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 >>2022年

高分SCI文摘

2022年

No.6

发布时间:2022-08-19 浏览次数:
字号: + - 14

Filters applied: from 2022/6/1 - 2022/6/30

1. Science. 2022 Jun 24;376(6600):eabh2841. doi: 10.1126/science.abh2841. Epub 2022

Jun 24.

 

Tumor necrosis factor induces pathogenic mitochondrial ROS in tuberculosis

through reverse electron transport.

 

Roca FJ(1), Whitworth LJ(1)(2), Prag HA(3), Murphy MP(1)(3), Ramakrishnan

L(1)(2).

 

Author information:

(1)Molecular Immunity Unit, Cambridge Institute of Therapeutic Immunology and

Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge

CB2 0AW, UK.

(2)MRC Laboratory of Molecular Biology, Cambridge CB2 0QH, UK.

(3)MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY,

UK.

 

Tumor necrosis factor (TNF) is a critical host resistance factor against

tuberculosis. However, excess TNF produces susceptibility by increasing

mitochondrial reactive oxygen species (mROS), which initiate a signaling cascade

to cause pathogenic necrosis of mycobacterium-infected macrophages. In

zebrafish, we identified the mechanism of TNF-induced mROS in tuberculosis.

Excess TNF in mycobacterium-infected macrophages elevates mROS production by

reverse electron transport (RET) through complex I. TNF-activated cellular

glutamine uptake leads to an increased concentration of succinate, a Krebs cycle

intermediate. Oxidation of this elevated succinate by complex II drives RET,

thereby generating the mROS superoxide at complex I. The complex I inhibitor

metformin, a widely used antidiabetic drug, prevents TNF-induced mROS and

necrosis of Mycobacterium tuberculosis-infected zebrafish and human macrophages;

metformin may therefore be useful in tuberculosis therapy.

 

DOI: 10.1126/science.abh2841

PMCID: PMC7612974

PMID: 35737799 [Indexed for MEDLINE]

 

2. CA Cancer J Clin. 2022 Jun 23. doi: 10.3322/caac.21731. Online ahead of print.

 

Cancer treatment and survivorship statistics, 2022.

 

Miller KD(1), Nogueira L(2), Devasia T(3), Mariotto AB(4), Yabroff KR(2), Jemal

A(5), Kramer J(6), Siegel RL(1).

 

Author information:

(1)Surveillance Research, American Cancer Society, Atlanta, Georgia.

(2)Health Services Research, American Cancer Society, Atlanta, Georgia.

(3)Data Analytics Branch, Surveillance Research Program, Division of Cancer

Control and Population Sciences, National Cancer Institute, Bethesda, Maryland.

(4)Surveillance Research Program, Division of Cancer Control and Population

Sciences, National Cancer Institute, Bethesda, Maryland.

(5)Surveillance and Health Equity Science, American Cancer Society, Atlanta,

Georgia.

(6)Department of Hematology and Medical Oncology, Emory University, Atlanta,

Georgia.

 

The number of cancer survivors continues to increase in the United States due to

the growth and aging of the population as well as advances in early detection

and treatment. To assist the public health community in better serving these

individuals, the American Cancer Society and the National Cancer Institute

collaborate triennially to estimate cancer prevalence in the United States using

incidence and survival data from the Surveillance, Epidemiology, and End Results

cancer registries, vital statistics from the Centers for Disease Control and

Prevention's National Center for Health Statistics, and population projections

from the US Census Bureau. Current treatment patterns based on information in

the National Cancer Database are presented for the most prevalent cancer types

by race, and cancer-related and treatment-related side-effects are also briefly

described. More than 18 million Americans (8.3 million males and 9.7 million

females) with a history of cancer were alive on January 1, 2022. The 3 most

prevalent cancers are prostate (3,523,230), melanoma of the skin (760,640), and

colon and rectum (726,450) among males and breast (4,055,770), uterine corpus

(891,560), and thyroid (823,800) among females. More than one-half (53%) of

survivors were diagnosed within the past 10 years, and two-thirds (67%) were

aged 65 years or older. One of the largest racial disparities in treatment is

for rectal cancer, for which 41% of Black patients with stage I disease receive

proctectomy or proctocolectomy compared to 66% of White patients. Surgical

receipt is also substantially lower among Black patients with non-small cell

lung cancer, 49% for stages I-II and 16% for stage III versus 55% and 22% for

White patients, respectively. These treatment disparities are exacerbated by the

fact that Black patients continue to be less likely to be diagnosed with stage I

disease than White patients for most cancers, with some of the largest

disparities for female breast (53% vs 68%) and endometrial (59% vs 73%).

Although there are a growing number of tools that can assist patients,

caregivers, and clinicians in navigating the various phases of cancer

survivorship, further evidence-based strategies and equitable access to

available resources are needed to mitigate disparities for communities of color

and optimize care for people with a history of cancer. CA Cancer J Clin.

2022;72:000-000.

 

© 2022 The Authors. CA: A Cancer Journal for Clinicians published by Wiley

Periodicals LLC on behalf of American Cancer Society.

