1
|
Backhus LM, Chang CF, Sakoda LC, Chambers SR, Henderson LM, Henschke CI, Hollenbeck GJ, Jacobson FL, Martin LW, Proctor ED, Schiller JH, Siegfried JM, Wisnivesky JP, Wolf AS, Jemal A, Kelly K, Sandler KL, Watkins PN, Smith RA, Rivera MP. The American Cancer Society National Lung Cancer Roundtable strategic plan: Lung cancer in women. Cancer 2024; 130:3985-3995. [PMID: 39302237 DOI: 10.1002/cncr.35083] [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] [Indexed: 09/22/2024]
Abstract
Lung cancer in women is a modern epidemic and represents a global health crisis. Cigarette smoking remains the most important risk factor for lung cancer in all patients and, among women globally, rates of smoking continue to increase. Although some data exist supporting sex-based differences across the continuum of lung cancer, there is currently a dearth of research exploring the differences in risk, biology, and treatment outcomes in women. Consequently, the American Cancer Society National Lung Cancer Roundtable recognizes the urgent need to promote awareness and future research that will close the knowledge gaps regarding lung cancer in women. To this end, the American Cancer Society National Lung Cancer Roundtable Task Group on Lung Cancer in Women convened a summit undertaking the following to: (1) summarize existing evidence and identify knowledge gaps surrounding the epidemiology, risk factors, biologic differences, and outcomes of lung cancer in women; (2) develop and prioritize research topics and questions that address research gaps and advance knowledge to improve quality of care of lung cancer in women; and (3) propose strategies for future research. PLAIN LANGUAGE SUMMARY: Lung cancer is the leading cause of cancer mortality in women, and, despite comparatively lower exposures to occupational and environmental carcinogens compared with men, disproportionately higher lung cancer rates in women who ever smoked and women who never smoked call for increased awareness and research that will close the knowledge gaps regarding lung cancer in women.
Collapse
Affiliation(s)
- Leah M Backhus
- Department of Cardiothoracic Surgery, Division of Thoracic Surgery, Stanford University, Stanford, California, USA
| | - Ching-Fei Chang
- Department of Pulmonary, Critical Care, and Sleep Medicine, University of Southern California, Keck School of Medicine, Los Angeles, California, USA
| | - Lori C Sakoda
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Shonta R Chambers
- Department of Health Equity, Patient Advocate Foundation, Hampton, Virginia, USA
| | - Louise M Henderson
- Department of Radiology, The University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Claudia I Henschke
- Department of Radiology, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona, USA
| | | | - Francine L Jacobson
- Department of Radiology, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Linda W Martin
- Department of Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Elridge D Proctor
- Government Affairs, GO2 Foundation for Lung Cancer, Washington, District of Columbia, USA
| | | | - Jill M Siegfried
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Juan P Wisnivesky
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Andrea S Wolf
- Department Thoracic Surgery, The Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ahmedin Jemal
- Surveillance and Health Equity Science, American Cancer Society, Atlanta, Georgia, USA
| | - Karen Kelly
- Department of Internal Medicine, Division of Hematology and Oncology, The University of California at Davis, Sacramento, California, USA
| | - Kim L Sandler
- Department of Radiology and Radiological Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Robert A Smith
- Center for Early Cancer Detection Science, American Cancer Society, Atlanta, Georgia, USA
| | - M Patricia Rivera
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Wilmot Cancer Institute, The University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
2
|
Kurzrock R, Chaudhuri AA, Feller-Kopman D, Florez N, Gorden J, Wistuba II. Healthcare disparities, screening, and molecular testing in the changing landscape of non-small cell lung cancer in the United States: a review. Cancer Metastasis Rev 2024; 43:1217-1231. [PMID: 38750337 PMCID: PMC11554720 DOI: 10.1007/s10555-024-10187-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 04/04/2024] [Indexed: 11/13/2024]
Abstract
Inequitable access to care continues to hinder improvements in diagnosis and treatment of lung cancer. This review describes healthcare disparities in the changing landscape of non-small cell lung cancer (NSCLC) in the United States, focusing on racial, ethnic, sex-based, and socioeconomic trends. Furthermore, strategies to address disparities, overcome challenges, and improve patient outcomes are proposed. Barriers exist across lung cancer screening, diagnosis, and treatment regimens, varying by sex, age, race and ethnicity, geography, and socioeconomic status. Incidence and mortality rates of lung cancer are higher among Black men than White men, and incidences in young women are substantially greater than in young men. Disparities may be attributed to geographic differences in screening access, with correlating higher incidence and mortality rates in rural versus urban areas. Lower socioeconomic status is also linked to lower survival rates. Several strategies could help reduce disparities and improve outcomes. Current guidelines could improve screening eligibility by incorporating sex, race, and socioeconomic status variables. Patient and clinician education on screening guidelines and patient-level barriers to care are key, and biomarker testing is critical since ~ 70% of patients with NSCLC have an actionable biomarker. Timely diagnosis, staging, and comprehensive biomarker testing, including cell-free DNA liquid biopsy, may provide valuable treatment guidance for patients with NSCLC. Efforts to improve lung cancer screening and biomarker testing access, decrease bias, and improve education about screening and testing are needed to reduce healthcare disparities in NSCLC.
Collapse
Affiliation(s)
- Razelle Kurzrock
- Medical College of Wisconsin Cancer Center, Froedtert and Medical College of Wisconsin, 8701 Watertown Plank Rd, Milwaukee, WI, 53226, USA.
| | - Aadel A Chaudhuri
- Department of Radiation Oncology, Washington University School of Medicine, 4511 Forest Park Ave, St. Louis, MO, 63108, USA
- Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - David Feller-Kopman
- Geisel School of Medicine, Dartmouth Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH, 03756, USA
| | - Narjust Florez
- Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Ave - DA1230, Boston, MA, 02215, USA
| | - Jed Gorden
- Department of Thoracic Surgery and Interventional Pulmonology, Swedish Cancer Institute, 1101 Madison St, Suite 900, Seattle, WA, 98104, USA
| | - Ignacio I Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX, 77030, USA
| |
Collapse
|
3
|
Volk RJ, Myers RE, Arenberg D, Caverly TJ, Hoffman RM, Katki HA, Mazzone PJ, Moulton BW, Reuland DS, Tanner NT, Smith RA, Wiener RS. The American Cancer Society National Lung Cancer Roundtable strategic plan: Current challenges and future directions for shared decision making for lung cancer screening. Cancer 2024; 130:3996-4011. [PMID: 39302231 DOI: 10.1002/cncr.35382] [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: 03/03/2024] [Revised: 04/07/2024] [Accepted: 04/12/2024] [Indexed: 09/22/2024]
Abstract
Shared decision making (SDM) between health care professionals and patients is essential to help patients make well informed choices about lung cancer screening (LCS). Patients who participate in SDM have greater LCS knowledge, reduced decisional conflict, and improved adherence to annual screening compared with patients who do not participate in SDM. SDM tools are acceptable to patients and clinicians. The importance of SDM in LCS is emphasized in recommendations from professional organizations and highlighted as a priority in the 2022 President's Cancer Panel Report. The updated 2022 national coverage determination from the Centers for Medicare & Medicaid Services reaffirms the value of SDM in offering LCS to eligible beneficiaries. The Shared Decision-Making Task Group of the American Cancer Society National Lung Cancer Roundtable undertook a group consensus process to identify priorities for research and implementation related to SDM for LCS and then evaluated current knowledge in these areas. Priority areas included: (1) developing feasible, adaptable SDM training programs for health care professionals; (2) understanding the impact of alternative health system LCS models on SDM practice and outcomes; (3) developing and evaluating new patient decision aids for use with diverse populations and in varied settings; (4) offering conceptual clarity about what constitutes a high-quality decision and developing appropriate quality measures; and (5) studying the use of prediction-augmented screening to support SDM in practice. Gaps in current research in all areas were observed. The authors conclude with a research and implementation agenda to advance the quality and implementation of SDM for persons who might benefit from LCS.
Collapse
Affiliation(s)
- Robert J Volk
- The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ronald E Myers
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | | | - Tanner J Caverly
- Veterans Affairs Ann Arbor Center for Clinical Management Research, University of Michigan Medical School, Institute for Health Policy Innovation, Ann Arbor, Michigan, USA
| | - Richard M Hoffman
- University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
- University of Iowa Holden Comprehensive Cancer Center, Iowa City, Iowa, USA
| | - Hormuzd A Katki
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Peter J Mazzone
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | - Daniel S Reuland
- Division of General Medicine and Clinical Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Nichole T Tanner
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, South Carolina, USA
- Medical University of South Carolina, Charleston, South Carolina, USA
| | - Robert A Smith
- Center for Early Cancer Detection Science, American Cancer Society, Atlanta, Georgia, USA
| | - Renda Soylemez Wiener
- Center for Healthcare Organization & Implementation Research, VA Boston Healthcare System, Boston, Massachusetts, USA
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
- National Center for Lung Cancer Screening, Veterans Health Administration, Washington, District of Columbia, USA
| |
Collapse
|
4
|
Lewis JA, Klein DE, Eberth JM, Carter-Bawa L, Studts JL, Tong BC, Smith RA, Kazerooni EA, Houston TP. The American Cancer Society National Lung Cancer Roundtable strategic plan: Provider engagement and outreach. Cancer 2024; 130:3973-3984. [PMID: 39302232 DOI: 10.1002/cncr.34555] [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] [Indexed: 09/22/2024]
Abstract
The American Cancer Society National Lung Cancer Roundtable strategic plan for provider engagement and outreach addresses barriers to the uptake of lung cancer screening, including lack of provider awareness and guideline knowledge about screening, concerns about potential harms from false-positive examinations, lack of time to implement workflows within busy primary care practices, insufficient infrastructure and administrative support to manage a screening program and patient follow-up, and implicit bias based on sex, race/ethnicity, social class, and smoking status. Strategies to facilitate screening include educational programming, clinical reminder systems within the electronic medical record, decision support aids, and tools to track nodules that can be implemented across a diversity of practices and health care organizational structures. PLAIN LANGUAGE SUMMARY: The American Cancer Society National Lung Cancer Roundtable strategic plan to reduce deaths from lung cancer includes strategies designed to support health care professionals, to better understand lung cancer screening, and to support adults who are eligible for lung cancer screening by providing counseling, referral, and follow-up.
Collapse
Affiliation(s)
- Jennifer A Lewis
- Veterans Health Administration-Tennessee Valley Healthcare System Geriatric Research, Education and Clinical Center, Nashville, Tennessee, USA
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee, USA
| | - Deborah E Klein
- Swedish Primary Care, Swedish Medical Center, Seattle, Washington, USA
| | - Jan M Eberth
- Dornsife School of Public Health, Drexel University, Philadelphia, Pennsylvania, USA
| | - Lisa Carter-Bawa
- Department of Psychiatry & Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Jamie L Studts
- Department of Medicine, Division of Medical Oncology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Betty C Tong
- Division of Cardiovascular and Thoracic Surgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Robert A Smith
- Center for Early Cancer Detection Science, American Cancer Society, Atlanta, Georgia, USA
| | - Ella A Kazerooni
- Division of Cardiothoracic Radiology, Department of Radiology, University of Michigan, Ann Arbor, Michigan, USA
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Thomas P Houston
- Department of Family Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
| |
Collapse
|
5
|
Fathi JT, Barry AM, Greenberg GM, Henschke CI, Kazerooni EA, Kim JJ, Mazzone PJ, Mulshine JL, Pyenson BS, Shockney LD, Smith RA, Wiener RS, White CS, Thomson CC. The American Cancer Society National Lung Cancer Roundtable strategic plan: Implementation of high-quality lung cancer screening. Cancer 2024; 130:3961-3972. [PMID: 39302235 DOI: 10.1002/cncr.34621] [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] [Indexed: 09/22/2024]
Abstract
More than a decade has passed since researchers in the Early Lung Cancer Action Project and the National Lung Screening Trial demonstrated the ability to save lives of high-risk individuals from lung cancer through regular screening by low dose computed tomography scan. The emergence of the most recent findings in the Dutch-Belgian lung-cancer screening trial (Nederlands-Leuvens Longkanker Screenings Onderzoek [NELSON]) further strengthens and expands on this evidence. These studies demonstrate the benefit of integrating lung cancer screening into clinical practice, yet lung cancer continues to lead cancer mortality rates in the United States. Fewer than 20% of screen eligible individuals are enrolled in lung cancer screening, leaving millions of qualified individuals without the standard of care and benefit they deserve. This article, part of the American Cancer Society National Lung Cancer Roundtable (ACS NLCRT) strategic plan, examines the impediments to successful adoption, dissemination, and implementation of lung cancer screening. Proposed solutions identified by the ACS NLCRT Implementation Strategies Task Group and work currently underway to address these challenges to improve uptake of lung cancer screening are discussed. PLAIN LANGUAGE SUMMARY: The evidence supporting the benefit of lung cancer screening in adults who previously or currently smoke has led to widespread endorsement and coverage by health plans. Lung cancer screening programs should be designed to promote high uptake rates of screening among eligible adults, and to deliver high-quality screening and follow-up care.
Collapse
Affiliation(s)
- Joelle T Fathi
- Department of Biobehavioral Nursing and Health Informatics, School of Nursing, University of Washington, Seattle, Washington, USA
- GO2 for Lung Cancer, Washington, District of Columbia, USA
| | - Angela M Barry
- GO2 for Lung Cancer, Washington, District of Columbia, USA
| | - Grant M Greenberg
- Department of Family Medicine, Lehigh Valley Health Network, Allentown, Pennsylvania, USA
| | - Claudia I Henschke
- Department of Radiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Phoenix Veterans Health Care System, Phoenix, Arizona, USA
| | - Ella A Kazerooni
- Department of Radiology, Michigan Medicine/University of Michigan, Ann Arbor, Michigan, USA
- Department of Internal Medicine, Michigan Medicine/University of Michigan, Ann Arbor, Michigan, USA
| | - Jane J Kim
- Department of Veterans Affairs, National Center for Health Promotion and Disease Prevention, Durham, North Carolina, USA
| | - Peter J Mazzone
- Respiratory Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - James L Mulshine
- Department of Internal Medicine, Rush University Medical College, Chicago, Illinois, USA
| | | | - Lillie D Shockney
- Surgical Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Robert A Smith
- Center for Early Cancer Detection Science, American Cancer Society, Atlanta, Georgia, USA
| | - Renda Soylemez Wiener
- Center for Healthcare Organization and Implementation Research, VA Boston Healthcare System, The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Charles S White
- Department of Radiology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Carey C Thomson
- Department of Medicine, Division of Pulmonary and Critical Care, Mount Auburn Hospital/Beth Israel Lahey Health, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Qazi SU, Hamid A, Ansari HUH, Khouri MG, Anker MS, Hall ME, Anker SD, Butler J, Khan MS. Trends in cancer and heart failure related mortality in adult US population: A CDC WONDER database analysis from 1999 to 2020. Am Heart J 2024; 278:170-180. [PMID: 39299631 DOI: 10.1016/j.ahj.2024.09.002] [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] [Received: 04/13/2024] [Revised: 08/17/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND With the advent of novel chemotherapy, survival of patients with cancer has improved. However, people with cancer have an increased risk of heart failure (HF). Conversely, HF-related mortality may undermine survival among people with cancer. We aim to analyze the trends of mortality in people with HF and cancer in the adult US population. METHODS We conducted an examination of death certificates sourced from the CDC WONDER (Centers for Disease Control and Prevention Wide-Ranging Online Data for Epidemiologic Research) database, from the years 1999 to 2020. Mortality in adults with HF and cancer was assessed. Age-adjusted mortality rates (AAMRs) per 100,000 persons and annual percent change were reported. RESULTS Between 1999 and 2020, 621,783 deaths occurred from HF in people with cancer. The AAMR declined from 16.4 in 1999 to 11.9 in 2017, after which an increase to 14.5 was observed in 2020. Men had consistently higher overall AAMR as compared to women (men = 18.1 vs women = 9.9). Similar AAMR was observed between non-Hispanic (NH) Blacks/African Americans (13.9) and NH Whites (13.3), with lower in American Indian/Alaska Native (9.6) and Hispanics (7.4). Asian/Pacific Islanders reported the lowest AAMR (5.7). The Midwestern region reported the highest AAMR (14.8). We observed the highest AAMR amongst the older population (61.4). CONCLUSION The mortality rates of people with HF and cancer are increasing in the adult U.S. POPULATION This underscores the need for increased screening, aggressive management, and subsequent surveillance of people at risk or with manifested HF in people with cancer.
