherapy) or chemotherapy alone. The primary

outcome was progression-free survival according to blinded independent central

review. Patients in the chemotherapy group who had disease progression were

allowed to cross over to receive amivantamab monotherapy.

RESULTS: A total of 308 patients underwent randomization (153 to receive

amivantamab-chemotherapy and 155 to receive chemotherapy alone).

Progression-free survival was significantly longer in the

amivantamab-chemotherapy group than in the chemotherapy group (median, 11.4

months and 6.7 months, respectively; hazard ratio for disease progression or

death, 0.40; 95% confidence interval [CI], 0.30 to 0.53; P<0.001). At 18 months,

progression-free survival was reported in 31% of the patients in the

amivantamab-chemotherapy group and in 3% in the chemotherapy group; a complete

or partial response at data cutoff was reported in 73% and 47%, respectively

(rate ratio, 1.50; 95% CI, 1.32 to 1.68; P<0.001). In the interim overall

survival analysis (33% maturity), the hazard ratio for death for

amivantamab-chemotherapy as compared with chemotherapy was 0.67 (95% CI, 0.42 to

1.09; P = 0.11). The predominant adverse events associated with

amivantamab-chemotherapy were reversible hematologic and EGFR-related toxic

effects; 7% of patients discontinued amivantamab owing to adverse reactions.

CONCLUSIONS: The use of amivantamab-chemotherapy resulted in superior efficacy

as compared with chemotherapy alone as first-line treatment of patients with

advanced NSCLC with EGFR exon 20 insertions. (Funded by Janssen Research and

Development; PAPILLON ClinicalTrials.gov number, NCT04538664.).

 

Copyright © 2023 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2306441

PMID: 37870976

 

2. N Engl J Med. 2023 Nov 2;389(18):1672-1684. doi: 10.1056/NEJMoa2304875. Epub

2023 Oct 23.

 

Perioperative Durvalumab for Resectable Non-Small-Cell Lung Cancer.

 

Heymach JV(1), Harpole D(1), Mitsudomi T(1), Taube JM(1), Galffy G(1), Hochmair

M(1), Winder T(1), Zukov R(1), Garbaos G(1), Gao S(1), Kuroda H(1), Ostoros

G(1), Tran TV(1), …

 

Author information:

(1)From the Department of Thoracic-Head and Neck Medical Oncology, University of

Texas M.D. Anderson Cancer Center, Houston (J.V.H.), and US Oncology Research,

the Woodlands (A.S.) - both in Texas; the Department of Surgery, Duke University

Medical Center (D.H.), and Duke Cancer Institute (J.C.) - both in Durham, NC;

the Division of Thoracic Surgery, Department of Surgery, Kindai University

Faculty of Medicine, Osaka-Sayama (T.M.), …

 

BACKGROUND: Neoadjuvant or adjuvant immunotherapy can improve outcomes in

patients with resectable non-small-cell lung cancer (NSCLC). Perioperative

regimens may combine benefits of both to improve long-term outcomes.

METHODS: We randomly assigned patients with resectable NSCLC (stage II to IIIB

[N2 node stage] according to the eighth edition of the AJCC Cancer Staging

Manual) to receive platinum-based chemotherapy plus durvalumab or placebo

administered intravenously every 3 weeks for 4 cycles before surgery, followed

by adjuvant durvalumab or placebo intravenously every 4 weeks for 12 cycles.

Randomization was stratified according to disease stage (II or III) and

programmed death ligand 1 (PD-L1) expression (≥1% or<1%). Primary end points

were event-free survival (defined as the time to the earliest occurrence of

progressive disease that precluded surgery or prevented completion of surgery,

disease recurrence [assessed in a blinded fashion by independent central

review], or death from any cause) and pathological complete response (evaluated

centrally).

RESULTS: A total of 802 patients were randomly assigned to receive durvalumab

(400 patients) or placebo (402 patients). The duration of event-free survival

was significantly longer with durvalumab than with placebo; the stratified

hazard ratio for disease progression, recurrence, or death was 0.68 (95%

confidence interval [CI], 0.53 to 0.88; P = 0.004) at the first interim

analysis. At the 12-month landmark analysis, event-free survival was observed in

73.4% of the patients who received durvalumab (95% CI, 67.9 to 78.1), as

compared with 64.5% of the patients who received placebo (95% CI, 58.8 to 69.6).

The incidence of pathological complete response was significantly greater with

durvalumab than with placebo (17.2% vs. 4.3% at the final analysis; difference,

13.0 percentage points; 95% CI, 8.7 to 17.6; P<0.001 at interim analysis of data

from 402 patients). Event-free survival and pathological complete response

benefit were observed regardless of stage and PD-L1 expression. Adverse events

of maximum grade 3 or 4 occurred in 42.4% of patients with durvalumab and in

43.2% with placebo. Data from 62 patients with documented EGFR or ALK

alterations were excluded from the efficacy analyses in the modified

intention-to-treat population.

CONCLUSIONS: In patients with resectable NSCLC, perioperative durvalumab plus

neoadjuvant chemotherapy was associated with significantly greater event-free

survival and pathological complete response than neoadjuvant chemotherapy alone,

with a safety profile that was consistent with the individual agents. (Funded by

AstraZeneca; AEGEAN ClinicalTrials.gov number, NCT03800134.).

 

Copyright © 2023 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2304875

PMID: 37870974 [Indexed for MEDLINE]

 

3. N Engl J Med. 2023 Nov 16;389(20):1839-1850. doi: 10.1056/NEJMoa2309457. Epub

2023 Oct 21.

 

First-Line Selpercatinib or Chemotherapy and Pembrolizumab in RET

Fusion-Positive NSCLC.

 

Zhou C(1), Solomon B(1), Loong HH(1), Park K(1), Pérol M(1), Arriola E(1),

Novello S(1), Han B(1), Zhou J(1), Ardizzoni A(1), Mak MP(1), Santini FC(1),

Elamin YY(1), Drilon A(1), Wolf J(1), Payakachat N(1), Uh MK(1), Rajakumar D(1),

Han H(1), Puri T(1), Soldatenkova V(1), Lin AB(1), Lin BK(1), Goto K(1);

LIBRETTO-431 Trial Investigators.