 

DOI: 10.3322/caac.21731

PMID: 35736631

 

3. N Engl J Med. 2022 Jul 14;387(2):120-131. doi: 10.1056/NEJMoa2204619. Epub 2022

Jun 3.

 

Adagrasib in Non-Small-Cell Lung Cancer Harboring a KRAS(G12C) Mutation.

 

Jänne PA(1), Riely GJ(1), Gadgeel SM(1), Heist RS(1), Ou SI(1), Pacheco JM(1),

Johnson ML(1), Sabari JK(1), Leventakos K(1), Yau E(1), Bazhenova L(1), Negrao

MV(1), Pennell NA(1), Zhang J(1), Anderes K(1), Der-Torossian H(1), Kheoh T(1),

Velastegui K(1), Yan X(1), Christensen JG(1), Chao RC(1), Spira AI(1).

 

Author information:

(1)From the Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute

(P.A.J.), and Massachusetts General Hospital (R.S.H.) - both in Boston; the

Thoracic Oncology Service, Division of Solid Tumor Oncology, Department of

Medicine, Memorial Sloan Kettering Cancer Center, and Weill Cornell Medical

College (G.J.R.), and Perlmutter Cancer Center, New York University Langone

Health (J.K.S.), New York, and the Department of Medicine, Roswell Park

Comprehensive Cancer Center, Buffalo (E.Y.) - all in New York; the Henry Ford

Cancer Institute, Detroit (S.M.G.); the University of California Irvine School

of Medicine, Chao Family Comprehensive Cancer Center, Orange (S.-H.I.O.),…

 

Comment in

    N Engl J Med. 2022 Jul 14;387(2):180-183.

    N Engl J Med. 2022 Jul 14;387(2):184-186.

 

BACKGROUND: Adagrasib, a KRASG12C inhibitor, irreversibly and selectively binds

KRASG12C, locking it in its inactive state. Adagrasib showed clinical activity

and had an acceptable adverse-event profile in the phase 1-1b part of the

KRYSTAL-1 phase 1-2 study.

METHODS: In a registrational phase 2 cohort, we evaluated adagrasib (600 mg

orally twice daily) in patients with KRASG12C -mutated non-small-cell lung

cancer (NSCLC) previously treated with platinum-based chemotherapy and

anti-programmed death 1 or programmed death ligand 1 therapy. The primary end

point was objective response assessed by blinded independent central review.

Secondary end points included the duration of response, progression-free

survival, overall survival, and safety.

RESULTS: As of October 15, 2021, a total of 116 patients with KRASG12C -mutated

NSCLC had been treated (median follow-up, 12.9 months); 98.3% had previously

received both chemotherapy and immunotherapy. Of 112 patients with measurable

disease at baseline, 48 (42.9%) had a confirmed objective response. The median

duration of response was 8.5 months (95% confidence interval [CI], 6.2 to 13.8),

and the median progression-free survival was 6.5 months (95% CI, 4.7 to 8.4). As

of January 15, 2022 (median follow-up, 15.6 months), the median overall survival

was 12.6 months (95% CI, 9.2 to 19.2). Among 33 patients with previously

treated, stable central nervous system metastases, the intracranial confirmed

objective response rate was 33.3% (95% CI, 18.0 to 51.8). Treatment-related

adverse events occurred in 97.4% of the patients - grade 1 or 2 in 52.6% and

grade 3 or higher in 44.8% (including two grade 5 events) - and resulted in drug

discontinuation in 6.9% of patients.

CONCLUSIONS: In patients with previously treated KRASG12C -mutated NSCLC,

adagrasib showed clinical efficacy without new safety signals. (Funded by Mirati

Therapeutics; ClinicalTrials.gov number, NCT03785249.).

 

Copyright © 2022 Massachusetts Medical Society.

 

DOI: 10.1056/NEJMoa2204619

PMID: 35658005 [Indexed for MEDLINE]

 

4. Cancer Cell. 2022 Jun 13;40(6):603-605. doi: 10.1016/j.ccell.2022.05.010. Epub

2022 Jun 2.

 

When immunotherapy meets surgery in non-small cell lung cancer.

 

Herbst RS(1), Wang M(2), Chen L(3).

 

Author information:

(1)Yale Comprehensive Cancer Center, Yale University School of Medicine, New

Haven, CT 06520-8028, USA. Electronic address: roy.herbst@yale.edu.

(2)Yale Comprehensive Cancer Center, Yale University School of Medicine, New

Haven, CT 06520-8028, USA.

(3)Yale Comprehensive Cancer Center, Yale University School of Medicine, New

Haven, CT 06520-8028, USA; Department of Immunobiology, Yale University School

of Medicine, New Haven, CT 06520, USA.

 

Comment on

    N Engl J Med. 2022 May 26;386(21):1973-1985.

 

The results of the most recent Checkmate-816 trial in The New England Journal of

Medicine using combination neoadjuvant immunotherapy with platinum-based

chemotherapy in resectable non-small cell lung cancer demonstrate the

effectiveness of neoadjuvant immunotherapy and provide further support that

biology and personalized therapy represent the foundation of lung cancer

treatment.