Collapse
Affiliation(s)
- Shurjeel Uddin Qazi
- Department of Medicine, Dow University of Health Sciences, Karachi, Pakistan
| | - Arsalan Hamid
- Division of Cardiology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | | | - Michel G Khouri
- Division of Cardiology, Department of Medicine, Duke University, Durham, NC, USA
| | - Markus S Anker
- Department of Cardiology CBF German Heart Center Charité, DZHK, BCRT, University Medicine Berlin FU and HU, Berlin, Germany
| | - Michael E Hall
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Stefan D Anker
- Department of Cardiology (CVK), Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Centre for Cardiovascular Research (DZHK) partner site Berlin, Charité Universitäts medizin Berlin, Germany
| | - Javed Butler
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA; Baylor Scott and White Research Institute, Dallas, Texas, USA
| | - Muhammad Shahzeb Khan
- Baylor Scott and White Research Institute, Dallas, Texas, USA; Division of Cardiology, Baylor Scott and White The Heart Hospital, Plano, TX, USA; Department of Medicine, Baylor College of Medicine, Temple, TX, USA.
| |
Collapse
|
7
|
Kazerooni EA, Wood DE, Rosenthal LS, Smith RA. The American Cancer Society National Lung Cancer Roundtable strategic plan: Introduction. Cancer 2024; 130:3948-3960. [PMID: 39302215 DOI: 10.1002/cncr.35385] [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] [Indexed: 09/22/2024]
Abstract
Lung cancer is the leading cause of cancer death in the United States and across the world. The American Cancer Society National Lung Cancer Roundtable (ACS NLCRT) was established in 2017 as a consortium of public, private, and voluntary organizations with a mission to lower the impact of lung cancer via prevention, early detection, and optimal therapy. The ACS NLCRT supports a comprehensive scope of work that covers the lung cancer continuum, from risk reduction, tobacco prevention and control, and early detection (screening and incidental lung nodule management) to guideline-based staging, biomarker testing, treatment, and survivorship and overarching issues such as stigma and nihilism, health equity, and tactical approaches such as state coalition efforts and policy initiatives. Applying a multidimensional and multisector approach, over 220 public, private, and government agency member organizations and 250 volunteer experts, patients, and caregiver advocate representatives collaborate to address challenges across the lung cancer continuum by catalyzing action to conceive, build, and strengthen innovative solutions. The wide-ranging membership allows the ACS NLCRT to harness the collective power and expertise of the entire lung cancer community by connecting leaders, communities, and systems to improve equity and access. These national, state, and local relationships provide partnerships for the dissemination of ACS NLCRT-developed tools and resources. This article describes the ACS NLCRT and introduces the series of accompanying and future articles that together make up the ACS NLCRT strategic plan, which provides a roadmap for future research, investment, and collaboration to reduce lung cancer mortality and lung cancer-related stigma and enhance survivorship.
Collapse
Affiliation(s)
- Ella A Kazerooni
- Departments of Radiology and Internal Medicine, Michigan Medicine and University of Michigan, Ann Arbor, Michigan, USA
| | - Douglas E Wood
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Lauren S Rosenthal
- Patient Support Department, American Cancer Society National Lung Cancer Roundtable, American Cancer Society, Atlanta, Georgia, USA
| | - Robert A Smith
- American Cancer Society Center for Early Cancer Detection Science, American Cancer Society, Atlanta, Georgia, USA
| |
Collapse
|
8
|
Sonawane K, Garg A, Toll BA, Deshmukh AA, Silvestri GA. Lung Cancer Screening Communication in the US, 2022. JAMA Netw Open 2024; 7:e2442811. [PMID: 39495515 PMCID: PMC11536220 DOI: 10.1001/jamanetworkopen.2024.42811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 09/11/2024] [Indexed: 11/05/2024] Open
Abstract
This cross-sectional study examines lung cancer screening communication between US clinicians and patients by smoking status and demographic, socioeconomic, and clinical characteristics.
Collapse
Affiliation(s)
- Kalyani Sonawane
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston
- Medical University of South Carolina Hollings Cancer Center, Charleston
| | - Ashvita Garg
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston
- Medical University of South Carolina Hollings Cancer Center, Charleston
| | - Benjamin A. Toll
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston
- Medical University of South Carolina Hollings Cancer Center, Charleston
| | - Ashish A. Deshmukh
- Department of Public Health Sciences, College of Medicine, Medical University of South Carolina, Charleston
- Medical University of South Carolina Hollings Cancer Center, Charleston
| | | |
Collapse
|
9
|
Sun H, Sun Z, Wang W, Cha X, Jiang Q, Wang X, Li Q, Liu S, Liu H, Chen Q, Yuan W, Xiao Y. The value of T1- and FST2-Weighted-based radiomics nomogram in differentiating pleomorphic adenoma and Warthin tumor. Transl Oncol 2024; 49:102087. [PMID: 39159554 PMCID: PMC11380391 DOI: 10.1016/j.tranon.2024.102087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 07/23/2024] [Accepted: 08/11/2024] [Indexed: 08/21/2024] Open
Abstract
PURPOSE To establish a radiomics nomogram based on MRI radiomics features combined with clinical characteristics for distinguishing pleomorphic adenoma (PA) from warthin tumor (WT). METHODS 294 patients with PA (n = 159) and WT (n = 135) confirmed by histopathology were included in this study between July 2017 and June 2023. Clinical factors including clinical data and MRI features were analyzed to establish clinical model. 10 MRI radiomics features were extracted and selected from T1WI and FS-T2WI, used to establish radiomics model and calculate radiomics scores (Rad-scores). Clinical factors and Rad-scores were combined to serve as crucial parameters for combined model. Through Receiver operator characteristics (ROC) curve and decision curve analysis (DCA), the discriminative values of the three models were qualified and compared, the best-performing combined model was visualized in the form of a radiomics nomogram. RESULTS The combined model demonstrated excellent discriminative performance for PA and WT in the training set (AUC=0.998) and testing set (AUC=0.993) and performed better compared with the clinical model and radiomics model in the training set (AUC=0.996, 0.952) and testing model (AUC=0.954, 0.849). The DCA showed that the combined model provided more overall clinical usefulness in distinguishing parotid PA from WT than another two models. CONCLUSION An analytical radiomics nomogram based on MRI radiomics features, incorporating clinical factors, can effectively distinguish between PA and WT.
Collapse
Affiliation(s)
- Hongbiao Sun
- Department of Radiology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Zuoheng Sun
- Department of Otolaryngology, Changzheng Hospital, Navy Medical University, Shanghai, China; Department of Otolaryngology, Naval Specialty Medical Center, Naval Medical University, Shanghai, China
| | - Wenwen Wang
- Department of Neurology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Xudong Cha
- Department of Otolaryngology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Qinling Jiang
- Department of Radiology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Xiang Wang
- Department of Radiology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Qingchu Li
- Department of Radiology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Shiyuan Liu
- Department of Radiology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Huanhai Liu
- Department of Otolaryngology, Changzheng Hospital, Navy Medical University, Shanghai, China
| | - Qi Chen
- Department of Radiology, Changzheng Hospital, Navy Medical University, Shanghai, China; Department of Radiology, Kunshan Third People's Hospital, Kunshan, Jiangsu, China
| | - Weimin Yuan
- Department of Radiology, Changzheng Hospital, Navy Medical University, Shanghai, China; Department of Radiology, Qingdao Special Servicemen Recuperation Center of PLA Navy, Qingdao, China
| | - Yi Xiao
- Department of Radiology, Changzheng Hospital, Navy Medical University, Shanghai, China.
| |
Collapse
|
10
|
Christensen NL, Gouliaev A, McPhail S, Lyratzopoulos G, Rasmussen TR, Jensen H. Routes to Diagnosis in Danish Lung Cancer Patients: Emergency Presentation, Age and Smoking History-A Population-Based Cohort Study. Clin Lung Cancer 2024; 25:e348-e356. [PMID: 38890094 DOI: 10.1016/j.cllc.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/16/2024] [Accepted: 05/24/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND The fast-track cancer pathway aims to expedite diagnosis of lung cancer and treatment and is the preferred route to diagnosis. Diagnosis following an unplanned admission (unplanned route) has been associated with poor outcomes. OBJECTIVE This study explores factors associated with lung cancer diagnosis following unplanned admissions, focusing on the elderly population. METHODS A retrospective cohort study using population-based data from Danish registries. Factors such as age, comorbidity, performance status, smoking history, socioeconomic parameters and treatment modality were analyzed in relation to route to diagnosis and prognosis. RESULTS Among 17,835 patients, 16% were elderly (≥ 80 years). The unplanned route constituted 28% of diagnostic routes, with higher proportion among the elderly (33%). Poor performance status and advanced disease stage were associated with the unplanned route. Married patients were less likely to undergo an unplanned route to diagnosis. Smoking did not significantly influence diagnostic route. The adjusted odds ratio for curative treatment and dying 12 months after diagnosis, following unplanned route to diagnosis were 0.68 (95% CI, 0.61-0.76) and 1.48 (95% CI, 1.36-1.61), respectively. CONCLUSION Frailty (poor performance status and high burden of comorbidity) in addition to unfavorable socioeconomic factors, which all were more prevalent among elderly patients, were associated with undergoing an unplanned route to diagnosis. However, age itself was not. Diagnosis following unplanned admission correlated with reduced likelihood of curative treatment and poorer prognosis. Expanding screening initiatives to include frail elderly individuals living alone, along with alertness by primary care clinicians, is warranted to improve outcomes for these patients.
Collapse
Affiliation(s)
- Niels Lyhne Christensen
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; Research Unit for General Practice, Aarhus, Denmark
| | - Anja Gouliaev
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Sean McPhail
- National Disease Registration Service, NHS England, Leeds, UK
| | - Georgios Lyratzopoulos
- Epidemiology of Cancer Healthcare and Outcomes, Department of Behavioural Science and Health, Institute of Epidemiology and Health Care (IEHC), University College London, London, UK
| | - Torben Riis Rasmussen
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Henry Jensen
- The Danish Clinical Quality Program, National Clinical Registries (RKKP), Aarhus, Denmark
| |
Collapse
|
11
|
Henderson LM, Durham DD, Gruden J, Pritchard M, Lane L, Long J, Bellinger C, Rivera MP. Comparing characteristics of individuals screened for lung cancer with 2021 vs 2013 US Preventive Services Task Force recommendations. J Natl Cancer Inst 2024; 116:1825-1829. [PMID: 38913873 PMCID: PMC11542990 DOI: 10.1093/jnci/djae141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/29/2024] [Accepted: 06/13/2024] [Indexed: 06/26/2024] Open
Abstract
We conducted a cross-sectional, multicenter study to compare the demographics, clinical characteristics, and lung cancer screening results among individuals eligible for lung cancer screening per 2013 vs 2021 US Preventive Services Task Force recommendations. Statistical tests are 2 sided, with P less than .05 considered statistically significant. Among 17 702 screened individuals (85.2% 2013 eligible, 14.8% 2021 newly eligible), a higher proportion of individuals screened per 2021 vs 2013 criteria were female (56.1% vs 48.1%, P < .001) and non-Hispanic Black (19.3% vs 13.4%, P < .001). The risk of developing and dying from lung cancer per 1000 people was statistically significantly higher among individuals eligible per 2013 vs 2021 criteria. A higher proportion of lung cancer screening exams had an increased suspicion of lung cancer in the 2013 vs 2021 criteria groups. Our data suggest that, as intended, updated 2021 US Preventive Services Task Force recommendations are leading to a higher proportion of lung cancer screening exams among non-Hispanic Black individuals and women.
Collapse
Affiliation(s)
- Louise M Henderson
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Danielle D Durham
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - James Gruden
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Michael Pritchard
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Lindsay Lane
- Department of Radiology, University of North Carolina, Chapel Hill, NC, USA
| | - Jason Long
- Department of Surgery, University of North Carolina, Chapel Hill, NC, USA
| | - Christina Bellinger
- Department of Internal Medicine, Section on Pulmonary, Critical Care, Allergy and Immunology Diseases, Winston-Salem, NC, USA
| | - M Patricia Rivera
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA
| |
Collapse
|
12
|
Boeri M, Sabia F, Ledda RE, Balbi M, Suatoni P, Segale M, Zanghì A, Cantarutti A, Rolli L, Valsecchi C, Corrao G, Marchianò A, Pastorino U, Sozzi G. Blood microRNA testing in participants with suspicious low-dose CT findings: follow-up of the BioMILD lung cancer screening trial. THE LANCET REGIONAL HEALTH. EUROPE 2024; 46:101070. [PMID: 39319217 PMCID: PMC11421266 DOI: 10.1016/j.lanepe.2024.101070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/22/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024]
Abstract
Background The proper management of suspicious radiologic findings is crucial to optimize the effectiveness of low-dose computed tomography (LDCT) lung cancer screening trials. In the BioMILD study, we evaluated the utility of combining a plasma 24-microRNA signature classifier (MSC) and LDCT to define the individual risk and personalize screening strategies. Here we aim to assess the utility of repeated MSC testing during annual screening rounds in 1024 participants with suspicious LDCT findings. Methods The primary outcome was two-year lung cancer incidence in relation to MSC test results, reported as relative risk (RR) with 95% confidence interval (CI). Lung cancer incidence and mortality were estimated using extended Cox models for time-dependent covariates, yielding the respective hazard ratios (HR). Clinicaltrials.gov ID: NCT02247453. Findings With a median follow-up of 8.5 years, the full study set included 1403 indeterminate LDCT (CTind) and 584 positive LDCT (CT+) results. A lung cancer RR increase in MSC+ compared to MSC- participants was observed in both the CTind (RR: 2.5; 95% CI: 1.4-4.32) and CT+ (RR: 2.6; 95% CI: 1.81-3.74) groups and was maintained when considering stage I or resectable tumors only. A 98% negative predictive value in CTind/MSC- and a 30% positive predictive value in CT+/MSC+ lesions were recorded. At seven years' follow-up, MSC+ participants had a cumulative HR of 4.4 (95% CI: 3.0-6.4) for lung cancer incidence and of 8.1 (95% CI: 2.7-24.5) for lung cancer mortality. Interpretation Our study shows that MSC can be reliably performed during LDCT screening rounds to increase the accuracy of lung cancer risk and mortality prediction and supports its clinical utility in the management of LDCT findings of uncertain malignancy. Funding Italian Association for Cancer Research; Italian Ministry of Health; Horizon2020; National Cancer Institute (NCI); Gensignia LifeScience.