 

Author information:

(1)From Shanghai Pulmonary Hospital, Tongji University School of Medicine

(C.Z.), and the Department of Respiratory and Critical Care Medicine, Shanghai

Chest Hospital, School of Medicine, Shanghai Jiao Tong University (B.H.),

Shanghai, the Chinese University of Hong Kong, Hong Kong (H.H.L.), …

 

BACKGROUND: Selpercatinib, a highly selective potent and brain-penetrant RET

inhibitor, was shown to have efficacy in patients with advanced RET

fusion-positive non-small-cell lung cancer (NSCLC) in a nonrandomized phase 1-2

study.

METHODS: In a randomized phase 3 trial, we evaluated the efficacy and safety of

first-line selpercatinib as compared with control treatment that consisted of

platinum-based chemotherapy with or without pembrolizumab at the investigator's

discretion. The primary end point was progression-free survival assessed by

blinded independent central review in both the intention-to-treat-pembrolizumab

population (i.e., patients whose physicians had planned to treat them with

pembrolizumab in the event that they were assigned to the control group) and the

overall intention-to-treat population. Crossover from the control group to the

selpercatinib group was allowed if disease progression as assessed by blinded

independent central review occurred during receipt of control treatment.

RESULTS: In total, 212 patients underwent randomization in the

intention-to-treat-pembrolizumab population. At the time of the preplanned

interim efficacy analysis, median progression-free survival was 24.8 months (95%

confidence interval [CI], 16.9 to not estimable) with selpercatinib and 11.2

months (95% CI, 8.8 to 16.8) with control treatment (hazard ratio for

progression or death, 0.46; 95% CI, 0.31 to 0.70; P<0.001). The percentage of

patients with an objective response was 84% (95% CI, 76 to 90) with

selpercatinib and 65% (95% CI, 54 to 75) with control treatment. The

cause-specific hazard ratio for the time to progression affecting the central

nervous system was 0.28 (95% CI, 0.12 to 0.68). Efficacy results in the overall

intention-to-treat population (261 patients) were similar to those in the

intention-to-treat-pembrolizumab population. The adverse events that occurred

with selpercatinib and control treatment were consistent with those previously

reported.

CONCLUSIONS: Treatment with selpercatinib led to significantly longer

progression-free survival than platinum-based chemotherapy with or without

pembrolizumab among patients with advanced RET fusion-positive NSCLC. (Funded by

Eli Lilly and others; ClinicalTrials.gov number, NCT04194944.).

 

Copyright © 2023 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2309457

PMID: 37870973

 

4. Cell. 2023 Nov 9;186(23):5135-5150.e28. doi: 10.1016/j.cell.2023.09.016. Epub

2023 Oct 20.

 

Mechanopathology of biofilm-like Mycobacterium tuberculosis cords.

 

Mishra R(1), Hannebelle M(2), Patil VP(3), Dubois A(4), Garcia-Mouton C(5),

Kirsch GM(1), Jan M(6), Sharma K(1), Guex N(6), Sordet-Dessimoz J(7), Perez-Gil

J(5), Prakash M(3), Knott GW(4), Dhar N(1), McKinney JD(1), Thacker VV(8).

 

Author information:

(1)Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015

Lausanne, Switzerland.

(2)Global Health Institute, École Polytechnique Fédérale de Lausanne, 1015

Lausanne, Switzerland; Department of Bioengineering, Stanford University,

Stanford, CA 94305, USA.

(3)Department of Bioengineering, Stanford University, Stanford, CA 94305, USA.

Comment in

    Cell. 2023 Nov 9;186(23):4994-4995.

 

Mycobacterium tuberculosis (Mtb) cultured axenically without detergent forms

biofilm-like cords, a clinical identifier of virulence. In lung-on-chip (LoC)

and mouse models, cords in alveolar cells contribute to suppression of innate

immune signaling via nuclear compression. Thereafter, extracellular cords cause

contact-dependent phagocyte death but grow intercellularly between epithelial

cells. The absence of these mechanopathological mechanisms explains the greater

proportion of alveolar lesions with increased immune infiltration and

dissemination defects in cording-deficient Mtb infections. Compression of Mtb

lipid monolayers induces a phase transition that enables mechanical energy

storage. Agent-based simulations demonstrate that the increased energy storage

capacity is sufficient for the formation of cords that maintain structural

integrity despite mechanical perturbation. Bacteria in cords remain

translationally active despite antibiotic exposure and regrow rapidly upon

cessation of treatment. This study provides a conceptual framework for the

biophysics and function in tuberculosis infection and therapy of cord

architectures independent of mechanisms ascribed to single bacteria.

 

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

 

DOI: 10.1016/j.cell.2023.09.016

PMCID: PMC10642369

PMID: 37865090 [Indexed for MEDLINE]

 

5. N Engl J Med. 2023 Oct 20. doi: 10.1056/NEJMoa2307980. Online ahead of print.

 

Tarlatamab for Patients with Previously Treated Small-Cell Lung Cancer.

 

Ahn MJ(1), Cho BC(1), Felip E(1), Korantzis I(1), Ohashi K(1), Majem M(1),

Juan-Vidal O(1), Handzhiev S(1), Izumi H(1), Lee JS(1), Dziadziuszko R(1), Wolf

J(1), Blackhall F(1), Reck M(1), Bustamante Alvarez J(1), Hummel HD(1),

Dingemans AC(1), Sands J(1), Akamatsu H(1), Owonikoko TK(1), Ramalingam SS(1),

Borghaei H(1), Johnson ML(1), Huang S(1), Mukherjee S(1), Minocha M(1), Jiang

T(1), Martinez P(1), Anderson ES(1), Paz-Ares L(1); DeLLphi-301 Investigators.

 

Author information:

(1)From Samsung Medical Center, Sungkyunkwan University School of Medicine

(M.-J.A.), and Yonsei Cancer Center, Yonsei University College of Medicine

(B.C.C.), Seoul, and Seoul National University Bundang Hospital, Seongnam

(J.-S.L.) - all in South Korea; Vall d'Hebron University Hospital and Vall

d'Hebron Institute of Oncology (E.F.) …

 

BACKGROUND: Tarlatamab, a bispecific T-cell engager immunotherapy targeting

delta-like ligand 3 and CD3, showed promising antitumor activity in a phase 1

trial in patients with previously treated small-cell lung cancer.

METHODS: In this phase 2 trial, we evaluated the antitumor activity and safety

of tarlatamab, administered intravenously every 2 weeks at a dose of 10 mg or

100 mg, in patients with previously treated small-cell lung cancer. The primary

end point was objective response (complete or partial response), as assessed by

blinded independent central review according to the Response Evaluation Criteria

in Solid Tumors, version 1.1.