 

Copyright © 2022 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2022.05.010

PMID: 35660136 [Indexed for MEDLINE]

 

5. Lancet Oncol. 2022 Jun;23(6):781-792. doi: 10.1016/S1470-2045(22)00226-1. Epub

2022 May 13.

 

Tiragolumab plus atezolizumab versus placebo plus atezolizumab as a first-line

treatment for PD-L1-selected non-small-cell lung cancer (CITYSCAPE): primary and

follow-up analyses of a randomised, double-blind, phase 2 study.

 

Cho BC(1), Abreu DR(2), Hussein M(3), Cobo M(4), Patel AJ(5), Secen N(6), Lee

KH(7), Massuti B(8), Hiret S(9), Yang JCH(10), Barlesi F(11), Lee DH(12), Ares

LP(13), Hsieh RW(14), Patil NS(14), Twomey P(14), Yang X(14), Meng R(14),

Johnson ML(15).

 

Author information:

(1)Yonsei Cancer Centre, Yonsei University College of Medicine, Seoul, South

Korea. Electronic address: cbc1971@yuhs.ac.

(2)Hospital Universitario Insular de Gran Canaria, Las Palmas, Spain.

(3)SCRI Florida Cancer Specialists, Leesburg, FL, USA.

Comment in

    Nat Rev Clin Oncol. 2022 Jul;19(7):428.

    Lancet Oncol. 2022 Jun;23(6):695-697.

 

BACKGROUND: Targeted inhibition of the PD-L1-PD-1 pathway might be further

amplified through combination of PD-1 or PD-L1 inhibitors with novel anti-TIGIT

inhibitory immune checkpoint agents, such as tiragolumab. In the CITYSCAPE

trial, we aimed to assess the preliminary efficacy and safety of tiragolumab

plus atezolizumab (anti-PD-L1) therapy as first-line treatment for

non-small-cell lung cancer (NSCLC).

METHODS: CITYSCAPE is a phase 2, randomised, double-blind, placebo-controlled

trial. Patients with chemotherapy-naive, PD-L1-positive (defined as a tumour

proportion score of ≥1% by 22C3 immunohistochemistry pharmDx assay; Dako,

Agilent Technologies, Santa Clara, CA, USA) recurrent or metastatic NSCLC with

measurable disease, Eastern Cooperative Oncology Group performance status of 0

or 1, and no EGFR or ALK alterations were enrolled from 41 clinics in Europe,

Asia, and the USA. Patients were randomly assigned (1:1), via an interactive

voice or web-based response system, to receive tiragolumab (600 mg) plus

atezolizumab (1200 mg) or placebo plus atezolizumab intravenously once every 3

weeks. Investigators and patients were masked to treatment assignment. The

co-primary endpoints were investigator-assessed objective response rate and

progression-free survival as per Response Evaluation Criteria in Solid Tumors

version 1.1 in the intention-to-treat population, analysed after approximately

80 progression-free survival events had been observed in the primary population.

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

drug. This trial is registered with ClinicalTrials.gov, NCT03563716, and is

ongoing.

FINDINGS: Patients were enrolled between Aug 10, 2018, and March 20, 2019. At

data cutoff for the primary analysis (June 30, 2019), 135 of 275 patients

assessed for eligibility were randomly assigned to receive tiragolumab plus

atezolizumab (67 [50%]) or placebo plus atezolizumab (68 [50%]). In this primary

analysis, after a median follow-up of 5·9 months (4·6-7·6, in the

intention-to-treat population, 21 patients (31·3% [95% CI 19·5-43·2]) in the

tiragolumab plus atezolizumab group versus 11 patients (16·2% [6·7-25·7]) in the

placebo plus atezolizumab group had an objective response (p=0·031). Median

progression-free survival was 5·4 months (95% CI 4·2-not estimable) in the

tiragolumab plus atezolizumab group versus 3·6 months (2·7-4·4) in the placebo

plus atezolizumab group (stratified hazard ratio 0·57 [95% CI 0·37-0·90],

p=0·015). 14 (21%) patients receiving tiragolumab plus atezolizumab and 12 (18%)

patients receiving placebo plus atezolizumab had serious treatment-related

adverse events. The most frequently reported grade 3 or worse treatment-related

adverse event was lipase increase (in six [9%] patients in the tiragolumab plus

atezolizumab group vs two [3%] in the placebo plus atezolizumab group). Two

treatment-related deaths (of pyrexia and infection) occurred in the tiragolumab

plus atezolizumab group.

INTERPRETATION: Tiragolumab plus atezolizumab showed a clinically meaningful

improvement in objective response rate and progression-free survival compared

with placebo plus atezolizumab in patients with chemotherapy-naive,

PD-L1-positive, recurrent or metastatic NSCLC. Tiragolumab plus atezolizumab was

well tolerated, with a safety profile generally similar to that of atezolizumab

alone. These findings demonstrate that tiragolumab plus atezolizumab is a

promising immunotherapy combination for the treatment of previously untreated,

locally advanced unresectable or metastatic NSCLC.

