1
|
Mazzilli SA, Rahal Z, Rouhani MJ, Janes SM, Kadara H, Dubinett SM, Spira AE. Translating premalignant biology to accelerate non-small-cell lung cancer interception. Nat Rev Cancer 2025; 25:379-392. [PMID: 39994467 DOI: 10.1038/s41568-025-00791-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/02/2025] [Indexed: 02/26/2025]
Abstract
Over the past decade, substantial progress has been made in the development of targeted and immune-based therapies for patients with advanced non-small-cell lung cancer. To further improve outcomes for patients with lung cancer, identifying and intercepting disease at the earliest and most curable stages are crucial next steps. With the recent implementation of low-dose computed tomography scan screening in populations at high risk, there is an emerging unmet need for new diagnostic, prognostic and therapeutic tools to help treat patients suspected of harbouring premalignant lesions and minimally invasive non-small-cell lung cancer. Continued advances in the identification of the earliest drivers of lung carcinogenesis are poised to address these unmet needs. Employing multimodal approaches to chart the temporal and spatial maps of the molecular events driving lung premalignant lesion progression will refine our understanding of early carcinogenesis. Elucidating the molecular drivers of premalignancy is critical to the development of biomarkers to detect those incubating a premalignant lesion, to stratify risk for progression to invasive cancer and to identify novel therapeutic targets to intercept that process. In this Review, we summarize emerging insights into the earliest cellular and molecular events associated with lung squamous and adenocarcinoma carcinogenesis and highlight the growing opportunity for translating these insights into clinical tools for early detection and disease interception to transform the outcomes for those at risk for lung cancer.
Collapse
Affiliation(s)
- Sarah A Mazzilli
- Sectional Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
| | - Zahraa Rahal
- Division of Pathology-Lab Medicine, Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Maral J Rouhani
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Sam M Janes
- Lungs for Living Research Centre, UCL Respiratory, University College London, London, UK
| | - Humam Kadara
- Division of Pathology-Lab Medicine, Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX, USA
| | - Steven M Dubinett
- Division of Pulmonary and Critical Care, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, and Greater Los Angeles Healthcare System, Los Angeles, CA, USA
| | - Avrum E Spira
- Sectional Computational Biomedicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA.
- Johnson & Johnson Innovative Medicine, Boston, MA, USA.
| |
Collapse
|
2
|
Brik A, Wichert K, Weber DG, Szafranski K, Rozynek P, Meier S, Ko YD, Büttner R, Gerwert K, Behrens T, Brüning T, Johnen G. Assessment of MYC and TERT copy number variations in lung cancer using digital PCR. BMC Res Notes 2023; 16:279. [PMID: 37858127 PMCID: PMC10585721 DOI: 10.1186/s13104-023-06566-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/11/2023] [Indexed: 10/21/2023] Open
Abstract
OBJECTIVE Lung cancer is the second most frequent cancer type and the most common cause of cancer-related deaths worldwide. Alteration of gene copy numbers are associated with lung cancer and the determination of copy number variations (CNV) is appropriate for the discrimination between tumor and non-tumor tissue in lung cancer. As telomerase reverse transcriptase (TERT) and v-myc avian myelocytomatosis viral oncogene homolog (MYC) play a role in lung cancer the aims of this study were the verification of our recent results analyzing MYC CNV in tumor and non-tumor tissue of lung cancer patients using an independent study group and the assessment of TERT CNV as an additional marker. RESULTS TERT and MYC status was analyzed using digital PCR (dPCR) in tumor and adjacent non-tumor tissue samples of 114 lung cancer patients. The difference between tumor and non-tumor samples were statistically significant (p < 0.0001) for TERT and MYC. Using a predefined specificity of 99% a sensitivity of 41% and 51% was observed for TERT and MYC, respectively. For the combination of TERT and MYC the overall sensitivity increased to 60% at 99% specificity. We demonstrated that a combination of markers increases the performance in comparison to individual markers. Additionally, the determination of CNV using dPCR might be an appropriate tool in precision medicine.
Collapse
Affiliation(s)
- Alexander Brik
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany.
| | - Katharina Wichert
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Daniel G Weber
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Katja Szafranski
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Peter Rozynek
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Swetlana Meier
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Yon-Dschun Ko
- Department of Internal Medicine, Johanniter-Kliniken Bonn GmbH, Bonn, Germany
| | - Reinhard Büttner
- Institute of Pathology, Medical Faculty and Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Klaus Gerwert
- Center for Protein Diagnostics (PRODI), Department of Biophysics, Ruhr University Bochum, Bochum, Germany
| | - Thomas Behrens
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Thomas Brüning
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| | - Georg Johnen
- Institute for Prevention and Occupational Medicine of the German Social Accident Insurance - Institute of the Ruhr University Bochum (IPA), Bochum, Germany
| |
Collapse
|
3
|
Jonsson S, Franklin WA, Varella-Garcia M, Kennedy TC, Merrick D, Matney KD, Oskarsdottir GN, Saemundsson A, Keith RL, Bunn PA, Miller YE. Prevalence, molecular markers, and outcome of bronchial squamous carcinoma in situ in high-risk subjects. APMIS 2023; 131:513-527. [PMID: 37608782 DOI: 10.1111/apm.13345] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 07/14/2023] [Indexed: 08/24/2023]
Abstract
Bronchial squamous carcinoma in situ (CIS) is a preinvasive lesion that is thought to precede invasive carcinoma. We conducted prospective autofluorescence and white light bronchoscopy trials between 1992 and 2016 to assess the prevalence, molecular markers, and outcome of individuals with CIS and other preneoplastic bronchial lesions. Biopsies were evaluated at multiple levels and selected biopsies were tested for aneuploidy and DNA sequenced for TP53 mutation. Thirty-one individuals with CIS were identified. Twenty-two cases of CIS occurred in association with concurrent invasive carcinomas. Seven of the invasive tumors were radiographically occult. In two cases, CIS spread from the focus of invasive carcinoma into contralateral lung lobes, forming secondary invasive tumors. In nine cases, CIS occurred as isolated lesions and one progressed to invasive squamous carcinoma at the same site 40 months after discovery. In a second case, CIS was a precursor of carcinoma at a separate site in a different lobe. In seven cases CIS regressed to a lower grade or disappeared. High level chromosomal aneusomy was often associated with TP53 mutation and with invasive carcinoma. CIS most often occurs in association with invasive squamous carcinoma and may extend along the airways into distant lobes. In rare cases, CIS may be observed to directly transform into invasive carcinoma. CIS may be indicative of invasive tumor at a separate distant site. Isolated CIS may regress. Molecular changes parallel histological changes in CIS and may be used to map clonal expansion in the airways.
Collapse
Affiliation(s)
- Steinn Jonsson
- Department of Medicine, University of Colorado Health Sciences Center, Denver, CO, USA
- Department of Medicine, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Wilbur A Franklin
- Department of Pathology, University of Colorado Health Sciences Center, Denver, CO, USA
| | | | - Timothy C Kennedy
- Department of Medicine, Presbyterian/St Luke's Health One Medical Center, Denver, CO, USA
| | - Daniel Merrick
- Department of Pathology, University of Colorado Health Sciences Center, Denver, CO, USA
| | - Kathryn D Matney
- Department of Pathology, University of Colorado Health Sciences Center, Denver, CO, USA
| | - Gudrun N Oskarsdottir
- Department of Medicine, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Arni Saemundsson
- Department of Medicine, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Robert L Keith
- Department of Medicine, University of Colorado Health Sciences Center, Denver, CO, USA
- Pulmonary Division, Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA
| | - Paul A Bunn
- Department of Medicine, University of Colorado Health Sciences Center, Denver, CO, USA
| | - York E Miller
- Department of Medicine, University of Colorado Health Sciences Center, Denver, CO, USA
- Pulmonary Division, Department of Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO, USA
| |
Collapse
|
4
|
Keith RL, Miller YE, Ghosh M, Franklin WA, Nakachi I, Merrick DT. Lung cancer: Premalignant biology and medical prevention. Semin Oncol 2022; 49:254-260. [PMID: 35305831 DOI: 10.1053/j.seminoncol.2022.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 02/08/2022] [Indexed: 11/11/2022]
Abstract
Lung cancer (both adenocarcinoma and squamous cell) progress through a series of pre-malignant histologic changes before the development of invasive disease. Each of these carcinogenic cascades is defined by genetic and epigenetic alterations in pulmonary epithelial cells. Additionally, alterations in the immune response, progenitor cell function, mutational burden, and microenvironmental mediated survival of mutated clones contribute to the risk of pre-malignant lesions progressing to cancer. Medical preventions studies have been completed and current and future trials are informed by the improved understanding of pre-malignancy. This will lead to precision chemoprevention trials based on lesional biology and histologic characteristics.