RESULTS: Overall, 220 patients received tarlatamab; patients had previously

received a median of two lines of treatment. Among patients evaluated for

antitumor activity and survival, the median follow-up was 10.6 months in the

10-mg group and 10.3 months in the 100-mg group. An objective response occurred

in 40% (97.5% confidence interval [CI], 29 to 52) of the patients in the 10-mg

group and in 32% (97.5% CI, 21 to 44) of those in the 100-mg group. Among

patients with an objective response, the duration of response was at least 6

months in 59% (40 of 68 patients). Objective responses at the time of data

cutoff were ongoing in 22 of 40 patients (55%) in the 10-mg group and in 16 of

28 patients (57%) in the 100-mg group. The median progression-free survival was

4.9 months (95% CI, 2.9 to 6.7) in the 10-mg group and 3.9 months (95% CI, 2.6

to 4.4) in the 100-mg group; the estimates of overall survival at 9 months were

68% and 66% of patients, respectively. The most common adverse events were

cytokine-release syndrome (in 51% of the patients in the 10-mg group and in 61%

of those in the 100-mg group), decreased appetite (in 29% and 44%,

respectively), and pyrexia (in 35% and 33%). Cytokine-release syndrome occurred

primarily during treatment cycle 1, and events in most of the patients were

grade 1 or 2 in severity. Grade 3 cytokine-release syndrome occurred less

frequently in the 10-mg group (in 1% of the patients) than in the 100-mg group

(in 6%). A low percentage of patients (3%) discontinued tarlatamab because of

treatment-related adverse events.

CONCLUSIONS: Tarlatamab, administered as a 10-mg dose every 2 weeks, showed

antitumor activity with durable objective responses and promising survival

outcomes in patients with previously treated small-cell lung cancer. No new

safety signals were identified. (Funded by Amgen; DeLLphi-301 ClinicalTrials.gov

number, NCT05060016.).

 

Copyright © 2023 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2307980

PMID: 37861218

 

6. Nat Med. 2023 Oct 30. doi: 10.1038/s41591-023-02660-6. Online ahead of print.

 

Association between pathologic response and survival after neoadjuvant therapy

in lung cancer.

 

Deutsch JS(1), Cimino-Mathews A(1), Thompson E(1), Provencio M(2), Forde PM(1),

Spicer J(3), Girard N(4), Wang D(1), Anders RA(1), Gabrielson E(1), Illei P(1),

Jedrych J(1), Danilova L(1), Sunshine J(1), Kerr KM(5), Tran M(6), Bushong J(6),

Cai J(6), Devas V(6), Neely J(6), Balli D(6), Cottrell TR(7), Baras AS(1), Taube

JM(8)(9).

 

Author information:

(1)Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University

School of Medicine, Baltimore, Maryland, USA.

(2)Hospital Universitario Puerta de Hierro, Madrid, Spain.

(3)McGill University Health Center, Montreal, Québec, Canada.

Neoadjuvant immunotherapy plus chemotherapy improves event-free survival (EFS)

and pathologic complete response (pCR, 0% residual viable tumor [RVT] in primary

tumor [PT]+lymph nodes [LNs]), and is approved for treatment of resectable lung

cancer. Pathologic response assessment after neoadjuvant therapy is the

potential analog to radiographic response for advanced disease. However, %RVT

thresholds beyond pCR and major pathologic response (≤10% RVT) have not been

explored. Pathologic response was prospectively assessed in the randomized,

phase 3 CheckMate 816 trial (NCT02998528), which evaluated neoadjuvant nivolumab

(anti-PD-1) plus chemotherapy in patients with resectable lung cancer. RVT,

regression, and necrosis were quantified (0%-100%) in PT and LNs using a

pan-tumor scoring system and tested for association with EFS in a prespecified

exploratory analysis. Regardless of LN involvement, EFS improved with 0% versus

>0% RVT-PT (HR = 0.18). RVT-PT predicted EFS for nivolumab plus chemotherapy

(AUC = 0.74); 2-year EFS rates were 90%, 60%, 57%, and 39% for patients with

0%-5%, >5%-30%, >30%-80%, and >80% RVT, respectively. Each 1% RVT associated

with a 0.017 HR increase for EFS. Combining pathologic response from PT+LNs

helped differentiate outcomes. When compared to radiographic response and ctDNA

clearance, %RVT best approximated EFS. These findings support pathologic

response as an emerging survival surrogate. Further assessment of the full

spectrum of %RVT in lung cancer and other tumor types is warranted.

 

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

 

DOI: 10.1038/s41591-023-02660-6

PMID: 37903504

 

7. Cancer Cell. 2023 Oct 9;41(10):1763-1773.e4. doi: 10.1016/j.ccell.2023.09.007.

 

Dynamic circulating tumor DNA during chemoradiotherapy predicts clinical

outcomes for locally advanced non-small cell lung cancer patients.

 

Pan Y(1), Zhang JT(1), Gao X(2), Chen ZY(1), Yan B(2), Tan PX(1), Yang XR(1),

Gao W(2), Gong Y(2), Tian Z(2), Liu SM(3), Lin H(1), Sun H(1), Huang J(1), Liu

SY(1), Yan HH(1), Dong S(1), Xu CR(1), Chen HJ(1), Wang Z(1), Li P(2), Guan

Y(2), Wang BC(1), Yang JJ(1), Tu HY(1), Yang XN(1), Zhong WZ(1), Xia X(2), Yi

X(4), Zhou Q(5), Wu YL(6).

 

Author information:

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

(Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou,

Guangdong, China.

(2)Geneplus-Beijing Institute, Beijing, China.

(3)Department of Hematology, First Affiliated Hospital, Institute of Hematology,

School of Medicine; Key Laboratory for Regenerative Medicine of Ministry of

Education, Jinan University, Guangzhou, Guangdong, China; Chinese Thoracic

Oncology Group (CTONG), Guangzhou, Guangdong, China.

Comment in

    Cancer Cell. 2023 Oct 9;41(10):1699-1701.

 

The value of circulating tumor DNA (ctDNA) during chemoradiotherapy (CRT)

remains unclear but is critical for detecting molecular residual disease (MRD).