FUNDING: F Hoffmann-La Roche and Genentech.

 

Copyright © 2022 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1470-2045(22)00226-1

PMID: 35576957 [Indexed for MEDLINE]

 

6. Lancet Oncol. 2022 Jun;23(6):739-747. doi: 10.1016/S1470-2045(22)00224-8. Epub

2022 May 13.

 

Adebrelimab or placebo plus carboplatin and etoposide as first-line treatment

for extensive-stage small-cell lung cancer (CAPSTONE-1): a multicentre,

randomised, double-blind, placebo-controlled, phase 3 trial.

 

Wang J(1), Zhou C(2), Yao W(3), Wang Q(4), Min X(5), Chen G(6), Xu X(7), Li

X(8), Xu F(9), Fang Y(10), Yang R(11), Yu G(12), Gong Y(13), Zhao J(14), Fan

Y(15), Liu Q(16), Cao L(17), Yao Y(18), Liu Y(19), Li X(20), Wu J(21), He Z(22),

Lu K(23), Jiang L(24), Hu C(25), Zhao W(26), Zhang B(27), Shi W(27), Zhang

X(27), Cheng Y(28); CAPSTONE-1 Study Group.

 

Collaborators: Cheng Y, Wang J, Zhou C, Yao W, Wang Q, Min X, Chen G, Xu X, Li

X, Xu F, Fang Y, Yang R, Yu G, Gong Y, Zhao J, Fan Y, Liu Q, Cao L, Yao Y, Liu

Y, Li X, Wu J, He Z, Lu K, Jiang L, Hu C, Zhao W, Yu H, Zhao J, Wu G, Huang D,

Chen C, Ding C, Zhang B, Wang X, Luo H, Li B, Zhang S, Lu H, Shi M, Chen X, Guo

Y, Liu H, Liu J, Gao H, Hu S, Hong Q, Li Q, Zhang B, Shi W, Zhang X.

 

Author information:

(1)Department of Medical Oncology, National Cancer Center, National Clinical

Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences

& Peking Union Medical College, Beijing, China.

(2)Department of Medical Oncology, Shanghai Pulmonary Hospital, Shanghai, China.

(3)Department of Thoracic Oncology, Sichuan Cancer Hospital & Institute,

Chengdu, China.

Comment in

    Lancet Oncol. 2022 Jun;23(6):692-693.

 

BACKGROUND: Extensive-stage small-cell lung cancer (ES-SCLC) is associated with

poor prognosis and treatment options are scarce. Immunotherapy has shown robust

clinical activity in ES-SCLC in previous phase 3 trials. We aimed to assess the

efficacy and safety of adebrelimab (SHR-1316), a novel anti-PD-L1 antibody, with

standard chemotherapy as a first-line treatment for ES-SCLC.

METHODS: The CAPSTONE-1 study was a randomised, double-blind,

placebo-controlled, phase 3 trial, done in 47 tertiary hospitals in China. Key

inclusion criteria were patients aged 18-75 years, with previously untreated

histologically or cytologically confirmed ES-SCLC and an Eastern Cooperative

Oncology Group (ECOG) performance status of 0-1. Eligible patients were randomly

assigned (1:1) to receive four to six cycles of carboplatin (area under the

curve of 5 mg/mL per min, day 1 of each cycle) and etoposide (100 mg/m2 of

body-surface area, on days 1-3 of each cycle) with either adebrelimab (20 mg/kg,

day 1 of each cycle) or matching placebo, followed by maintenance therapy with

adebrelimab or placebo. All treatments were given intravenously in 21-day

cycles. Randomisation was done using a centralised interactive web response

system with a block size of four, stratified by liver metastases, brain

metastases, and lactate dehydrogenase concentration. The primary endpoint was

overall survival in patients who received at least one dose of study medication.

Safety was analysed in the as-treated population. This study is complete and

registered with ClinicalTrials.gov, NCT03711305.

FINDINGS: Between Dec 26, 2018, and Sept 4, 2020, 462 eligible patients were

enrolled and randomly assigned: 230 (50%) patients received adebrelimab plus

chemotherapy (adebrelimab group) and 232 (50%) patients received placebo plus

chemotherapy (placebo group). At data cutoff (Oct 8, 2021), median follow-up was

13·5 months (IQR 8·9-20·1). Median overall survival was significantly improved

in the adebrelimab group (median 15·3 months [95% CI 13·2-17·5]) compared with

the placebo group (12·8 months [11·3-13·7]; hazard ratio 0·72 [95% CI

0·58-0·90]; one-sided p=0·0017). The most common treatment-related grade 3 or 4

adverse events were decreased neutrophil count (174 [76%] patients in the

adebrelimab group and 175 [75%] patients in the placebo group), decreased white

blood cell count (106 [46%] and 88 [38%]), decreased platelet count (88 [38%]

and 78 [34%]), and anaemia (64 [28%] and 66 [28%]). Treatment-related serious

adverse events occurred in 89 (39%) patients in the adebrelimab group and 66

(28%) patients in the placebo group. Four treatment-related deaths were

reported: two each in the adebrelimab group (respiratory failure and

interstitial lung disease and pneumonia) and placebo group (multiple organ

dysfunction and unknown cause of death).