Collapse
Affiliation(s)
- R L Keith
- Division of Pulmonary Sciences and Critical Care Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO.
| | - Y E Miller
- Division of Pulmonary Sciences and Critical Care Medicine, Rocky Mountain Regional Veterans Affairs Medical Center, Aurora, CO; Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - M Ghosh
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - Wilbur A Franklin
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO
| | - I Nakachi
- Department of Pulmonary Medicine, Keio University, Tokyo, Japan
| | - D T Merrick
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
5
|
Kammer MN, Deppen SA, Antic S, Jamshedur Rahman S, Eisenberg R, Maldonado F, Aldrich MC, Sandler KL, Landman B, Massion PP, Grogan EL. The impact of the lung EDRN-CVC on Phase 1, 2, & 3 biomarker validation studies. Cancer Biomark 2022; 33:449-465. [DOI: 10.3233/cbm-210382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The Early Detection Research Network’s (EDRN) purpose is to discover, develop and validate biomarkers and imaging methods to detect early-stage cancers or at-risk individuals. The EDRN is composed of sites that fall into four categories: Biomarker Developmental Laboratories (BDL), Biomarker Reference Laboratories (BRL), Clinical Validation Centers (CVC) and Data Management and Coordinating Centers. Each component has a crucial role to play within the mission of the EDRN. The primary role of the CVCs is to support biomarker developers through validation trials on promising biomarkers discovered by both EDRN and non-EDRN investigators. The second round of funding for the EDRN Lung CVC at Vanderbilt University Medical Center (VUMC) was funded in October 2016 and we intended to accomplish the three missions of the CVCs: To conduct innovative research on the validation of candidate biomarkers for early cancer detection and risk assessment of lung cancer in an observational study; to compare biomarker performance; and to serve as a resource center for collaborative research within the Network and partner with established EDRN BDLs and BRLs, new laboratories and industry partners. This report outlines the impact of the VUMC EDRN Lung CVC and describes the role in promoting and validating biological and imaging biomarkers.
Collapse
Affiliation(s)
- Michael N. Kammer
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Stephen A. Deppen
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA
| | - Sanja Antic
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - S.M. Jamshedur Rahman
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rosana Eisenberg
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fabien Maldonado
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Melinda C. Aldrich
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kim L. Sandler
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bennett Landman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Pierre P. Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Eric L. Grogan
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
- Tennessee Valley Healthcare System, Veterans Affairs, Nashville, TN, USA
| |
Collapse
|
6
|
Keshavarz-Fathi M, Rezaei N. Cancer Immunoprevention: Current Status and Future Directions. Arch Immunol Ther Exp (Warsz) 2021; 69:3. [PMID: 33638703 DOI: 10.1007/s00005-021-00604-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 02/06/2021] [Indexed: 12/24/2022]
Abstract
Cancer is one of the most serious diseases affecting health and the second leading cause of death worldwide. Despite the development of various therapeutic modalities to deal with cancer, limited improvement in overall survival of patients has been yielded. Since there is no certain cure for cancer, detection of premalignant lesions, and prevention of their progression are vital to the decline of high morbidity and mortality of cancer. Among approaches to cancer prevention, immunoprevention has gained further attention in recent years. Deep understanding of the tumor/immune system interplay and successful prevention of virally-induced malignancies by vaccines have paved the way toward broadening cancer immunoprevention application. The identification of tumor antigens in premalignant lesions was the turning point in cancer immunoprevention that led to designing preventive vaccines for various malignancies including multiple myeloma, colorectal, and breast cancer. In addition to vaccines, immune checkpoint inhibitors are also being tested for the prevention of oral squamous cell carcinoma (SCC), and imiquimod which is an established drug for the prevention of skin SCC, is a non-specific immunomodulator. Herein, to provide a bench-to-bedside understanding of cancer immunoprevention, we will review the role of the immune system in suppression and promotion of tumors, immunoprevention of virally-induced cancers, identification of tumor antigens in premalignant lesions, and clinical advances of cancer immunoprevention.
Collapse
Affiliation(s)
- Mahsa Keshavarz-Fathi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Dr. Qarib St, Keshavarz Blvd, 14194, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| |
Collapse
|
7
|
Katz RL, Zaidi TM, Pujara D, Shanbhag ND, Truong D, Patil S, Mehran RJ, El-Zein RA, Shete SS, Kuban JD. Identification of circulating tumor cells using 4-color fluorescence in situ hybridization: Validation of a noninvasive aid for ruling out lung cancer in patients with low-dose computed tomography-detected lung nodules. Cancer Cytopathol 2020; 128:553-562. [PMID: 32320527 DOI: 10.1002/cncy.22278] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 01/09/2020] [Accepted: 02/10/2020] [Indexed: 02/03/2023]
Abstract
BACKGROUND Approximately one third of needle biopsies that are performed to rule out malignancy of indeterminate pulmonary nodules detected radiologically during lung cancer screening are negative, thus exposing cancer-free patients to risks of pneumothorax, bleeding, and infection. A noninvasive confirmatory tool (eg, liquid biopsy) is urgently needed in the lung cancer diagnosis setting to stratify patients who should receive biopsy versus those who should be monitored. METHODS A novel antigen-independent, 4-color fluorescence in situ hybridization (FISH)-based method was developed to detect circulating tumor cells (CTCs) with abnormalities in gene copy numbers in mononuclear cell-enriched peripheral blood samples from patients with (n = 107) and without (n = 100) lung cancer. RESULTS Identification of CTCs using FISH probes at 10q22.3/CEP10 and 3p22.1/3q29 detected lung cancer cases with 94.2% accuracy, 89% sensitivity, and 100% specificity compared with biopsy. CONCLUSION The high accuracy of this liquid biopsy method suggests that it may be used as a noninvasive decision tool to reduce the frequency of unnecessary needle biopsy in patients with benign pulmonary lesions.
Collapse
Affiliation(s)
- Ruth L Katz
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Tanweer M Zaidi
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Deep Pujara
- Department of Neurology, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Namita D Shanbhag
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Duy Truong
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shekhar Patil
- Department of Internal Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, Texas
| | - Reza J Mehran
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Randa A El-Zein
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Radiology, Houston Methodist Research Institute, Houston, Texas
| | - Sanjay S Shete
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joshua D Kuban
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.,Department of Interventional Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
8
|
Beane JE, Mazzilli SA, Campbell JD, Duclos G, Krysan K, Moy C, Perdomo C, Schaffer M, Liu G, Zhang S, Liu H, Vick J, Dhillon SS, Platero SJ, Dubinett SM, Stevenson C, Reid ME, Lenburg ME, Spira AE. Molecular subtyping reveals immune alterations associated with progression of bronchial premalignant lesions. Nat Commun 2019; 10:1856. [PMID: 31015447 PMCID: PMC6478943 DOI: 10.1038/s41467-019-09834-2] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 03/28/2019] [Indexed: 12/13/2022] Open
Abstract
Bronchial premalignant lesions (PMLs) are precursors of lung squamous cell carcinoma, but have variable outcome, and we lack tools to identify and treat PMLs at risk for progression to cancer. Here we report the identification of four molecular subtypes of PMLs with distinct differences in epithelial and immune processes based on RNA-Seq profiling of endobronchial biopsies from high-risk smokers. The Proliferative subtype is enriched with bronchial dysplasia and exhibits up-regulation of metabolic and cell cycle pathways. A Proliferative subtype-associated gene signature identifies subjects with Proliferative PMLs from normal-appearing uninvolved large airway brushings with high specificity. In progressive/persistent Proliferative lesions expression of interferon signaling and antigen processing/presentation pathways decrease and immunofluorescence indicates a depletion of innate and adaptive immune cells compared with regressive lesions. Molecular biomarkers measured in PMLs or the uninvolved airway can enhance histopathological grading and suggest immunoprevention strategies for intercepting the progression of PMLs to lung cancer.