In this prospective study, we sequenced 761 blood samples from 139 patients with

locally advanced non-small cell lung cancer treated with definitive radiation

therapy (RT). ctDNA concentrations showed a significantly declining trend as CRT

progressed at on-RT and after-RT time points versus baseline. Thirty-eight

(27.3%) patients with early undetectable ctDNA at both on-RT (RT reached 40 Gy)

and after-RT time points, indicating early response to CRT, had better survival

outcomes for both with or without consolidation immune checkpoint inhibitors.

Longitudinal undetectable MRD was found in 20.1% patients. The 2-year

cancer-specific progression-free survival of these patients was 88.4%,

corresponding to a potentially cured population. Further analysis revealed that

pretreatment ctDNA variants serve as an essential MRD informed source. These

data provide clinical insights for ctDNA-MRD detection.

 

Copyright © 2023 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2023.09.007

PMID: 37816331 [Indexed for MEDLINE]

 

8. Nat Med. 2023 Oct;29(10):2559-2569. doi: 10.1038/s41591-023-02598-9. Epub 2023

Oct 9.

 

ctDNA response after pembrolizumab in non-small cell lung cancer: phase 2

adaptive trial results.

 

Anagnostou V(1)(2), Ho C(3), Nicholas G(4), Juergens RA(5), Sacher A(6), Fung

AS(7), Wheatley-Price P(4), Laurie SA(4), Levy B(8), Brahmer JR(8)(9), Balan

A(8), Niknafs N(8), Avrutin E(10), Zhu L(10), Sausen M(11), Bradbury PA(6),

O'Donnell-Tormey J(12), Gaudreau PO(10), Ding K(10), Dancey J(13).

 

Author information:

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

Medicine, Baltimore, MD, USA. vanagno1@jhmi.edu.

(2)Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University

School of Medicine, Baltimore, MD, USA. vanagno1@jhmi.edu.

(3)BCCA-Vancouver Cancer Centre, Vancouver, BC, Canada.

Circulating tumor DNA (ctDNA) has shown promise in capturing primary resistance

to immunotherapy. BR.36 is a multi-center, randomized, ctDNA-directed, phase 2

trial of molecular response-adaptive immuno-chemotherapy for patients with lung

cancer. In the first of two independent stages, 50 patients with advanced

non-small cell lung cancer received pembrolizumab as standard of care. The

primary objectives of stage 1 were to ascertain ctDNA response and determine

optimal timing and concordance with radiologic Response Evaluation Criteria in

Solid Tumors (RECIST) response. Secondary endpoints included the evaluation of

time to ctDNA response and correlation with progression-free and overall

survival. Maximal mutant allele fraction clearance at the third cycle of

pembrolizumab signified molecular response (mR). The trial met its primary

endpoint, with a sensitivity of ctDNA response for RECIST response of 82% (90%

confidence interval (CI): 52-97%) and a specificity of 75% (90% CI: 56.5-88.5%).

Median time to ctDNA response was 2.1 months (90% CI: 1.5-2.6), and patients

with mR attained longer progression-free survival (5.03 months versus

2.6 months) and overall survival (not reached versus 7.23 months). These

findings are incorporated into the ctDNA-driven interventional molecular

response-adaptive second stage of the BR.36 trial in which patients at risk of

progression are randomized to treatment intensification or continuation of

therapy. ClinicalTrials.gov ID: NCT04093167 .

 

© 2023. The Author(s).

 

DOI: 10.1038/s41591-023-02598-9

PMCID: PMC10579094

PMID: 37814061 [Indexed for MEDLINE]

 

9. Lancet Infect Dis. 2023 Oct 30:S1473-3099(23)00491-7. doi:

10.1016/S1473-3099(23)00491-7. Online ahead of print.

 

Diagnostic accuracy of a three-gene Mycobacterium tuberculosis host response

cartridge using fingerstick blood for childhood tuberculosis: a multicentre

prospective study in low-income and middle-income countries.

 

Olbrich L(1), Verghese VP(2), Franckling-Smith Z(3), Sabi I(4), Ntinginya NE(4),

Mfinanga A(4), Banze D(5), Viegas S(5), Khosa C(5), Semphere R(6), Nliwasa M(6),

McHugh TD(7), Larsson L(8), Razid A(8), Song R(9), Corbett EL(10), Nabeta P(11),

Trollip A(11), Graham SM(12), Hoelscher M(13), Geldmacher C(14), Zar HJ(3),

Michael JS(15), Heinrich N(16); RaPaed-TB consortium.

 

Author information:

(1)Division of Infectious Diseases and Tropical Medicine, LMU University

Hospital, LMU Munich, Munich, Germany; German Centre for Infection Research

(DZIF), Partner Site Munich, Munich, Germany; Fraunhofer Institute ITMP,

Immunology, Infection and Pandemic Research, Munich, Germany; Oxford Vaccine

Group, Department of Paediatrics and the NIHR Oxford Biomedical Research Centre,

University of Oxford, Oxford, UK.

(2)Pediatric Infectious Diseases, Department of Pediatrics, Christian Medical

College, Vellore, India.

(3)Department of Paediatrics and Child Health, SA-MRC Unit on Child and

Adolescent Health, University of Cape Town, Cape Town, South Africa.

BACKGROUND: Childhood tuberculosis remains a major cause of morbidity and

mortality in part due to missed diagnosis. Diagnostic methods with enhanced

sensitivity using easy-to-obtain specimens are needed. We aimed to assess the

diagnostic accuracy of the Cepheid Mycobacterium tuberculosis Host Response

prototype cartridge (MTB-HR), a candidate test measuring a three-gene

transcriptomic signature from fingerstick blood, in children with presumptive

tuberculosis disease.

METHODS: RaPaed-TB was a prospective diagnostic accuracy study conducted at four

sites in African countries (Malawi, Mozambique, South Africa, and Tanzania) and

one site in India. Children younger than 15 years with presumptive pulmonary or

extrapulmonary tuberculosis were enrolled between Jan 21, 2019, and June 30,

2021. MTB-HR was performed at baseline and at 1 month in all children and was

repeated at 3 months and 6 months in children on tuberculosis treatment.

Accuracy was compared with tuberculosis status based on standardised

microbiological, radiological, and clinical data.