Collapse
Affiliation(s)
- Mattia Boeri
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Federica Sabia
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Roberta E. Ledda
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
- Department of Medicine and Surgery (DiMeC), Section of Radiology, Unit of Surgical Sciences, University of Parma, Parma, 43121, Italy
| | - Maurizio Balbi
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
- Department of Oncology, Radiology Unit, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10043, Italy
| | - Paola Suatoni
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Miriam Segale
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Anna Zanghì
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Anna Cantarutti
- Division of Biostatistics, Department of Statistics and Quantitative Methods, Epidemiology and Public Health, University of Milano-Bicocca, Milan, 20126, Italy
| | - Luigi Rolli
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Camilla Valsecchi
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Giovanni Corrao
- Division of Biostatistics, Department of Statistics and Quantitative Methods, Epidemiology and Public Health, University of Milano-Bicocca, Milan, 20126, Italy
| | - Alfonso Marchianò
- Department of Radiology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Ugo Pastorino
- Unit of Thoracic Surgery, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| | - Gabriella Sozzi
- Unit of Epigenomics & Biomarkers of Solid Tumors, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, 20133, Italy
| |
Collapse
|
13
|
Mazzone PJ, Bach PB, Carey J, Schonewolf CA, Bognar K, Ahluwalia MS, Cruz-Correa M, Gierada D, Kotagiri S, Lloyd K, Maldonado F, Ortendahl JD, Sequist LV, Silvestri GA, Tanner N, Thompson JC, Vachani A, Wong KK, Zaidi AH, Catallini J, Gershman A, Lumbard K, Millberg LK, Nawrocki J, Portwood C, Rangnekar A, Sheridan CC, Trivedi N, Wu T, Zong Y, Cotton L, Ryan A, Cisar C, Leal A, Dracopoli N, Scharpf RB, Velculescu VE, Pike LRG. Clinical Validation of a Cell-Free DNA Fragmentome Assay for Augmentation of Lung Cancer Early Detection. Cancer Discov 2024; 14:2224-2242. [PMID: 38829053 PMCID: PMC11528203 DOI: 10.1158/2159-8290.cd-24-0519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 05/24/2024] [Accepted: 06/01/2024] [Indexed: 06/05/2024]
Abstract
Lung cancer screening via annual low-dose computed tomography has poor adoption. We conducted a prospective case-control study among 958 individuals eligible for lung cancer screening to develop a blood-based lung cancer detection test that when positive is followed by a low-dose computed tomography. Changes in genome-wide cell-free DNA fragmentation profiles (fragmentomes) in peripheral blood reflected genomic and chromatin characteristics of lung cancer. We applied machine learning to fragmentome features to identify individuals who were more or less likely to have lung cancer. We trained the classifier using 576 cases and controls from study samples and validated it in a held-out group of 382 cases and controls. The validation demonstrated high sensitivity for lung cancer and consistency across demographic groups and comorbid conditions. Applying test performance to the screening eligible population in a 5-year model with modest utilization assumptions suggested the potential to prevent thousands of lung cancer deaths. Significance: Lung cancer screening has poor adoption. Our study describes the development and validation of a novel blood-based lung cancer screening test utilizing a highly affordable, low-coverage genome-wide sequencing platform to analyze cell-free DNA fragmentation patterns. The test could improve lung cancer screening rates leading to substantial public health benefits. See related commentary by Haber and Skates, p. 2025.
Collapse
Affiliation(s)
| | | | | | | | - Katalin Bognar
- Medicus Economics, LLC, Formerly PHAR, San Francisco, California
| | | | | | - David Gierada
- Washington University at St. Louis, St. Louis, Missouri
| | | | | | | | | | | | | | - Nichole Tanner
- Department of Veterans Affairs, Charleston, South Carolina
| | - Jeffrey C. Thompson
- Division of Pulmonary, Allergy and Critical Care Medicine, Thoracic Oncology Group, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anil Vachani
- Division of Pulmonary, Allergy and Critical Care Medicine, Thoracic Oncology Group, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kwok-Kin Wong
- New York University Langone Health, New York, New York
| | - Ali H. Zaidi
- Allegheny Health Network, Pittsburgh, Pennsylvania
| | | | | | | | | | | | | | | | | | | | - Tony Wu
- DELFI Diagnostics, Baltimore, Maryland
| | | | | | | | | | | | | | - Robert B. Scharpf
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Victor E. Velculescu
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | |
Collapse
|
14
|
Snider M, Salama JK, Boyer M. Survival and recurrence rates following SBRT or surgery in medically operable Stage I NSCLC. Lung Cancer 2024; 197:107962. [PMID: 39366309 DOI: 10.1016/j.lungcan.2024.107962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 09/17/2024] [Accepted: 09/21/2024] [Indexed: 10/06/2024]
Abstract
OBJECTIVES Surgery is the standard of care for early-stage non-small cell lung cancer (NSCLC), with SBRT reserved for patients who are not surgical candidates. We hypothesized overall survival (OS), lung cancer-specific survival (LCSS), progression free survival (PFS), and recurrence rates following SBRT or surgery in medically operable patients with Stage I NSCLC from the Veterans' Health Care System (VAHS) would be equivalent. MATERIALS AND METHODS Medically operable patients diagnosed with Stage I NSCLC between 2000-2020 from the VAHS, determined by an FEV1 or DLCO > 60 % of predicted and Charlson comorbidity index (CCI) of 0 or 1, treated with SBRT or surgery were identified. SBRT patients were propensity score matched in a 1:1:1 ratio to those undergoing resection (SBRT:lobectomy:sub-lobar resection). OS, LCSS, and PFS and site of recurrence were determined. RESULTS 103 patients were included in each cohort. With a median follow-up of 7.9 years 5-year OS for all patients was 51 % (95 % CI 46-57 %). After propensity score matching, OS (HR 2.08, 1.59), LCSS (HR 2.28, 1.97), and PFS (1.97, 1.45) were significantly worse with SBRT compared to either lobectomy or sub-lobar resection, respectively, (p < 0.05 for each comparison). Regional recurrence was significantly higher following SBRT (15.5 % vs 6.8 % or 4.9 %; p < 0.05), but there was no significant difference in local (28.2 % vs 21.4 % or 21.4 %; p > 0.05) or distant recurrence (10.7 % vs 9.7 % or 13.6 %; p > 0.05) when compared to lobectomy or sub-lobar resection, respectively. CONCLUSION In medically operable patients, OS, LCSS, and PFS following either lobectomy or sub-lobar resection were superior to that for SBRT for Stage I NSCLC, likely due in part to higher regional recurrence following SBRT. This suggests that pulmonary function test results and CCI alone are insufficient to define a cohort of medically operable patients suited for SBRT. These data support strategies to overcome regional recurrences seen with SBRT.
Collapse
Affiliation(s)
- Michael Snider
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, United States
| | - Joseph K Salama
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, United States; Radiation Oncology Clinical Service, Durham VA Health Care System, Durham, NC, United States
| | - Matthew Boyer
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, United States; Radiation Oncology Clinical Service, Durham VA Health Care System, Durham, NC, United States.
| |
Collapse
|
15
|
Abdipourbozorgbaghi M, Vancura A, Radpour R, Haefliger S. Circulating miRNA panels as a novel non-invasive diagnostic, prognostic, and potential predictive biomarkers in non-small cell lung cancer (NSCLC). Br J Cancer 2024; 131:1350-1362. [PMID: 39215192 PMCID: PMC11473829 DOI: 10.1038/s41416-024-02831-3] [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: 05/02/2024] [Revised: 08/13/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is characterised by its aggressiveness and poor prognosis. Early detection and accurate prediction of therapeutic responses remain critical for improving patient outcomes. In the present study, we investigated the potential of circulating microRNA (miRNA) as non-invasive biomarkers in patients with NSCLC. METHODS We quantified miRNA expression in plasma from 122 participants (78 NSCLC; 44 healthy controls). Bioinformatic tools were employed to identify miRNA panels for accurate NSCLC diagnosis. Validation was performed using an independent publicly available dataset of more than 4000 NSCLC patients. Next, we correlated miRNA expression with clinicopathological information to identify independent prognostic miRNAs and those predictive of anti-PD-1 treatment response. RESULTS We identified miRNA panels for lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC) diagnosis. The LUAD panel consists of seven circulating miRNAs (miR-9-3p, miR-96-5p, miR-147b-3p, miR-196a-5p, miR-708-3p, miR-708-5p, miR-4652-5p), while the LUSC panel comprises nine miRNAs (miR-130b-3p, miR-269-3p, miR-301a-5p, miR-301b-5p, miR-744-3p, miR-760, miR-767-5p, miR-4652-5p, miR-6499-3p). Additionally, miR-135b-5p, miR-196a-5p, miR-31-5p (LUAD), and miR-205 (LUSC) serve as independent prognostic markers for survival. Furthermore, two miRNA clusters, namely miR-183/96/182 and miR-767/105, exhibit predictive potential in anti-PD-1-treated LUAD patients. CONCLUSIONS Circulating miRNA signatures demonstrate diagnostic and prognostic value for NSCLC and may guide treatment decisions in clinical practice.
Collapse
Affiliation(s)
- Maryam Abdipourbozorgbaghi
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Adrienne Vancura
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Ramin Radpour
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| | - Simon Haefliger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
- Department of BioMedical Research (DBMR), University of Bern, Bern, Switzerland.
| |
Collapse
|
16
|
Gandomi A, Hasan E, Chusid J, Paul S, Inra M, Makhnevich A, Raoof S, Silvestri G, Bade BC, Cohen SL. Evaluating the accuracy of lung-RADS score extraction from radiology reports: Manual entry versus natural language processing. Int J Med Inform 2024; 191:105580. [PMID: 39096594 DOI: 10.1016/j.ijmedinf.2024.105580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 07/16/2024] [Accepted: 07/27/2024] [Indexed: 08/05/2024]
Abstract
INTRODUCTION Radiology scoring systems are critical to the success of lung cancer screening (LCS) programs, impacting patient care, adherence to follow-up, data management and reporting, and program evaluation. LungCT ScreeningReporting and Data System (Lung-RADS) is a structured radiology scoring system that provides recommendations for LCS follow-up that are utilized (a) in clinical care and (b) by LCS programs monitoring rates of adherence to follow-up. Thus, accurate reporting and reliable collection of Lung-RADS scores are fundamental components of LCS program evaluation and improvement. Unfortunately, due to variability in radiology reports, extraction of Lung-RADS scores is non-trivial, and best practices do not exist. The purpose of this project is to compare mechanisms to extract Lung-RADS scores from free-text radiology reports. METHODS We retrospectively analyzed reports of LCS low-dose computed tomography (LDCT) examinations performed at a multihospital integrated healthcare network in New York State between January 2016 and July 2023. We compared three methods of Lung-RADS score extraction: manual physician entry at time of report creation, manual LCS specialist entry after report creation, and an internally developed, rule-based natural language processing (NLP) algorithm. Accuracy, recall, precision, and completeness (i.e., the proportion of LCS exams to which a Lung-RADS score has been assigned) were compared between the three methods. RESULTS The dataset includes 24,060 LCS examinations on 14,243 unique patients. The mean patient age was 65 years, and most patients were male (54 %) and white (75 %). Completeness rate was 65 %, 68 %, and 99 % for radiologists' manual entry, LCS specialists' entry, and NLP algorithm, respectively. Accuracy, recall, and precision were high across all extraction methods (>94 %), though the NLP-based approach was consistently higher than both manual entries in all metrics. DISCUSSION An NLP-based method of LCS score determination is an efficient and more accurate means of extracting Lung-RADS scores than manual review and data entry. NLP-based methods should be considered best practice for extracting structured Lung-RADS scores from free-text radiology reports.
Collapse
Affiliation(s)
- Amir Gandomi
- Northwell, New Hyde Park, NY, USA; Institute of Health System Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA; Frank G. Zarb School of Business, Hofstra University, Hempstead, NY, USA
| | - Eusha Hasan
- Northwell, New Hyde Park, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Jesse Chusid
- Northwell, New Hyde Park, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; North Shore University Hospital, Northwell, Manhasset, NY, USA
| | - Subroto Paul
- Northwell, New Hyde Park, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Lenox Hill Hospital, Northwell, New York, NY, USA
| | - Matthew Inra
- Northwell, New Hyde Park, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Lenox Hill Hospital, Northwell, New York, NY, USA
| | - Alex Makhnevich
- Northwell, New Hyde Park, NY, USA; Institute of Health System Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; North Shore University Hospital, Northwell, Manhasset, NY, USA
| | - Suhail Raoof
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; North Shore University Hospital, Northwell, Manhasset, NY, USA; Lenox Hill Hospital, Northwell, New York, NY, USA
| | | | - Brett C Bade
- Northwell, New Hyde Park, NY, USA; Institute of Health System Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; Lenox Hill Hospital, Northwell, New York, NY, USA.
| | - Stuart L Cohen
- Northwell, New Hyde Park, NY, USA; Institute of Health System Science, Feinstein Institutes for Medical Research, Manhasset, NY, USA; Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA; North Shore University Hospital, Northwell, Manhasset, NY, USA
| |
Collapse
|
17
|
Xu F, Wang C, Li H, Yu B, Chang L, Wang F, Long C, Bai L, Zhao H, Che N. Evaluation of cfDNA fragmentation characteristics in plasma for the diagnosis of lung cancer: A prospective cohort study. Cancer Sci 2024. [PMID: 39466000 DOI: 10.1111/cas.16360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/12/2024] [Accepted: 09/15/2024] [Indexed: 10/29/2024] Open
Abstract
Lung cancer is one of the most prevalent cancers worldwide, yet only approximately 16% of patients are diagnosed in early stage, highlighting the urgent need for novel, highly accurate detection models. In our study, patients with suspected lung cancer or lung disease, as identified through radiographic imaging, along with healthy individuals, were consecutively recruited from Beijing Chest Hospital. Circulating free DNA (cfDNA) was extracted from plasma samples, and low-depth whole-genome sequencing was performed to identify fragmentomic features for model construction. A total of 265 participants were prospectively enrolled, comprising 124 lung cancer patients and 141 noncancer individuals. The model we developed was based on four cfDNA fragmentation characteristics, including 20-bp breakpoint nucleotides motif, fragmentation size coverage, fragmentation size distribution, and copy number variation. In an independent test cohort, the model achieved an area under the curve (AUC) of 0.861 (95% CI: 0.781-0.942) and demonstrated a sensitivity of 70% (95% CI: 53.5%-83.4%) at a specificity of 89.4% (95% CI: 76.9%-96.5%). Notably, the model was also effective in detecting early-stage cancer, with an AUC of 0.808 (95% CI: 0.69-0.925). In summary, our lung cancer detection model shows strong screening capabilities by leveraging four cfDNA fragmentation characteristics, exhibiting robust performance in early cancer diagnosis.
Collapse
Affiliation(s)
- Fudong Xu
- Department of Pathology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Chong Wang
- Thoracic Minimally Invasive Treatment Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Hongxia Li
- Department of Medical Oncology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Bo Yu
- Department of Medicine, Beijing USCI Medical Laboratory, Beijing, China
| | - Luyuan Chang
- Department of Medicine, Beijing USCI Medical Laboratory, Beijing, China
| | - Feng Wang
- Thoracic Minimally Invasive Treatment Center, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Chaolian Long
- Department of Pathology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| | - Ling Bai
- Department of Medicine, Beijing USCI Medical Laboratory, Beijing, China
| | - Hanqing Zhao
- Department of Medicine, Beijing USCI Medical Laboratory, Beijing, China
| | - Nanying Che
- Department of Pathology, Beijing Chest Hospital, Capital Medical University, Beijing, China
- Beijing Tuberculosis & Thoracic Tumor Research Institute, Beijing, China
| |
Collapse
|
18
|
Thien Nguyen CV, Hanh Nguyen TH, Vo DH, Vi Van TT, Huong Nguyen GT, Tran TH, Nguyen TH, Khoa Huynh LA, Nguyen TD, Tran NH, Thi Ha TM, Quynh Le PT, Truong XL, Nguyen HDL, Tran UV, Hoang TQ, Nguyen VB, Le VC, Nguyen XC, Phuong Nguyen TM, Nguyen VH, Nhat Tran NT, Quynh Dang TN, Tran MH, Nguyen PN, Dao TH, Phuc Nguyen HT, Tran NT, Phan TV, Nguyen DS, Tang HS, Giang H, Phan MD, Nguyen HN, Tran LS. Evaluation of a multimodal ctDNA-based assay for detection of aggressive cancers lacking standard screening tests. Future Oncol 2024:1-11. [PMID: 39431470 DOI: 10.1080/14796694.2024.2413266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/03/2024] [Indexed: 10/22/2024] Open
Abstract
Aim: Cancers lacking standard screening (LSS) options account for approximately 70% of cancer-related deaths due to late-stage diagnosis. Circulating tumor DNA (ctDNA) is a promising biomarker for multi-cancer early detection. We previously developed SPOT-MAS, a multimodal ctDNA-based assay analyzing methylation and fragmentomic profiles, effective in detecting common cancers (breast, colorectal, liver, lung and gastric). This study extends the analysis to five LSS cancers: endometrial, esophageal, head and neck, ovarian and pancreatic.Methods: SPOT-MAS was applied to profile cfDNA methylation and fragmentomic patterns in 739 healthy individuals and 135 LSS cancer patients.Results: We identified 347 differentially methylated regions and observed genome-wide hypomethylation across all five LSS cancers. Esophageal and head and neck cancers showed an enrichment of short cfDNA fragments (<150 bp). Eleven 4-mer end motifs were consistently altered in cfDNA fragments across all LSS cancers. Many significant signatures were consistent with previous observations in common cancers. Notably, SPOT-MAS achieved 96.2% specificity and 74.8% overall sensitivity, with a lower sensitivity of 60.7% in early-stage cancers.Conclusion: This proof-of-concept study demonstrates that SPOT-MAS a non-invasive test trained on five common cancer types, could detect a number of LSS cancer cases, potentially complementing existing screening programs.