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/pharmacology
- Antineoplastic Agents, Immunological/therapeutic use
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/immunology
- Biopsy
- Bronchi/diagnostic imaging
- Bronchi/immunology
- Bronchi/pathology
- Bronchoscopy
- Carcinoma, Bronchogenic/genetics
- Carcinoma, Bronchogenic/immunology
- Carcinoma, Bronchogenic/pathology
- Carcinoma, Bronchogenic/prevention & control
- Cohort Studies
- Datasets as Topic
- Disease Progression
- Early Detection of Cancer/methods
- Gene Expression Profiling
- Gene Expression Regulation, Neoplastic/immunology
- Gene Regulatory Networks/genetics
- Gene Regulatory Networks/immunology
- Humans
- Immunity, Cellular/drug effects
- Immunity, Cellular/genetics
- Lung Neoplasms/genetics
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Mass Screening/methods
- Middle Aged
- Precancerous Conditions/diagnostic imaging
- Precancerous Conditions/genetics
- Precancerous Conditions/immunology
- Precancerous Conditions/pathology
- RNA, Messenger/genetics
- Respiratory Mucosa/cytology
- Respiratory Mucosa/diagnostic imaging
- Respiratory Mucosa/immunology
- Respiratory Mucosa/pathology
- Sequence Analysis, RNA
- T-Lymphocytes/immunology
- Tomography, X-Ray Computed
- Up-Regulation
Collapse
Affiliation(s)
| | | | | | - Grant Duclos
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Kostyantyn Krysan
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | | | | | | | - Gang Liu
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Sherry Zhang
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Hanqiao Liu
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Jessica Vick
- Boston University School of Medicine, Boston, MA, 02118, USA
| | | | | | - Steven M Dubinett
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | | | - Mary E Reid
- Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14203, USA
| | - Marc E Lenburg
- Boston University School of Medicine, Boston, MA, 02118, USA
| | - Avrum E Spira
- Boston University School of Medicine, Boston, MA, 02118, USA
- Johnson and Johnson Innovation, Cambridge, MA, 02142, USA
| |
Collapse
|
9
|
Merrick DT, Edwards MG, Franklin WA, Sugita M, Keith RL, Miller YE, Friedman MB, Dwyer-Nield LD, Tennis MA, O'Keefe MC, Donald EJ, Malloy JM, van Bokhoven A, Wilson S, Koch PJ, O'Shea C, Coldren C, Orlicky DJ, Lu X, Baron AE, Hickey G, Kennedy TC, Powell R, Heasley L, Bunn PA, Geraci M, Nemenoff RA. Altered Cell-Cycle Control, Inflammation, and Adhesion in High-Risk Persistent Bronchial Dysplasia. Cancer Res 2018; 78:4971-4983. [PMID: 29997230 PMCID: PMC6147150 DOI: 10.1158/0008-5472.can-17-3822] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 05/16/2018] [Accepted: 07/06/2018] [Indexed: 01/01/2023]
Abstract
Persistent bronchial dysplasia is associated with increased risk of developing invasive squamous cell carcinoma (SCC) of the lung. In this study, we hypothesized that differences in gene expression profiles between persistent and regressive bronchial dysplasia would identify cellular processes that underlie progression to SCC. RNA expression arrays comparing baseline biopsies from 32 bronchial sites that persisted/progressed to 31 regressive sites showed 395 differentially expressed genes [ANOVA, FDR ≤ 0.05). Thirty-one pathways showed significantly altered activity between the two groups, many of which were associated with cell-cycle control and proliferation, inflammation, or epithelial differentiation/cell-cell adhesion. Cultured persistent bronchial dysplasia cells exhibited increased expression of Polo-like kinase 1 (PLK1), which was associated with multiple cell-cycle pathways. Treatment with PLK1 inhibitor induced apoptosis and G2-M arrest and decreased proliferation compared with untreated cells; these effects were not seen in normal or regressive bronchial dysplasia cultures. Inflammatory pathway activity was decreased in persistent bronchial dysplasia, and the presence of an inflammatory infiltrate was more common in regressive bronchial dysplasia. Regressive bronchial dysplasia was also associated with trends toward overall increases in macrophages and T lymphocytes and altered polarization of these inflammatory cell subsets. Increased desmoglein 3 and plakoglobin expression was associated with higher grade and persistence of bronchial dysplasia. These results identify alterations in the persistent subset of bronchial dysplasia that are associated with high risk for progression to invasive SCC. These alterations may serve as strong markers of risk and as effective targets for lung cancer prevention.Significance: Gene expression profiling of high-risk persistent bronchial dysplasia reveals changes in cell-cycle control, inflammatory activity, and epithelial differentiation/cell-cell adhesion that may underlie progression to invasive SCC. Cancer Res; 78(17); 4971-83. ©2018 AACR.
Collapse
Affiliation(s)
- Daniel T Merrick
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Michael G Edwards
- Department of Medicine/Division of Pulmonary Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Wilbur A Franklin
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Michio Sugita
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Robert L Keith
- Department of Medicine/Division of Pulmonary Medicine, Denver Veterans Affairs Medical Center, Aurora, Colorado
- Department of Medicine/Division of Pulmonary Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - York E Miller
- Department of Medicine/Division of Pulmonary Medicine, Denver Veterans Affairs Medical Center, Aurora, Colorado
- Department of Medicine/Division of Pulmonary Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Micah B Friedman
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lori D Dwyer-Nield
- Department of Medicine/Division of Pulmonary Medicine, Denver Veterans Affairs Medical Center, Aurora, Colorado
- School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Meredith A Tennis
- Department of Medicine/Division of Pulmonary Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mary C O'Keefe
- Department of Pathology, Denver Health Medical Center, Denver, Colorado
| | - Elizabeth J Donald
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jessica M Malloy
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Adrie van Bokhoven
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Storey Wilson
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Peter J Koch
- Department of Regenerative Medicine and Stem Cell Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Charlene O'Shea
- Department of Regenerative Medicine and Stem Cell Research, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - David J Orlicky
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Xian Lu
- Department of Biostatistics and Informatics, Colorado School of Public Health, Denver, Colorado
| | - Anna E Baron
- Department of Biostatistics and Informatics, Colorado School of Public Health, Denver, Colorado
| | - Greg Hickey
- Department of Medicine/Division of Pulmonary Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Timothy C Kennedy
- Department of Medicine/Division of Pulmonary Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Roger Powell
- School of Pharmacy, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lynn Heasley
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Paul A Bunn
- Department of Medicine/Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mark Geraci
- Department of Medicine, Indiana University, Bloomington, Indiana
| | - Raphael A Nemenoff
- Department of Medicine/Division of Pulmonary Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Department of Medicine, Division of Renal Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
10
|
Preinvasive disease of the airway. Cancer Treat Rev 2017; 58:77-90. [DOI: 10.1016/j.ctrv.2017.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/23/2017] [Accepted: 05/27/2017] [Indexed: 01/20/2023]
|
11
|
Barón AE, Kako S, Feser WJ, Malinowski H, Merrick D, Garg K, Malkoski S, Pretzel S, Siegfried JM, Franklin WA, Miller Y, Wolf HJ, Varella-Garcia M. Clinical Utility of Chromosomal Aneusomy in Individuals at High Risk of Lung Cancer. J Thorac Oncol 2017. [PMID: 28634123 DOI: 10.1016/j.jtho.2017.06.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Low-dose computed tomography screening for lung cancer has a high false-positive rate with frequent discovery of indeterminate pulmonary nodules. Noninvasive biomarkers are needed to reduce false positives and improve risk stratification. A retrospective longitudinal evaluation was performed to assess chromosomal aneusomy in sputum by fluorescence in situ hybridization (CA-FISH) in four nested case-control studies. METHODS Receiver operating characteristic analysis resulted in two grouped cohorts: a high-risk cohort (Colorado High-Risk Cohort and Colorado Nodule Cohort [68 case patients and 69 controls]) and a screening cohort (American College of Radiology Imaging Network/National Lung Screening Trial and Pittsburgh Lung Screening Study [97 case patients and 185 controls]). The CA-FISH assay was a four-target DNA panel encompassing the EGFR and v-myc avian myelocytomatosis viral oncogene homolog (MYC) genes, and the 5p15 and centromere 6 regions or the fibroblast growth factor 1 gene (FGFR1) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha gene (PIK3CA). A four-category scale (normal, probably normal, probably abnormal, and abnormal) was applied. Sensitivity, specificity, and positive and negative likelihood ratios (LRs) (with 95% confidence intervals [CIs]) were estimated for each cohort. RESULTS Sensitivity and specificity were, respectively, 0.67 (95% CI: 0.55-0.78) and 0.94 (95% CI: 0.85-0.98) for high-risk participants and 0.20 (95% CI: 0.13-0.30) and 0.84 (95% CI: 0.78-0.89) for screening participants. The positive and negative LRs were, respectively, 11.66 (95% CI: 4.44-30.63) and 0.34 (95% CI: 0.24-0.48) for high-risk participants and 1.36 (95% CI: 0.81-2.28) and 0.93 (95% CI: 0.83-1.05) for screening participants. CONCLUSION The high positive LR of sputum CA-FISH indicates that it could be a useful adjunct to low-dose computed tomography for lung cancer in high-risk settings. For screening, however, its low positive LR limits clinical utility. Prospective assessment of CA-FISH in the incidentally identified indeterminate nodule setting is ongoing in the Colorado Pulmonary Nodule Biomarker Trial.
Collapse
Affiliation(s)
- Anna E Barón
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Severine Kako
- Department of Medicine, Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - William J Feser
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Heather Malinowski
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel Merrick
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Kavita Garg
- Department of Radiology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Stephen Malkoski
- Department of Pulmonary and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Shannon Pretzel
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jill M Siegfried
- Department of Pharmacology, Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Wilbur A Franklin
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - York Miller
- Department of Pulmonary and Critical Care Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Veterans Affairs Medical Center, Denver, Colorado
| | - Holly J Wolf
- Department of Community and Behavioral Health, Colorado School of Public Health, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Marileila Varella-Garcia
- Department of Medicine, Division of Medical Oncology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado; Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
12
|
Correia LL, Johnson JA, McErlean P, Bauer J, Farah H, Rassl DM, Rintoul RC, Sethi T, Lavender P, Rawlins EL, Littlewood TD, Evan GI, McCaughan FM. SOX2 Drives Bronchial Dysplasia in a Novel Organotypic Model of Early Human Squamous Lung Cancer. Am J Respir Crit Care Med 2017; 195:1494-1508. [PMID: 28199128 PMCID: PMC5470746 DOI: 10.1164/rccm.201510-2084oc] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 02/15/2017] [Indexed: 12/25/2022] Open
Abstract
RATIONALE Improving the early detection and chemoprevention of lung cancer are key to improving outcomes. The pathobiology of early squamous lung cancer is poorly understood. We have shown that amplification of sex-determining region Y-box 2 (SOX2) is an early and consistent event in the pathogenesis of this disease, but its functional oncogenic potential remains uncertain. We tested the impact of deregulated SOX2 expression in a novel organotypic system that recreates the molecular and microenvironmental context in which squamous carcinogenesis occurs. OBJECTIVES (1) To develop an in vitro model of bronchial dysplasia that recapitulates key molecular and phenotypic characteristics of the human disease; (2) to test the hypothesis that SOX2 deregulation is a key early event in the pathogenesis of bronchial dysplasia; and (3) to use the model for studies on pathogenesis and chemoprevention. METHODS We engineered the inducible activation of oncogenes in immortalized bronchial epithelial cells. We used three-dimensional tissue culture to build an organotypic model of bronchial dysplasia. MEASUREMENTS AND MAIN RESULTS We recapitulated human bronchial dysplasia in vitro. SOX2 deregulation drives dysplasia, and loss of tumor promoter 53 is a cooperating genetic event that potentiates the dysplastic phenotype. Deregulated SOX2 alters critical genes implicated in hallmarks of cancer progression. Targeted inhibition of AKT prevents the initiation of the dysplastic phenotype. CONCLUSIONS In the appropriate genetic and microenvironmental context, acute deregulation of SOX2 drives bronchial dysplasia. This confirms its oncogenic potential in human cells and affords novel insights into the impact of SOX2 deregulation. This model can be used to test therapeutic agents aimed at chemoprevention.