FINDINGS: 5313 potentially eligible children were screened, of whom 975 were

eligible. 784 children had MTB-HR test results, of whom 639 had a diagnostic

classification and were included in the analysis. MTB-HR differentiated children

with culture-confirmed tuberculosis from those with unlikely tuberculosis with a

sensitivity of 59·8% (95% CI 50·8-68·4). Using any microbiological confirmation

(culture, Xpert MTB/RIF Ultra, or both), sensitivity was 41·6% (34·7-48·7), and using 

a composite clinical reference standard, sensitivity was 29·6%

(25·4-34·2). Specificity for all three reference standards was 90·3% (95% CI

85·5-94·0). Performance was similar in different age groups and by malnutrition

status. Among children living with HIV, accuracy against the strict reference

standard tended to be lower (sensitivity 50·0%, 15·7-84·3) compared with those

without HIV (61·0%, 51·6-69·9), although the difference did not reach

statistical significance. Combining baseline MTB-HR result with one Ultra result

identified 71·2% of children with microbiologically confirmed tuberculosis.

INTERPRETATION: MTB-HR showed promising diagnostic accuracy for

culture-confirmed tuberculosis in this large, geographically diverse, paediatric

cohort and hard-to-diagnose subgroups.

FUNDING: European and Developing Countries Clinical Trials Partnership, UK

Medical Research Council, Swedish International Development Cooperation Agency,

Bundesministerium für Bildung und Forschung; German Center for Infection

Research (DZIF).

 

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

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

rights reserved.

 

DOI: 10.1016/S1473-3099(23)00491-7

PMID: 37918414

 

10. J Clin Oncol. 2023 Oct 20:101200JCO2301891. doi: 10.1200/JCO.23.01891. Online

ahead of print.

 

A Phase 3, Randomized study of atezolizumab plus bevacizumab and chemotherapy in

patients with EGFR or ALK mutated in non-small cell lung cancer (ATTLAS,

KCSG-LU19-04).

 

Park S(1), Kim TM(2), Han JY(3), Lee GW(4), Shim BY(5), Lee YG(6), Kim SW(7),

Kim IH(8), Lee S(9), Kim YJ(10), Park JH(11), Park SG(12), Lee KH(13), Kang

EJ(14), Kim JW(15), Shin SH(16), Ock CY(17), Nam BH(18), Lee J(19), Jung HA(1),

Sun JM(1), Lee SH(1), Ahn JS(1), Ahn MJ(1).

 

Author information:

(1)Division of Hematology-Oncology, Department of Medicine, Samsung Medical

Center, Sungkyunkwan University School of Medicine, Seoul, Korea.

(2)Department of Internal Medicine, Seoul National University Hospital, Seoul

National University Cancer Research Institute, Seoul, Korea.

(3)Center for Lung Cancer, Research Institute and Hospital, National Cancer

Center, Goyang-si Gyeonggi-do, Korea.

PURPOSE: In the treatment of non-small cell lung cancer (NSCLC) with a driver

mutation, the role of anti-PD-(L)1 antibody following tyrosine kinase inhibitor

(TKI) remains unclear. This randomized, open-label, multicenter, phase 3 study

evaluates the efficacy of atezolizumab plus bevacizumab, paclitaxel and

carboplatin (ABCP) in EGFR or ALK-mutated NSCLC that progressed prior to TKI

therapy.

METHODS: We compared the clinical efficacy of ABCP followed by maintenance

therapy with atezolizumab plus bevacizumab with pemetrexed plus carboplatin or

cisplatin (PC arm) followed by pemetrexed maintenance. The primary endpoint was

progression-free survival (PFS).

RESULTS: A total of 228 patients with activating EGFR mutation (n=215) or ALK

translocation (n=13) were enrolled from 16 sites in the Republic of Korea and

randomized at 2:1 ratio to either ABCP (n=154) or PC arm (n=74). The median

follow-up duration was 26.1 months (95%CI 24.7-28.2). Objective response rates

(69.5% vs 41.9%, P<0.001) and median PFS (8.48 vs. 5.62 months, hazard ratio

[HR] 0.62 [0.45-0.86], P=0.004) were significantly better in the ABCP than PC

arm. PFS benefit increased as PD-L1 expression increased, with HR of 0.47, 0.41,

and 0.24 for PD-L1 ≥1%, ≥10% and ≥50%, respectively. Overall survival was

similar between ABCP and PC arm (20.63 vs. 20.27 months, HR 1.01 [0.69-1.46],

P=0.975). The safety profile of the ABCP arm was comparable to that previously

reported, with no additional safety signals, but higher rates of

treatment-related adverse events were observed compared to the PC arm.

CONCLUSION: This study is the first randomized phase 3 study to demonstrate the

clinical benefit of anti-PD-L1 antibody in combination with bevacizumab and

chemotherapy in EGFR or ALK mutated NSCLC who have progressed on relevant

targeted therapy.

 

DOI: 10.1200/JCO.23.01891

PMID: 37861993

 

11. Cell Host Microbe. 2023 Nov 8;31(11):1820-1836.e10. doi:

10.1016/j.chom.2023.09.010. Epub 2023 Oct 16.

 

Mycobacterium tuberculosis suppresses host DNA repair to boost its intracellular

survival.

 

Liu S(1), Guan L(1), Peng C(1), Cheng Y(1), Cheng H(1), Wang F(1), Ma M(1),

Zheng R(1), Ji Z(2), Cui P(1), Ren Y(1), Li L(1), Shi C(1), Wang J(3), Huang

X(3), Cai X(4), Qu D(4), Zhang H(5), Mao Z(5), Liu H(6), Wang P(7), Sha W(7),

Yang H(8), Wang L(9), Ge B(10).

 

Author information:

(1)Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key

Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji

University School of Medicine, Shanghai 200433, P.R. China; Department of

Microbiology and Immunology, Tongji University School of Medicine, Shanghai

200092, P.R. China.

(2)Department of Microbiology and Immunology, Tongji University School of

Medicine, Shanghai 200092, P.R. China.

(3)Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Key

Laboratory of Pathogen-Host Interaction, Ministry of Education, Tongji

University School of Medicine, Shanghai 200433, P.R. China.

Mycobacterium tuberculosis (Mtb) triggers distinct changes in macrophages,

resulting in the formation of lipid droplets that serve as a nutrient source. We

discover that Mtb promotes lipid droplets by inhibiting DNA repair responses,

resulting in the activation of the type-I IFN pathway and scavenger receptor-A1

(SR-A1)-mediated lipid droplet formation. Bacterial urease C (UreC, Rv1850)

inhibits host DNA repair by interacting with RuvB-like protein 2 (RUVBL2) and

impeding the formation of the RUVBL1-RUVBL2-RAD51 DNA repair complex. The

suppression of this repair pathway increases the abundance of micronuclei that

trigger the cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes

(STING) pathway and subsequent interferon-β (IFN-β) production. UreC-mediated

activation of the IFN-β pathway upregulates the expression of SR-A1 to form

lipid droplets that facilitate Mtb replication. UreC inhibition via a urease

inhibitor impaired Mtb growth within macrophages and in vivo. Thus, our findings

identify mechanisms by which Mtb triggers a cascade of cellular events that

establish a nutrient-rich replicative niche.

