您好!欢迎来到首都医科大学附属北京胸科医院官方网站!

高分SCI文摘

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

高分SCI文摘

2021年

No.4

来源:tushuguan 发布时间:2021-04-15 浏览次数:
字号: + - 14

  Medical Abstracts

 Filters applied: from 2021/3/1 - 2021/3/31.

 

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

2021 Mar 23.

 

A non-canonical type 2 immune response coordinates tuberculous granuloma

formation and epithelialization.

 

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

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

 

Author information:

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

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

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

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

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

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

Medicine, Durham, NC 27710, USA.

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

complex host immune structure that dictates infection trajectory and physiology.

Granuloma macrophages undergo a dramatic transition in which entire epithelial

modules are induced and define granuloma architecture. In tuberculosis,

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

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

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

zebrafish granulomas and analysis of Mycobacterium tuberculosis-infected

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

countervailing type 2 signals associate with macrophage epithelialization. We

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

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

cell autonomously within macrophages, where it is required for epithelioid

transformation and incorporation into necrotic granulomas. These findings

establish the signaling pathway that produces the hallmark structure of

mycobacterial infection.

 

Copyright © 2021 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.cell.2021.02.046

PMID: 33761328

 

2. JAMA. 2021 Mar 9;325(10):962-970. doi: 10.1001/jama.2021.1117.

 

Screening for Lung Cancer: US Preventive Services Task Force Recommendation

Statement.

 

US Preventive Services Task Force, Krist AH(1)(2), Davidson KW(3), Mangione

CM(4), Barry MJ(5), Cabana M(6), Caughey AB(7), Davis EM(8), Donahue KE(9),

Doubeni CA(10), Kubik M(11), Landefeld CS(12), Li L(13), Ogedegbe G(14), Owens

DK(15), Pbert L(16), Silverstein M(17), Stevermer J(18), Tseng CW(19)(20), Wong

JB(21).

 

Author information:

(1)Fairfax Family Practice Residency, Fairfax, Virginia.

(2)Virginia Commonwealth University, Richmond.

(3)Feinstein Institute for Medical Research at Northwell Health, Manhasset, New

York.

Comment in

    JAMA. 2021 Mar 9;325(10):939-941.

 

Summary for patients in

    JAMA. 2021 Mar 9;325(10):1016.

 

IMPORTANCE: Lung cancer is the second most common cancer and the leading cause

of cancer death in the US. In 2020, an estimated 228?820 persons were diagnosed

with lung cancer, and 135?720 persons died of the disease. The most important

risk factor for lung cancer is smoking. Increasing age is also a risk factor for

lung cancer. Lung cancer has a generally poor prognosis, with an overall 5-year

survival rate of 20.5%. However, early-stage lung cancer has a better prognosis

and is more amenable to treatment.

OBJECTIVE: To update its 2013 recommendation, the US Preventive Services Task

Force (USPSTF) commissioned a systematic review on the accuracy of screening for

lung cancer with low-dose computed tomography (LDCT) and on the benefits and

harms of screening for lung cancer and commissioned a collaborative modeling

study to provide information about the optimum age at which to begin and end

screening, the optimal screening interval, and the relative benefits and harms

of different screening strategies compared with modified versions of

multivariate risk prediction models.

POPULATION: This recommendation statement applies to adults aged 50 to 80 years

who have a 20 pack-year smoking history and currently smoke or have quit within

the past 15 years.

EVIDENCE ASSESSMENT: The USPSTF concludes with moderate certainty that annual

screening for lung cancer with LDCT has a moderate net benefit in persons at

high risk of lung cancer based on age, total cumulative exposure to tobacco

smoke, and years since quitting smoking.

RECOMMENDATION: The USPSTF recommends annual screening for lung cancer with LDCT

in adults aged 50 to 80 years who have a 20 pack-year smoking history and

currently smoke or have quit within the past 15 years. Screening should be

discontinued once a person has not smoked for 15 years or develops a health

problem that substantially limits life expectancy or the ability or willingness

to have curative lung surgery. (B recommendation) This recommendation replaces

the 2013 USPSTF statement that recommended annual screening for lung cancer with

LDCT in adults aged 55 to 80 years who have a 30 pack-year smoking history and

currently smoke or have quit within the past 15 years.

 

DOI: 10.1001/jama.2021.1117

PMID: 33687470 [Indexed for MEDLINE]

 

3. Nat Med. 2021 Mar;27(3):504-514. doi: 10.1038/s41591-020-01224-2. Epub 2021 Feb

18.

 

Neoadjuvant nivolumab or nivolumab plus ipilimumab in operable non-small cell

lung cancer: the phase 2 randomized NEOSTAR trial.

 

Cascone T(1), William WN Jr(2)(3), Weissferdt A(4)(5), Leung CH(6), Lin HY(6),

Pataer A(5), Godoy MCB(7), Carter BW(7), Federico L(8), Reuben A(2), …

 

Author information:

(1)Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson

Cancer Center, Houston, TX, USA. tcascone@mdanderson.org.

(2)Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson

Cancer Center, Houston, TX, USA.

(3)Oncology Center, Hospital BP, a Beneficencia Portuguesa de São Paulo, São

Paulo, Brazil.

Ipilimumab improves clinical outcomes when combined with nivolumab in metastatic

non-small cell lung cancer (NSCLC), but its efficacy and impact on the immune

microenvironment in operable NSCLC remain unclear. We report the results of the

phase 2 randomized NEOSTAR trial (NCT03158129) of neoadjuvant nivolumab or

nivolumab+ipilimumab followed by surgery in 44 patients with operable NSCLC,

using major pathologic response (MPR) as the primary endpoint. The MPR rate for

each treatment arm was tested against historical controls of neoadjuvant

chemotherapy. The nivolumab+ipilimumab arm met the prespecified primary

endpoint threshold of 6 MPRs in 21 patients, achieving a 38% MPR rate (8/21). We

observed a 22% MPR rate (5/23) in the nivolumab arm. In 37 patients resected on

trial, nivolumab and nivolumab+ipilimumab produced MPR rates of 24% (5/21) and

50% (8/16), respectively. Compared with nivolumab, nivolumab+ipilimumab

resulted in higher pathologic complete response rates (10% versus 38%), less

viable tumor (median 50% versus 9%), and greater frequencies of effector,

tissue-resident memory and effector memory T cells. Increased abundance of gut

Ruminococcus and Akkermansia spp. was associated with MPR to dual therapy. Our

data indicate that neoadjuvant nivolumab+ipilimumab-based therapy enhances

pathologic responses, tumor immune infiltrates and immunologic memory, and

merits further investigation in operable NSCLC.

 

DOI: 10.1038/s41591-020-01224-2

PMID: 33603241

 

4. Cancer Cell. 2021 Mar 8;39(3):346-360.e7. doi: 10.1016/j.ccell.2020.12.014. Epub

2021 Jan 21.

 

Patterns of transcription factor programs and immune pathway activation define

four major subtypes of SCLC with distinct therapeutic vulnerabilities.

 

Gay CM(1), Stewart CA(1), Park EM(1), Diao L(2), Groves SM(3), Heeke S(1), Nabet

BY(4), Fujimoto J(5), Solis LM(5), Lu W(5), Xi Y(2), Cardnell RJ(1), Wang Q(2),

Fabbri G(6), Cargill KR(1), Vokes NI(1), Ramkumar K(1), Zhang B(1), Della Corte

CM(7), Robson P(8), Swisher SG(9), Roth JA(9), Glisson BS(1), Shames DS(4),

Wistuba II(5), Wang J(2), Quaranta V(3), Minna J(10), Heymach JV(1), Byers

LA(11).

