1
|
Wang J, Li B, Zhang L, Wang Z, Shen J. Safety and local efficacy of computed tomography-guided microwave ablation for treating early-stage non-small cell lung cancer adjacent to bronchovascular bundles. Eur Radiol 2024; 34:236-246. [PMID: 37505251 DOI: 10.1007/s00330-023-09997-z] [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: 10/29/2022] [Revised: 05/24/2023] [Accepted: 06/03/2023] [Indexed: 07/29/2023]
Abstract
OBJECTIVES To retrospectively evaluate the safety and efficacy of computed tomography (CT)-guided percutaneous microwave ablation in treating early-stage non-small cell lung cancer (NSCLC) adjacent to bronchovascular bundles. METHODS Two hundred and thirty-one patients with early-stage NSCLC who underwent CT-guided microwave ablation of the tumor were included for analysis. Among these, 66 lesions were located adjacent to the bronchovascular bundle. Achievement of the specific ablation range (defined as the ablation zone encompassing the tumor and the adjacent vessel) was assessed after ablation. Complications and tumor progression after treatment were examined and compared between the bronchovascular bundle and non-bronchovascular bundle groups. RESULTS A total of 231 patients were included. Overall, 1-, 2-, and 3-year local progression-free survival (LPFS) was 77.4%, 70.5%, and 63.8%, respectively. Bronchovascular bundle proximity, pure-solid tumor, tumor size, and ablation margin < 5 mm were independent risk factors for local progression in multivariate analysis. In the bronchovascular bundle group, the 1-, 2- and 3-year LPFS rates were 63.0%, 50.7%, and 43.4%, respectively; vessel proximity and specific ablation range failure were independent risk factors for local progression. Overall survival in the entire cohort was 93.0% at 1 year, 76.1% at 2 years, and 55.0% at 3 years. The incidence of postoperative complications did not significantly differ between the two groups (p > 0.05). The most common complication was pneumothorax. Severe hemoptysis did not occur. CONCLUSION Tumor location near the bronchovascular bundles was a significant risk factor for local progression after microwave ablation. Achieving a specific ablation range may increase LPFS for these lesions. CLINICAL RELEVANCE STATEMENT Achieving the specific ablation range may improve local efficacy for early-stage non-small cell lung cancer located adjacent to the bronchovascular bundle. KEY POINTS • Local efficacy of percutaneous microwave ablation in treating early-stage non-small cell lung cancer was affected by bronchovascular bundle proximity. • Achieving the specific ablation range may improve local efficacy for lesions located adjacent to the bronchovascular bundle.
Collapse
Affiliation(s)
- Jun Wang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Rd, Pudong, Shanghai, 200127, China
| | - Bo Li
- Department of Medical Imaging, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Rd, Pudong, Shanghai, 200127, China
| | - Liang Zhang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Rd, Pudong, Shanghai, 200127, China
| | - Zhi Wang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Rd, Pudong, Shanghai, 200127, China
| | - Jialin Shen
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, No. 160 Pujian Rd, Pudong, Shanghai, 200127, China.
| |
Collapse
|
2
|
Dolgalev I, Zhou H, Murrell N, Le H, Sakellaropoulos T, Coudray N, Zhu K, Vasudevaraja V, Yeaton A, Goparaju C, Li Y, Sulaiman I, Tsay JCJ, Meyn P, Mohamed H, Sydney I, Shiomi T, Ramaswami S, Narula N, Kulicke R, Davis FP, Stransky N, Smolen GA, Cheng WY, Cai J, Punekar S, Velcheti V, Sterman DH, Poirier JT, Neel B, Wong KK, Chiriboga L, Heguy A, Papagiannakopoulos T, Nadorp B, Snuderl M, Segal LN, Moreira AL, Pass HI, Tsirigos A. Inflammation in the tumor-adjacent lung as a predictor of clinical outcome in lung adenocarcinoma. Nat Commun 2023; 14:6764. [PMID: 37938580 PMCID: PMC10632519 DOI: 10.1038/s41467-023-42327-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: 10/28/2022] [Accepted: 10/06/2023] [Indexed: 11/09/2023] Open
Abstract
Approximately 30% of early-stage lung adenocarcinoma patients present with disease progression after successful surgical resection. Despite efforts of mapping the genetic landscape, there has been limited success in discovering predictive biomarkers of disease outcomes. Here we performed a systematic multi-omic assessment of 143 tumors and matched tumor-adjacent, histologically-normal lung tissue with long-term patient follow-up. Through histologic, mutational, and transcriptomic profiling of tumor and adjacent-normal tissue, we identified an inflammatory gene signature in tumor-adjacent tissue as the strongest clinical predictor of disease progression. Single-cell transcriptomic analysis demonstrated the progression-associated inflammatory signature was expressed in both immune and non-immune cells, and cell type-specific profiling in monocytes further improved outcome predictions. Additional analyses of tumor-adjacent transcriptomic data from The Cancer Genome Atlas validated the association of the inflammatory signature with worse outcomes across cancers. Collectively, our study suggests that molecular profiling of tumor-adjacent tissue can identify patients at high risk for disease progression.