Collapse
Affiliation(s)
| | | | - Peter McErlean
- Department of Asthma, Allergy, and Lung Biology, Guy’s Hospital, King’s College London, London, United Kingdom; and
| | - Julien Bauer
- Cambridge Genomic Services, Department of Pathology, and
| | - Hassan Farah
- Department of Asthma, Allergy, and Lung Biology, Guy’s Hospital, King’s College London, London, United Kingdom; and
| | | | - Robert C. Rintoul
- Department of Thoracic Oncology, Papworth Hospital Foundation National Health Service Trust, Papworth Everard, Cambridge, United Kingdom
| | - Tariq Sethi
- Department of Asthma, Allergy, and Lung Biology, Guy’s Hospital, King’s College London, London, United Kingdom; and
| | - Paul Lavender
- Department of Asthma, Allergy, and Lung Biology, Guy’s Hospital, King’s College London, London, United Kingdom; and
| | | | | | | | - Frank M. McCaughan
- Department of Biochemistry
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
- Department of Asthma, Allergy, and Lung Biology, Guy’s Hospital, King’s College London, London, United Kingdom; and
| |
Collapse
|
13
|
Beane J, Mazzilli SA, Tassinari AM, Liu G, Zhang X, Liu H, Buncio AD, Dhillon SS, Platero SJ, Lenburg ME, Reid ME, Lam S, Spira AE. Detecting the Presence and Progression of Premalignant Lung Lesions via Airway Gene Expression. Clin Cancer Res 2017; 23:5091-5100. [PMID: 28533227 DOI: 10.1158/1078-0432.ccr-16-2540] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 03/23/2017] [Accepted: 05/17/2017] [Indexed: 11/16/2022]
Abstract
Purpose: Lung cancer is the leading cause of cancer-related death in the United States. The molecular events preceding the onset of disease are poorly understood, and no effective tools exist to identify smokers with premalignant lesions (PMLs) that will progress to invasive cancer. Prior work identified molecular alterations in the smoke-exposed airway field of injury associated with lung cancer. Here, we focus on an earlier stage in the disease process leveraging the airway field of injury to study PMLs and its utility in lung cancer chemoprevention.Experimental Design: Bronchial epithelial cells from normal appearing bronchial mucosa were profiled by mRNA-Seq from subjects with (n = 50) and without (n = 25) PMLs. Using surrogate variable and gene set enrichment analysis, we identified genes, pathways, and lung cancer-related gene sets differentially expressed between subjects with and without PMLs. A computational pipeline was developed to build and test a chemoprevention-relevant biomarker.Results: We identified 280 genes in the airway field associated with the presence of PMLs. Among the upregulated genes, oxidative phosphorylation was strongly enriched, and IHC and bioenergetics studies confirmed pathway findings in PMLs. The relationship between PMLs and squamous cell carcinomas (SCC) was also confirmed using published lung cancer datasets. The biomarker performed well predicting the presence of PMLs (AUC = 0.92, n = 17), and changes in the biomarker score associated with progression/stability versus regression of PMLs (AUC = 0.75, n = 51).Conclusions: Transcriptomic alterations in the airway field of smokers with PMLs reflect metabolic and early lung SCC alterations and may be leveraged to stratify smokers at high risk for PML progression and monitor outcome in chemoprevention trials. Clin Cancer Res; 23(17); 5091-100. ©2017 AACR.
Collapse
Affiliation(s)
- Jennifer Beane
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts.
| | - Sarah A Mazzilli
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Anna M Tassinari
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Gang Liu
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Xiaohui Zhang
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Hanqiao Liu
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Anne Dy Buncio
- Department of Medicine, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Samjot S Dhillon
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Suso J Platero
- Janssen Research and Development, Spring House, Pennsylvania
| | - Marc E Lenburg
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| | - Mary E Reid
- Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York
| | - Stephen Lam
- Department of Medicine, BC Cancer Research Centre, Vancouver, British Columbia, Canada
| | - Avrum E Spira
- Department of Medicine, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
14
|
Mandrioli D, Belpoggi F, Silbergeld EK, Perry MJ. Aneuploidy: a common and early evidence-based biomarker for carcinogens and reproductive toxicants. Environ Health 2016; 15:97. [PMID: 27729050 PMCID: PMC5059969 DOI: 10.1186/s12940-016-0180-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 09/28/2016] [Indexed: 05/29/2023]
Abstract
Aneuploidy, defined as structural and numerical aberrations of chromosomes, continues to draw attention as an informative effect biomarker for carcinogens and male reproductive toxicants. It has been well documented that aneuploidy is a hallmark of cancer. Aneuploidies in oocytes and spermatozoa contribute to infertility, pregnancy loss and a number of congenital abnormalities, and sperm aneuploidy is associated with testicular cancer. It is striking that several carcinogens induce aneuploidy in somatic cells, and also adversely affect the chromosome compliment of germ cells. In this paper we review 1) the contributions of aneuploidy to cancer, infertility, and developmental abnormalities; 2) techniques for assessing aneuploidy in precancerous and malignant lesions and in sperm; and 3) the utility of aneuploidy as a biomarker for integrated chemical assessments of carcinogenicity, and reproductive and developmental toxicity.
Collapse
Affiliation(s)
- Daniele Mandrioli
- Cesare Maltoni Cancer Research Center, Ramazzini Institute, 40010 Bentivoglio, Bologna, Italy
| | - Fiorella Belpoggi
- Cesare Maltoni Cancer Research Center, Ramazzini Institute, 40010 Bentivoglio, Bologna, Italy
| | - Ellen K. Silbergeld
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, 21205 Baltimore, MD USA
| | - Melissa J. Perry
- Department of Environmental and Occupational Health, Milken Institute School of Public Health, The George Washington University, 950 New Hampshire Ave. NW, 4th Floor, Washington, DC 20052 USA
| |
Collapse
|
15
|
Merrick DT, Gao D, Miller YE, Keith RL, Baron AE, Feser W, Kennedy TC, Blatchford PJ, Braudrick S, Hirsch FR, Heasley L, Bunn PA, Franklin WA. Persistence of Bronchial Dysplasia Is Associated with Development of Invasive Squamous Cell Carcinoma. Cancer Prev Res (Phila) 2016; 9:96-104. [PMID: 26542061 PMCID: PMC4706769 DOI: 10.1158/1940-6207.capr-15-0305] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 10/26/2015] [Indexed: 12/22/2022]
Abstract
Bronchial dysplasia (BD), a presumed precursor of pulmonary squamous cell carcinoma (SCC), rarely progresses to invasive cancer. A high-risk cohort at the University of Colorado provided an opportunity to directly sample airway epithelium at mapped sites on successive bronchoscopies. We have hypothesized that persistent dysplastic lesions showing a similar or higher level of dysplasia on follow-up biopsy, are associated with increased risk for the development of SCC. Endoscopic biopsies from 188 high-risk subjects were histologically classified according to the current WHO classification for BD using a numeric histology score ranging from 1 to 8 representing normal bronchial mucosa through invasive lung cancer. Differences in follow-up histology scores were compared between sites classified by clinical, histologic, and immunohistochemical variables. Subjects with a higher frequency of sites that persist or progress to high-grade dysplasia (≥37.5% persist/progress, N = 35 versus <37.5% persist/progress, N = 114) show a significant association with development of incident invasive SCC (adjusted HR, 7.84; 95% confidence interval, 1.56-39.39), and those with incident lung SCC have adjusted mean follow-up histology scores 1.55 U higher than in subjects without lung cancer. Current smoking, elevated Ki67 growth fraction, histologic features of angiogenic squamous dysplasia (ASD) and higher histology score in baseline biopsies are significantly associated with increased follow-up histology scores. These results show that persistent BD is associated with the development of invasive SCC. Furthermore, increased expression of Ki67, the presence of angiogenic change and degree of baseline atypia are associated with persistence of BD.