 

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

 

DOI: 10.1016/j.chom.2023.09.010

PMID: 37848028 [Indexed for MEDLINE]

 

12. J Clin Oncol. 2023 Oct 11:JCO2301435. doi: 10.1200/JCO.23.01435. Online ahead of print.

 

Systemic Therapy for Small-Cell Lung Cancer: ASCO-Ontario Health (Cancer Care

Ontario) Guideline.

 

Khurshid H(1), Ismaila N(2), Bian J(3), Dabney R(4), Das M(5), Ellis P(6),

Feldman J(7), Hann C(8), Kulkarni S(9), Laskin J(10), Manochakian R(11), Mishra

DR(12), Preeshagul I(13), Reddy P(14), Saxena A(15), Weinberg F(16), Kalemkerian

GP(17).

 

Author information:

(1)Brown University, Providence, RI.

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

(3)Maine Health, South Portland, ME.

PURPOSE: To provide evidence-based recommendations to practicing clinicians on

the management of patients with small-cell lung cancer.

METHODS: An Expert Panel of medical oncology, thoracic surgery, radiation

oncology, pulmonary, community oncology, research methodology, and advocacy

experts were convened to conduct a literature search, which included systematic

reviews, meta-analyses, and randomized controlled trials published from 1990

through 2022. Outcomes of interest included response rates, overall survival,

disease-free survival or recurrence-free survival, and quality of life. Expert

Panel members used available evidence and informal consensus to develop

evidence-based guideline recommendations.

RESULTS: The literature search identified 95 relevant studies to inform the

evidence base for this guideline.

RECOMMENDATIONS: Evidence-based recommendations were developed to address

systemic therapy options, timing of therapy, treatment in patients who are older

or with poor performance status, role of biomarkers, and use of

myeloid-supporting agents in patients with small-cell lung cancer.Additional

information is available at www.asco.org/thoracic-cancer-guidelines.

 

DOI: 10.1200/JCO.23.01435

PMID: 37820295

 

13. Nat Commun. 2023 Oct 4;14(1):6182. doi: 10.1038/s41467-023-41937-9.

 

Global burden of disease due to rifampicin-resistant tuberculosis: a

mathematical modeling analysis.

 

Menzies NA(1)(2), Allwood BW(#)(3), Dean AS(#)(4), Dodd PJ(#)(5), Houben

RMGJ(#)(6)(7), James LP(#)(8)(9), Knight GM(#)(10), Meghji J(#)(11), Nguyen

LN(#)(4), Rachow A(#)(12)(13)(14), Schumacher SG(#)(4), Mirzayev F(4), Cohen

T(15).

 

Author information:

(1)Department of Global Health and Population, Harvard T. H. Chan School of

Public Health, Boston, USA. nmenzies@hsph.harvard.edu.

(2)Center for Health Decision Science, Harvard T. H. Chan School of Public

Health, Boston, USA. nmenzies@hsph.harvard.edu.

(3)Division of Pulmonology, Department of Medicine, Stellenbosch University &

Tygerberg Hospital, Cape Town, South Africa.

In 2020, almost half a million individuals developed rifampicin-resistant

tuberculosis (RR-TB). We estimated the global burden of RR-TB over the lifetime

of affected individuals. We synthesized data on incidence, case detection, and

treatment outcomes in 192 countries (99.99% of global tuberculosis). Using a

mathematical model, we projected disability-adjusted life years (DALYs) over the

lifetime for individuals developing tuberculosis in 2020 stratified by country,

age, sex, HIV, and rifampicin resistance. Here we show that incident RR-TB in

2020 was responsible for an estimated 6.9 (95% uncertainty interval: 5.5, 8.5)

million DALYs, 44% (31, 54) of which accrued among TB survivors. We estimated an

average of 17 (14, 21) DALYs per person developing RR-TB, 34% (12, 56) greater

than for rifampicin-susceptible tuberculosis. RR-TB burden per 100,000 was

highest in former Soviet Union countries and southern African countries. While

RR-TB causes substantial short-term morbidity and mortality, nearly half of the

overall disease burden of RR-TB accrues among tuberculosis survivors. The

substantial long-term health impacts among those surviving RR-TB disease suggest

the need for improved post-treatment care and further justify increased health

expenditures to prevent RR-TB transmission.

 

© 2023. Springer Nature Limited.

 

DOI: 10.1038/s41467-023-41937-9

PMCID: PMC10550952

PMID: 37794037 [Indexed for MEDLINE]

 

14. J Clin Oncol. 2023 Oct 3:JCO2300910. doi: 10.1200/JCO.23.00910. Online ahead of print.

 

Canakinumab as Adjuvant Therapy in Patients With Completely Resected

Non-Small-Cell Lung Cancer: Results From the CANOPY-A Double-Blind, Randomized

Clinical Trial.

 

Garon EB(1), Lu S(2), Goto Y(3), De Marchi P(4), Paz-Ares L(5), Spigel DR(6),

Thomas M(7), Yang JC(8), Ardizzoni A(9), Barlesi F(10)(11), Orlov S(12),

Yoshioka H(13), Mountzios G(14), Khanna S(15), Bossen C(16), Carbini M(16),

Turri S(15), Myers A(17), Cho BC(18).

 

Author information:

(1)David Geffen School of Medicine at UCLA/TRIO-US/TRIO-Global Network, Los

Angeles, CA.

(2)Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University,

Shanghai, China.

(3)National Cancer Center, Tokyo, Japan.

PURPOSE: Effective treatments for resectable non-small-cell lung cancer (NSCLC)

are limited and relapse rates are high. The interleukin (IL)-1β pathway has been

linked with tumor development and progression, including in the Canakinumab

Anti-Inflammatory Thrombosis Outcomes cardiovascular study in which IL-1β

pathway inhibition with canakinumab reduced lung cancer incidence and mortality

in an exploratory analysis.