INTERPRETATION: Adding adebrelimab to chemotherapy significantly improved

overall survival with an acceptable safety profile in patients with ES-SCLC,

supporting this combination as a new first-line treatment option for this

population.

FUNDING: Jiangsu Hengrui Pharmaceuticals.

 

Copyright © 2022 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1470-2045(22)00224-8

PMID: 35576956 [Indexed for MEDLINE]

 

7. Lancet Infect Dis. 2022 Jun 27:S1473-3099(22)00222-5. doi:

10.1016/S1473-3099(22)00222-5. Online ahead of print.

 

Safety and immunogenicity of VPM1002 versus BCG in South African newborn babies:

a randomised, phase 2 non-inferiority double-blind controlled trial.

 

Cotton MF(1), Madhi SA(2), Luabeya AK(3), Tameris M(3), Hesseling AC(4), Shenje

J(3), Schoeman E(3), Hatherill M(5), Desai S(6), Kapse D(6), Brückner S(7), Koen

A(2), Jose L(2), Moultrie A(2), Bhikha S(2), Walzl G(8), Gutschmidt A(8), Kotze

LA(8), Allies DL(8), Loxton AG(8), Shaligram U(6), Abraham M(9), Johnstone

H(10), Grode L(7), Kaufmann SHE(11), Kulkarni PS(12).

 

Author information:

(1)Tygerberg Academic Hospital, Parow Valley, South Africa.

(2)South African Medical Research Council Vaccines and Infectious Diseases

Analytical Research Unit, Faculty of Health Sciences, Johannesburg, South

Africa; Department of Science/National Research Foundation-Vaccine Preventable

Diseases, Johannesburg, South Africa; University of the Witwatersrand,

Johannesburg, South Africa.

(3)South African Tuberculosis Vaccine Initiative (SATVI), Institute of

Infectious Disease and Molecular Medicine and Department of Pathology,

University of Cape Town, Cape Town, South Africa.

BACKGROUND: Tuberculosis is a major public health problem worldwide.

Immunisation with Mycobacterium bovis BCG vaccine is partially effective in

infants, reducing the incidence of miliary and tuberculosis meningitis, but is

less effective against pulmonary tuberculosis. We aimed to compare safety and

immunogenicity of VPM1002-a recombinant BCG vaccine developed to address this

gap-with BCG in HIV exposed and HIV unexposed newborn babies.

METHODS: This double-blind, randomised, active controlled phase 2 study was

conducted at four health centres in South Africa. Eligible neonates were aged 12

days or younger with a birthweight of 2·5-4·2 kg, and could be HIV exposed

(seropositive mothers) or unexposed (seronegative mothers). Newborn babies were

excluded if they had acute or chronic illness, fever, hypothermia, sepsis,

cancer, or congenital malformation, or if they received blood products or

immunosuppressive therapy. Participants were excluded if their mothers (aged ≥18

years) had active tuberculosis disease, diabetes, a history of immunodeficiency

except for HIV, hepatitis B or syphilis seropositivity, received blood products

in the preceding 6 months, any acute infectious disease, or any suspected

substance abuse. Participants were randomly assigned to VPM1002 or BCG

vaccination in a 3:1 ratio, stratified by HIV status using the random number

generator function in SAS, using a block size of eight paticipants. The primary

outcome was non-inferiority (margin 15%) of VPM1002 to BCG vaccine in terms of

incidence of grade 3-4 adverse drug reactions or ipsilateral or generalised

lymphadenopathy of 10 mm or greater in diameter by 12 months. The primary

outcome was assessed in all vaccinated participants (safety population) at

regular follow-up visits until 12 months after vaccination. Secondary

immunogenicity outcomes were interferon-γ levels and percentages of

multifunctional CD4+ and CD8+ T cells among all lymphocytes across the 12 month

study period. The study was registered with ClinicalTrials.gov, NCT02391415.

FINDINGS: Between June 4, 2015 and Oct 16, 2017, 416 eligible newborn babies

were randomly assigned and received study vaccine. Seven (2%) of 312

participants in the VPM1002 group had a grade 3-4 vaccine-related adverse

reaction or lymphadenopathy of 10 mm or greater in diameter compared with 34

(33%) of 104 participants in the BCG group (risk difference -30·45% [95% CI

-39·61% to -21·28%]; pnon-inferiority<0·0001); VPM1002 was thus non-inferior to

BCG for the primary outcome. Incidence of severe injection site reactions was

lower with VPM1002 than BCG: scarring occurred in 65 (21%) participants in the

VPM1002 group versus 77 (74%) participants in the BCG group (p<0·0001);

ulceration occurred in one (<1%) versus 15 (14%; p<0·0001); and abscess

formation occurred in five (2%) versus 23 (22%; p<0·0001). Restimulated IFNγ

concentrations were lower in the VPM1002 group than the BCG group at week 6,

week 12, month 6, and month 12. The percentage of multifunctional CD4+ T cells

was higher in the VPM1002 group than the BCG group at day 14 but lower at week

6, week 12, month 6, and month 12. The percentage of multifunctional CD8+ T

cells was lower in the VPM1002 group than the BCG group at week 6, week 12, and

month 6, but did not differ at other timepoints.