Collapse
Affiliation(s)
- Daniel T Merrick
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
| | - Dexiang Gao
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - York E Miller
- Division of Pulmonary Medicine, Department of Medicine, Denver Veterans Affairs Medical Center, Denver, Colorado. Division of Pulmonary Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Robert L Keith
- Division of Pulmonary Medicine, Department of Medicine, Denver Veterans Affairs Medical Center, Denver, Colorado. Division of Pulmonary Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Anna E Baron
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado
| | - William Feser
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado
| | - Timothy C Kennedy
- Division of Pulmonary Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Patrick J Blatchford
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Sarah Braudrick
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, Colorado
| | - Fred R Hirsch
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lynn Heasley
- Department of Craniofacial Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Paul A Bunn
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Wilbur A Franklin
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
16
|
Cao Y, Liu Y, Yang X, Liu X, Han N, Zhang K, Lin D. Estimation of the Survival of Patients With Lung Squamous Cell Carcinoma Using Genomic Copy Number Aberrations. Clin Lung Cancer 2015; 17:68-74.e5. [PMID: 26427646 DOI: 10.1016/j.cllc.2015.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/04/2015] [Accepted: 08/11/2015] [Indexed: 01/25/2023]
Abstract
BACKGROUND Estimation of the survival of patients with lung squamous cell carcinoma (SCC) on the basis of histopathology is inadequate. The aim of this study was to identify genomic regions with potential value for estimating the prognosis of these patients. PATIENTS AND METHODS Depending on their survival time, 100 patients with primary lung SCC were separated into high- or low-risk prognostic groups, and their copy number aberrations (CNAs) were analyzed using array-comparative genomic hybridization (array-CGH). RESULTS We identified 123 CNA regions that were significantly associated with survival. Among these regions, some have been reported previously (eg, amplifications of 8p12, 3q27.1, and loss of 9p21.3 and 13q34) but others have never been reported. For example, gains of 3q27.1, 5p13.2, and 5p13.3 were found to be associated with a favorable prognosis, but patients harboring gains of 11q23.3, 11q13.1, and 14q32.3, and deletions of 3p21.3 and 9p21.3 tended to have poor survival. Among the 123 CNA regions, 41 were further selected to construct a survival estimation model that could effectively separate SCC patients into high- or low-risk groups with an accuracy of 92%, sensitivity of 90%, and specificity of 94%. The results of the array-CGH were further validated in an independent cohort of 45 formalin-fixed, paraffin-embedded specimens using real-time polymerase chain reaction. CONCLUSION A number of CNA regions were found to be associated with the survival of SCC patients, and we were able to construct a model to estimate prognosis on the basis of these regions. Assessment of these CNAs could potentially assist in clinical decision-making regarding adjuvant therapy after surgery.
Collapse
Affiliation(s)
- Yan Cao
- Department of Pathology, Plastic Surgery Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Yu Liu
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Xin Yang
- Department of Pathology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, P.R. China; Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, P.R. China
| | - XiangYang Liu
- Department of Thoracic Surgical Oncology, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Naijun Han
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Kaitai Zhang
- State Key Laboratory of Molecular Oncology, Department of Etiology and Carcinogenesis, Cancer Institute (Hospital), Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, P.R. China
| | - Dongmei Lin
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Pathology, Peking University Cancer Hospital and Institute, Beijing, P.R. China.
| |
Collapse
|
17
|
Hosgood HD, Song M, Hsiung CA, Yin Z, Shu XO, Wang Z, Chatterjee N, Zheng W, Caporaso N, Burdette L, Yeager M, Berndt SI, Landi MT, Chen CJ, Chang GC, Hsiao CF, Tsai YH, Chien LH, Chen KY, Huang MS, Su WC, Chen YM, Chen CH, Yang TY, Wang CL, Hung JY, Lin CC, Perng RP, Chen CY, Chen KC, Li YJ, Yu CJ, Chen YS, Chen YH, Tsai FY, Kim C, Seow WJ, Bassig BA, Wu W, Guan P, He Q, Gao YT, Cai Q, Chow WH, Xiang YB, Lin D, Wu C, Wu YL, Shin MH, Hong YC, Matsuo K, Chen K, Wong MP, Lu D, Jin L, Wang JC, Seow A, Wu T, Shen H, Fraumeni JF, Yang PC, Chang IS, Zhou B, Chanock SJ, Rothman N, Lan Q. Interactions between household air pollution and GWAS-identified lung cancer susceptibility markers in the Female Lung Cancer Consortium in Asia (FLCCA). Hum Genet 2015; 134:333-41. [PMID: 25566987 PMCID: PMC5537621 DOI: 10.1007/s00439-014-1528-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/29/2014] [Indexed: 10/24/2022]
Abstract
We previously carried out a multi-stage genome-wide association study (GWAS) on lung cancer among never smokers in the Female Lung Cancer Consortium in Asia (FLCCA) (6,609 cases, 7,457 controls) that identified novel susceptibility loci at 10q25.2, 6q22.2, and 6p21.32, and confirmed two previously identified loci at 5p15.33 and 3q28. Household air pollution (HAP) attributed to solid fuel burning for heating and cooking, is the leading cause of the overall disease burden in Southeast Asia, and is known to contain lung carcinogens. To evaluate the gene-HAP interactions associated with lung cancer in loci independent of smoking, we analyzed data from studies participating in FLCCA with fuel use information available (n = 3; 1,731 cases; 1,349 controls). Coal use was associated with a 30% increased risk of lung cancer (OR 1.3, 95% CI 1.0-1.6). Among the five a priori SNPs identified by our GWAS, two showed a significant interaction with coal use (HLA Class II rs2395185, p = 0.02; TP63 rs4488809 (rs4600802), p = 0.04). The risk of lung cancer associated with coal exposure varied with the respective alleles for these two SNPs. Our observations provide evidence that genetic variation in HLA Class II and TP63 may modify the association between HAP and lung cancer risk. The roles played in the cell cycle and inflammation pathways by the proteins encoded by these two genes provide biological plausibility for these interactions; however, additional replication studies are needed in other non-smoking populations.
Collapse
Affiliation(s)
- H Dean Hosgood
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, 1300 Morris Park Ave. Belfer 1309, 10461, Bronx, NY, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Figg WD, Chau CH, Price DK, Till C, Goodman PJ, Cho Y, Varella-Garcia M, Reichardt JKV, Tangen CM, Leach RJ, van Bokhoven A, Thompson IM, Lucia MS. Androgen receptor CAG repeat length and TMPRSS2:ETS prostate cancer risk: results From the Prostate Cancer Prevention Trial. Urology 2014; 84:127-31. [PMID: 24824408 DOI: 10.1016/j.urology.2014.03.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 03/14/2014] [Accepted: 03/17/2014] [Indexed: 11/18/2022]
Abstract
OBJECTIVE To investigate the association between the length of the polymorphic trinucleotide CAG microsatellite repeats in exon 1 of the AR gene and the risk of prostate cancer containing TMPRSS2:ETS fusion genes. METHODS This nested case-control study came from subjects enrolled in the Prostate Cancer Prevention Trial and included 195 biopsy-proven prostate cancer cases with a known TMPRSS2:ETS status and 1344 matched controls. RESULTS There was no association between the CAG repeat length and the risk of TMPRSS2:ETS-positive (odds ratio, 0.97; 95% confidence interval, 0.91-1.04) or TMPRSS2:ETS-negative prostate cancer (odds ratio, 1.04; 95% confidence interval, 0.97-1.11) and in patients with low- or high-grade disease. CONCLUSION Our findings suggested that AR CAG repeats are not associated with TMPRSS2:ETS formation in prostate cancer.
Collapse
Affiliation(s)
- William D Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD.
| | - Cindy H Chau
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Douglas K Price
- Genitourinary Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Cathee Till
- SWOG Statistical Center, the Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Phyllis J Goodman
- SWOG Statistical Center, the Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Yonggon Cho
- Chonbuk National University Medical School, Jeonju, South Korea
| | | | - Juergen K V Reichardt
- School of Pharmacy and Molecular Sciences, James Cook University, Townsville, Queensland, Australia
| | - Catherine M Tangen
- SWOG Statistical Center, the Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Robin J Leach
- Department of Urology and Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Adrie van Bokhoven
- Department of Pathology, University of Colorado Denver School of Medicine, Aurora, CO
| | - Ian M Thompson
- Department of Urology and Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - M Scott Lucia
- Department of Pathology, University of Colorado Denver School of Medicine, Aurora, CO
| |
Collapse
|
19
|
Ooi AT, Gower AC, Zhang KX, Vick JL, Hong L, Nagao B, Wallace WD, Elashoff DA, Walser TC, Dubinett SM, Pellegrini M, Lenburg ME, Spira A, Gomperts BN. Molecular profiling of premalignant lesions in lung squamous cell carcinomas identifies mechanisms involved in stepwise carcinogenesis. Cancer Prev Res (Phila) 2014; 7:487-95. [PMID: 24618292 DOI: 10.1158/1940-6207.capr-13-0372] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lung squamous cell carcinoma (SCC) is thought to arise from premalignant lesions in the airway epithelium; therefore, studying these lesions is critical for understanding lung carcinogenesis. Previous microarray and sequencing studies designed to discover early biomarkers and therapeutic targets for lung SCC had limited success identifying key driver events in lung carcinogenesis, mostly due to the cellular heterogeneity of patient samples examined and the interindividual variability associated with difficult to obtain airway premalignant lesions and appropriate normal control samples within the same patient. We performed RNA sequencing on laser-microdissected representative cell populations along the SCC pathologic continuum of patient-matched normal basal cells, premalignant lesions, and tumor cells. We discovered transcriptomic changes and identified genomic pathways altered with initiation and progression of SCC within individual patients. We used immunofluorescent staining to confirm gene expression changes in premalignant lesions and tumor cells, including increased expression of SLC2A1, CEACAM5, and PTBP3 at the protein level and increased activation of MYC via nuclear translocation. Cytoband enrichment analysis revealed coordinated loss and gain of expression in chromosome 3p and 3q regions, respectively, during carcinogenesis. This is the first gene expression profiling study of airway premalignant lesions with patient-matched SCC tumor samples. Our results provide much needed information about the biology of premalignant lesions and the molecular changes that occur during stepwise carcinogenesis of SCC, and it highlights a novel approach for identifying some of the earliest molecular changes associated with initiation and progression of lung carcinogenesis within individual patients.