Collapse
Affiliation(s)
| | | | - Dac Ho Vo
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | | | | | | | | | | | - Thi Minh Thi Ha
- Department of Medical Genetics, University of Medicine & Pharmacy, Hue University, Vietnam
| | - Phan Tuong Quynh Le
- Department of Medical Genetics, University of Medicine & Pharmacy, Hue University, Vietnam
| | - Xuan Long Truong
- Department of Internal Medicine, University of Medicine & Pharmacy, Hue University, Vietnam
| | | | - Uyen Vu Tran
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Thi Van Phan
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | - Hoa Giang
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| | | | | | - Le Son Tran
- Medical Genetics Institute, Ho Chi Minh, Vietnam
| |
Collapse
|
19
|
Atlas SJ, Gallagher KL, McGovern SE, Wint AJ, Smith RE, Aman DG, Zhao W, Burdick TE, Orav EJ, Zhou L, Wright A, Tosteson ANA, Haas JS. Patient Perceptions on the Follow-Up of Abnormal Cancer Screening Test Results. J Gen Intern Med 2024:10.1007/s11606-024-09128-4. [PMID: 39424768 DOI: 10.1007/s11606-024-09128-4] [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: 05/30/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Timely follow-up after an abnormal cancer screening test result is needed to maximize the benefits of screening, but is frequently not achieved. Little is known about patient experiences with the process of following up abnormal screening results. OBJECTIVE Assess patient experiences and perceptions regarding the process of a diagnostic workup following abnormal breast, cervical, or colorectal cancer screening results. DESIGN Survey of participating patients between April 2021 and June 2022 after reaching the primary outcome time point in a randomized controlled trial to improve follow-up of overdue abnormal screening results. PARTICIPANTS Patients from 44 participating practices in three primary care practice networks. MAIN MEASURES Self-reported ease of scheduling follow-up, perceived barriers or concerns, provider trust, and satisfaction with communication and care received for the follow-up of abnormal screening results. RESULTS Overall, 241 (25.0%) patients completed the survey including 66 (32.8%) with breast, 79 (25.3%) with cervical, and 96 (21.3%) with colorectal screening test; median age 55 years, 79.7% women, 80.5% non-Hispanic white, and 51.0% did not complete recommended follow-up. Most patients were worried that the test would find cancer (63.1%), but fewer worried about discomfort or side effects (34.4%), and neither were associated with completing follow-up. However, 17% of patients did not think they needed follow-up tests or appointments and were less likely to complete follow-up (10.5% vs. 24.0%, respectively, p-value 0.009). Most patients were very satisfied with their overall care (71.0%), but only 50.2% strongly agreed that they trusted their provider to put their medical needs above all else when making recommendations. CONCLUSIONS Patients with overdue abnormal breast, cervical, and colorectal cancer screening test results reported important deficiencies in the management of recommended follow-up. Addressing patient concerns about fear of cancer and effectively communicating the need for follow-up procedures may improve timely follow-up after an abnormal cancer screening result. TRIAL REGISTRATION ClinicalTrials.gov NCT03979495.
Collapse
Affiliation(s)
- Steven J Atlas
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA.
| | - Katherine L Gallagher
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Sydney E McGovern
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Amy J Wint
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Rebecca E Smith
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine, Hanover, NH, USA
- Department of Community and Family Medicine, Dartmouth Health, Lebanon, NH, USA
| | - David G Aman
- Research Computing, Dartmouth College, Lebanon, NH, USA
| | - Wenyan Zhao
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine, Hanover, NH, USA
| | - Timothy E Burdick
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine, Hanover, NH, USA
- Department of Community and Family Medicine, Dartmouth Health, Lebanon, NH, USA
- SYNERGY CTSI Research Informatics, Dartmouth Health, Lebanon, NH, USA
- Department of Biomedical Data Science, Geisel School of Medicine, Hanover, NH, USA
| | - E John Orav
- Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Li Zhou
- Division of General Internal Medicine and Primary Care, Brigham and Women's Hospital, Boston, MA, USA
| | - Adam Wright
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anna N A Tosteson
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine, Hanover, NH, USA
- Department of Community and Family Medicine, Dartmouth Health, Lebanon, NH, USA
- Dartmouth Cancer Center, Dartmouth Health and Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Jennifer S Haas
- Division of General Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| |
Collapse
|
20
|
Powers JC, Rothberg MB, Kovach JD, Casacchia NJ, Stanley E, Martinez KA. Clinician response to the 2021 USPSTF recommendation for colorectal cancer screening in average risk adults aged 45-49 years. Am J Prev Med 2024:S0749-3797(24)00348-9. [PMID: 39424207 DOI: 10.1016/j.amepre.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/03/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
INTRODUCTION In 2021, the USPSTF lowered the recommended age of colorectal cancer (CRC) screening initiation from 50 to 45 years. We assessed clinician response to the updated guideline in a major health system. METHODS This was a retrospective cohort study of average-risk, CRC screening-naïve adults aged 45-50 years with a primary care appointment between July 2018 and February 2023. We defined the pre-guideline change period as July 2018-February 2020 (pre-period) and the post-guideline change period as July 2021-February 2023 (post-period). Clinician ordering of any CRC screening type was assessed. Mixed effects Poisson regression was used to model the incidence rate ratio (IRR) of a patient receiving a screening order, including an interaction between age (45-49 years versus 50 years) and time-period (pre- versus post-guideline change.) Variation in screening orders were also described by calendar quarter and clinician. RESULTS There were 28,114 patients in the pre-period and 22,509 in the post-period. Compared to patients aged 40-49 years in the pre-period, those in the post-period were more likely to have screening ordered (IRR:12.1; 95%CI:11.3-13.0). The screening ordering rate increased for 50-year-olds from the pre- to the post-period (IRR:1.08;95%CI:1.01-1.16) and was slightly higher than that of 45-49-year-olds in the post-period (IRR:1.08; 95%CI:1.02-1.14). All clinicians increased their ordering rate for patients aged 45-49 years. Within five months of the guideline change, the ordering rate for 45-49-year-olds and 50-year-olds was nearly the same. CONCLUSIONS Rapidly following the guideline change, clinicians increased their screening ordering rate for 45-49-year-olds, indicating almost complete uptake of the recommendation.
Collapse
|
21
|
Wiesel O, Suharev T, Awad A, Abzah L, Laser-Azogui A, Mark Danieli M. The Potential Benefit of a Novel Urine Biosensor Platform for Lung Cancer Detection in the Decision-Making Process: From the Bench to the Bedside. J Clin Med 2024; 13:6164. [PMID: 39458114 PMCID: PMC11508546 DOI: 10.3390/jcm13206164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Lung cancer is the leading cause of cancer-related mortality worldwide. Lung cancer screening and early detection resulted in a decrease in cancer-specific mortality; however, it introduced additional dilemmas and adherence barriers for patients and providers. Methods: Innovations such as biomolecular diagnosis and biosensor-based technology improve the detection and stratification of high-risk patients and might assist in overcoming adherence barriers, hence providing new horizons for better selection of screened populations. Conclusions: In the present manuscript, we discuss some of the dilemmas clinicians are currently facing during the diagnosis and treatment processes. We further highlight the potential benefits of a novel biosensor platform for lung cancer detection during the decision making process surrounding lung cancer.
Collapse
Affiliation(s)
- Ory Wiesel
- Baruch Padeh—Tzafon Medical Center, Affiliated with the Azrieli Faculty of Medicine, Bar-Ilan University, Zefad 1528001, Israel; (O.W.); (A.A.); (L.A.)
| | | | - Alaa Awad
- Baruch Padeh—Tzafon Medical Center, Affiliated with the Azrieli Faculty of Medicine, Bar-Ilan University, Zefad 1528001, Israel; (O.W.); (A.A.); (L.A.)
| | - Lina Abzah
- Baruch Padeh—Tzafon Medical Center, Affiliated with the Azrieli Faculty of Medicine, Bar-Ilan University, Zefad 1528001, Israel; (O.W.); (A.A.); (L.A.)
| | | | | |
Collapse
|
22
|
Kim BG, Nam H, Hwang I, Choi YL, Hwang JH, Lee HY, Park KM, Shin SH, Jeong BH, Lee K, Kim H, Kim HK, Um SW. The Growth of Screening-Detected Pure Ground-Glass Nodules Following 10 Years of Stability. Chest 2024:S0012-3692(24)05298-X. [PMID: 39389342 DOI: 10.1016/j.chest.2024.09.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 09/02/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
BACKGROUND It remains uncertain how long pure ground-glass nodules (pGGNs) detected on low-dose CT (LDCT) imaging should be followed up. Further studies with longer follow-up periods are needed to determine the optimal follow-up duration for pGGNs. RESEARCH QUESTION What is the percentage of enlarging nodules among pGGNs that have remained stable for 10 years? STUDY DESIGN AND METHODS This was a retrospective cohort study originating from participants with pGGNs detected on LDCT scans between 1997 and 2006 whose natural courses were reported in 2013. We re-analyzed all the follow-up data until July 2022. The study participants were followed up per our institutional guidelines until they were no longer a candidate for definitive treatment. The growth of the pGGNs was defined as an increase in the diameter of the entire nodule by ≥ 2 mm or the appearance of new solid portions within the nodules. RESULTS A total of 89 patients with 135 pGGNs were followed up for a median of 193 months. Of 135 pGGNs, 23 (17.0%) increased in size, and the median time to the first detection of a size change was 71 months. Of the 23 growing pGGNs, 122 were detected on the first LDCT scan and 13 were newly detected on the follow-up CT scan. An increase in size was observed within 5 years in 8 nodules (34.8%), between 5 and 10 years in 12 nodules (52.2%), and following 10 years in 3 nodules (13.0%). Fifteen nodules were histologically confirmed as adenocarcinoma by surgery. Among the 76 pGGNs stable for 10 years, 3 (3.9%) increased in size. INTERPRETATION Among pGGNs that remained stable for 10 years, 3.9% eventually grew, indicating that some pGGNs can grow even following a long period of stability. We suggest that pGGNs may need to be followed up for > 10 years to confirm growth.
Collapse
Affiliation(s)
- Bo-Guen Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Division of Pulmonary Medicine, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyunseung Nam
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Inwoo Hwang
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jung Hye Hwang
- Center for Health Promotion, Samsung Medical Center, Seoul, Republic of Korea
| | - Ho Yun Lee
- Department of Radiology and Center for Imaging Science, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea
| | - Kyung-Mi Park
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sun Hye Shin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Byeong-Ho Jeong
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyungjong Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hojoong Kim
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hong Kwan Kim
- Department of Thoracic and Cardiovascular Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Sang-Won Um
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
23
|
Li C, Chen J, Chen Y, Zhang C, Yang H, Yu S, Song H, Fu P, Zeng X. The association between patterns of exposure to adverse life events and the risk of chronic kidney disease: a prospective cohort study of 140,997 individuals. Transl Psychiatry 2024; 14:424. [PMID: 39375339 PMCID: PMC11458756 DOI: 10.1038/s41398-024-03114-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
Exposure to adverse life events is linked to somatic disorders. The study aims to evaluate the association between adverse events at varying life stages and the risk of chronic kidney disease (CKD), a condition affecting about 10% population worldwide. This prospective cohort study included 140,997 participants from the UK Biobank. Using survey items related to childhood maltreatment, adulthood adversity and catastrophic trauma, we performed latent class analysis to summarize five distinct patterns of exposure to adverse life events, namely "low-level exposure", "childhood exposure", "adulthood exposure", "sexual abuse" and "child-to-adulthood exposure". We used Cox proportional hazard regression to evaluate the association of patterns of exposure to adverse life events with CKD, regression-based mediation analysis to decompose the total effect, and gene-environment-wide interaction study (GEWIS) to identify interactions between genetic loci and adverse life events. During a median follow-up of 5.98 years, 2734 cases of incident CKD were identified. Compared with the "low-level exposure" pattern, "child-to-adulthood exposure" was associated with increased risk of CKD (hazard ratio 1.37, 95% CI 1.14 to 1.65). BMI, smoking and hypertension mediated 11.45%, 9.79%, and 4.50% of this total effect, respectively. Other patterns did not show significant results. GEWIS and subsequent analyses indicated that the magnitude of the association between adverse life events and CKD differed according to genetic polymorphisms, and identified potential underlying pathways (e.g., interleukin 1 receptor activity). These findings underscore the importance of incorporating an individual's psychological encounters and genetic profiles into the precision prevention of CKD.
Collapse
Affiliation(s)
- Chunyang Li
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Chen
- Central Laboratory, Sichuan Academy of Medical Science and Sichuan Provincial Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yilong Chen
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Chao Zhang
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huazhen Yang
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Shaobin Yu
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huan Song
- Center of Mental Health, West China Hospital, Sichuan University, Chengdu, China
- Center of Public Health Sciences, Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | - Ping Fu
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoxi Zeng
- Department of Nephrology and Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
24
|
Marshall HM, Fong KM. Lung cancer screening - Time for an update? Lung Cancer 2024; 196:107956. [PMID: 39321555 DOI: 10.1016/j.lungcan.2024.107956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/27/2024]
Abstract
Lung cancer screening can reduce the mortality of lung cancer, the leading cause of cancer death worldwide. Real world screening experience highlights areas for improvement in a complex and changing world, particularly ethnic disparity, and the potential for new and emerging risk factors, in addition to well known risk of smoking and asbestos exposure. Biomarkers offer the promise of objective risk assessment but are not yet ready for clinical practice. This review discusses some of the major issues faced by lung cancer screening and the potential role for biomarkers.