 

Author information:

(1)Department of Thoracic/Head & Neck Medical Oncology, the University of Texas

MD Anderson Cancer Center, Houston, TX, USA.

(2)Department of Bioinformatics and Computational Biology, the University of

Texas MD Anderson Cancer Center, Houston, TX, USA.

(3)Department of Biochemistry, Vanderbilt University Medical Center, Nashville,

TN, USA.

Despite molecular and clinical heterogeneity, small cell lung cancer (SCLC) is

treated as a single entity with predictably poor results. Using tumor expression

data and non-negative matrix factorization, we identify four SCLC subtypes

defined largely by differential expression of transcription factors ASCL1,

NEUROD1, and POU2F3 or low expression of all three transcription factor

signatures accompanied by an Inflamed gene signature (SCLC-A, N, P, and I,

respectively). SCLC-I experiences the greatest benefit from the addition of

immunotherapy to chemotherapy, while the other subtypes each have distinct

vulnerabilities, including to inhibitors of PARP, Aurora kinases, or BCL-2.

Cisplatin treatment of SCLC-A patient-derived xenografts induces intratumoral

shifts toward SCLC-I, supporting subtype switching as a mechanism of acquired

platinum resistance. We propose that matching baseline tumor subtype to therapy,

as well as manipulating subtype switching on therapy, may enhance depth and

duration of response for SCLC patients.

 

Copyright © 2020 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.ccell.2020.12.014

PMID: 33482121

 

5. Lancet Infect Dis. 2021 Mar 23:S1473-3099(20)30792-1. doi:

10.1016/S1473-3099(20)30792-1. Online ahead of print.

 

Food for thought: addressing undernutrition to end tuberculosis.

 

Sinha P(1), Lönnroth K(2), Bhargava A(3), Heysell SK(4), Sarkar S(5), Salgame

P(6), Rudgard W(7), Boccia D(8), Van Aartsen D(4), Hochberg NS(9).

 

Author information:

(1)Section of Infectious Diseases, Department of Medicine, Boston University

School of Medicine, Boston University, MA, USA. Electronic address:

sinha.pranay@pm.me.

(2)Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden.

(3)Department of Medicine, Yenepoya Medical College, and Center for Nutrition

Studies, Yenepoya (Deemed to be University), Mangalore, India; Department of

Medicine, McGill University, Montreal, QC, Canada.

Tuberculosis is the leading cause of deaths from an infectious disease

worldwide. WHO's End TB Strategy is falling short of several 2020 targets.

Undernutrition is the leading population-level risk factor for tuberculosis.

Studies have consistently found that undernutrition is associated with increased

tuberculosis incidence, increased severity, worse treatment outcomes, and

increased mortality. Modelling studies support implementing nutritional

interventions for people living with tuberculosis and those at risk of

tuberculosis disease to ensure the success of the End TB Strategy. In this

Personal View, we highlight nutrition-related immunocompromisation, implications

of undernutrition for tuberculosis treatment and prevention, the role of

nutritional supplementation, pharmacokinetics and pharmacodynamics of

antimycobacterial medications in undernourished people with tuberculosis, and

the role of social protection interventions in addressing undernutrition as a

tuberculosis risk factor. To catalyse action on this insufficiently addressed

accelerant of the global tuberculosis epidemic, research should be prioritised

to understand the immunological pathways that are impaired by nutrient

deficiencies, develop tools to diagnose clinical and subclinical tuberculosis in

people who are undernourished, and understand how nutritional status affects the

efficacy of tuberculosis vaccine and therapy. Through primary research,

modelling, and implementation research, policy change should also be

accelerated, particularly in countries with a high burden of tuberculosis.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(20)30792-1

PMID: 33770535

 

6. Lancet Infect Dis. 2021 Mar 23:S1473-3099(20)30732-5. doi:

10.1016/S1473-3099(20)30732-5. Online ahead of print.

 

Measuring health-care delays among privately insured patients with tuberculosis

in the USA: an observational cohort study.

 

El Halabi J(1), Palmer N(1), McDuffie M(1), Golub JJ(2), Fox K(1), Kohane I(1),

Farhat MR(3).

 

Author information:

(1)Department of Biomedical Informatics, Harvard Medical School, Boston, MA,

USA.

(2)Center for Tuberculosis Research, Johns Hopkins University, Baltimore, MD,

USA.

(3)Department of Biomedical Informatics, Harvard Medical School, Boston, MA,

USA; Division of Pulmonary and Critical Care Medicine, Massachusetts General

Hospital, Boston, MA, USA. Electronic address: maha_farhat@hms.harvard.edu.

 

BACKGROUND: A high index of suspicion is needed to initiate appropriate testing

for tuberculosis due to its protean symptoms, yet health-care providers in

low-incidence settings are becoming less familiar with the disease as rates

decline. We aimed to estimate delays in tuberculosis diagnosis and treatment at

the US national level between 2008 and 2016.

METHODS: In this retrospective observational cohort study, we repurposed private

insurance claims data provided by Aetna (Connecticut, USA), to measure

health-care delays in tuberculosis diagnosis in the USA in 2008-16. Active

tuberculosis was determined by diagnosis codes and the filling of

anti-tuberculosis treatment prescriptions. Health-care delays were defined as

the duration between the first health-care visit for a tuberculosis symptom and

the initiation of anti-tuberculosis treatment. We assessed if delays varied over

time, and by patient and system variables, using multivariable regression. We

estimated household tuberculosis transmission and respiratory complications

after treatment initiation.

FINDINGS: We confirmed 738 active tuberculosis cases (incidence 1·45 per 100000

person-years) with a median health-care delay of 24 days (IQR 10-45).

Multivariable regression analysis showed that longer delays were associated with

older age (8·4% per 10 year increase [95% CI 4·0 to 13·1]; p<0·0086) and non-HIV

immunosuppression (19·2% [15·1 to 60·0]; p=0·0432). Presenting with three or

more symptoms was associated with a shorter delay (-22·5% [-39·1 to -2·0];

p=0·0415), relative to presenting with one symptom, as did use of chest imaging

(-24·9% [-37·9 to -8·9]; p<0·0098), tuberculosis nucleic acid amplification

tests (-19·2% [-32·7 to -3·1]; p=0·0241), and care by a tuberculosis specialist

provider (-17·2% [-33·1 to -22·3]; p<0·0087). Longer delays were associated with

an increased rate of respiratory complications even after controlling for

patient characteristics, and an increased rate of secondary tuberculosis among

dependents.

INTERPRETATION: In the USA, the median health-care delay for privately insured

patients with tuberculosis exceeds WHO-recommended levels of 21 days (3 weeks).

The results suggest the need for health-care provider education on best

practices in tuberculosis diagnosis, including the use of molecular tests and

the maintenance of a high index of suspicion for the disease.

FUNDING: US National Institutes of Health.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(20)30732-5

PMID: 33770534

 

7. Am J Respir Crit Care Med. 2021 Mar 24. doi: 10.1164/rccm.202011-4239PP. Online

ahead of print.

 

Latent Tuberculosis: Two Centuries of Confusion.

 

Behr MA(1), Kaufmann E(2), Duffin J(3), Edelstein PH(4), Ramakrishnan L(5).

 

Author information:

(1)McGill University, 5620, Division of Infectious Diseases, Montreal, Quebec,

Canada; marcel.behr@mcgill.ca.