Collapse
Affiliation(s)
- Aik T Ooi
- Mattel Children's Hospital, University of California, Los Angeles, 10833 Le Conte Avenue A2-410MDCC, Los Angeles, CA 90095.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Nakachi I, Rice JL, Coldren CD, Edwards MG, Stearman RS, Glidewell SC, Varella-Garcia M, Franklin WA, Keith RL, Lewis MT, Gao B, Merrick DT, Miller YE, Geraci MW. Application of SNP microarrays to the genome-wide analysis of chromosomal instability in premalignant airway lesions. Cancer Prev Res (Phila) 2013; 7:255-65. [PMID: 24346345 DOI: 10.1158/1940-6207.capr-12-0485] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Chromosomal instability is central to the process of carcinogenesis. The genome-wide detection of somatic chromosomal alterations (SCA) in small premalignant lesions remains challenging because sample heterogeneity dilutes the aberrant cell information. To overcome this hurdle, we focused on the B allele frequency data from single-nucleotide polymorphism microarrays (SNP arrays). The difference of allelic fractions between paired tumor and normal samples from the same patient (delta-θ) provides a simple but sensitive detection of SCA in the affected tissue. We applied the delta-θ approach to small, heterogeneous clinical specimens, including endobronchial biopsies and brushings. Regions identified by delta-θ were validated by FISH and quantitative PCR in heterogeneous samples. Distinctive genomic variations were successfully detected across the whole genome in all invasive cancer cases (6 of 6), carcinoma in situ (3 of 3), and high-grade dysplasia (severe or moderate; 3 of 11). Not only well-described SCAs in lung squamous cell carcinoma, but also several novel chromosomal alterations were frequently found across the preinvasive dysplastic cases. Within these novel regions, losses of putative tumor suppressors (RNF20 and SSBP2) and an amplification of RASGRP3 gene with oncogenic activity were observed. Widespread sampling of the airway during bronchoscopy demonstrated that field cancerization reflected by SCAs at multiple sites was detectable. SNP arrays combined with delta-θ analysis can detect SCAs in heterogeneous clinical sample and expand our ability to assess genomic instability in the airway epithelium as a biomarker of lung cancer risk.
Collapse
Affiliation(s)
- Ichiro Nakachi
- University of Colorado, Anschutz Medical Campus, 12700, East 19th Avenue, RC2 9th Floor, Aurora, CO 80045.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Li C, Hong W. Research status and funding trends of lung cancer biomarkers. J Thorac Dis 2013; 5:698-705. [PMID: 24255784 DOI: 10.3978/j.issn.2072-1439.2013.10.10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Accepted: 10/11/2013] [Indexed: 01/11/2023]
Abstract
Lung cancer is one of malignant tumors with the highest morbidity and mortality in the world. At present, research of early diagnosis, treatment, prognosis, and metastasis associated biomarkers is most active. This article reviewed the research status of lung cancer biomarkers and analyzed the funding situation in the field of lung cancer markers in recent 10 years in China and abroad, to provide a reference for the future basic and clinical translational research of lung cancer biomarkers.
Collapse
Affiliation(s)
- Cui Li
- Department of Health Science, National Natural Science Fundation of China, Beijing 100083, China
| | | |
Collapse
|
22
|
van Boerdonk RAA, Daniels JMA, Snijders PJF, Grünberg K, Thunnissen E, van de Wiel MA, Ylstra B, Postmus PE, Meijer CJLM, Meijer GA, Smit EF, Sutedja TG, Heideman DAM. DNA copy number aberrations in endobronchial lesions: a validated predictor for cancer. Thorax 2013; 69:451-7. [PMID: 24227199 DOI: 10.1136/thoraxjnl-2013-203821] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
We recently identified a DNA copy number aberration (CNA)-based classifier, including changes at 3p26.3-p11.1, 3q26.2-29, and 6p25.3-24.3, as a risk predictor for cancer in individuals presenting with endobronchial squamous metaplasia. The current study was set out to validate the prediction accuracy of this classifier in an independent series of endobronchial squamous metaplastic and dysplastic lesions. The study included 36 high-risk subjects who had endobronchial lesions of various histological grades that were identified and biopsied by autofluorescence bronchoscopy and were subjected to arrayCGH in a nested case-control design. Of the 36 patients, 12 had a carcinoma in situ or invasive carcinoma at the same site at follow-up (median 11 months, range 4-24), while 24 controls remained cancer free (78 months, range 21-142). The previously defined CNA-based classifier demonstrated 92% (95% CI 77% to 98%) accuracy for cancer (in situ) prediction. All nine subjects with CNA-based classifier-positive endobronchial lesions at baseline experienced cancer outcome, whereas all 24 controls and 3 cases were classified as being low risk. In conclusion, CNAs prove to be a highly accurate biomarker for assessing the progression risk of endobronchial squamous metaplastic and dysplastic lesions. This classifier could assist in selecting subjects with endobronchial lesions who might benefit from more aggressive therapeutic intervention or surveillance.
Collapse
|
23
|
Hu QY, Jin TB, Wang L, Zhang L, Geng T, Liang G, Kang LL. Genetic variation in the TP63 gene is associated with lung cancer risk in the Han population. Tumour Biol 2013; 35:1863-6. [DOI: 10.1007/s13277-013-1248-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/23/2013] [Indexed: 12/30/2022] Open
|
24
|
Hensing TA, Salgia R. Molecular biomarkers for future screening of lung cancer. J Surg Oncol 2013; 108:327-33. [PMID: 23893423 DOI: 10.1002/jso.23382] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 06/28/2013] [Indexed: 12/28/2022]
Abstract
The Landmark National Lung Screening Trial established the potential for low dose CT screening (LDCT) to reduce lung cancer-specific mortality in high-risk patients as defined by smoking history and age. However, the prevalence of lung cancer in asymptomatic smokers selected based on the NLST criteria is low. Recent advances have facilitated biomarker discovery for early diagnosis of lung cancer through the analysis of surrogate tissues, including airway epithelium, sputum, exhaled breath, and blood. Although a number of candidate diagnostic biomarkers have been described, none have been validated for use in the clinical setting. The NLST ACRIN biomarker repository is a valuable resource of annotated biological specimens that were collected during the NLST trial, which has the potential to facilitate validation of candidate biomarkers for early diagnosis identified in discovery trials. It will be important to perform retrospective and prospective analysis of biomarkers to screen for lung cancer. The review below summarizes some of our understanding of biomarkers in screening.
Collapse
Affiliation(s)
- Thomas A Hensing
- NorthShore University HealthSystem, Clinical Associate Professor of Medicine, University of Chicago Pritzker, Chicago, Illinois
| | | |
Collapse
|
25
|
Quintans JSS, Antoniolli AR, Onofre FMB, Onofre ASC. Detection of lung cancer using multiple genetic markers--a systematic review. Diagn Cytopathol 2013; 41:834-42. [PMID: 23513001 DOI: 10.1002/dc.22978] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 01/01/2013] [Indexed: 11/08/2022]
Abstract
Lung cancer is the leading cause of cancer deaths worldwide, and has one of the lowest survival rates of any solid tumor. In recent years, several attempts have been conducted to improve an early or accelerated diagnosis due to better overall prognosis after therapy. The aim of this study was evaluating the use of genetic markers for diagnosis of lung cancer. This study was conducted in accordance to Transparent Reporting of Systematic Reviews and Meta-Analyses. Three Internet sources were used to search: MEDLINE-PubMed, EMBASE, and LILACS. The databases were searched for studies conducted in the period up to and including May 10, 2011. The following inclusion criteria were applied: lung cancer studies, and the use of genetic markers for diagnosis. Studies using animal models, review articles, meta-analyses, abstracts, conference proceedings, editorials/letters, case reports, incorrect study population, inadequate data, and cytology was not obtained, were excluded. A total of 1,901 abstracts/citations were identified for preliminary review. From 24 final selected studies, 17 referred to chromosomal markers diagnosis, eight to genes as marker, and one to both subjects. Fluorescence in situ hybridization (FISH) was applied in all studies. Despite the limitations of this study, application of genetic markers to lung cancer diagnosis seems to have prognosis value irrespective of detection methodology used. FISH was the main technique applied to diagnose genetics alterations and revealed a high specificity, although some authors reported low sensitivity.
Collapse
|
26
|
SH3GL2 is frequently deleted in non-small cell lung cancer and downregulates tumor growth by modulating EGFR signaling. J Mol Med (Berl) 2012; 91:381-93. [PMID: 22968441 DOI: 10.1007/s00109-012-0955-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 08/12/2012] [Accepted: 08/29/2012] [Indexed: 01/22/2023]
Abstract
The purpose of this study was to identify key genetic pathways involved in non-small cell lung cancer (NSCLC) and understand their role in tumor progression. We performed a genome wide scanning using paired tumors and corresponding 16 mucosal biopsies from four follow-up lung cancer patients on Affymetrix 250K-NSpI array platform. We found that a single gene SH3GL2 located on human chromosome 9p22 was most frequently deleted in all the tumors and corresponding mucosal biopsies. We further validated the alteration pattern of SH3GL2 in a substantial number of primary NSCLC tumors at DNA and protein level. We also overexpressed wild-type SH3GL2 in three NSCLC cell lines to understand its role in NSCLC progression. Validation in 116 primary NSCLC tumors confirmed frequent loss of heterozygosity of SH3GL2 in overall 51 % (49/97) of the informative cases. We found significantly low (p = 0.0015) SH3GL2 protein expression in 71 % (43/60) primary tumors. Forced overexpression of wild-type (wt) SH3GL2 in three NSCLC cell lines resulted in a marked reduction of active epidermal growth factor receptor (EGFR) expression and an increase in EGFR internalization and degradation. Significantly decreased in vitro (p = 0.0015-0.030) and in vivo (p = 0.016) cellular growth, invasion (p = 0.029-0.049), and colony formation (p = 0.023-0.039) were also evident in the wt-SH3GL2-transfected cells accompanied by markedly low expression of activated AKT(Ser(473)), STAT3 (Tyr(705)), and PI3K. Downregulation of SH3GL2 interactor USP9X and activated ß-catenin was also evident in the SH3GL2-transfected cells. Our results indicate that SH3GL2 is frequently deleted in NSCLC and regulates cellular growth and invasion by modulating EGFR function.