Collapse
Affiliation(s)
- Henry M Marshall
- The University of Queensland Thoracic Research Centre and the Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| | - Kwun M Fong
- The University of Queensland Thoracic Research Centre and the Department of Thoracic Medicine, The Prince Charles Hospital, Brisbane, Australia
| |
Collapse
|
25
|
Resnick K, Shah A, Mason J, Kuhn P, Nieva J, Shishido SN. Circulation of rare events in the liquid biopsy for early detection of lung mass lesions. Thorac Cancer 2024; 15:2100-2109. [PMID: 39233479 PMCID: PMC11471425 DOI: 10.1111/1759-7714.15429] [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: 05/14/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Lung cancer screening with low-dose computed tomography (CT) scans (LDCT) has reduced mortality for patients with high-risk smoking histories, but it has significant limitations: LDCT screening implementation remains low, high rates of false-positive scans, and current guidelines exclude those without smoking histories. We sought to explore the utility of liquid biopsy (LBx) in early cancer screening and diagnosis of lung cancer. METHODS Using the high-definition single-cell assay workflow, we analyzed 99 peripheral blood samples from three cohorts: normal donors (NDs) with no known pathology (n = 50), screening CT patients (n = 25) with Lung-RADS score of 1-2, and biopsy (BX) patients (n = 24) with abnormal CT scans requiring tissue biopsy. RESULTS For CT and BX patients, demographic information was roughly equivalent; however, average pack-years smoked differed. A total of 14 (58%) BX patients were diagnosed with primary lung cancer (BX+). The comparison of the rare event enumerations among the cohorts revealed a greater incidence of total events, rare cells, and oncosomes, as well as specific cellular phenotypes in the CT and BX cohorts compared with the ND cohort. LBx analytes were also significantly elevated in the BX compared with the CT samples, but there was no difference between BX+ and BX- samples. CONCLUSIONS The data support the utility of the LBx in distinguishing patients with an alveolar lesion from those without, providing a potential avenue for prescreening before LDCT.
Collapse
Affiliation(s)
- Karen Resnick
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Anya Shah
- Convergent Science Institute for Cancer, Michelson Center, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jeremy Mason
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Convergent Science Institute for Cancer, Michelson Center, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Institute of Urology, Catherine & Joseph Aresty Department of UrologyKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Peter Kuhn
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Convergent Science Institute for Cancer, Michelson Center, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Institute of Urology, Catherine & Joseph Aresty Department of UrologyKeck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Biomedical EngineeringViterbi School of Engineering, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Aerospace and Mechanical EngineeringViterbi School of Engineering, University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Biological SciencesDornsife College of Letters, Arts, and Sciences, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jorge Nieva
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Stephanie N. Shishido
- Convergent Science Institute for Cancer, Michelson Center, University of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
26
|
Ezenwankwo E, Nguyen DT, Akpabio IU, Eberth JM. Expanding reach, enhancing capacity: embracing the role of primary care in lung cancer screening and smoking cessation in the United States. LANCET REGIONAL HEALTH. AMERICAS 2024; 38:100870. [PMID: 39253709 PMCID: PMC11382108 DOI: 10.1016/j.lana.2024.100870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024]
Affiliation(s)
- Elochukwu Ezenwankwo
- Drexel University Dornsife School of Public Health, Nesbitt Building, 3215 Market Street, Philadelphia, PA, 19104, United States
| | - Duong Thuy Nguyen
- Drexel University Dornsife School of Public Health, Nesbitt Building, 3215 Market Street, Philadelphia, PA, 19104, United States
| | | | - Jan M Eberth
- Drexel University Dornsife School of Public Health, Nesbitt Building, 3215 Market Street, Philadelphia, PA, 19104, United States
| |
Collapse
|
27
|
Shah SK, Krishnan V, Khan AA, Fass L, Chaudhry T, Seder CW, Geissen NM, Liptay MJ, Alex GC. Women are Underrepresented in Non-small Cell Lung Cancer Clinical Trials: A Systematic Review. Ann Surg Oncol 2024; 31:6673-6679. [PMID: 38987373 DOI: 10.1245/s10434-024-15720-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 06/17/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE To perform a systematic review of clinical trials examining non-small cell lung cancer (NSCLC) to better understand the equity afforded to women in the study of lung cancer. METHODS An electronic search was conducted for all NSCLC clinical trials published between 2010 and 2020 with included words "carcinoma, non-small cell, lung" and "non-small cell lung cancer." Studies from PubMed, Cochrane, and SCOPUS were included and were uploaded into Covidence to assist with systematic review. All articles were screened by two separate individuals and reviewed for location, study type, cancer stage, field of study of the research team, and percentage of females included. Student's t-test was used to compare the means of males and females. RESULTS Across the 269 studies that met inclusion criteria, fewer females than males were enrolled (38.7% vs. 61.1%; p < 0.0001). Compared with studies from 2010 to 2015, those from 2016 to 2020 had greater representation of females (36.7% vs. 41.4%, p = 0.0091, respectively). Both nonsurgical and surgical studies enrolled fewer female than male patients (38.1% vs. 61.7%, p < 0.0001; 43.1% vs. 57.2%, p = 0.0002, respectively). Clinical trials from the USA had the least difference between sexes with an average of 46.7% females enrolled. Less females compared with males were enrolled in early-stage NSCLC (37.6% female vs. 62.6% male, p < 0.0001) and late-stage NSCLC trials (37.6% female vs. 62.0% male, p < 0.0001). CONCLUSIONS Despite recent improvement, there continues to be significant underrepresentation of females compared with males in NSCLC clinical trials.
Collapse
Affiliation(s)
- Savan K Shah
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Vaishnavi Krishnan
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Arsalan A Khan
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Lucas Fass
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Talib Chaudhry
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Christopher W Seder
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Nicole M Geissen
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Michael J Liptay
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Gillian C Alex
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
28
|
Mascalchi M, Puliti D, Cavigli E, Cortés-Ibáñez FO, Picozzi G, Carrozzi L, Gorini G, Delorme S, Zompatori M, Raffaella De Luca G, Diciotti S, Eva Comin C, Alì G, Kaaks R. Large cell carcinoma of the lung: LDCT features and survival in screen-detected cases. Eur J Radiol 2024; 179:111679. [PMID: 39163805 DOI: 10.1016/j.ejrad.2024.111679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2024] [Revised: 07/17/2024] [Accepted: 08/08/2024] [Indexed: 08/22/2024]
Abstract
PURPOSE To investigate the early radiological features and survival of Large Cell Carcinoma (LCC) cases diagnosed in low-dose computed tomography (LDCT) screening trials. METHODS Two radiologists jointly reviewed the radiological features of screen-detected LCCs observed in NLST, ITALUNG, and LUSI trials between 2002 and 2016, comprising a total of 29,744 subjects who underwent 3-5 annual screening LDCT examinations. Survival or causes of death were established according to the mortality registries extending more than 12 years since randomization. RESULTS LCC was diagnosed in 30 (4 %) of 750 subjects with screen-detected lung cancer (LC), including 15 prevalent and 15 incident cases. Three additional LCCs occurred as interval cancers during the screening period. LDCT images were available for 29 cases of screen-detected LCCs, and 28 showed a single, peripheral, and well-defined solid nodule or mass with regularly smooth (39 %), lobulated (43 %), or spiculated (18 %) margins. One case presented as hilar mass. In 9 incident LCCs, smaller solid nodules were identified in prior LDCT examinations, allowing us to calculate a mean Volume Doubling Time (VDT) of 98.7 ± 47.8 days. The overall five-year survival rate was 50 %, with a significant (p = 0.0001) difference between stages I-II (75 % alive) and stages III-IV (10 % alive). CONCLUSIONS LCC is a fast-growing neoplasm that can escape detection by annual LDCT screening. LCC typically presents as a single solid peripheral nodule or mass, often with lobulated margins, and exhibits a short VDT. The 5-year survival reflects the stage at diagnosis.
Collapse
Affiliation(s)
- Mario Mascalchi
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio", University of Florence, Florence, Italy; Division of Cancer Epidemiology (C020), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany.
| | - Donella Puliti
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Edoardo Cavigli
- Department of Radiology, Emergency Radiology AOU Careggi, Florence, Italy
| | - Francisco O Cortés-Ibáñez
- Division of Cancer Epidemiology (C020), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Giulia Picozzi
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Laura Carrozzi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy; Pulmonary Unit, Cardiothoracic and Vascular Department, Pisa University Hospital, Pisa, Italy
| | - Giuseppe Gorini
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Stefan Delorme
- Division of Radiology (E010), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | | | - Giulia Raffaella De Luca
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, Italy
| | - Stefano Diciotti
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, Italy
| | - Camilla Eva Comin
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Greta Alì
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Italy
| | - Rudolf Kaaks
- Division of Cancer Epidemiology (C020), German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany; Translational Lung Research Center Heidelberg (TLRC-H), the German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
29
|
Schattner A, Dubin I, Uliel L. Unexpected Hyperamylasemia. Am J Med 2024; 137:e184-e185. [PMID: 38925499 DOI: 10.1016/j.amjmed.2024.06.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024]
Affiliation(s)
- Ami Schattner
- Department of Medicine, Laniado University Hospital, Sanz Medical Center Netanya, Israel; The Faculty of Medicine, Hebrew University Hadassah Medical School, Jerusalem, Israel; Adelson School of Medicine, Ariel University, Israel.
| | - Ina Dubin
- Department of Medicine, Laniado University Hospital, Sanz Medical Center Netanya, Israel; Adelson School of Medicine, Ariel University, Israel
| | - Livnat Uliel
- Adelson School of Medicine, Ariel University, Israel; Department of Imaging, Laniado University Hospital, Sanz Medical Center, Netanya, Israel
| |
Collapse
|
30
|
Niranjan SJ, Tipre M, Hardy CM, Bowman T, Baskin ML. Empowering minoritized Alabamians screened for lung cancer-The Alabama Lung Cancer Awareness Screening and Education (ALCASE) project. Cancer Med 2024; 13:e70213. [PMID: 39400521 PMCID: PMC11472649 DOI: 10.1002/cam4.70213] [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: 10/11/2023] [Revised: 06/06/2024] [Accepted: 08/28/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND In Alabama only 4% of those eligible have been screened for lung cancer. The ALCASE project focused on navigating eligible individuals to lung cancer screening. METHODS Trained local staff enrolled screen eligible individuals from seven rural counties and one urban county. Demographics and knowledge of and barriers to lung cancer screening were collected using questionnaires. Education was provided and individuals were navigated to undergo screening. Descriptive statistics for enrolled and screened participants were computed using SAS 9.4. Debriefing interviews were conducted with the ALCASE staff regarding facilitators/barriers to implementing this project and lessons learned. Using NVivo, themes were identified through a combined deductive and inductive process. RESULTS In total, 447 people were contacted of which 257 were enrolled. Participants were predominantly African American (86.8%), female (56.8%), and 86.4% had health insurance. Study participants acknowledged the need for more education of lung cancer/screening procedures; help navigating clinics for screening services and having healthcare facilities close to home. The top five barriers to getting screened were transportation, financial issues, emotional concerns, healthcare insurance, and COVID-19. Of the 257, 106 participants (41%) completed a primary care referral and were screened for lung cancer. Debriefing interviews revealed: (i) Overall impressions of implementing ALCASE were positive. (ii) Barriers in implementing ALCASE were identified predominantly at the physician and institutional level. (iii) Facilitators in implementing ALCASE were identified at multiple levels. (iv) Suggestions on improving lung cancer screening leaned toward mitigating barriers at the institutional and structural level. CONCLUSION Ability to get screened is severely challenged by both personal and structural barriers.
Collapse
Affiliation(s)
- Soumya J. Niranjan
- Department of Health Services Administration, School of Health ProfessionsUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Meghan Tipre
- Division of Hematology and Oncology, School of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| | - Claudia M. Hardy
- Division of Hematology/Oncology O'Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Tara Bowman
- O'Neal Comprehensive Cancer CenterUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Monica L. Baskin
- Division of Hematology and Oncology, School of MedicineUniversity of PittsburghPittsburghPennsylvaniaUSA
| |
Collapse
|
31
|
Bando JM, Tashkin DP, Barjaktarevic IZ. Impact of Marijuana Use on Lung Health. Semin Respir Crit Care Med 2024; 45:548-559. [PMID: 38968961 DOI: 10.1055/s-0044-1785679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
The widespread use of marijuana in the context of increasing legalization has both short- and long-term health implications. Although various modes of marijuana use-smoked, vaped, or ingested-may lead to a wide scope of potential systemic effects, we focus here on inhalational use of marijuana as the most common mode with the lung as the organ that is most directly exposed to its effects. Smoked marijuana has been associated with symptoms of chronic bronchitis and histopathologic changes in airway epithelium, but without consistent evidence of long-term decline in pulmonary function. Its role in immunomodulation, both for risk of infection and protection against a hyperinflammatory host response to infection, has been suggested in animal models and in vitro without conclusive extrapolation to humans. Marijuana smoke contains carcinogens like those found in tobacco, raising concern about its role in lung cancer, but evidence is mixed and made challenging by concurrent tobacco use. Vaping may offer a potential degree of harm reduction when compared with smoking marijuana with reduction of exposure to several toxins, including carbon monoxide, and reduction in chronic respiratory symptoms. However, these potential benefits are counterbalanced by risks including vaping-associated lung injury, potentially more intense drug exposure, and other yet not well-understood toxicities. As more states legalize marijuana and the federal government considers changing this from a Schedule I to a Schedule III controlled substance, we anticipate an increase in prospective medical studies concerning the risks related to marijuana use. This review is based on currently available data concerning the impact of inhaled marijuana on lung health.
Collapse
Affiliation(s)
- Joanne M Bando
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Donald P Tashkin
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | - Igor Z Barjaktarevic
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| |
Collapse
|
32
|
Levi M, Lazebnik T, Kushnir S, Yosef N, Shlomi D. Machine learning computational model to predict lung cancer using electronic medical records. Cancer Epidemiol 2024; 92:102631. [PMID: 39053365 DOI: 10.1016/j.canep.2024.102631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 07/06/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Lung cancer (LC) screening using low-dose computed tomography (CT) is recommended according to standard risk criteria or personalized risk calculators. Machine learning (ML) models that can predict disease risk are an emerging method in medicine for identifying hidden associations that are personally unique. MATERIALS AND METHODS Using the tree-based pipeline optimization tool (TPOT), we developed an ML-based model, which is an ensemble of the Random Forest and XGboost models, based on known risk factors for LC, as part of a larger trial for ML prediction using electronic medical records and chest CT. We used data from patients with LC vs. controls (1:2) of patients aged ≥ 35 years. We developed a model for all LC patients as well as for patients with and without a smoking background. We included age, gender, body mass index (BMI), smoking history, socioeconomic status (SES), history of chronic obstructive pulmonary disease (COPD)/emphysema/chronic bronchitis (CB), interstitial lung disease (ILD)/pulmonary fibrosis (PF), and family history of LC. RESULTS Of the 4076 patients, 1428 (35 %) were in the LC group and 2648 (65 %) were in the control group. For the entire study population, our model achieved an accuracy of 71.2 %, with a sensitivity of 69 % and a positive predictive value (PPV) of 74 %. Higher accuracy was achieved for the two subgroups. An accuracy of 74.8 % (sensitivity 72 %, PPV 76 %) and 73.0 % (sensitivity 76 %, PPV 72 %) was achieved for the smoking and never-smoking cohorts, respectively. For the entire population and smoker cohort, COPD/emphysema/CB were the most important contributors, followed by BMI and age, while in the never-smoking cohort, BMI, age and SES were the most important contributors. CONCLUSION Known risk factors for LC could be used in ML models to modestly predict LC. Further studies are needed to confirm these results in new patients and to improve them.