INTERPRETATION: VPM1002 was less reactogenic than BCG and was not associated

with any serious safety concern. Both vaccines were immunogenic, although

responses were higher with the BCG vaccine. VPM1002 is currently being studied

for efficacy and safety in a multicentric phase 3 clinical trial in babies in

sub-Saharan Africa.

FUNDING: Serum Institute of India.

 

Copyright © 2022 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(22)00222-5

PMID: 35772447

 

8. Nat Commun. 2022 Jun 17;13(1):3500. doi: 10.1038/s41467-022-30908-1.

 

Addressing challenges with real-world synthetic control arms to demonstrate the

comparative effectiveness of Pralsetinib in non-small cell lung cancer.

 

Popat S(1), Liu SV(2), Scheuer N(3), Hsu GG(4), Lockhart A(4), Ramagopalan

SV(5), Griesinger F(6), Subbiah V(7).

 

Author information:

(1)Royal Marsden Hospital and Institute of Cancer Research, London, UK.

(2)Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC,

USA.

(3)Roche Products Ltd, Welwyn Garden City, UK.

(4)Cytel, Inc, Waltham, MA, USA.

(5)Global Access, F. Hoffmann-La Roche, Basel, Switzerland.

sreeram.ramagopalan@roche.com.

(6)Department of Medical Oncology, Pius-Hospital Oldenburg, Oldenburg, Germany.

(7)The University of Texas MD Anderson Cancer Center, Houston, TX, USA.

vsubbiah@mdanderson.org.

 

As advanced non-small cell lung cancer (aNSCLC) is being increasingly divided

into rare oncogene-driven subsets, conducting randomised trials becomes

challenging. Using real-world data (RWD) to construct control arms for

single-arm trials provides an option for comparative data. However,

non-randomised treatment comparisons have the potential to be biased and cause

concern for decision-makers. Using the example of pralsetinib from a RET

fusion-positive aNSCLC single-arm trial (NCT03037385), we demonstrate a relative

survival benefit when compared to pembrolizumab monotherapy and pembrolizumab

with chemotherapy RWD cohorts. Quantitative bias analyses show that results for

the RWD-trial comparisons are robust to data missingness, potential poorer

outcomes in RWD and residual confounding. Overall, the study provides evidence

in favour of pralsetinib as a first-line treatment for RET fusion-positive

aNSCLC. The quantification of potential bias performed in this study can be used

as a template for future studies of this nature.

 

© 2022. The Author(s).

 

DOI: 10.1038/s41467-022-30908-1

PMCID: PMC9205915

PMID: 35715405 [Indexed for MEDLINE]

 

9. J Clin Oncol. 2022 Jun 17:JCO2200873. doi: 10.1200/JCO.22.00873. Online ahead of print.

 

Neoadjuvant Chemotherapy Plus Immunotherapy in Early-Stage Resectable

Non-Small-Cell Lung Cancer.

 

Passaro A(1), Attili I(1), de Marinis F(1).

 

Author information:

(1)Division of Thoracic Oncology, European Institute of Oncology IRCCS, Milan,

Italy.

 

The Oncology Grand Rounds series is designed to place original reports published

in the Journal into clinical context. A case presentation is followed by a

description of diagnostic and management challenges, a review of the relevant

literature, and a summary of the authors' suggested management approaches. The

goal of this series is to help readers better understand how to apply the

results of key studies, including those published in the Journal of Clinical

Oncology, to patients seen in their own clinical practice.

 

DOI: 10.1200/JCO.22.00873

PMID: 35714306

 

10. Annu Rev Microbiol. 2022 Jun 16. doi: 10.1146/annurev-micro-121321-093031.

Online ahead of print.

 

Evolution of Tuberculosis Pathogenesis.

 

Pepperell CS(1).

 

Author information:

(1)Division of Infectious Diseases, Department of Medicine, and Department of

Medical Microbiology and Immunology, School of Medicine and Public Health,

University of Wisconsin-Madison, Madison, Wisconsin, USA; email:

pepperell@wisc.edu.

 

Mycobacterium tuberculosis is a globally distributed, lethal pathogen of humans.

The virulence armamentarium of M. tuberculosis appears to have been developed on

a scaffold of antiphagocytic defenses found among diverse, mostly free-living

species of Mycobacterium. Pathoadaptation was further aided by the modularity,

flexibility, and interactivity characterizing mycobacterial effectors and their

regulators. During emergence of M. tuberculosis, novel genetic material was

acquired, created, and integrated with existing tools. The major mutational

mechanisms underlying these adaptations are discussed in this review, with

examples. During its evolution, M. tuberculosis lost the ability and/or

opportunity to engage in lateral gene transfer, but despite this it has retained

the adaptability that characterizes mycobacteria. M. tuberculosis exemplifies

the evolutionary genomic mechanisms underlying adoption of the pathogenic niche,

and studies of its evolution have uncovered a rich array of discoveries about

how new pathogens are made. Expected final online publication date for the

Annual Review of Microbiology, Volume 76 is September 2022. Please see

http://www.annualreviews.org/page/journal/pubdates for revised estimates.