(2)McGill University, 5620, Montreal, Quebec, Canada.

(3)Queen's University, 4257, Kingston, Ontario, Canada.

(4)University of Pennsylvania, 6572, Philadelphia, Pennsylvania, United States.

(5)Cambridge University, 2152, Department of Medicine, Cambridge, United Kingdom

of Great Britain and Northern Ireland.

 

The term latent tuberculosis (TB) was coined two centuries ago to describe

post-mortem tuberculous pathology in the absence of ante-mortem tuberculosis

manifestations. However, the meaning of the term has changed with each passing

century, engendering confusion. In the early 20th century, with the advent of

microbiological assays for live tubercle bacteria, latent TB switched from the

host to refer to the bacteria from post-mortem tissues of nontuberculous hosts.

Then in the late 20th century, the definition of latent TB infection returned to

the host, this time referring to those with immunoreactivity to Mycobacterium

tuberculosis antigens. Based on this new definition, latent TB infection is

unique among bacterial infectious diseases, in that supportive evidence of the

infection state is sought by the absence of the causative bacterium and its

clinical manifestations. The use of indirect bedside and laboratory tests to

denote infection creates clinical and research confusion, as the tests for

immunoreactivity suffer from recognized limitations in sensitivity and

specificity. We propose that the concept of latent TB infection be separated

from that of tuberculous immunoreactivity in the interest of correct diagnosis

and focused treatment, correct formulation and interpretation of research

questions and better allocation of programmatic resources for TB elimination. To

this end, we suggest new terminology to course-correct our thinking about

tuberculous infection (TBI) which is subdivided into tuberculous infection-no

disease (TBInd) and the long-accepted term for the disease, tuberculosis (TB).

 

DOI: 10.1164/rccm.202011-4239PP

PMID: 33761302

 

8. Nat Commun. 2021 Mar 19;12(1):1780. doi: 10.1038/s41467-021-22057-8.

 

Clinical impact of subclonal EGFR T790M mutations in advanced-stage EGFR-mutant

non-small-cell lung cancers.

 

Vaclova T(1), Grazini U(2), Ward L(3), O'Neill D(3), Markovets A(4), Huang X(5),

Chmielecki J(4), Hartmaier R(4), Thress KS(4)(6), Smith PD(2), Barrett JC(4),

Downward J(7), de Bruin EC(8).

 

Author information:

(1)Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, UK.

(2)Bioscience, Oncology R&D, AstraZeneca, Cambridge, UK.

(3)Discovery Science, BioPharmaceutical R&D, AstraZeneca, Cambridge, UK.

(4)Translational Medicine, Oncology R&D, AstraZeneca, Boston, MA, USA.

(5)Biometrics Oncology, Oncology R&D, AstraZeneca, Cambridge, UK.

(6)Global Marketing Diagnostics, Oncology Business, AstraZeneca, Gaithersburg,

MD, USA.

(7)Oncogene Biology, Francis Crick Institute, London, UK.

(8)Translational Medicine, Oncology R&D, AstraZeneca, Cambridge, UK.

elza.de-bruin@astrazeneca.com.

 

Advanced non-small-cell lung cancer (NSCLC) patients with EGFR T790M-positive

tumours benefit from osimertinib, an epidermal growth factor receptor-tyrosine

kinase inhibitor (EGFR-TKI). Here we show that the size of the EGFR

T790M-positive clone impacts response to osimertinib. T790M subclonality, as

assessed by a retrospective NGS analysis of 289 baseline plasma ctDNA samples

from T790M-positive advanced NSCLC patients from the AURA3 phase III trial, is

associated with shorter progression-free survival (PFS), both in the osimertinib

and the chemotherapy-treated patients. Both baseline and longitudinal ctDNA

profiling indicate that the T790M subclonal tumours are enriched for PIK3CA

alterations, which we demonstrate to confer resistance to osimertinib in vitro

that can be partially reversed by PI3K pathway inhibitors. Overall, our results

elucidate the impact of tumour heterogeneity on response to osimertinib in

advanced stage NSCLC patients and could help define appropriate combination

therapies in these patients.

 

DOI: 10.1038/s41467-021-22057-8

PMID: 33741979 [Indexed for MEDLINE]

 

9. Immunol Rev. 2021 Mar 12. doi: 10.1111/imr.12959. Online ahead of print.

 

The double-edged sword of Tregs in M tuberculosis, M avium, and M absessus

infection.

 

Verma D(1), Chan ED(2)(3)(4), Ordway DJ(1).

 

Author information:

(1)Mycobacteria Research Laboratory, Department of Microbiology, Immunology, and

Pathology, Colorado State University, Fort Collins, CO, USA.

(2)Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical

Center, Denver, CO, USA.

(3)Departments of Medicine and Academic Affairs, National Jewish Health, Denver,

CO, USA.

(4)Division of Pulmonary Sciences and Critical Care Medicine, University of

Colorado Anschutz Medical Campus, Denver, CO, USA.

 

Immunity against different Mycobacteria species targeting the lung requires

distinctly different pulmonary immune responses for bacterial clearance. Many

parameters of acquired and regulatory immune responses differ quantitatively and

qualitatively from immunity during infection with Mycobacteria species.

Nontuberculosis Mycobacteria species (NTM) Mycobacterium avium- (M avium),

Mycobacterium abscessus-(M abscessus), and the Mycobacteria species

Mycobacterium tuberculosis-(Mtb). Herein, we discuss the potential implications

of acquired and regulatory immune responses in the context of animal and human

studies, as well as future directions for efforts to treat Mycobacteria

diseases.

 

© 2021 John Wiley & Sons A/S. Published by John Wiley & Sons Ltd.

 

DOI: 10.1111/imr.12959

PMID: 33713043

 

10. Cell Host Microbe. 2021 Mar 5:S1931-3128(21)00084-6. doi:

10.1016/j.chom.2021.02.005. Online ahead of print.

 

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

tuberculous granuloma.

 

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

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

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

 

Author information:

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

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

Washington, Seattle, WA 98195, USA.

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

Institute, Seattle, WA 98109, USA.

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

CD4 T cell effector function is required for optimal containment of

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

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

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

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

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

rapidly extinguished within the granuloma but not within unaffected lung

regions, suggesting localized immunosuppression. We identified a signature of

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

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

accumulation of terminally differentiated effector CD4 T cells, improved IFN?

production within granulomas, and reduced bacterial burdens. These findings

uncover a spatially localized immunosuppressive mechanism associated with Mtb

infection and provide potential targets for host-directed therapy.

 

Copyright © 2021. Published by Elsevier Inc.

 

DOI: 10.1016/j.chom.2021.02.005

PMID: 33711270

 

11. Immunol Rev. 2021 Mar 12. doi: 10.1111/imr.12965. Online ahead of print.

 

BCG-induced protection against Mycobacterium tuberculosis infection: Evidence,

mechanisms, and implications for next-generation vaccines.

 

Foster M(1), Hill PC(2), Setiabudiawan TP(3), Koeken VACM(3)(4), Alisjahbana

B(5), van Crevel R(3).

 

Author information:

(1)Department of Microbiology and Immunology, University of Otago, Dunedin, New

Zealand.

(2)Centre for International Health, University of Otago, Dunedin, New Zealand.

(3)Department of Internal Medicine and Radboud Center for Infectious Diseases

(RCI), Radboud University Medical Center, Nijmegen, The Netherlands.

(4)Department of Computational Biology for Individualised Infection Medicine,

Centre for Individualised Infection Medicine (CiiM) & TWINCORE, Joint Ventures

between The Helmholtz-Centre for Infection Research (HZI) and The Hannover

Medical School (MHH), Hannover, Germany.