Collapse
Affiliation(s)
- Matanel Levi
- Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Teddy Lazebnik
- Department of Mathematics, Ariel University, Ariel, Israel; Department of Cancer Biology, Cancer Institute, University College London, London, UK
| | - Shiri Kushnir
- Research Authority, Rabin Medical Center, Beilinson Campus, Petah-Tiqwa, Israel
| | - Noga Yosef
- Research Unit, Dan, Petah-Tiqwa District, Clalit Health Services Community Division, Ramat-Gan, Israel
| | - Dekel Shlomi
- Adelson School of Medicine, Ariel University, Ariel, Israel; Pulmonary Clinic, Dan, Petah-Tiqwa District, Clalit Health Services Community Division, Ramat-Gan, Israel.
| |
Collapse
|
33
|
Shah SK, Kim S, Khan AA, Krishnan V, Lally AM, Shah PN, Alex GC, Seder CW, Liptay MJ, Geissen NM. Examination of Firefighting as an Occupational Exposure Criteria for Lung Cancer Screening. Lung 2024; 202:649-655. [PMID: 39164595 DOI: 10.1007/s00408-024-00736-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/30/2024] [Indexed: 08/22/2024]
Abstract
PURPOSE Firefighting is known to be carcinogenic to humans. However, current lung cancer screening guidelines do not account for occupational exposure. We hypothesize that firefighting is an independent risk factor associated with the development of high-risk lung nodules on low-dose CT (LDCT). METHODS Members of a firefighter's union underwent LDCT at a single institution between April 2022 and June 2023 within a lung cancer screening program. Results were interpreted by designated chest radiologists and reported using the Lung-RADS scoring system. Demographic and radiographic data were recorded, and summary statistics are reported. RESULTS 1347 individuals underwent lung cancer screening, with a median age of 51 years (IQR 42-58), including 56 (4.2%) females. Overall, 899 (66.7%) were never smokers, 345 (25.6%) were former smokers, and 103 (7.7%) were current smokers. There were 41 firefighters (3.0%) who had high-risk (Lung-RADS 3 or 4) nodules requiring intervention or surveillance, of which 21 (1.5%) were Lung-RADS 3 and 20 (1.5%) that were Lung-RADS 4. Of the firefighters with high-risk nodules, only 6 (14.6%) were eligible for LDCT based on current screening guidelines. There were 7 high-risk nodules (0.5%) that required procedural intervention, 6 (85.7%) of which were from the non-screening eligible cohort. There were also 20 never-smoking firefighters (57.1%) with high-risk nodules that were non-screening eligible. CONCLUSION Firefighting, even in the absence of smoking history, may be associated with the development of high-risk lung nodules on LDCT. Carefully selected occupational exposures should be considered in the development of future lung cancer screening guidelines.
Collapse
Affiliation(s)
- Savan K Shah
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA
| | - Seungjun Kim
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA
| | - Arsalan A Khan
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA
| | - Vaishnavi Krishnan
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA
| | - Ann M Lally
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA
| | - Palmi N Shah
- Division of Body Imaging Radiology, Department of Diagnostic Radiology and Nuclear Medicine, Rush University Medical Center, Chicago, IL, USA
| | - Gillian C Alex
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA
| | - Christopher W Seder
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA
| | - Michael J Liptay
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA
| | - Nicole M Geissen
- Department of Cardiovascular and Thoracic Surgery, Rush University Medical Center, 1725 W Harrison St, Suite 774, Chicago, IL, 60612, USA.
| |
Collapse
|
34
|
Lim WH, Lee JH, Park H, Park CM, Yoon SH. Effect of smoking on the diagnostic results and complication rates of percutaneous transthoracic needle biopsy. Eur Radiol 2024; 34:6514-6526. [PMID: 38528137 PMCID: PMC11399209 DOI: 10.1007/s00330-024-10705-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 02/01/2024] [Accepted: 02/25/2024] [Indexed: 03/27/2024]
Abstract
OBJECTIVE To investigate the association of smoking with the outcomes of percutaneous transthoracic needle biopsy (PTNB). METHODS In total, 4668 PTNBs for pulmonary lesions were retrospectively identified. The associations of smoking status (never, former, current smokers) and smoking intensity (≤ 20, 21-40, > 40 pack-years) with diagnostic results (malignancy, non-diagnostic pathologies, and false-negative results in non-diagnostic pathologies) and complications (pneumothorax and hemoptysis) were assessed using multivariable logistic regression analysis. RESULTS Among the 4668 PTNBs (median age of the patients, 66 years [interquartile range, 58-74]; 2715 men), malignancies, non-diagnostic pathologies, and specific benign pathologies were identified in 3054 (65.4%), 1282 (27.5%), and 332 PTNBs (7.1%), respectively. False-negative results for malignancy occurred in 20.5% (236/1153) of non-diagnostic pathologies with decidable reference standards. Current smoking was associated with malignancy (adjusted odds ratio [OR], 1.31; 95% confidence interval [CI]: 1.02-1.69; p = 0.03) and false-negative results (OR, 2.64; 95% CI: 1.32-5.28; p = 0.006), while heavy smoking (> 40 pack-years) was associated with non-diagnostic pathologies (OR, 1.69; 95% CI: 1.19-2.40; p = 0.003) and false-negative results (OR, 2.12; 95% CI: 1.17-3.92; p = 0.02). Pneumothorax and hemoptysis occurred in 21.8% (1018/4668) and 10.6% (495/4668) of PTNBs, respectively. Heavy smoking was associated with pneumothorax (OR, 1.33; 95% CI: 1.01-1.74; p = 0.04), while heavy smoking (OR, 0.64; 95% CI: 0.40-0.99; p = 0.048) and current smoking (OR, 0.64; 95% CI: 0.42-0.96; p = 0.04) were inversely associated with hemoptysis. CONCLUSION Smoking history was associated with the outcomes of PTNBs. Current and heavy smoking increased false-negative results and changed the complication rates of PTNBs. CLINICAL RELEVANCE STATEMENT Smoking status and intensity were independently associated with the outcomes of PTNBs. Non-diagnostic pathologies should be interpreted cautiously in current or heavy smokers. A patient's smoking history should be ascertained before PTNB to predict and manage complications. KEY POINTS • Smoking status and intensity might independently contribute to the diagnostic results and complications of PTNBs. • Current and heavy smoking (> 40 pack-years) were independently associated with the outcomes of PTNBs. • Operators need to recognize the association between smoking history and the outcomes of PTNBs.
Collapse
Affiliation(s)
- Woo Hyeon Lim
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Radiology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Jong Hyuk Lee
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Radiology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Hyungin Park
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Chang Min Park
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Department of Radiology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
| | - Soon Ho Yoon
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Department of Radiology, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
| |
Collapse
|
35
|
Milch HS, Haramati LB. The science and practice of imaging-based screening: What the radiologist needs to know. Clin Imaging 2024; 114:110266. [PMID: 39216274 DOI: 10.1016/j.clinimag.2024.110266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Imaging-based screening is an important public health focus and a fundamental part of Diagnostic Radiology. Hence, radiologists should be familiar with the concepts that drive imaging-based screening practice including goals, risks, biases and clinical trials. This review article discusses an array of imaging-based screening exams including the key epidemiology and evidence that drive screening guidelines for abdominal aortic aneurysm, breast cancer, carotid artery disease, colorectal cancer, coronary artery disease, lung cancer, osteoporosis, and thyroid cancer. We will provide an overview on societal interests in screening, screening-related inequities, and opportunities to address them. Emerging evidence for opportunistic screening and the role of AI in imaging-based screening will be explored. In-depth knowledge and formalized training in imaging-based screening strengthens radiologists as clinician scientists and has the potential to broaden our public health leadership opportunities. SUMMARY SENTENCE: An overview of key screening concepts, the evidence that drives today's imaging-based screening practices, and the need for radiologist leadership in screening policies and evidence development.
Collapse
Affiliation(s)
- Hannah S Milch
- Department of Radiology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America.
| | - Linda B Haramati
- Department of Radiology and Biomedical Imaging, Yale School of Medicine, New Haven, CT, United States of America
| |
Collapse
|
36
|
Kunitomo Y, Sather P, Killam J, Pisani MA, Slade MD, Tanoue LT. Impact of Structured Reporting For Lung Cancer Screening Low-Dose CT Scan Incidental Findings on Physician Management. Chest 2024; 166:896-898. [PMID: 38346557 DOI: 10.1016/j.chest.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Affiliation(s)
- Yukiko Kunitomo
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Polly Sather
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Jonathan Killam
- Department of Radiology, Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Margaret A Pisani
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Martin D Slade
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Lynn T Tanoue
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT.
| |
Collapse
|
37
|
Ebrahimpour L, Després P, Manem V. Differential Radiomics-Based Signature Predicts Lung Cancer Risk Accounting for Imaging Parameters in NLST Cohort. Cancer Med 2024; 13:e70359. [PMID: 39463128 PMCID: PMC11513548 DOI: 10.1002/cam4.70359] [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: 07/18/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVE Lung cancer remains the leading cause of cancer-related mortality worldwide, with most cases diagnosed at advanced stages. Hence, there is a need to develop effective predictive models for early detection. This study aims to investigate the impact of imaging parameters and delta radiomic features from temporal scans on lung cancer risk prediction. METHODS Using the National Lung Screening Trial (NLST) within a nested case-control study involving 462 positive screenings, radiomic features were extracted from temporal computed tomography (CT) scans and harmonized with ComBat method to adjust variations in slice thickness category (TC) and reconstruction kernel type (KT). Both harmonized and non-harmonized features from baseline (T0), delta features between T0 and a year later (T1), and combined T0 and delta features were utilized for the analysis. Feature reduction was done using LASSO, followed by five feature selection (FS) methods and nine machine learning (ML) models, evaluated with 5-fold cross-validation repeated 10 times. Synthetic Minority Oversampling Technique (SMOTE) was applied to address class imbalances for lung cancer risk prediction. RESULTS Models using delta features outperformed baseline features, with SMOTE consistently boosting performance when using combination of baseline and delta features. TC-based harmonized features improved performance with SMOTE, but overall, harmonization did not significantly enhance the model performance. The highest test score of 0.76 was achieved in three scenarios: delta features with a Gradient Boosting (GB) model (TC-based harmonization and MultiSurf FS); and T0 + delta features, with both a Support Vector Classifier (SVC) model (KT-based harmonization and F-test FS), and an XGBoost (XGB) model (TC-based harmonization and Mutual Information (MI) FS), all using SMOTE. CONCLUSIONS This study underscores the significance of delta radiomic features and balanced datasets to improve lung cancer prediction. While our findings are based on a subsample of NLST data, they provide a valuable foundation for further exploration. Further research is needed to assess the impact of harmonization on imaging-derived models. Future investigations should explore advanced harmonization techniques and additional imaging parameters to develop robust radiomics-based biomarkers of lung cancer risk.
Collapse
Affiliation(s)
- Leyla Ebrahimpour
- Centre de Recherche du CHU de QuébecUniversité LavalCanada
- Department of Radiology and Imaging SciencesEmory UniversityAtlantaGeorgiaUSA
- Département de Physique, de Génie Physique et D'optiqueUniversité LavalCanada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de QuébecCanada
| | - Philippe Després
- Département de Physique, de Génie Physique et D'optiqueUniversité LavalCanada
- Centre de Recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de QuébecCanada
- Big Data Research CenterUniversité LavalCanada
| | - Venkata S. K. Manem
- Centre de Recherche du CHU de QuébecUniversité LavalCanada
- Big Data Research CenterUniversité LavalCanada
- Cancer Research CenterUniversité LavalCanada
- Département de Biologie Moléculaire, Biochimie Médicale et PathologieUniversité LavalCanada
| |
Collapse
|
38
|
Sun CY, Cao D, Wang YN, Weng NQ, Ren QN, Wang SC, Zhang MY, Mai SJ, Wang HY. Cholesterol inhibition enhances antitumor response of gilteritinib in lung cancer cells. Cell Death Dis 2024; 15:704. [PMID: 39349433 PMCID: PMC11443066 DOI: 10.1038/s41419-024-07082-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: 03/19/2024] [Revised: 09/07/2024] [Accepted: 09/16/2024] [Indexed: 10/02/2024]
Abstract
Repositioning approved antitumor drugs for different cancers is a cost-effective approach. Gilteritinib was FDA-approved for the treatment of FLT3-mutated acute myeloid leukemia in 2018. However, the therapeutic effects and mechanism of Gilteritinib on other malignancies remain to be defined. In this study, we identified that gilteritinib has an inhibitory effect on lung cancer cells (LCCs) without FLT3 mutation in vitro and in vivo. Unexpectedly, we found that gilteritinib induces cholesterol accumulation in LCCs via upregulating cholesterol biosynthetic genes and inhibiting cholesterol efflux. This gilteritinib-induced cholesterol accumulation not only attenuates the antitumor effect of gilteritinib but also induces gilteritinib-resistance in LCCs. However, when cholesterol synthesis was prevented by squalene epoxidase (SQLE) inhibitor NB-598, both LCCs and gilteritinib-resistant LCCs became sensitive to gilteritinib. More importantly, the natural cholesterol inhibitor 25-hydroxycholesterol (25HC) can suppress cholesterol biosynthesis and increase cholesterol efflux in LCCs. Consequently, 25HC treatment significantly increases the cytotoxicity of gilteritinib on LCCs, which can be rescued by the addition of exogenous cholesterol. In a xenograft model, the combination of gilteritinib and 25HC showed significantly better efficacy than either monotherapy in suppressing lung cancer growth, without obvious general toxicity. Thus, our findings identify an increase in cholesterol induced by gilteritinib as a mechanism for LCC survival, and highlight the potential of combining gilteritinib with cholesterol-lowering drugs to treat lung cancer.
Collapse
Affiliation(s)
- Chao-Yue Sun
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P.R. China
- College of Biological and Pharmaceutical Engineering, West Anhui University, Lu'an, 237012, China
| | - Di Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Medical Imaging, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
| | - Yue-Ning Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Nuo-Qing Weng
- Department of Gastrointestinal Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Qian-Nan Ren
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P.R. China
- Department of Radiation Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Shuo-Cheng Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Mei-Yin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Shi-Juan Mai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P.R. China.
| | - Hui-Yun Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, P.R. China.
| |
Collapse
|
39
|
Barta JA, Farjah F, Thomson CC, Dyer DS, Wiener RS, Slatore CG, Smith-Bindman R, Rosenthal LS, Silvestri GA, Smith RA, Gould MK. The American Cancer Society National Lung Cancer Roundtable strategic plan: Optimizing strategies for lung nodule evaluation and management. Cancer 2024. [PMID: 39347601 DOI: 10.1002/cncr.35181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Lung nodules are frequently detected on low-dose computed tomography scans performed for lung cancer screening and incidentally detected on imaging performed for other reasons. There is wide variability in how lung nodules are managed by general practitioners and subspecialists, with high rates of guideline-discordant care. This may be due in part to the level of evidence underlying current practice guideline recommendations (primarily based on findings from uncontrolled studies of diagnostic accuracy). The primary aims of lung nodule management are to minimize harms of diagnostic evaluations while expediting the evaluation, diagnosis, and treatment of lung cancer. Potentially useful tools such as lung cancer probability calculators, automated methods to identify patients with nodules in the electronic health record, and multidisciplinary team evaluation are often underused due to limited availability, accessibility, and/or provider knowledge. Finally, relatively little attention has been paid to identifying and reducing disparities among individuals with screening-detected or incidentally detected lung nodules. This contribution to the American Cancer Society National Lung Cancer Roundtable Strategic Plan aims to identify and describe these knowledge gaps in lung nodule management and propose recommendations to advance clinical practice and research. Major themes that are addressed include improving the quality of evidence supporting lung nodule evaluation guidelines, strategically leveraging information technology, and placing emphasis on equitable approaches to nodule management. The recommendations outlined in this strategic plan, when carried out through interdisciplinary efforts with a focus on health equity, ultimately aim to improve early detection and reduce the morbidity and mortality of lung cancer. PLAIN LANGUAGE SUMMARY: Lung nodules may be identified on chest scans of individuals who undergo lung cancer screening (screening-detected nodules) or among patients for whom a scan was performed for another reason (incidental nodules). Although the vast majority of lung nodules are not lung cancer, it is important to have evidence-based, standardized approaches to the evaluation and management of a lung nodule. The primary aims of lung nodule management are to diagnose lung cancer while it is still in an early stage and to avoid unnecessary procedures and other harms.