 

DOI: 10.1146/annurev-micro-121321-093031

PMID: 35709500

 

11. Nat Commun. 2022 Jun 7;13(1):3155. doi: 10.1038/s41467-022-30914-3.

 

Viral coinfection promotes tuberculosis immunopathogenesis by type I IFN

signaling-dependent impediment of Th1 cell pulmonary influx.

 

Kang TG(#)(1)(2), Kwon KW(#)(3), Kim K(1)(4), Lee I(5), Kim MJ(1)(2), Ha

SJ(6)(7), Shin SJ(8)(9).

 

Author information:

(1)Department of Biochemistry, College of Life Science & Biotechnology, Yonsei

University, Seoul, 03722, Republic of Korea.

(2)Brain Korea 21 (BK21) FOUR Program, Yonsei Education & Research Center for

Biosystems, Yonsei University, Seoul, 03722, Republic of Korea.

(3)Department of Microbiology, Graduate School of Medical Science, Brain Korea

21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of

Korea.

Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), is often

exacerbated upon coinfection, but the underlying immunological mechanisms remain

unclear. Here, to elucidate these mechanisms, we use an Mtb and lymphocytic

choriomeningitis virus coinfection model. Viral coinfection significantly

suppresses Mtb-specific IFN-γ production, with elevated bacterial loads and

hyperinflammation in the lungs. Type I IFN signaling blockade rescues the

Mtb-specific IFN-γ response and ameliorates lung immunopathology. Single-cell

sequencing, tissue immunofluorescence staining, and adoptive transfer

experiments indicate that viral infection-induced type I IFN signaling could

inhibit CXCL9/10 production in myeloid cells, ultimately impairing pulmonary

migration of Mtb-specific CD4+ T cells. Thus, our study suggests that augmented

and sustained type I IFNs by virus coinfection prior to the pulmonary

localization of Mtb-specific Th1 cells exacerbates TB immunopathogenesis by

impeding the Mtb-specific Th1 cell influx. Our study highlights a negative

function of viral coinfection-induced type I IFN responses in delaying

Mtb-specific Th1 responses in the lung.

 

© 2022. The Author(s).

 

DOI: 10.1038/s41467-022-30914-3

PMCID: PMC9174268

PMID: 35672321 [Indexed for MEDLINE]

 

12. J Exp Med. 2022 Jun 6;219(6):e20220445. doi: 10.1084/jem.20220445. Epub 2022 May 11.

 

Drug development challenges in nontuberculous mycobacterial lung disease: TB to

the rescue.

 

Dartois V(1)(2), Dick T(1)(2)(3).

 

Author information:

(1)Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ.

(2)Department of Medical Sciences, Hackensack Meridian School of Medicine,

Nutley, NJ.

(3)Department of Microbiology and Immunology, Georgetown University, Washington,

DC.

 

Nontuberculous mycobacterial pulmonary disease (NTM-PD) is treated with multiple

repurposed drugs. Chemotherapy is long and often toxic, includes parenteral

drugs, and suffers from poor cure rates. There is an urgent need for more

efficacious, tolerated, and oral antibiotics optimized towards the treatment of

NTM-PD, adapted to the spectrum of disease. In contrast to the empty NTM

pipeline, drug development for the related tuberculosis lung disease has

experienced a renaissance. Here, we argue that applying lessons learned from

tuberculosis will facilitate the discovery of curative oral regimens for NTM-PD.

 

© 2022 Dartois and Dick.

 

DOI: 10.1084/jem.20220445

PMCID: PMC9098649

PMID: 35543723 [Indexed for MEDLINE]

 

13. J Clin Oncol. 2022 Jun 10;40(17):1916-1928. doi: 10.1200/JCO.21.02010. Epub 2022

Mar 10.

 

Artificial Intelligence-Powered Spatial Analysis of Tumor-Infiltrating

Lymphocytes as Complementary Biomarker for Immune Checkpoint Inhibition in

Non-Small-Cell Lung Cancer.

 

Park S(1), Ock CY(2), Kim H(3), Pereira S(2), Park S(2), Ma M(2), Choi S(4), Kim

S(5), Shin S(2), Aum BJ(2), Paeng K(2), Yoo D(2), Cha H(1), Park S(1), Suh

KJ(6), Jung HA(1), Kim SH(6), Kim YJ(6), Sun JM(1), Chung JH(3), Ahn JS(1), Ahn

MJ(1), Lee JS(6), Park K(1), Song SY(4), Bang YJ(7), Choi YL(4), Mok TS(8), Lee

SH(1)(9).

 

Author information:

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

Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of

Korea.

(2)Lunit, Seoul, Republic of Korea.