Collapse
|
27
|
Hassanein M, Callison JC, Callaway-Lane C, Aldrich MC, Grogan EL, Massion PP. The state of molecular biomarkers for the early detection of lung cancer. Cancer Prev Res (Phila) 2012; 5:992-1006. [PMID: 22689914 DOI: 10.1158/1940-6207.capr-11-0441] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Using biomarkers to select the most at-risk population, to detect the disease while measurable and yet not clinically apparent has been the goal of many investigations. Recent advances in molecular strategies and analytic platforms, including genomics, epigenomics, proteomics, and metabolomics, have identified increasing numbers of potential biomarkers in the blood, urine, exhaled breath condensate, bronchial specimens, saliva, and sputum, but none have yet moved to the clinical setting. Therefore, there is a recognized gap between the promise and the product delivery in the cancer biomarker field. In this review, we define clinical contexts where risk and diagnostic biomarkers may have use in the management of lung cancer, identify the most relevant candidate biomarkers of early detection, provide their state of development, and finally discuss critical aspects of study design in molecular biomarkers for early detection of lung cancer.
Collapse
Affiliation(s)
- Mohamed Hassanein
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt-Ingram Cancer Center, Nashville TN 37232, USA
| | | | | | | | | | | |
Collapse
|
28
|
Jantus-Lewintre E, Usó M, Sanmartín E, Camps C. Update on biomarkers for the detection of lung cancer. LUNG CANCER-TARGETS AND THERAPY 2012; 3:21-29. [PMID: 28210122 DOI: 10.2147/lctt.s23424] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Patients at risk for lung cancer may have subclinical disease for years before presentation. The diagnosis of this disease is primarily based on symptoms, and detection often occurs after curative intervention is no longer possible. At present, no lung cancer early-detection biomarker is clinically available. This study reviews the most recent advances in early detection and molecular diagnostic biomarkers for the detection of lung cancer. This review includes an overview of the various biological specimens and matrices in which these biomarkers could be analyzed, as well as the diverse strategies and approaches for identifying new biomarkers that are currently being explored. Several novel and attractive biomarker candidates for the early detection of lung cancer exist. A remarkable shift is taking place from research based on single markers to analyzing signatures that are more complex in order to take advantage of new high-throughput technologies. However, it is still necessary to validate the most promising markers and the standardization of procedures that will lead to specific clinical applications.
Collapse
Affiliation(s)
- Eloisa Jantus-Lewintre
- Molecular Oncology Laboratory, Fundación para la Investigación del Hospital General Universitario, Valencia, Spain
| | - Marta Usó
- Molecular Oncology Laboratory, Fundación para la Investigación del Hospital General Universitario, Valencia, Spain
| | - Elena Sanmartín
- Molecular Oncology Laboratory, Fundación para la Investigación del Hospital General Universitario, Valencia, Spain
| | - Carlos Camps
- Molecular Oncology Laboratory, Fundación para la Investigación del Hospital General Universitario, Valencia, Spain; Deparment of Medical Oncology, Consorcio Hospital General Universitario, Valencia, Spain; Department of Medicine, Universitat de València, Valencia, Spain
| |
Collapse
|
29
|
Dietrich D, Hasinger O, Liebenberg V, Field JK, Kristiansen G, Soltermann A. DNA methylation of the homeobox genes PITX2 and SHOX2 predicts outcome in non-small-cell lung cancer patients. DIAGNOSTIC MOLECULAR PATHOLOGY : THE AMERICAN JOURNAL OF SURGICAL PATHOLOGY, PART B 2012; 21:93-104. [PMID: 22555092 DOI: 10.1097/pdm.0b013e318240503b] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Biomarkers that facilitate prediction of disease progression in lung cancer patients might be clinically valuable in optimizing individualized therapy. In this study, the ability of the DNA methylation biomarkers PITX2 and SHOX2 to predict disease outcome in lung cancer patients has been evaluated. Quantitative, methylation-specific (HeavyMethyl), real-time polymerase chain reaction assays were used to measure DNA methylation of PITX2 and SHOX2 in bisulfite-converted DNA from formalin-fixed, paraffin-embedded tissues from 474 non-small-cell lung cancer patients. In univariate Cox Proportional Hazard analysis, high methylation of SHOX2 and PITX2 was a significant predictor of progression-free survival [SHOX2: n=465, hazard ratio (HR)=1.395 (1.130 to 1.721), P=0.002; PITX2: n=445, HR=1.312 (1.059 to 1.625), P=0.013]. Patients with low methylation of either PITX2 and/or SHOX2 (n=319) showed a significantly higher risk of disease progression as compared with patients with higher methylation of both genes [n=126; HR=1.555 (1.210 to 1.999), P=0.001]. This was particularly true for the subgroup of patients receiving no adjuvant radiotherapy or chemotherapy [n=258, HR=1.838 (1.252 to 2.698), P=0.002]. In multivariate analysis, both biomarkers added significant independent prognostic information to pT, pN, pM, and grade. Another interesting finding of this study was that SHOX2 and PITX2 DNA methylation was shown to be inversely correlated with TTF1 (also known as NKX2-1) expression (PITX2: P=0.018, SHOX2: P<0.001). TFF1 expression was previously found to be associated with improved survival in the same patient cohort. DNA methylation of PITX2 and SHOX2 is an independent prognostic biomarker for disease progression in non-small-cell lung cancer patients.
Collapse
Affiliation(s)
- Dimo Dietrich
- Institute of Pathology, University Hospital Bonn, Bonn, Germany.
| | | | | | | | | | | |
Collapse
|
30
|
Genetic variant in TP63 on locus 3q28 is associated with risk of lung adenocarcinoma among never-smoking females in Asia. Hum Genet 2012; 131:1197-203. [PMID: 22367405 DOI: 10.1007/s00439-012-1144-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 02/04/2012] [Indexed: 12/21/2022]
Abstract
A recent genome-wide association study (GWAS) of subjects from Japan and South Korea reported a novel association between the TP63 locus on chromosome 3q28 and risk of lung adenocarcinoma (p = 7.3 × 10(-12)); however, this association did not achieve genome-wide significance (p ≤ 10(-7)) among never-smoking males or females. To determine if this association with lung cancer risk is independent of tobacco use, we genotyped the TP63 SNPs reported by the previous GWAS (rs10937405 and rs4488809) in 3,467 never-smoking female lung cancer cases and 3,787 never-smoking female controls from 10 studies conducted in Taiwan, Mainland China, South Korea, and Singapore. Genetic variation in rs10937405 was associated with risk of lung adenocarcinoma [n = 2,529 cases; p = 7.1 × 10(-8); allelic risk = 0.80, 95% confidence interval (CI) = 0.74-0.87]. There was also evidence of association with squamous cell carcinoma of the lung (n = 302 cases; p = 0.037; allelic risk = 0.82, 95% CI = 0.67-0.99). Our findings provide strong evidence that genetic variation in TP63 is associated with the risk of lung adenocarcinoma among Asian females in the absence of tobacco smoking.
Collapse
|
31
|
Coldren CD, Miller YE. Progressive endobronchial premalignancy: marked by original CIN. Am J Respir Crit Care Med 2011; 184:869-70. [PMID: 22003147 DOI: 10.1164/rccm.201108-1476ed] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
32
|
Abstract
In the decade since the last Lancet Seminar on lung cancer there have been advances in many aspects of the classification, diagnosis, and treatment of non-small-cell lung cancer (NSCLC). An international panel of experts has been brought together to focus on changes in the epidemiology and pathological classification of NSCLC, the role of CT screening and other techniques that could allow earlier diagnosis and more effective treatment of the disease, and the recently introduced seventh edition of the TNM classification and its relation to other prognostic factors such as biological markers. We also describe advances in treatment that have seen the introduction of a new generation of chemotherapy agents, a proven advantage to adjuvant chemotherapy after complete resection for specific stage groups, new techniques for the planning and administration of radiotherapy, and new surgical approaches to assess and reduce the risks of surgical treatment.