Collapse
Affiliation(s)
- Julie A Barta
- Division of Pulmonary and Critical Care Medicine, Jane and Leonard Korman Respiratory Institute, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Farhood Farjah
- Department of Surgery, University of Washington, Seattle, Washington, USA
| | - Carey Conley Thomson
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Beth Israel Lahey Health/Mount Auburn Hospital, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Debra S Dyer
- Department of Radiology, National Jewish Health, Denver, Colorado, USA
| | - Renda Soylemez Wiener
- Center for Healthcare Organization & Implementation Research, VA Boston Healthcare System, Boston, Massachusetts, USA
- National Center for Lung Cancer Screening, Veterans Health Administration, Washington, DC, USA
- The Pulmonary Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Christopher G Slatore
- Division of Pulmonary and Critical Care Medicine, Oregon Health and Science University, Portland, Oregon, USA
| | - Rebecca Smith-Bindman
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California, USA
| | - Lauren S Rosenthal
- American Cancer Society National Lung Cancer Roundtable, Atlanta, Georgia, USA
| | - Gerard A Silvestri
- Division of Pulmonary and Critical Care Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Robert A Smith
- Center for Early Cancer Detection Science, American Cancer Society, Atlanta, Georgia, USA
| | - Michael K Gould
- Department of Health Systems Science, Kaiser Permanente, Bernard J. Tyson School of Medicine, Pasadena, California, USA
| |
Collapse
|
40
|
Lee S, Park EH, Jang BY, Kang YJ, Jung KW, Cha HS, Choi KS. Survival of lung cancer patients according to screening eligibility using Korean Lung Cancer Registry 2014-2016. Sci Rep 2024; 14:22585. [PMID: 39343824 PMCID: PMC11439945 DOI: 10.1038/s41598-024-69994-0] [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: 03/19/2024] [Accepted: 08/12/2024] [Indexed: 10/01/2024] Open
Abstract
This study assessed survival for lung cancer patients meeting criteria for the National Lung Cancer Screening Program in Korea launched in 2019 and updated guideline reported by the US Preventive Service Task Force (USPSTF). We assessed all-cause mortality based on the Korean Lung Cancer Registry (KLCR), including lung cancer patients diagnosed in 2014-2016. We compared survival among lung cancer patients eligible for extended USPSTF criteria (age 50-80 years and ≥ 20 pack-years) and those meeting current criteria (age 54-74 years and ≥ 30 pack-years, current or within the past 15 years). The nearest neighbour propensity-score matching was performed to generate a matched set. Kaplan-Meier curves were generated to compare survival among groups; differences in survival were analyzed using the stratified log-rank test. The mortality risk was estimated based on a Cox proportional hazards regression model and the robust standard error was calculated. Of 8110 patients, 37.4% and 24.3% met the extended USPSTF eligibility criteria and National Lung Cancer Screening Program (NLCSP) criteria, respectively. Overall mortality risk was not significantly different between the extended younger age group and the NLCSP group (hazard ratio [HR] [95% confidence interval (CI)]: 0.78 [0.59-1.02]). The extended older age group had a significantly higher mortality risk (HR [95% CI]: 1.41 [1.26-1.58]). Mortality risk was not significantly different between patients who smoked 20-29 pack-years and those who smoked ≥ 30 pack-years (HR [95% CI]: 0.90 [0.79-1.03]). Lung cancer patients aged 50-53 years and those with a 20-29 pack-years smoking history exhibited similar mortality risk to individuals meeting current criteria, while patients aged 75-80 years were at a higher risk of death. Although we verified similar or higher mortality risks in extended subgroups, a careful assessment of the benefits and harms of the screening tests is necessary when contemplating the extension of criteria.
Collapse
Affiliation(s)
- Sangwon Lee
- Cancer Data Center, National Cancer Control Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Eun Hye Park
- Cancer Data Center, National Cancer Control Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
- Department of Medical Information Management, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Bo Yun Jang
- Cancer Data Center, National Cancer Control Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
- Department of Medical Information Management, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Ye Ji Kang
- Cancer Data Center, National Cancer Control Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Kyu-Won Jung
- Division of Cancer Registration and Surveillance, National Cancer Control Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Hyo Soung Cha
- Cancer Data Center, National Cancer Control Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea.
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea.
| | - Kui Son Choi
- Cancer Data Center, National Cancer Control Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea.
- Department of Cancer Control and Population Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea.
| |
Collapse
|
41
|
Pignone M, Chang P, Kluz N, Altillo B, Fekete A, Martinez A, Medbery R, Queralt Y, Shah K, Vanin L. Achieving Equitable Lung Cancer Screening Implementation in a Texas Safety Net Health System. Am J Prev Med 2024:S0749-3797(24)00332-5. [PMID: 39343325 DOI: 10.1016/j.amepre.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/01/2024]
Abstract
INTRODUCTION A lung cancer screening program using low dose CT (LDCT) in a Federally Qualified Health Center (FQHC) in Central Texas was developed and assessed for equitable implementation. METHODS From 11/2020-8/2023, patients aged 55-77 years who currently smoked or quit within 15 years with ≥20 pack-years of exposure were identified through EHR query and mailed outreach, or through direct provider referrals. A bilingual social worker confirmed eligibility, provided telecare shared decision-making (SDM), coordinated screening, and offered smoking cessation. To assess equity, LDCT completion across demographics was compared, in 2023. RESULTS A total of 6,486 patients were mailed outreach materials; 479 patients responded, of whom 108 (22.5%) were eligible and 71 (65.7%) participated in SDM. 629 eligible patients were referred internally; 579 (92.0%) completed SDM. Of the 650 patients who completed SDM, 636 (97.8%) agreed to screening. Mean age was 61.7 years; 38.1% were female. The population was diverse: 35.8% identified as Latino, 17.8% as African-American, 26.8% had Medicare or Medicaid, 48.0% used the county medical assistance program, 14.2% were uninsured, and 76.7% currently smoked. Overall, 528 (83.0%) patients completed LDCT. There were no statistically significant differences in completion by age, gender, race/ethnicity, or insurance status. Spanish-speaking patients were more likely to complete the CT than English speakers (OR 2.22, 95% CI=1.22, 4.41) and those who formerly smoked were more likely to complete the CT than patients who currently smoked (OR 1.93, 95% CI=1.12, 3.51). CONCLUSIONS The navigator-centered program achieved equitable implementation of lung cancer screening in a diverse FQHC system.
Collapse
Affiliation(s)
- Michael Pignone
- Department of Medicine, Duke University School of Medicine, Durham, North Carolina; Department of Population Health, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Patrick Chang
- Department of Population Health, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Nicole Kluz
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas.
| | - Brandon Altillo
- Department of Population Health, Dell Medical School, The University of Texas at Austin, Austin, Texas; Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas; CommUnityCare Health Centers, Austin, Texas
| | - Andrea Fekete
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas; Central Health East Multispecialty Clinic, Austin, Texas
| | - Amaris Martinez
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Rachel Medbery
- Department of Surgery and Perioperative Care, Dell Medical School, The University of Texas at Austin, Austin, Texas; Cardiothoracic and Vascular Surgeons, Austin, Texas
| | - Yvonne Queralt
- Department of Diagnostic Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas; Austin Radiological Association, Austin, Texas
| | - Koonj Shah
- Department of Internal Medicine, Dell Medical School, The University of Texas at Austin, Austin, Texas; Dell Seton Medical Center, Ascension, Austin, Texas
| | | |
Collapse
|
42
|
Zhang J, Rui H, Hu H. Noninvasive multi-cancer detection using blood-based cell-free microRNAs. Sci Rep 2024; 14:22136. [PMID: 39333750 PMCID: PMC11436735 DOI: 10.1038/s41598-024-73783-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Patients diagnosed with early-stage cancers have a substantially higher chance of survival than those with late-stage diseases. However, the option for early cancer screening is limited, with most cancer types lacking an effective screening tool. Here we report a miRNA-based blood test for multi-cancer early detection based on examination of serum microRNA microarray data from cancer patients and controls. First, a large multi-cancer training set that included 1,408 patients across 7 cancer types and 1,408 age- and gender-matched non-cancer controls was used to develop a 4-microRNA diagnostic model using 10-fold cross-validation. In three independent validation sets comprising a total of 4,875 cancer patients across 13 cancer types and 3,722 non-cancer participants, the 4-microRNA model achieved greater than 90% sensitivity for 9 cancer types (lung, biliary tract, bladder, colorectal, esophageal, gastric, glioma, pancreatic, and prostate cancers) and 75-84% sensitivity for 3 cancer types (sarcoma, liver, and ovarian cancer), while maintaining greater than 99% specificity. The sensitivity remained to be > 99% for patients with stage 1 lung cancer. Our study provided novel evidence to support the development of an inexpensive and accurate miRNA-based blood test for multi-cancer early detection.
Collapse
Affiliation(s)
| | - Hallgeir Rui
- Department of Pharmacology, Physiology & Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA
| | - Hai Hu
- Chan Soon-Shiong Institute of Molecular Medicine at Windber, 620 7th Street, 15963, Windber, PA, USA.
- miRoncol Diagnostics, Inc, Philadelphia, PA, USA.
| |
Collapse
|
43
|
Salem DP, Bortolin LT, Gusenleitner D, Grosha J, Zabroski IO, Biette KM, Banerjee S, Sedlak CR, Byrne DM, Hamzeh BF, King MS, Cuoco LT, Santos-Heiman T, Barcaskey GN, Yang KS, Duff PA, Winn-Deen ES, Guettouche T, Mattoon DR, Huang EK, Schekman RW, Couvillon AD, Sedlak JC. Colocalization of Cancer-Associated Biomarkers on Single Extracellular Vesicles for Early Detection of Cancer. J Mol Diagn 2024:S1525-1578(24)00209-5. [PMID: 39326670 DOI: 10.1016/j.jmoldx.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 07/16/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
Detection of cancer early, when it is most treatable, remains a significant challenge because of the lack of diagnostic methods sufficiently sensitive to detect nascent tumors. Early-stage tumors are small relative to their tissue of origin, heterogeneous, and infrequently manifest in clinical symptoms. Detection of their presence is made more difficult by a lack of abundant tumor-specific indicators (ie, protein biomarkers, circulating tumor DNA) that would enable detection using a noninvasive diagnostic assay. To overcome these obstacles, we have developed a liquid biopsy assay that interrogates circulating extracellular vesicles (EVs) to detect tumor-specific biomarkers colocalized on the surface of individual EVs. We demonstrate the technical feasibility of this approach in human cancer cell line-derived EVs, where we show strong correlations between assay signal and cell line gene/protein expression for the ovarian cancer-associated biomarkers bone marrow stromal antigen-2, folate receptor-α, and mucin-1. Furthermore, we demonstrate that detecting distinct colocalized biomarkers on the surface of EVs significantly improves discrimination performance relative to single biomarker measurements. Using this approach, we observe promising discrimination of high-grade serous ovarian cancer versus benign ovarian masses and healthy women in a proof-of-concept clinical study.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Christopher R Sedlak
- Mercy BioAnalytics Inc., Waltham, Massachusetts; Former Mercy BioAnalytics employee
| | | | | | | | | | | | | | | | | | | | | | | | | | - Randy W Schekman
- HHMI Investigator, Department of Molecular and Cell Biology, Li Ka Shing Center, University of California Berkeley, Berkeley, California
| | | | | |
Collapse
|
44
|
Wang C, Shao J, He Y, Wu J, Liu X, Yang L, Wei Y, Zhou XS, Zhan Y, Shi F, Shen D, Li W. Data-driven risk stratification and precision management of pulmonary nodules detected on chest computed tomography. Nat Med 2024:10.1038/s41591-024-03211-3. [PMID: 39289570 DOI: 10.1038/s41591-024-03211-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 07/22/2024] [Indexed: 09/19/2024]
Abstract
The widespread implementation of low-dose computed tomography (LDCT) in lung cancer screening has led to the increasing detection of pulmonary nodules. However, precisely evaluating the malignancy risk of pulmonary nodules remains a formidable challenge. Here we propose a triage-driven Chinese Lung Nodules Reporting and Data System (C-Lung-RADS) utilizing a medical checkup cohort of 45,064 cases. The system was operated in a stepwise fashion, initially distinguishing low-, mid-, high- and extremely high-risk nodules based on their size and density. Subsequently, it progressively integrated imaging information, demographic characteristics and follow-up data to pinpoint suspicious malignant nodules and refine the risk scale. The multidimensional system achieved a state-of-the-art performance with an area under the curve (AUC) of 0.918 (95% confidence interval (CI) 0.918-0.919) on the internal testing dataset, outperforming the single-dimensional approach (AUC of 0.881, 95% CI 0.880-0.882). Moreover, C-Lung-RADS exhibited a superior sensitivity compared with Lung-RADS v2022 (87.1% versus 63.3%) in an independent cohort, which was screened using mobile computed tomography scanners to broaden screening accessibility in resource-constrained settings. With its foundation in precise risk stratification and tailored management, this system has minimized unnecessary invasive procedures for low-risk cases and recommended prompt intervention for extremely high-risk nodules to avert diagnostic delays. This approach has the potential to enhance the decision-making paradigm and facilitate a more efficient diagnosis of lung cancer during routine checkups as well as screening scenarios.
Collapse
Affiliation(s)
- Chengdi Wang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| | - Jun Shao
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Yichu He
- Department of Research and Development, United Imaging Intelligence, Shanghai, China
| | - Jiaojiao Wu
- Department of Research and Development, United Imaging Intelligence, Shanghai, China
| | - Xingting Liu
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Liuqing Yang
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Ying Wei
- Department of Research and Development, United Imaging Intelligence, Shanghai, China
| | - Xiang Sean Zhou
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China
| | - Yiqiang Zhan
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China
| | - Feng Shi
- Department of Research and Development, United Imaging Intelligence, Shanghai, China.
| | - Dinggang Shen
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai, China.
- Shanghai Clinical Research and Trial Center, Shanghai, China.
| | - Weimin Li
- Department of Pulmonary and Critical Care Medicine, Targeted Tracer Research and Development Laboratory, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, China.
| |
Collapse
|
45
|
Mascalchi M, Cavigli E, Picozzi G, Cozzi D, De Luca GR, Diciotti S. The Azygos Esophageal Recess Is Not to Be Missed in Screening Lung Cancer With LDCT. J Thorac Imaging 2024:00005382-990000000-00151. [PMID: 39267479 DOI: 10.1097/rti.0000000000000813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
PURPOSE Lesion overlooking and late diagnostic workup can compromise the efficacy of low-dose CT (LDCT) screening of lung cancer (LC), implying more advanced and less curable disease stages. We hypothesized that the azygos esophageal recess (AER) of the right lower lobe (RLL) might be an area prone to lesion overlooking in LC screening. MATERIALS AND METHODS Two radiologists reviewed the LDCT examinations of all the screen-detected incident LCs observed in the active arm of 2 randomized clinical trials: ITALUNG and national lung screening trial. Those in the AER were compared with those in the remainder of the RLL for possible differences in diagnostic lag according to the Lung-RADS 1.1 recommendations, size, stage, and mortality. RESULTS Six (11.7%) of 51 screen-detected incident LCs of the RLL were located in the AER. The diagnostic lag time was significantly longer (P=0.046) in the AER LC (mean 14±9 mo) than in the LC in the remaining RLL (mean 7.3±1 mo). Size and stage at diagnosis were not significantly different. All 6 subjects with LC in the AER and 16 (35.5%) of 45 subjects with LC in the remaining RLL (P=0.004) died of LC after a median follow-up of 12 years. CONCLUSION Our retrospective study indicates that AER might represent a lung region of the RLL prone to have early LC overlooked due to detection or interpretation errors with possible detrimental consequences for the subject undergoing LC screening.