(5)Tuberculosis Working Group, Faculty of Medicine, Universitas Padjadjaran,

Bandung, Indonesia.

 

The tuberculosis (TB) vaccine Bacillus Calmette-Guérin (BCG) was introduced 100

years ago, but as it provides insufficient protection against TB disease,

especially in adults, new vaccines are being developed and evaluated. The

discovery that BCG protects humans from becoming infected with Mycobacterium

tuberculosis (Mtb) and not just from progressing to TB disease provides

justification for considering Mtb infection as an endpoint in vaccine trials.

Such trials would require fewer participants than those with disease as an

endpoint. In this review, we first define Mtb infection and disease phenotypes

that can be used for mechanistic studies and/or endpoints for vaccine trials.

Secondly, we review the evidence for BCG-induced protection against Mtb

infection from observational and BCG re-vaccination studies, and discuss

limitations and variation of this protection. Thirdly, we review possible

underlying mechanisms for BCG efficacy against Mtb infection, including

alternative T cell responses, antibody-mediated protection, and innate immune

mechanisms, with a specific focus on BCG-induced trained immunity, which

involves epigenetic and metabolic reprogramming of innate immune cells. Finally,

we discuss the implications for further studies of BCG efficacy against Mtb

infection, including for mechanistic research, and their relevance to the design

and evaluation of new TB vaccines.

 

© 2021 The Authors. Immunological Reviews published by John Wiley & Sons Ltd.

 

DOI: 10.1111/imr.12965

PMID: 33709421

 

12. Nat Commun. 2021 Mar 11;12(1):1606. doi: 10.1038/s41467-021-21748-6.

 

Biofilm formation in the lung contributes to virulence and drug tolerance of

Mycobacterium tuberculosis.

 

Chakraborty P(1), Bajeli S(1), Kaushal D(2), Radotra BD(3), Kumar A(4)(5).

 

Author information:

(1)Council of Scientific and Industrial Research, Institute of Microbial

Technology, Chandigarh, India.

(2)Southwest National Primate Research Center, Texas Biomedical Research

Institute, San Antonio, TX, USA.

(3)Department of Histopathology, Postgraduate Institute of Medical Education and

Research, Chandigarh, India.

(4)Council of Scientific and Industrial Research, Institute of Microbial

Technology, Chandigarh, India. ashwanik@imtech.res.in.

(5)Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar

Pradesh, India. ashwanik@imtech.res.in.

 

Tuberculosis is a chronic disease that displays several features commonly

associated with biofilm-associated infections: immune system evasion, antibiotic

treatment failures, and recurrence of infection. However, although Mycobacterium

tuberculosis (Mtb) can form cellulose-containing biofilms in vitro, it remains

unclear whether biofilms are formed during infection in vivo. Here, we

demonstrate the formation of Mtb biofilms in animal models of infection and in

patients, and that biofilm formation can contribute to drug tolerance. First, we

show that cellulose is also a structural component of the extracellular matrix

of in vitro biofilms of fast and slow-growing nontuberculous mycobacteria. Then,

we use cellulose as a biomarker to detect Mtb biofilms in the lungs of

experimentally infected mice and non-human primates, as well as in lung tissue

sections obtained from patients with tuberculosis. Mtb strains defective in

biofilm formation are attenuated for survival in mice, suggesting that biofilms

protect bacilli from the host immune system. Furthermore, the administration of

nebulized cellulase enhances the antimycobacterial activity of isoniazid and

rifampicin in infected mice, supporting a role for biofilms in phenotypic drug

tolerance. Our findings thus indicate that Mtb biofilms are relevant to human

tuberculosis.

 

DOI: 10.1038/s41467-021-21748-6

PMCID: PMC7952908

PMID: 33707445 [Indexed for MEDLINE]

 

13. Cancer Discov. 2021 Mar 11:candisc.1385.2020. doi: 10.1158/2159-8290.CD-20-1385. Online ahead of print.

 

Genetic determinants of EGFR-Driven Lung Cancer Growth and Therapeutic Response

In Vivo.

 

Foggetti G(1), Li C(2), Cai H(3), Hellyer JA(4), Lin WY(3), Ayeni D(5), Hastings

K(6), Choi J(7), Wurtz A(8), Andrejka L(9), Maghini DG(10), Rashleigh N(8), Levy

S(8), Homer R(11), Gettinger SN(12), Diehn M(13), Wakelee HA(14), Petrov DA(15),

Winslow MM(3), Politi K(16).

 

Author information:

(1)Medical Oncology, Yale School of Medicine.

(2)Biology, Stanford University.

(3)Department of Genetics, Stanford University School of Medicine.

In lung adenocarcinoma, oncogenic EGFR mutations co-occur with many tumor

suppressor gene alterations, however the extent to which these contribute to

tumor growth and response to therapy in vivo remains largely unknown. By

quantifying the effects of inactivating ten putative tumor suppressor genes in a

mouse model of EGFR-driven Trp53-deficient lung adenocarcinoma, we found that

Apc, Rb1, or Rbm10 inactivation strongly promoted tumor growth. Unexpectedly,

inactivation of Lkb1 or Setd2 - the strongest drivers of growth in a Kras-driven

model - reduced EGFR-driven tumor growth. These results are consistent with

mutational frequencies in human EGFR- and KRAS-driven lung adenocarcinomas.

Furthermore, Keap1 inactivation reduced the sensitivity of EGFR-driven tumors to

the EGFR inhibitor osimertinib and mutations in the KEAP1 pathway were

associated with decreased time on tyrosine kinase inhibitor treatment in

patients. Our study highlights how the impact of genetic alterations differ

across oncogenic contexts and that the fitness landscape shifts upon treatment.

 

Copyright ©2021, American Association for Cancer Research.

 

DOI: 10.1158/2159-8290.CD-20-1385

PMID: 33707235

 

14. Am J Respir Crit Care Med. 2021 Mar 11. doi: 10.1164/rccm.202007-2634OC. Online

ahead of print.

 

Glycemic Trajectories After Tuberculosis Diagnosis and Treatment Outcomes of New

Tuberculosis Patients: A Prospective Study in Eastern China.

 

Liu Q(1), You N(1), Pan H(2), Shen Y(3), Lu P(4), Wang J(5), Lu W(1), Zhu L(6),

Martinez L(7).

 

Author information:

(1)Center for Disease Control and Prevention of Jiangsu Province, Department of

Chronic Communicable Disease, Nanjing, China.

(2)The Third People's Hospital of Zhenjiang Affiliated to Jiangsu University,

Department of Tuberculosis, Zhenjiang, China.

(3)University of Georgia, Department of Epidemiology and Biostatistics, Athens,

Georgia, United States.

Rationale Newly diagnosed tuberculosis patients often have inconsistent glycemic

measurements during and after treatment. Distinct glycemic trajectories after

tuberculosis diagnosis are not well characterized and whether patients with

stress hyperglycemia have poor treatment outcomes is not known. Objectives To

identify distinct glycemic trajectories from tuberculosis diagnosis to

post-treatment and to assess the relationship between glycemic trajectories and

tuberculosis treatment outcomes. Methods Newly diagnosed, drug-susceptible

tuberculosis patients with at least three fasting plasma glucose (FPG) tests at

tuberculosis diagnosis and during the 3rd and 6th month of treatment were

identified and included from Jiangsu Province, China. Patients were also given

an additional FPG test at two and four months post-treatment. Measurements and

Main Results Several distinct glycemic trajectories from tuberculosis diagnosis

to post-treatment were found including consistently normal glycemic testing

(43%), transient hyperglycemia (24%), erratic glycemic instability (12%),

diabetes (16%), and consistently hyperglycemic but without diabetes (6%).