Collapse
Affiliation(s)
- Peter Goldstraw
- Academic Department of Thoracic Surgery, Royal Brompton and Harefield NHS Foundation Trust, Imperial College School of Medicine, London, UK.
| | | | | | | | | | | | | |
Collapse
|
33
|
van Boerdonk RAA, Sutedja TG, Snijders PJF, Reinen E, Wilting SM, van de Wiel MA, Thunnissen FEBJM, Duin S, Kooi C, Ylstra B, Meijer CJLM, Meijer GA, Grünberg K, Daniels JMA, Postmus PE, Smit EF, Heideman DAM. DNA copy number alterations in endobronchial squamous metaplastic lesions predict lung cancer. Am J Respir Crit Care Med 2011; 184:948-56. [PMID: 21799074 DOI: 10.1164/rccm.201102-0218oc] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
RATIONALE Autofluorescence bronchoscopy (AFB) is a valid strategy for detecting premalignant endobronchial lesions. However, no biomarker can reliably predict lung cancer risk of subjects with AFB-visualized premalignant lesions. OBJECTIVES The present study set out to identify AFB-visualized squamous metaplastic (SqM) lesions with malignant potential by DNA copy number profiling. METHODS Regular AFB examinations in 474 subjects at risk of lung cancer identified six subjects with SqM lesions at baseline, and carcinoma in situ or carcinoma (carcinoma in situ or greater) at the initial SqM site at follow-up bronchoscopy. These progressive SqM lesions were compared for immunostaining pattern and array comparative genomic hybridization-based chromosomal profiles with 23 SqM lesions of subjects who remained cancer-free. Specific DNA copy number alterations (CNAs) linked to cancer risk were identified and accuracy of CNAs to predict endobronchial cancer in this series was determined. MEASUREMENTS AND MAIN RESULTS At baseline, p53, p63, and Ki-67 immunostaining were not predictive for a differential clinical outcome of SqM lesions. The mean number of CNAs in baseline SqM of cases was significantly higher compared with control subjects (P < 0.01). Chromosomal regions significantly more frequently altered in SqM of cases were 3p26.3-p11.1, 3q26.2-q29, 9p13.3-p13.2, and 17p13.3-p11.2 (family-wise error rate <0.10). CNAs were specifically detected at the site of future cancer. In cases, baseline-detected CNAs persisted in subsequent biopsies taken from the initial site, and levels increased toward cancer progression. In this series, a model based on CNAs at 3p26.3-p11.1, 3q26.2-29, and 6p25.3-24.3 predicted cancer with 97% accuracy. CONCLUSIONS The data suggest that the presence of specific CNAs in SqM lesions predict endobronchial cancer.
Collapse
Affiliation(s)
- Robert A A van Boerdonk
- VU University Medical Center, Department of Pathology, De Boelelaan 1117, Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
While no real improvement in the long term survival has been obtained in lung cancer, during this decade a significant improvement in cancer control has been obtained by biology driven targeted therapy as with anti EGFR tyrosine kinase. Two phases can be described in the knowledge of lung cancer biology: a first phase open in the 1980s describing the main molecular anomalies and impaired cell control mechanisms, and a second phase starting in the 2004-2005 giving rise to the therapeutic applications of this knowledge. A new molecular classification of lung cancer, particularly adenocarcinomas will soon be proposed for therapeutic application.
Collapse
|
35
|
Katz RL, He W, Khanna A, Fernandez RL, Zaidi TM, Krebs M, Caraway NP, Zhang HZ, Jiang F, Spitz MR, Blowers DP, Jimenez CA, Mehran RJ, Swisher SG, Roth JA, Morris JS, Etzel CJ, El-Zein R. Genetically abnormal circulating cells in lung cancer patients: an antigen-independent fluorescence in situ hybridization-based case-control study. Clin Cancer Res 2010; 16:3976-87. [PMID: 20651054 DOI: 10.1158/1078-0432.ccr-09-3358] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE We performed a study to determine if a fluorescence in situ hybridization (FISH)-based assay using isolated peripheral blood mononuclear cells (PBMCs) with DNA probes targeting specific sites on chromosomes known to have abnormalities in non-small cell lung cancer (NSCLC) cases could detect circulating genetically abnormal cells (CACs). EXPERIMENTAL DESIGN We evaluated 59 NSCLC cases with stage I through IV disease and 24 controls. PBMCs and matched tumors were hybridized with 2 two-color [3p22.1/CEP3 and 10q22.3 (SP-A)/CEP10) and 2 four-color [CEP3, CEP7, CEP17, and 9p21.3 (URO); and EGFR, c-MYC, 6p11-q11, and 5p15.2 (LAV)] FISH probes. Percentages of cytogenetically abnormal cells (CACs) in peripheral blood and in matched tumor specimens were quantified by using an automated fluorescent scanner. Numbers of CACs were calculated based on the percentage of CACs (defined as PBMCs with genetic abnormalities) per milliliter of blood and expressed per microliter of blood. RESULTS Patients with NSCLC had significantly higher numbers of CACs than controls. Mean number of CACs ranged from 7.23 +/- 1.32/microL for deletions of 10q22.3/CEP10 to 45.52 +/- 7.49/microL for deletions of 3p22.1/CEP3. Numbers of CACs with deletions of 3p22.1, 10q22.3, and 9p21.3, and gains of URO, increased significantly from early to advanced stage of disease. CONCLUSIONS We have developed a sensitive and quantitative antigen-independent FISH-based test for detecting CACs in peripheral blood of patients with NSCLC, which showed a significant correlation with the presence of cancer. If this pilot study can be validated in a larger study, CACs may have a role in the management of patients with NSCLC.
Collapse
Affiliation(s)
- Ruth L Katz
- Department of Pathology, University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Advances in the diagnosis of lung cancer: contribution of molecular biology to bronchoscopic diagnosis. Curr Opin Pulm Med 2010; 16:315-20. [DOI: 10.1097/mcp.0b013e328337f938] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Ishizumi T, McWilliams A, MacAulay C, Gazdar A, Lam S. Natural history of bronchial preinvasive lesions. Cancer Metastasis Rev 2010; 29:5-14. [PMID: 20112052 DOI: 10.1007/s10555-010-9214-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Preinvasive bronchial lesions defined as dysplasia and carcinoma in situ (CIS) have been considered as precursors of squamous cell carcinoma of the lung. The risk and rate of progression of preinvasive lesions to invasive squamous cell carcinoma as well as the mechanism of progression or regression are incompletely understood. While the evidence for the multistage, stepwise progression model is weak with relatively few documented lesions that progress through various grades of dysplasia to CIS and then to invasive carcinoma, the concept of field carcinogenesis is strongly supported. The presence of high-grade dysplasia or CIS is a risk marker for lung cancer both in the central airways and peripheral lung. Genetic alterations such as loss of heterozygosity in chromosome 3p or chromosomal aneusomy as well as host factors such as the inflammatory load and levels of anti-inflammatory proteins in the lung influence the progression or regression of preinvasive lesions. CIS is different than severe dysplasia at the molecular level and has different clinical outcome. Molecular analysis of dysplastic lesions that progress to CIS or invasive cancer and rare lesions that progress rapidly from hyperplasia or metaplasia to CIS or invasive cancer will shed light on the key molecular determinants driving development to an invasive phenotype versus those associated with tobacco smoke damage.
Collapse
Affiliation(s)
- Taichiro Ishizumi
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan.
| | | | | | | | | |
Collapse
|
38
|
McCaughan F, Pole JCM, Bankier AT, Konfortov BA, Carroll B, Falzon M, Rabbitts TH, George PJ, Dear PH, Rabbitts PH. Progressive 3q amplification consistently targets SOX2 in preinvasive squamous lung cancer. Am J Respir Crit Care Med 2010; 182:83-91. [PMID: 20299530 DOI: 10.1164/rccm.201001-0005oc] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Amplification of distal 3q is the most common genomic aberration in squamous lung cancer (SQC). SQC develops in a multistage progression from normal bronchial epithelium through dysplasia to invasive disease. Identifying the key driver events in the early pathogenesis of SQC will facilitate the search for predictive molecular biomarkers and the identification of novel molecular targets for chemoprevention and therapeutic strategies. For technical reasons, previous attempts to analyze 3q amplification in preinvasive lesions have focused on small numbers of predetermined candidate loci rather than an unbiased survey of copy-number variation. OBJECTIVES To perform a detailed analysis of the 3q amplicon in bronchial dysplasia of different histological grades. METHODS We use molecular copy-number counting (MCC) to analyze the structure of chromosome 3 in 19 preinvasive bronchial biopsy specimens from 15 patients and sequential biopsy specimens from 3 individuals. MEASUREMENTS AND MAIN RESULTS We demonstrate that no low-grade lesions, but all high-grade lesions, have 3q amplification. None of seven low-grade lesions progressed clinically, whereas 8 of 10 patients with high-grade disease progressed to cancer. We identify a minimum commonly amplified region on chromosome 3 consisting of 17 genes, including 2 known oncogenes, SOX2 and PIK3CA. We confirm that both genes are amplified in all high-grade dysplastic lesions tested. We further demonstrate, in three individuals, that the clinical progression of high-grade preinvasive disease is associated with incremental amplification of SOX2, suggesting this promotes malignant progression. CONCLUSIONS These findings demonstrate progressive 3q amplification in the evolution of preinvasive SQC and implicate SOX2 as a key target of this dynamic process.
Collapse
Affiliation(s)
- Frank McCaughan
- Centre for Respiratory Research, Royal Free and University College Medical School, London, United Kingdom.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Voss JS, Kipp BR, Halling KC, Henry MR, Jett JR, Clayton AC, Rickman OB. Fluorescencein SituHybridization Testing Algorithm Improves Lung Cancer Detection in Bronchial Brushing Specimens. Am J Respir Crit Care Med 2010; 181:478-85. [DOI: 10.1164/rccm.200907-1121oc] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
|