METHODS: CANOPY-A (ClinicalTrials.gov identifier: NCT03447769) is a phase III,

randomized, double-blind, multicenter study of canakinumab versus placebo for

adult patients with stage II-IIIA or IIIB (T >5 cm, N2-positives II-IIIB;

American Joint Committee on Cancer/Union for International Cancer Control

version 8), completely resected NSCLC who had received adjuvant cisplatin-based

chemotherapy. The primary end point was disease-free survival (DFS) and the key

secondary end point was overall survival (OS).

RESULTS: In total, 1,382 patients were randomized to 200 mg canakinumab (n =

693) or placebo (n = 689) once every 3 weeks for 18 cycles. Grade ≥3 adverse

events (AEs) were reported in 20.8% and 19.6% of patients receiving canakinumab

and placebo, respectively; AEs led to discontinuation in 4.3% and 4.1% of

patients in these groups, respectively. This study did not meet its primary end

point. Median DFS was 35.0 months (canakinumab arm) and 29.7 months (placebo

arm; hazard ratio, 0.94; 95% CI, 0.78 to 1.14; one-sided P = .258). DFS subgroup

analyses did not show any meaningful differences between arms. As expected,

because of canakinumab-driven IL-1β pathway inhibition, C-reactive protein and

IL-6 levels decreased in the canakinumab arm versus placebo arm, but had no

correlation with differential clinical outcomes. OS was not formally tested as

DFS was not statistically significant.

CONCLUSION: CANOPY-A did not show a DFS benefit of adding canakinumab after

surgery and adjuvant cisplatin-based chemotherapy in patients with resected,

stage II-III NSCLC. No new safety signals were identified with canakinumab.

 

DOI: 10.1200/JCO.23.00910

PMID: 37788412

 

15. PLoS Med. 2023 Oct 3;20(10):e1004287. doi: 10.1371/journal.pmed.1004287.

eCollection 2023 Oct.

 

Assessing eligibility for lung cancer screening using parsimonious ensemble

machine learning models: A development and validation study.

 

Callender T(1), Imrie F(2), Cebere B(3), Pashayan N(4), Navani N(1), van der

Schaar M(3)(5)(6), Janes SM(1).

 

Author information:

(1)Department of Respiratory Medicine, University College London, London, United

Kingdom.

(2)Department of Electrical and Computer Engineering, University of California,

Los Angeles, California, United States of America.

(3)Department of Applied Mathematics and Theoretical Physics, University of

Cambridge, Cambridge, United Kingdom.

(4)Department of Applied Health Research, University College London, London,

United Kingdom.

(5)Cambridge Centre for AI in Medicine, University of Cambridge, Cambridge,

United Kingdom.

(6)Alan Turing Institute, London, United Kingdom.

 

BACKGROUND: Risk-based screening for lung cancer is currently being considered

in several countries; however, the optimal approach to determine eligibility

remains unclear. Ensemble machine learning could support the development of

highly parsimonious prediction models that maintain the performance of more

complex models while maximising simplicity and generalisability, supporting the

widespread adoption of personalised screening. In this work, we aimed to develop

and validate ensemble machine learning models to determine eligibility for

risk-based lung cancer screening.

METHODS AND FINDINGS: For model development, we used data from 216,714

ever-smokers recruited between 2006 and 2010 to the UK Biobank prospective

cohort and 26,616 high-risk ever-smokers recruited between 2002 and 2004 to the

control arm of the US National Lung Screening (NLST) randomised controlled

trial. The NLST trial randomised high-risk smokers from 33 US centres with at

least a 30 pack-year smoking history and fewer than 15 quit-years to annual CT

or chest radiography screening for lung cancer. We externally validated our

models among 49,593 participants in the chest radiography arm and all 80,659

ever-smoking participants in the US Prostate, Lung, Colorectal and Ovarian

(PLCO) Screening Trial. The PLCO trial, recruiting from 1993 to 2001, analysed

the impact of chest radiography or no chest radiography for lung cancer

screening. We primarily validated in the PLCO chest radiography arm such that we

could benchmark against comparator models developed within the PLCO control arm.

Models were developed to predict the risk of 2 outcomes within 5 years from

baseline: diagnosis of lung cancer and death from lung cancer. We assessed model

discrimination (area under the receiver operating curve, AUC), calibration

(calibration curves and expected/observed ratio), overall performance (Brier

scores), and net benefit with decision curve analysis. Models predicting lung

cancer death (UCL-D) and incidence (UCL-I) using 3 variables-age, smoking

duration, and pack-years-achieved or exceeded parity in discrimination, overall

performance, and net benefit with comparators currently in use, despite

requiring only one-quarter of the predictors. In external validation in the PLCO

trial, UCL-D had an AUC of 0.803 (95% CI: 0.783, 0.824) and was well calibrated

with an expected/observed (E/O) ratio of 1.05 (95% CI: 0.95, 1.19). UCL-I had an

AUC of 0.787 (95% CI: 0.771, 0.802), an E/O ratio of 1.0 (95% CI: 0.92, 1.07).

The sensitivity of UCL-D was 85.5% and UCL-I was 83.9%, at 5-year risk

thresholds of 0.68% and 1.17%, respectively, 7.9% and 6.2% higher than the

USPSTF-2021 criteria at the same specificity. The main limitation of this study

is that the models have not been validated outside of UK and US cohorts.

CONCLUSIONS: We present parsimonious ensemble machine learning models to predict

the risk of lung cancer in ever-smokers, demonstrating a novel approach that

could simplify the implementation of risk-based lung cancer screening in

multiple settings.

 

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

under the terms of the Creative Commons Attribution License, which permits

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

original author and source are credited.

 

DOI: 10.1371/journal.pmed.1004287

PMCID: PMC10547178

PMID: 37788223 [Indexed for MEDLINE]

 

16. J Clin Invest. 2023 Oct 2;133(19):e173156. doi: 10.1172/JCI173156.

 

The Mycobacterium tuberculosis genome at 25 years: lessons and lingering

questions.

 

Koleske BN(1), Jacobs WR Jr(2), Bishai WR(1).

 

Author information:

(1)Center for Tuberculosis Research, Department of Medicine, Johns Hopkins

School of Medicine, Baltimore, Maryland, USA.

(2)Department of Microbiology and Immunology, Albert Einstein College of

Medicine, Bronx, New York, USA.