Compared to participants with a consistently normal glycemic trajectory,

patients were more likely to fail treatment if they had transient hyperglycemia

(Adjusted Odds Ratio [AOR], 4.20; 95% CI, 1.57-11.25, P=0.004) or erratic

glycemic instability (AOR, 5.98; 95% CI, 2.00-17.87; P=0.001). Patients living

with diabetes also had higher risk of treatment failure (AOR, 6.56; 95%

Confidence Interval [CI], 2.22-19.35, P=0.001), and this was modified by

glycemic control and metformin use. Conclusions Among tuberculosis patients

without diabetes, glycemic changes were common and may represent an important

marker for patient response to tuberculosis treatment.

 

DOI: 10.1164/rccm.202007-2634OC

PMID: 33705666

 

15. J Clin Oncol. 2021 Mar 8:JCO2002212. doi: 10.1200/JCO.20.02212. Online ahead of print.

 

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

Lung Cancer: CheckMate 451.

 

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

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

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

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

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

 

Author information:

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

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

South Korea.

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

Louis, MO.

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

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

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

plus ipilimumab and nivolumab monotherapy as maintenance therapy following

first-line chemotherapy for ED-SCLC.

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

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

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

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

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

progression or unacceptable toxicity. Primary end point was overall survival

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

hierarchically tested.

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

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

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

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

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

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

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

benefit with nivolumab plus ipilimumab was observed in patients with tumor

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

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

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

CONCLUSION: Maintenance therapy with nivolumab plus ipilimumab did not prolong

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

There were no new safety signals.

 

DOI: 10.1200/JCO.20.02212

PMID: 33683919

 

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

Mar 1.

 

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

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

 

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

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

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

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

 

Author information:

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

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

University of Manchester and The Christie NHS Foundation Trust, Manchester

Academic Health Science Centre, Manchester, United Kingdom.

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

Milano, Milan, Italy.

PURPOSE: Genetic rearrangements of the tyrosine receptor kinase ROS

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

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

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

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

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

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

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

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

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

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

intracranial PFS, and safety.

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

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

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

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

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

patients with measurable baseline CNS metastases by blinded independent central

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

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

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

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

no new safety signals were found.

CONCLUSION: Entrectinib continued to demonstrate a high level of clinical

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

CNS metastases.

 

DOI: 10.1200/JCO.20.03025

PMID: 33646820

 

17. J Clin Oncol. 2021 Mar 20;39(9):1040-1091. doi: 10.1200/JCO.20.03570. Epub 2021

Feb 16.

 

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

and OH (CCO) Joint Guideline Update.

 

Hanna NH(1), Robinson AG(2), Temin S(3), Baker S Jr(4), Brahmer JR(5), Ellis

PM(6), Gaspar LE(7)(8), Haddad RY(9), Hesketh PJ(10), Jain D(11), Jaiyesimi

I(12), Johnson DH(13), Leighl NB(14), Moffitt PR(15), Phillips T(16), Riely

GJ(17), Rosell R(18), Schiller JH(19), Schneider BJ(20), Singh N(21), Spigel

DR(22), Tashbar J(23), Masters G(24).

 

Author information:

(1)Indiana University Simon Comprehensive Cancer Center, Indianapolis, IN.

(2)Kingston General Hospital, School of Medicine, Queen's University, ON,

Canada.

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

PURPOSE: To provide evidence-based recommendations updating the 2017 ASCO

guideline on systemic therapy for patients with stage IV non-small-cell lung

cancer (NSCLC) with driver alterations. A guideline update for systemic therapy

for patients with stage IV NSCLC without driver alterations was published

separately.

METHODS: The American Society of Clinical Oncology and Ontario Health (Cancer

Care Ontario) NSCLC Expert Panel updated recommendations based on a systematic

review of randomized controlled trials (RCTs) from December 2015 to January 2020

and meeting abstracts from ASCO 2020.

RESULTS: This guideline update reflects changes in evidence since the previous

update. Twenty-seven RCTs, 26 observational studies, and one meta-analysis

provide the evidence base (total 54). Outcomes of interest included efficacy and

safety. Additional literature suggested by the Expert Panel is discussed.

RECOMMENDATIONS: All patients with nonsquamous NSCLC should have the results of

testing for potentially targetable mutations (alterations) before implementing

therapy for advanced lung cancer, regardless of smoking status recommendations,

when possible, following other existing high-quality testing guidelines. Most

patients should receive targeted therapy for these alterations: Targeted

therapies against ROS-1 fusions, BRAF V600e mutations, RET fusions, MET exon 14

skipping mutations, and NTRK fusions should be offered to patients, either as

initial or second-line therapy when not given in the first-line setting. New or

revised recommendations include the following: Osimertinib is the optimal

first-line treatment for patients with activating epidermal growth factor

receptor mutations (exon 19 deletion, exon 21 L858R, and exon 20 T790M);

alectinib or brigatinib is the optimal first-line treatment for patients with

anaplastic lymphoma kinase fusions. For the first time, to our knowledge, the

guideline includes recommendations regarding RET, MET, and NTRK alterations.

Chemotherapy is still an option at most stages.Additional information is

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

 

DOI: 10.1200/JCO.20.03570

PMID: 33591844

 

18. Immunity. 2021 Mar 9;54(3):526-541.e7. doi: 10.1016/j.immuni.2021.01.003. Epub

2021 Jan 29.

 

Early innate and adaptive immune perturbations determine long-term severity of

chronic virus and Mycobacterium tuberculosis coinfection.

 

Xu W(1), Snell LM(1), Guo M(2), Boukhaled G(1), Macleod BL(1), Li M(3), Tullius

MV(4), Guidos CJ(5), Tsao MS(1), Divangahi M(6), Horwitz MA(7), Liu J(8), Brooks

DG(9).

 

Author information:

(1)Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G

2M9, Canada.

(2)Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G

2M9, Canada; Department of Immunology, University of Toronto, Toronto, ON M5S

1A8, Canada.

(3)Princess Margaret Cancer Center, University Health Network, Toronto, ON M5G

2M9, Canada; Department of Molecular Genetics, University of Toronto, Toronto,

ON M5S 1A8, Canada.

Chronic viral infections increase severity of Mycobacterium tuberculosis (Mtb)

coinfection. Here, we examined how chronic viral infections alter the pulmonary

microenvironment to foster coinfection and worsen disease severity. We developed

a coordinated system of chronic virus and Mtb infection that induced central

clinical manifestations of coinfection, including increased Mtb burden,

extra-pulmonary dissemination, and heightened mortality. These disease states

were not due to chronic virus-induced immunosuppression or exhaustion; rather,

increased amounts of the cytokine TNFα initially arrested pulmonary Mtb growth,

impeding dendritic cell mediated antigen transportation to the lymph node and

subverting immune-surveillance, allowing bacterial sanctuary. The cryptic Mtb

replication delayed CD4 T cell priming, redirecting T helper (Th) 1 toward Th17

differentiation and increasing pulmonary neutrophilia, which diminished

long-term survival. Temporally restoring CD4 T cell induction overcame these

diverse disease sequelae to enhance Mtb control. Thus, Mtb co-opts TNFα from the

chronic inflammatory environment to subvert immune-surveillance, avert early

immune function, and foster long-term coinfection.