Collapse
Affiliation(s)
- Mario Mascalchi
- Department of Clinical and Experimental Biomedical Sciences "Mario Serio," University of Florence, Florence, Italy
| | - Edoardo Cavigli
- Radiology Division, Nuovo Ospedale S. Giovanni di Dio "Torregalli", Azienda Sanitaria Toscana Centro, Italy
- Department of Radiology, Emergency Radiology AOU Careggi, Florence, Italy
| | - Giulia Picozzi
- Clinical Epidemiology Unit, Institute for Cancer Research, Prevention and Clinical Network (ISPRO), Florence, Italy
| | - Diletta Cozzi
- Department of Radiology, Emergency Radiology AOU Careggi, Florence, Italy
| | - Giulia Raffaella De Luca
- Department of Electrical, Electronic, and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
| | - Stefano Diciotti
- Department of Electrical, Electronic, and Information Engineering 'Guglielmo Marconi', University of Bologna, Cesena, Italy
- Alma Mater Research Institute for Human-Centered Artificial Intelligence, University of Bologna, Bologna, Italy
| |
Collapse
|
46
|
Stewart GD, Godoy A, Farquhar F, Kitt J, Cartledge J, Kimuli M, Rossi SH, Shinkins B, Burbidge S, Burge SW, Caglic I, Collins E, Crosbie PAJ, Eckert C, Fraser S, Hancock N, Iball GR, Marshall C, Masson G, Neal RD, Rogerson S, Smith A, Sharp SJ, Simmonds I, Wallace T, Ward M, Callister MEJ, Usher-Smith JA. Abdominal Noncontrast Computed Tomography Scanning to Screen for Kidney Cancer and Other Abdominal Pathology Within Community-based Computed Tomography Screening for Lung Cancer: Results of the Yorkshire Kidney Screening Trial. Eur Urol 2024:S0302-2838(24)02567-3. [PMID: 39271419 DOI: 10.1016/j.eururo.2024.08.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND AND OBJECTIVE The Yorkshire Kidney Screening Trial (YKST) assessed the feasibility of adding abdominal noncontrast computed tomography (NCCT) to lung cancer screening to screen for kidney cancer and other abdominal pathology. METHODS A prospective diagnostic study offered abdominal NCCT to 55-80-yr-old ever-smokers attending a UK randomised lung cancer screening trial (May 2021 to October 2022). The exclusion criteria were dementia, frailty, previous kidney/lung cancer, and computed tomography (CT) of the abdomen and thorax within previous 6 and 12 mo, respectively. Six-month follow-up was undertaken. KEY FINDINGS AND LIMITATIONS A total of 4438 people attended lung screening, of whom 4309 (97%) were eligible for and 4019 (93%) accepted abdominal NCCT. Only 3.9% respondents regretted participating. The additional time to conduct the YKST processes was 13.3 min. Of the participants, 2586 (64%) had a normal abdominal NCCT, whilst 787 (20%) required an abdominal NCCT imaging review but no further action and 611 (15%) required further evaluation (investigations and/or clinic). Of the participants, 211 (5.3%) had a new serious finding, including 25 (0.62%) with a renal mass/complex cyst, of whom ten (0.25%) had histologically proven kidney cancer; ten (0.25%) with other cancers; and 60 (1.5%) with abdominal aortic aneurysms (AAAs). Twenty-five (0.62%) participants had treatment with curative intent. Of the participants, 1017 (25%) had nonserious findings, most commonly benign renal cysts (727 [18%]), whereas only 259 (6.4%) had nonserious findings requiring further tests. The number needed to screen to detect one serious abdominal finding was 18; it was 93 to detect one suspicious renal lesion and 402 to detect one histologically confirmed renal cancer. Limitations of the cohort were fixed age range and being prior lung cancer screening attendees. CONCLUSIONS AND CLINICAL IMPLICATIONS In this first prospective risk-stratified screening study of abdominal NCCT offered alongside CT thorax, uptake and participant satisfaction were high. The prevalence of serious findings, cancers, and AAAs, is in the range of established screening programmes such as bowel cancer. Longer-term outcomes and cost effectiveness should now be evaluated.
Collapse
Affiliation(s)
- Grant D Stewart
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK.
| | - Angela Godoy
- CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK; Department of Oncology, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Fiona Farquhar
- Research and Innovation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Jessica Kitt
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Jon Cartledge
- Department of Urology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Michael Kimuli
- Department of Urology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sabrina H Rossi
- Department of Surgery, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK; CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK
| | - Bethany Shinkins
- Division of Health Sciences, Warwick Medical School, University of Warwick, Coventry, UK; Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Simon Burbidge
- Department of Radiology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Sarah W Burge
- CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK; Department of Oncology, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | - Iztok Caglic
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Emma Collins
- Department of Endocrine Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Philip A J Crosbie
- Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Claire Eckert
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Sheila Fraser
- Department of Endocrine Surgery, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Neil Hancock
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Gareth R Iball
- School of AHP & Midwifery, Faculty of Health Studies, University of Bradford, Bradford, UK
| | | | | | - Richard D Neal
- Exeter Collaboration for Academic Primary Care (APEx), University of Exeter, Exeter, UK
| | - Suzanne Rogerson
- Research and Innovation, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Andrew Smith
- The Pancreas Unit, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Stephen J Sharp
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Irene Simmonds
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Tom Wallace
- Leeds Vascular Institute, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Matthew Ward
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK
| | - Matthew E J Callister
- Leeds Institute of Health Sciences, University of Leeds, Leeds, UK; Department of Respiratory Medicine, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Juliet A Usher-Smith
- CRUK Cambridge Centre, Cambridge Biomedical Campus, Cambridge, UK; Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Chen A, Wu E, Huang R, Shen B, Han R, Wen J, Zhang Z, Li Q. Development of Lung Cancer Risk Prediction Machine Learning Models for Equitable Learning Health System: Retrospective Study. JMIR AI 2024; 3:e56590. [PMID: 39259582 PMCID: PMC11425024 DOI: 10.2196/56590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 04/02/2024] [Accepted: 05/01/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND A significant proportion of young at-risk patients and nonsmokers are excluded by the current guidelines for lung cancer (LC) screening, resulting in low-screening adoption. The vision of the US National Academy of Medicine to transform health systems into learning health systems (LHS) holds promise for bringing necessary structural changes to health care, thereby addressing the exclusivity and adoption issues of LC screening. OBJECTIVE This study aims to realize the LHS vision by designing an equitable, machine learning (ML)-enabled LHS unit for LC screening. It focuses on developing an inclusive and practical LC risk prediction model, suitable for initializing the ML-enabled LHS (ML-LHS) unit. This model aims to empower primary physicians in a clinical research network, linking central hospitals and rural clinics, to routinely deliver risk-based screening for enhancing LC early detection in broader populations. METHODS We created a standardized data set of health factors from 1397 patients with LC and 1448 control patients, all aged 30 years and older, including both smokers and nonsmokers, from a hospital's electronic medical record system. Initially, a data-centric ML approach was used to create inclusive ML models for risk prediction from all available health factors. Subsequently, a quantitative distribution of LC health factors was used in feature engineering to refine the models into a more practical model with fewer variables. RESULTS The initial inclusive 250-variable XGBoost model for LC risk prediction achieved performance metrics of 0.86 recall, 0.90 precision, and 0.89 accuracy. Post feature refinement, a practical 29-variable XGBoost model was developed, displaying performance metrics of 0.80 recall, 0.82 precision, and 0.82 accuracy. This model met the criteria for initializing the ML-LHS unit for risk-based, inclusive LC screening within clinical research networks. CONCLUSIONS This study designed an innovative ML-LHS unit for a clinical research network, aiming to sustainably provide inclusive LC screening to all at-risk populations. It developed an inclusive and practical XGBoost model from hospital electronic medical record data, capable of initializing such an ML-LHS unit for community and rural clinics. The anticipated deployment of this ML-LHS unit is expected to significantly improve LC-screening rates and early detection among broader populations, including those typically overlooked by existing screening guidelines.
Collapse
Affiliation(s)
- Anjun Chen
- School of Public Health, Guilin Medical University, Guilin, China
| | - Erman Wu
- West China Hospital, Chengdu, China
| | | | | | | | - Jian Wen
- Department of Neurology, Guilin Medical University Affiliated Hospital, Guilin, Guangxi, China
| | - Zhiyong Zhang
- School of Public Health, Guilin Medical University, Guilin, China
| | - Qinghua Li
- Department of Neurology, Guilin Medical University Affiliated Hospital, Guilin, Guangxi, China
| |
Collapse
|
48
|
Graber-Naidich A, Choi E, Wu JT, Ellis-Caleo TJ, Neal J, Wakelee HA, Kurian AW, Han SS. Smoking and the Risk of Second Primary Lung Cancer Among Breast Cancer Survivors from the Population-Based UK Biobank Study. Clin Lung Cancer 2024:S1525-7304(24)00195-5. [PMID: 39332922 DOI: 10.1016/j.cllc.2024.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/29/2024]
Abstract
OBJECTIVE Long-term breast cancer (BC) survivors are known to develop second malignancies, with second primary lung cancer (SPLC) one common type. Smoking was identified as a main risk factor for SPLC among BC survivors. These findings were limited to the U.S. and focused on smoking status, not incorporating cumulative smoking exposures (eg, pack-years). We examine SPLC incidence and evaluate the associations between SPLC risk and cumulative cigarette smoking exposures and other potential factors among BC survivors in a prospective European cohort. METHODS Of 502,505 participants enrolled in the UK Biobank in 2006 to 2010, we identified 8429 patients diagnosed with BC between 2006 and 2016 and followed for second malignancies through 2016. Smoking information was collected at enrollment, and treatment data were collected using electronic health records. Multivariable cause-specific Cox regression (CSC) evaluated the association between each factor and SPLC risk. RESULTS Of 8429 BC patients, 40 (0.47%) developed SPLC over 45,376 person-years. The 10-year cumulative SPLC incidence was 0.48% (95% CI = 0.33%-0.62%). The CSC analysis confirmed the association between SPLC and ever-smoking status (adjusted hazard-ratio (aHR) = 3.46 (P < .001). The analysis showed a 24% increment in SPLC risk per 10 smoking pack-years among BC survivors (aHR = 1.24 per-10 pack-years, P = .01). The associations between SPLC and other variables remained statistically insignificant. We applied the USPSTF lung cancer screening eligibility criteria and found that 80% of the 40 BC survivors who developed SPLC would have been ineligible for lung cancer screening. CONCLUSION In a large, European cohort, cumulative smoking exposure is significantly associated with SPLC risk among BC survivors.
Collapse
Affiliation(s)
| | - Eunji Choi
- Quantitative Sciences Unit, Stanford University, Stanford, CA; Department of Population Health Sciences, Weill Cornell Medicine, New York, NY
| | - Julie T Wu
- Quantitative Sciences Unit, Stanford University, Stanford, CA
| | | | - Joel Neal
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA
| | - Heather A Wakelee
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA
| | - Allison W Kurian
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA
| | - Summer S Han
- Quantitative Sciences Unit, Stanford University, Stanford, CA; Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA.
| |
Collapse
|
49
|
Reddy A, Abe-Nornes F, Haskell A, Saito M, Schumacher M, Venkat A, Venkatasubramanian K, Woodhouse K, Zhang Y, Niktafar H, Leveque A, Kedroske B, Ramnath N, Cohn A. Developing a systems-focused tool for modeling lung cancer screening resource needs. COST EFFECTIVENESS AND RESOURCE ALLOCATION 2024; 22:63. [PMID: 39237997 PMCID: PMC11378520 DOI: 10.1186/s12962-024-00573-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/25/2024] [Indexed: 09/07/2024] Open
Abstract
BACKGROUND Early detection through screening dramatically improves lung cancer survival rates, including among war Veterans, who are at heightened risk. The effectiveness of low dose computed tomography scans in lung cancer screening (LCS) prompted the Veteran's Affairs Lung Precision Oncology Program (VA LPOP) to increase screening rates. We aimed to develop an adaptive population health tool to determine adequate resource allocation for the program, with a specific focus on primary care providers, nurse navigators, and radiologists. METHODS We developed a tool using C + + that uses inputs that represents the process of the VA LCS program in Ann Arbor, Michigan to calculate FTEs of human resource needs to screen a given population. Further, we performed a sensitivity analysis to understand how resource needs are impacted by changes in population, screening eligibility, and time allocated for the nurse navigators' tasks. RESULTS Using estimates from the VA LCS Program as demonstrative inputs, we determined that the greatest number of full-time equivalents required were for radiologists, followed by nurse navigators and then primary care providers, for a target population of 75,000. An increase in the population resulted in a linear increase of resource needs, with radiologists experiencing the greatest rate of increase, followed by nurse navigators and primary care providers. These resource requirements changed with primary care providers, nurse navigators and radiologists demonstrating the greatest increase when 1-20, 20-40 and > 40% of Veterans accepted to be screened respectively. Finally, when increasing the time allocated to check eligibility by the nurse navigator from zero to three minutes, there was a linear increase in the full-time equivalents required for the nurse navigator. CONCLUSION Variation of resource utilization demonstrated by our user facing tool emphasizes the importance of tailored strategies to accommodate specific population demographics and downstream work. We will continue to refine this tool by incorporating additional variability in system parameters, resource requirements following an abnormal test result, and resource distribution over time to reach steady state. While our tool is designed for a specific program in one center, it has wider applicability to other cancer screening programs.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Anthony Leveque
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Beth Kedroske
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| | - Nithya Ramnath
- Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
- Rogel Cancer Center, Michigan Medicine, Ann Arbor, MI, USA
| | - Amy Cohn
- University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
50
|
Bashour SI, Khan A, Song J, Chintalapani G, Kleinszig G, Sabath BF, Lin J, Grosu HB, Jimenez CA, Eapen GA, Ost DE, Sarkiss M, Casal RF. Improving Shape-Sensing Robotic-Assisted Bronchoscopy Outcomes with Mobile Cone-Beam Computed Tomography Guidance. Diagnostics (Basel) 2024; 14:1955. [PMID: 39272739 PMCID: PMC11394119 DOI: 10.3390/diagnostics14171955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Computed tomography to body divergence (CTBD) is one of the main barriers to bronchoscopic techniques for the diagnosis of peripherally located lung nodules. Cone-beam CT (CBCT) guidance is being rapidly adopted to correct for this phenomenon and to potentially increase diagnostic outcomes. In this trial, we hypothesized that the addition of mobile CBCT (m-CBCT) could improve the rate of tool in lesion (TIL) and the diagnostic yield of shape-sensing robotic-assisted bronchoscopy (SS-RAB). METHODS This was a prospective, single-arm study, which enrolled patients with peripheral lung nodules of 1-3 cm and compared the rate of TIL and the diagnostic yield of SS-RAB alone and combined with mCBCT. RESULTS A total of 67 subjects were enrolled, the median nodule size was 1.7 cm (range, 0.9-3 cm). TIL was achieved in 23 patients (34.3%) with SS-RAB alone, and 66 patients (98.6%) with the addition of mCBCT (p < 0.0001). The diagnostic yield of SS-RAB alone was 29.9% (95% CI, 29.3-42.3%) and it was 86.6% (95% CI, 76-93.7%) with the addition of mCBCT (p < 0.0001). There were no pneumothoraxes or any bronchoscopy-related complications, and the median total dose-area product (DAP) was 50.5 Gy-cm2. CONCLUSIONS The addition of mCBCT guidance to SS-RAB allows bronchoscopists to compensate for CTBD, leading to an increase in TIL and diagnostic yield, with acceptable radiation exposure.
Collapse
Affiliation(s)
- Sami I Bashour
- Department of Pulmonary and Critical Care Medicine, Michael E. DeBakey VA Medical Center, Houston, TX 77030, USA
| | - Asad Khan
- Department of Pulmonary and Critical Care Medicine, Ochsner Health Rush, Meridian, MS 39301, USA
| | - Juhee Song
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | - Bruce F Sabath
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Julie Lin
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Horiana B Grosu
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carlos A Jimenez
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Georgie A Eapen
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - David E Ost
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mona Sarkiss
- Department of Anesthesia and Peri-Operative Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Roberto F Casal
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|