 

First achieved in 1998 by Cole et al., the complete genome sequence of

Mycobacterium tuberculosis continues to provide an invaluable resource to

understand tuberculosis (TB), the leading cause of global infectious disease

mortality. At the 25-year anniversary of this accomplishment, we describe how

insights gleaned from the M. tuberculosis genome have led to vital tools for TB

research, epidemiology, and clinical practice. The increasing accessibility of

whole-genome sequencing across research and clinical settings has improved our

ability to predict antibacterial susceptibility, to track epidemics at the level

of individual outbreaks and wider historical trends, to query the efficacy of

the bacille Calmette-Guérin (BCG) vaccine, and to uncover targets for novel

antitubercular therapeutics. Likewise, we discuss several recent efforts to

extract further discoveries from this powerful resource.

 

DOI: 10.1172/JCI173156

PMCID: PMC10541200

PMID: 37781921 [Indexed for MEDLINE]

 

17. Nat Rev Clin Oncol. 2023 Oct;20(10):716-732. doi: 10.1038/s41571-023-00808-4. Epub 2023 Aug 17.

 

Emerging therapeutics and evolving assessment criteria for intracranial

metastases in patients with oncogene-driven non-small-cell lung cancer.

 

Pan K(1), Concannon K(1), Li J(2), Zhang J(3), Heymach JV(3), Le X(4).

 

Author information:

(1)Department of Cancer Medicine, University of Texas MD Anderson Cancer Center,

Houston, TX, USA.

(2)Department of Radiation Oncology, University of Texas MD Anderson Cancer

Center, Houston, TX, USA.

(3)Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD

Anderson Cancer Center, Houston, TX, USA.

(4)Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD

Anderson Cancer Center, Houston, TX, USA. xle1@mdanderson.org.

 

The improved survival outcomes of patients with non-small-cell lung cancer

(NSCLC), largely owing to the improved control of systemic disease provided by

immune-checkpoint inhibitors and novel targeted therapies, have highlighted the

challenges posed by central nervous system (CNS) metastases as a devastating yet

common complication, with up to 50% of patients developing such lesions during

the course of the disease. Early-generation tyrosine-kinase inhibitors (TKIs)

often provide robust systemic disease control in patients with oncogene-driven

NSCLCs, although these agents are usually unable to accumulate to

therapeutically relevant concentrations in the CNS owing to an inability to

cross the blood-brain barrier. However, the past few years have seen a paradigm

shift with the emergence of several novel or later-generation TKIs with improved

CNS penetrance. Such agents have promising levels of activity against brain

metastases, as demonstrated by data from preclinical and clinical studies. In

this Review, we describe current preclinical and clinical evidence of the

intracranial activity of TKIs targeting various oncogenic drivers in patients

with NSCLC, with a focus on newer agents with enhanced CNS penetration,

leptomeningeal disease and the need for intrathecal treatment options. We also

discuss evolving assessment criteria and regulatory considerations for future

clinical investigations.

 

© 2023. Springer Nature Limited.

 

DOI: 10.1038/s41571-023-00808-4

PMID: 37592034 [Indexed for MEDLINE]

 

18. J Clin Oncol. 2023 Oct 1;41(28):4472-4477. doi: 10.1200/JCO.23.00046. Epub 2023 Jun 16.

 

Intracranial Efficacy of Adagrasib in Patients From the KRYSTAL-1 Trial With

KRAS(G12C)-Mutated Non-Small-Cell Lung Cancer Who Have Untreated CNS Metastases.

 

Negrao MV(1), Spira AI(2)(3)(4), Heist RS(5), Jänne PA(6), Pacheco JM(7), Weiss

J(8), Gadgeel SM(9), Velastegui K(10), Yang W(10), Der-Torossian H(10),

Christensen JG(10), Sabari JK(11).

 

Author information:

(1)Department of Thoracic/Head & Neck Medical Oncology, MD Anderson Cancer

Center, University of Texas, Houston, TX.

(2)Virginia Cancer Specialists, Fairfax, VA.

(3)US Oncology Research, The Woodlands, TX.

Clinical trials frequently include multiple end points that mature at different

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

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

available. Clinical Trial Updates provide an opportunity to disseminate

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

primary end point has already been reported.Patients with Kirsten rat sarcoma

viral oncogene homolog (KRAS)-mutated non-small-cell lung cancer (NSCLC) and

untreated CNS metastases have a worse prognosis than similar patients without

KRAS mutations. Adagrasib has previously demonstrated CNS penetration

preclinically and cerebral spinal fluid penetration clinically. We evaluated

adagrasib in patients with KRASG12C-mutated NSCLC and untreated CNS metastases

from the KRYSTAL-1 trial (ClinicalTrials.gov identifier: NCT03785249; phase Ib

cohort), in which adagrasib 600 mg was administered orally, twice daily. Study

outcomes included the safety and clinical activity (intracranial [IC] and

systemic) by blinded independent central review. Twenty-five patients with

KRASG12C-mutated NSCLC and untreated CNS metastases were enrolled and evaluated

(median follow-up, 13.7 months); 19 patients were radiographically evaluable for

IC activity. Safety was consistent with previous reports of adagrasib, with

grade 3 treatment-related adverse events (TRAEs) in 10 patients (40%) and one

grade 4 (4%) and no grade 5 TRAEs. The most common CNS-specific TRAEs included

dysgeusia (24%) and dizziness (20%). Adagrasib demonstrated an IC objective

response rate of 42%, disease control rate of 90%, progression-free survival of

5.4 months, and median overall survival of 11.4 months. Adagrasib is the first

KRASG12C inhibitor to prospectively demonstrate IC activity in patients with

KRASG12C-mutated NSCLC and untreated CNS metastases, supporting further

investigation in this population.

 

DOI: 10.1200/JCO.23.00046

PMCID: PMC10553074

PMID: 37327468 [Indexed for MEDLINE]

 

19. J Clin Oncol. 2023 Oct 10;41(29):4678-4687. doi: 10.1200/JCO.23.00059. Epub 2023 Jun 16.

 

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

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

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

 

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

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

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

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

 

Author information:

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

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

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

Comment in

    doi: 10.1200/JCO.23.01207.

 

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

the safety, tolerability, and antitumor activity of datopotamab deruxtecan

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

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

cancer (NSCLC).

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

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

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

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

survival, and pharmacokinetics.

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

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

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

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

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

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

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

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

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

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

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

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

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

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

CONCLUSION: Promising antitumor activity and a manageable safety profile were

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

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

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

 

DOI: 10.1200/JCO.23.00059

PMCID: PMC10564307

PMID: 37327461 [Indexed for MEDLINE]

 

 

 


上一篇: No.11

下一篇: No.9