(3)Department of Pathology, Seoul National University Bundang Hospital,

Seongnam, Republic of Korea.

PURPOSE: Biomarkers on the basis of tumor-infiltrating lymphocytes (TIL) are

potentially valuable in predicting the effectiveness of immune checkpoint

inhibitors (ICI). However, clinical application remains challenging because of

methodologic limitations and laborious process involved in spatial analysis of

TIL distribution in whole-slide images (WSI).

METHODS: We have developed an artificial intelligence (AI)-powered WSI analyzer

of TIL in the tumor microenvironment that can define three immune phenotypes

(IPs): inflamed, immune-excluded, and immune-desert. These IPs were correlated

with tumor response to ICI and survival in two independent cohorts of patients

with advanced non-small-cell lung cancer (NSCLC).

RESULTS: Inflamed IP correlated with enrichment in local immune cytolytic

activity, higher response rate, and prolonged progression-free survival compared

with patients with immune-excluded or immune-desert phenotypes. At the WSI

level, there was significant positive correlation between tumor proportion score

(TPS) as determined by the AI model and control TPS analyzed by pathologists (P

< .001). Overall, 44.0% of tumors were inflamed, 37.1% were immune-excluded, and

18.9% were immune-desert. Incidence of inflamed IP in patients with programmed

death ligand-1 TPS at < 1%, 1%-49%, and ≥ 50% was 31.7%, 42.5%, and 56.8%,

respectively. Median progression-free survival and overall survival were,

respectively, 4.1 months and 24.8 months with inflamed IP, 2.2 months and 14.0

months with immune-excluded IP, and 2.4 months and 10.6 months with

immune-desert IP.

CONCLUSION: The AI-powered spatial analysis of TIL correlated with tumor

response and progression-free survival of ICI in advanced NSCLC. This is

potentially a supplementary biomarker to TPS as determined by a pathologist.

 

DOI: 10.1200/JCO.21.02010

PMCID: PMC9177249

PMID: 35271299 [Indexed for MEDLINE]

 

14. J Clin Oncol. 2022 Jun 1;40(16):1755-1762. doi: 10.1200/JCO.21.01745. Epub 2022

Feb 14.

 

Effect of an Antiracism Intervention on Racial Disparities in Time to Lung

Cancer Surgery.

 

Charlot M(1)(2)(3), Stein JN(1)(4), Damone E(5), Wood I(6), Forster M(7), Baker

S(3)(8), Emerson M(2)(5), Samuel-Ryals C(2)(3)(9), Yongue C(3)(10), Eng

E(2)(3)(4), Manning M(3)(11), Deal A(2), Cykert S(2)(3)(6).

 

Author information:

(1)Division of Oncology, Department of Medicine, University of North Carolina,

Chapel Hill, NC.

(2)University of North Carolina Lineberger Comprehensive Cancer Center, Chapel

Hill, NC.

(3)Greensboro Health Disparities Collaborative, Greensboro, NC.

 

Comment in

    J Clin Oncol. 2022 Jun 1;40(16):1718-1720.

 

PURPOSE: Timely lung cancer surgery is a metric of high-quality cancer care and

improves survival for early-stage non-small-cell lung cancer. Historically,

Black patients experience longer delays to surgery than White patients and have

lower survival rates. Antiracism interventions have shown benefits in reducing

racial disparities in lung cancer treatment.

METHODS: We conducted a secondary analysis of Accountability for Cancer Care

through Undoing Racism and Equity, an antiracism prospective pragmatic trial, at

five cancer centers to assess the impact on overall timeliness of lung cancer

surgery and racial disparities in timely surgery. The intervention consisted of

(1) a real-time warning system to identify unmet care milestones, (2)

race-specific feedback on lung cancer treatment rates, and (3) patient

navigation. The primary outcome was surgery within 8 weeks of diagnosis. Risk

ratios (RRs) and 95% CIs were estimated using log-binomial regression and

adjusted for clinical and demographic factors.

RESULTS: A total of 2,363 patients with stage I and II non-small-cell lung

cancer were included in the analyses: intervention (n = 263), retrospective

control (n = 1,798), and concurrent control (n = 302). 87.1% of Black patients

and 85.4% of White patients in the intervention group (P = .13) received surgery

within 8 weeks of diagnosis compared with 58.7% of Black patients and 75.0% of

White patients in the retrospective group (P < .01) and 64.9% of Black patients

and 73.2% of White patients (P = .29) in the concurrent group. Black patients in

the intervention group were more likely to receive timely surgery than Black

patients in the retrospective group (RR 1.43; 95% CI, 1.26 to 1.64). White

patients in the intervention group also had timelier surgery than White patients

in the retrospective group (RR 1.10; 95% CI, 1.02 to 1.18).

CONCLUSION: Accountability for Cancer Care through Undoing Racism and Equity is

associated with timelier lung cancer surgery and reduction of the racial gap in

timely surgery.

 

DOI: 10.1200/JCO.21.01745

PMCID: PMC9148687

PMID: 35157498 [Indexed for MEDLINE]

 

 


上一篇: No.7

下一篇: No.5