 

Copyright © 2021 Elsevier Inc. All rights reserved.

 

DOI: 10.1016/j.immuni.2021.01.003

PMCID: PMC7946746

PMID: 33515487

 

19. Lancet Infect Dis. 2021 Mar;21(3):354-365. doi: 10.1016/S1473-3099(20)30914-2.

Epub 2021 Jan 25.

 

Biomarker-guided tuberculosis preventive therapy (CORTIS): a randomised

controlled trial.

 

Scriba TJ(1), Fiore-Gartland A(2), Penn-Nicholson A(1), Mulenga H(1), Kimbung

Mbandi S(1), Borate B(2), Mendelsohn SC(1), Hadley K(1), Hikuam C(1), Kaskar

M(1), Musvosvi M(1), Bilek N(1), Self S(2), Sumner T(3), White RG(3), Erasmus

M(1), Jaxa L(1), Raphela R(1), Innes C(4), Brumskine W(4), Hiemstra A(5),

Malherbe ST(5), Hassan-Moosa R(6), Tameris M(1), Walzl G(5), Naidoo K(6),

Churchyard G(7), Hatherill M(8); CORTIS-01 Study Team.

 

Author information:

(1)South African Tuberculosis Vaccine Initiative, Institute of Infectious

Disease and Molecular Medicine, Division of Immunology, Department of Pathology,

University of Cape Town, Cape Town, South Africa.

(2)Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research

Center, Seattle, WA, USA.

(3)TB Modelling Group, TB Centre, Centre for Mathematical Modelling of

Infectious Diseases, Department of Infectious Disease Epidemiology, London

School of Hygiene & Tropical Medicine, London, UK.

Comment in

    Lancet Infect Dis. 2021 Mar;21(3):299-300.

BACKGROUND: Targeted preventive therapy for individuals at highest risk of

incident tuberculosis might impact the epidemic by interrupting transmission. We

tested performance of a transcriptomic signature of tuberculosis (RISK11) and

efficacy of signature-guided preventive therapy in parallel, using a hybrid

three-group study design.

METHODS: Adult volunteers aged 18-59 years were recruited at five geographically

distinct communities in South Africa. Whole blood was sampled for RISK11 by

quantitative RT-PCR assay from eligible volunteers without HIV, recent previous

tuberculosis (ie, <3 years before screening), or comorbidities at screening.

RISK11-positive participants were block randomised (1:2; block size 15) to

once-weekly, directly-observed, open-label isoniazid and rifapentine for 12

weeks (ie, RISK11 positive and 3HP positive), or no treatment (ie, RISK11

positive and 3HP negative). A subset of eligible RISK11-negative volunteers were

randomly assigned to no treatment (ie, RISK11 negative and 3HP negative).

Diagnostic discrimination of prevalent tuberculosis was tested in all

participants at baseline. Thereafter, prognostic discrimination of incident

tuberculosis was tested in the untreated RISK11-positive versus RISK11-negative

groups, and treatment efficacy in the 3HP-treated versus untreated

RISK11-positive groups, during active surveillance through 15 months. The

primary endpoint was microbiologically confirmed pulmonary tuberculosis. The

primary outcome measures were risk ratio [RR] for tuberculosis of

RISK11-positive to RISK11-negative participants, and treatment efficacy. This

trial is registered with ClinicalTrials.gov, NCT02735590.

FINDINGS: 20?207 volunteers were screened, and 2923 participants were enrolled,

including RISK11-positive participants randomly assigned to 3HP (n=375) or no

3HP (n=764), and 1784 RISK11-negative participants. Cumulative probability of

prevalent or incident tuberculosis disease was 0·066 (95% CI 0·049 to 0·084) in

RISK11-positive (3HP negative) participants and 0·018 (0·011 to 0·025) in

RISK11-negative participants (RR 3·69, 95% CI 2·25-6·05) over 15 months.

Tuberculosis prevalence was 47 (4·1%) of 1139 versus 14 (0·78%) of 1984 in

RISK11-positive compared with RISK11-negative participants, respectively

(diagnostic RR 5·13, 95% CI 2·93 to 9·43). Tuberculosis incidence over 15 months

was 2·09 (95% CI 0·97 to 3·19) vs 0·80 (0·30 to 1·30) per 100 person years in

RISK11-positive (3HP-negative) participants compared with RISK11-negative

participants (cumulative incidence ratio 2·6, 95% CI 1·2 to 5·9). Serious

adverse events related to 3HP included one hospitalisation for seizures

(unintentional isoniazid overdose) and one death of unknown cause (possibly

temporally related). Tuberculosis incidence over 15 months was 1·94 (95% CI 0·35

to 3·50) versus 2·09 (95% CI 0·97 to 3·19) per 100 person-years in 3HP-treated

RISK11-positive participants compared with untreated RISK11-positive

participants (efficacy 7·0%, 95% CI -145 to 65).

INTERPRETATION: The RISK11 signature discriminated between individuals with

prevalent tuberculosis, or progression to incident tuberculosis, and individuals

who remained healthy, but provision of 3HP to signature-positive individuals

after exclusion of baseline disease did not reduce progression to tuberculosis

over 15 months.

FUNDING: Bill and Melinda Gates Foundation, South African Medical Research

Council.

 

Copyright © 2021 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(20)30914-2

PMCID: PMC7907670

PMID: 33508224 [Indexed for MEDLINE]

 

20. Lancet Infect Dis. 2021 Mar;21(3):366-375. doi: 10.1016/S1473-3099(20)30928-2.

Epub 2021 Jan 25.

 

Transcriptomic signatures for diagnosing tuberculosis in clinical practice: a

prospective, multicentre cohort study.

 

Hoang LT(1), Jain P(1), Pillay TD(1), Tolosa-Wright M(1), Niazi U(2), Takwoingi

Y(3), Halliday A(4), Berrocal-Almanza LC(1), Deeks JJ(3), Beverley P(1), Kon

OM(5), Lalvani A(6).

 

Author information:

(1)Tuberculosis Research Centre, National Heart and Lung Institute, Imperial

College London, London, UK.

(2)Guy's and St Thomas' National Health Service Foundation Trust and King's

College London National Institute for Health Research Biomedical Research Centre

Translational Bioinformatics Platform, Guy's Hospital, London, UK.

(3)Test Evaluation Research Group, Institute of Applied Health Research,

University of Birmingham, Birmingham, UK; National Institute of Health Research

Birmingham Biomedical Research Centre, University Hospitals Birmingham National

Health Service Foundation Trust and University of Birmingham, Birmingham, UK.

BACKGROUND: Blood transcriptomic signatures for diagnosis of tuberculosis have

shown promise in case-control studies, but none have been prospectively designed

or validated in adults presenting with the full clinical spectrum of suspected

tuberculosis, including extrapulmonary tuberculosis and common differential

diagnoses that clinically resemble tuberculosis. We aimed to evaluate the

diagnostic accuracy of transcriptomic signatures in patients presenting with

clinically suspected tuberculosis in routine practice.

METHODS: The Validation of New Technologies for Diagnostic Evaluation of

Tuberculosis (VANTDET) study was nested within a prospective, multicentre cohort

study in secondary care in England (IDEA 11/H0722/8). Patients (aged ≥16 years)

suspected of having tuberculosis in the routine clinical inpatient and

outpatient setting were recruited at ten National Health Service hospitals in

England for IDEA and were included in VANTDET if they provided consent for

genomic analysis. Patients had whole blood taken for microarray analysis to

measure abundance of transcripts and were followed up for 6-12 months to

determine final diagnoses on the basis of predefined diagnostic criteria. The

diagnostic accuracy of six signatures derived from the cohort and three

previously published transcriptomic signatures with potentially high diagnostic

performance were assessed by calculating area under the receiver-operating

characteristic curves (AUC-ROCs), sensitivities, and specificities.

FINDINGS: Between Nov 25, 2011, and Dec 31, 2013, 1162 participants were

enrolled. 628 participants (aged ≥16 years) were included in the analysis, of

whom 212 (34%) had culture-confirmed tuberculosis, 89 (14%) had highly probable

tuberculosis, and 327 (52%) had tuberculosis excluded. The novel signature with

highest performance for identifying all active tuberculosis gave an AUC-ROC of

0·87 (95% CI 0·81-0·92), sensitivity of 77% (66-87), and specificity of 84%

(74-91). The best-performing published signature gave an AUC-ROC of 0·83

(0·80-0·86), sensitivity of 78% (73-83), and specificity of 76% (70-80). For

detecting highly probable tuberculosis, the best novel signature yielded results

of 0·86 (0·71-0·95), 77% (56-94%), and 77% (57-95%). None of the relevant

cohort-derived or previously published signatures achieved the WHO-defined

targets of paired sensitivity and specificity for a non-sputum-based diagnostic

test.

INTERPRETATION: In a clinically representative cohort in routine practice in a

low-incidence setting, transcriptomic signatures did not have adequate accuracy

for diagnosis of tuberculosis, including in patients with highly probable

tuberculosis where the unmet need is greatest. These findings suggest that

transcriptomic signatures have little clinical utility for diagnostic assessment

of suspected tuberculosis.

FUNDING: National Institute for Health Research.

 

Copyright © 2021 The Author(s). Published by Elsevier Ltd. This is an Open

Access article under the CC BY-NC-ND 4.0 license. Published by Elsevier Ltd..

All rights reserved.

 

DOI: 10.1016/S1473-3099(20)30928-2

PMCID: PMC7907671

PMID: 33508221 [Indexed for MEDLINE]

 

21. J Clin Oncol. 2021 Mar 10;39(8):931-939. doi: 10.1200/JCO.20.03364. Epub 2021

Jan 27.

 

Radiation Therapy for Small-Cell Lung Cancer: ASCO Guideline Endorsement of an

ASTRO Guideline.

 

Daly ME(1), Ismaila N(2), Decker RH(3), Higgins K(4), Owen D(5), Saxena A(6),

Franklin GE(7), Donaldson D(8), Schneider BJ(9).

 

Author information:

(1)University of California, Davis, CA.

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

(3)Yale School of Medicine, New Haven, CT.

(4)Emory University School of Medicine, Atlanta, GA.

(5)Mayo Clinic, Rochester, MN.

(6)Weill Cornell Medicine, New York, NY.

(7)New Mexico Cancer Center, Albuquerque, NM.

(8)Dusty Joy Foundation, High Point, NC.

(9)University of Michigan School of Medicine, Ann Arbor, MI.

 

PURPOSE: The American Society for Radiation Oncology (ASTRO) produced an

evidence-based guideline on radiation therapy (RT) for small-cell lung cancer

(SCLC). Because of the relevance of this topic to ASCO membership, ASCO reviewed

the guideline, applying a set of procedures and policies used to critically

examine guidelines developed by other organizations.

METHODS: The ASTRO guideline on RT for SCLC was reviewed for developmental rigor

by methodologists. Then, an ASCO Expert Panel reviewed the content and the

recommendations.

RESULTS: The ASCO Expert Panel determined that the recommendations from ASTRO

guideline on RT for SCLC, published in June 2020, are clear, thorough, and based

upon the most relevant scientific evidence. ASCO endorsed ASTRO guideline on RT

for SCLC with a few discussion points.

RECOMMENDATIONS: Recommendations addressed thoracic radiotherapy for

limited-stage SCLC, role of stereotactic body radiotherapy in stage I or II

node-negative SCLC, prophylactic cranial radiotherapy, and thoracic

consolidation for extensive-stage SCLC.Additional information is available at

www.asco.org/thoracic-cancer-guidelines.

 

DOI: 10.1200/JCO.20.03364

PMID: 33502911

 

22. J Clin Oncol. 2021 Mar 1;39(7):723-733. doi: 10.1200/JCO.20.01605. Epub 2021 Jan 15.

 

Five-Year Outcomes From the Randomized, Phase III Trials CheckMate 017 and 057:

Nivolumab Versus Docetaxel in Previously Treated Non-Small-Cell Lung Cancer.

 

Borghaei H(1), Gettinger S(2), Vokes EE(3), Chow LQM(4), Burgio MA(5), de Castro

Carpeno J(6), Pluzanski A(7), Arrieta O(8), Frontera OA(9), Chiari R(10), Butts

C(11), Wójcik-Tomaszewska J(12), Coudert B(13), Garassino MC(14), Ready N(15),

Felip E(16), García MA(17), Waterhouse D(18), Domine M(19), Barlesi F(20),

Antonia S(21), Wohlleber M(22), Gerber DE(23), Czyzewicz G(24), Spigel DR(25),

Crino L(5), Eberhardt WEE(26), Li A(27), Marimuthu S(27), Brahmer J(28).

 

Author information:

(1)Fox Chase Cancer Center, Philadelphia, PA.

(2)Yale Comprehensive Cancer Center, New Haven, CT.

(3)Univeristy of Chicago Medicine and Biologic Sciences Division, Chicago, IL.

Erratum in

    J Clin Oncol. 2021 Apr 1;39(10):1190.

 

PURPOSE: Immunotherapy has revolutionized the treatment of advanced

non-small-cell lung cancer (NSCLC). In two phase III trials (CheckMate 017 and

CheckMate 057), nivolumab showed an improvement in overall survival (OS) and

favorable safety versus docetaxel in patients with previously treated, advanced

squamous and nonsquamous NSCLC, respectively. We report 5-year pooled efficacy

and safety from these trials.

METHODS: Patients (N = 854; CheckMate 017/057 pooled) with advanced NSCLC, ECOG

PS ≤ 1, and progression during or after first-line platinum-based chemotherapy

were randomly assigned 1:1 to nivolumab (3 mg/kg once every 2 weeks) or

docetaxel (75 mg/m2 once every 3 weeks) until progression or unacceptable

toxicity. The primary end point for both trials was OS; secondary end points

included progression-free survival (PFS) and safety. Exploratory landmark

analyses were investigated.

RESULTS: After the minimum follow-up of 64.2 and 64.5 months for CheckMate 017

and 057, respectively, 50 nivolumab-treated patients and nine docetaxel-treated

patients were alive. Five-year pooled OS rates were 13.4% versus 2.6%,

respectively; 5-year PFS rates were 8.0% versus 0%, respectively.

Nivolumab-treated patients without disease progression at 2 and 3 years had an

82.0% and 93.0% chance of survival, respectively, and a 59.6% and 78.3% chance

of remaining progression-free at 5 years, respectively. Treatment-related

adverse events (TRAEs) were reported in 8 of 31 (25.8%) nivolumab-treated

patients between 3-5 years of follow-up, seven of whom experienced new events;

one (3.2%) TRAE was grade 3, and there were no grade 4 TRAEs.

CONCLUSION: At 5 years, nivolumab continued to demonstrate a survival benefit

versus docetaxel, exhibiting a five-fold increase in OS rate, with no new safety

signals. These data represent the first report of 5-year outcomes from

randomized phase III trials of a programmed death-1 inhibitor in previously

treated, advanced NSCLC.

 

DOI: 10.1200/JCO.20.01605

PMID: 33449799

 

23. J Clin Oncol. 2021 Mar 1;39(7):713-722. doi: 10.1200/JCO.20.01820. Epub 2020 Dec 17.

 

Gefitinib Versus Vinorelbine Plus Cisplatin as Adjuvant Treatment for Stage

II-IIIA (N1-N2) EGFR-Mutant NSCLC: Final Overall Survival Analysis of CTONG1104

Phase III Trial.

 

Zhong WZ(1), Wang Q(2), Mao WM(3), Xu ST(2), Wu L(4), Wei YC(5), Liu YY(6), Chen

C(7), Cheng Y(8), Yin R(9), Yang F(10), Ren SX(11), Li XF(12), Li J(13), Huang

C(14), Liu ZD(15), Xu S(16), Chen KN(17), Xu SD(18), Liu LX(19), Yu P(20), Wang

BH(21), Ma HT(22), Yang JJ(1), Yan HH(1), Yang XN(1), Liu SY(1), Zhou Q(1), Wu

YL(1).

 

Author information:

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

Guangdong Academy of Medical Sciences, Guangzhou, China.

(2)Fudan University Affiliated Zhongshan Hospital, Shanghai, China.

(3)Zhejiang Cancer Hospital, Hangzhou, China.

PURPOSE: ADJUVANT-CTONG1104 (ClinicalTrials.gov identifier: NCT01405079), a

randomized phase III trial, showed that adjuvant gefitinib treatment

significantly improved disease-free survival (DFS) versus vinorelbine plus

cisplatin (VP) in patients with epidermal growth factor receptor (EGFR)

mutation-positive resected stage II-IIIA (N1-N2) non-small-cell lung cancer

(NSCLC). Here, we report the final overall survival (OS) results.

METHODS: From September 2011 to April 2014, 222 patients from 27 sites were

randomly assigned 1:1 to adjuvant gefitinib (n = 111) or VP (n = 111). Patients

with resected stage II-IIIA (N1-N2) NSCLC and EGFR-activating mutation were

enrolled, receiving gefitinib for 24 months or VP every 3 weeks for four cycles.

The primary end point was DFS (intention-to-treat [ITT] population). Secondary

end points included OS, 3-, 5-year (y) DFS rates, and 5-year OS rate. Post hoc

analysis was conducted for subsequent therapy data.

RESULTS: Median follow-up was 80.0 months. Median OS (ITT) was 75.5 and 62.8

months with gefitinib and VP, respectively (hazard ratio [HR], 0.92; 95% CI,

0.62 to 1.36; P = .674); respective 5-year OS rates were 53.2% and 51.2% (P =

.784). Subsequent therapy was administered upon progression in 68.4% and 73.6%

of patients receiving gefitinib and VP, respectively. Subsequent targeted

therapy contributed most to OS (HR, 0.23; 95% CI, 0.14 to 0.38) compared with no

subsequent therapy. Updated 3y DFS rates were 39.6% and 32. 5% with gefitinib

and VP (P = .316) and 5y DFS rates were 22. 6% and 23.2% (P = .928),

respectively.

CONCLUSION: Adjuvant therapy with gefitinib in patients with early-stage NSCLC

and EGFR mutation demonstrated improved DFS over standard of care chemotherapy.

Although this DFS advantage did not translate to a significant OS difference, OS

with adjuvant gefitinib was one of the longest observed in this patient group

compared with historic data.

 

DOI: 10.1200/JCO.20.01820

PMID: 33332190

 

24. Lancet Infect Dis. 2021 Mar;21(3):376-384. doi: 10.1016/S1473-3099(20)30598-3.

Epub 2020 Dec 11.

 

Comparing accuracy of lipoarabinomannan urine tests for diagnosis of pulmonary

tuberculosis in children from four African countries: a cross-sectional study.

 

Nkereuwem E(1), Togun T(2), Gomez MP(1), Székely R(3), Macé A(3), Jobe D(1),

Schumacher SG(3), Kampmann B(4), Denkinger CM(5); Reach4KidsAfrica (R4KA)

Consortium.

 

Author information:

(1)Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at

London School of Hygiene & Tropical Medicine, Banjul, The Gambia.

(2)Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at

London School of Hygiene & Tropical Medicine, Banjul, The Gambia; Faculty of

Infectious and Tropical Diseases, London, UK.

(3)FIND (Foundation for Innovative New Diagnostics), Geneva, Switzerland.

Comment in

    Lancet Infect Dis. 2021 Mar;21(3):302-303.

 

BACKGROUND: A sensitive and specific non-sputum-based test would be

groundbreaking for the diagnosis of childhood tuberculosis. We assessed side by

side the diagnostic accuracy of the urine-based lipoarabinomannan assays

Fujifilm SILVAMP TB LAM (FujiLAM) and Alere Determine TB LAM Ag (AlereLAM) for

detection of childhood tuberculosis.

METHODS: In this cross-sectional study, we tested urine samples from children

younger than 15 years with presumed pulmonary tuberculosis. Children were

consecutively recruited from four dedicated outpatient childhood tuberculosis

clinics in The Gambia, Mali, Nigeria, and Tanzania. Biobanked urine samples were

thawed and tested using FujiLAM and AlereLAM assays. We measured diagnostic

performance against a microbiological reference standard (confirmed

tuberculosis) and a composite reference standard (confirmed and unconfirmed

tuberculosis). Sensitivity and specificity were estimated with bivariate

random-effects meta-analyses.

FINDINGS: Between July 1, 2017, and Dec 1, 2018, we obtained and stored urine

samples from 415 children. 63 (15%) children had confirmed tuberculosis, 113

(27%) had unconfirmed tuberculosis, and 239 (58%) were unlikely to have

tuberculosis. 61 children were HIV-positive (prevalence 15%). Using the

microbiological reference standard, the sensitivity of FujiLAM was 64·9% (95% CI

43·7-85·2; positive in 40 of 63 confirmed samples) and the sensitivity of

AlereLAM was 30·7% (8·6-61·6; 19 of 63). The specificity of FujiLAM was 83·8%

(95% CI 76·5-89·4; negative in 297 of 352 unconfirmed and unlikely samples) and

the specificity of AlereLAM was 87·8% (79·0-93·7; 312 of 352). Against the

composite reference standard, both assays had decreased sensitivity; the

sensitivity of FujiLAM was 32·9% (95% CI 24·6-41·9; positive in 58 of 176

confirmed and unconfirmed samples) and the sensitivity of AlereLAM was 20·2%

(12·3-29·4; 36 of 176). The specificity of FujiLAM was 83·3% (95% CI 71·8-91·7;

negative in 202 of 239 unlikely samples) and the specificity of AlereLAM was

90·0% (81·6-95·6; 216 of 239).

INTERPRETATION: By comparison with AlereLAM, FujiLAM showed higher sensitivity

and similar specificity. FujiLAM could potentially add value to the rapid

diagnosis of tuberculosis in children.

FUNDING: German Federal Ministry of Education and Research, the Global Health

Innovative Technology Fund, the UK Research and Innovation Global Challenges

Research Fund, and the UK Medical Research Council.

 

Copyright © 2021 Elsevier Ltd. All rights reserved.

 

DOI: 10.1016/S1473-3099(20)30598-3

PMID: 33316214 [Indexed for MEDLINE]

 

 

上一篇: No. 5

下一篇: No.3