Collapse
Affiliation(s)
- Igor Dolgalev
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, USA
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Hua Zhou
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, USA
| | - Nina Murrell
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, USA
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Hortense Le
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | | | - Nicolas Coudray
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, USA
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
- Department of Cell Biology, NYU Grossman School of Medicine, New York, USA
| | - Kelsey Zhu
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
| | | | - Anna Yeaton
- The Optical Profiling Platform at The Broad Institute of MIT And Harvard, Cambridge, USA
| | - Chandra Goparaju
- Department of Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, USA
| | - Yonghua Li
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, New York, USA
| | - Imran Sulaiman
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, New York, USA
| | - Jun-Chieh J Tsay
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, New York, USA
| | - Peter Meyn
- Genome Technology Center, Office of Science and Research, NYU Grossman School of Medicine, New York, USA
| | - Hussein Mohamed
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
| | - Iris Sydney
- Center for Biospecimen Research and Development, NYU Grossman School of Medicine, New York, USA
| | - Tomoe Shiomi
- Center for Biospecimen Research and Development, NYU Grossman School of Medicine, New York, USA
| | - Sitharam Ramaswami
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Genome Technology Center, Office of Science and Research, NYU Grossman School of Medicine, New York, USA
| | - Navneet Narula
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
| | - Ruth Kulicke
- Celsius Therapeutics, Cambridge, Massachusetts, USA
| | - Fred P Davis
- Celsius Therapeutics, Cambridge, Massachusetts, USA
| | | | | | - Wei-Yi Cheng
- Pharma Research & Early Development Informatics, Roche Innovation Center New York, New Jersey, USA
| | - James Cai
- Pharma Research & Early Development Informatics, Roche Innovation Center New York, New Jersey, USA
| | - Salman Punekar
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Vamsidhar Velcheti
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Daniel H Sterman
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, New York, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - J T Poirier
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Ben Neel
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Luis Chiriboga
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
| | - Adriana Heguy
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Genome Technology Center, Office of Science and Research, NYU Grossman School of Medicine, New York, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Thales Papagiannakopoulos
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Bettina Nadorp
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, USA
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA
| | - Matija Snuderl
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Leopoldo N Segal
- Division of Pulmonary, Critical Care and Sleep Medicine, NYU Grossman School of Medicine, New York, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Andre L Moreira
- Department of Pathology, NYU Grossman School of Medicine, New York, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA
| | - Harvey I Pass
- Department of Cardiothoracic Surgery, NYU Grossman School of Medicine, New York, USA.
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA.
| | - Aristotelis Tsirigos
- Department of Pathology, NYU Grossman School of Medicine, New York, USA.
- Applied Bioinformatics Laboratories, NYU Grossman School of Medicine, New York, USA.
- Division of Precision Medicine, Department of Medicine, NYU Grossman School of Medicine, New York, USA.
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York, NY, USA.
| |
Collapse
|
3
|
Jiménez Londoño GA, García Vicente AM, Bosque JJ, Amo-Salas M, Pérez-Beteta J, Honguero-Martinez AF, Pérez-García VM, Soriano Castrejón ÁM. SUVmax to tumor perimeter distance: a robust radiomics prognostic biomarker in resectable non-small cell lung cancer patients. Eur Radiol 2022; 32:3889-3902. [PMID: 35133484 DOI: 10.1007/s00330-021-08523-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 10/18/2021] [Accepted: 11/30/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVE The purpose of this study was to evaluate the prognostic value of novel geometric variables obtained from pre-treatment [18F]FDG PET/CT with respect to classical ones in patients with non-small cell lung cancer (NSCLC). METHODS Retrospective study including stage I-III NSCLC patients with baseline [18F]FDG PET/CT. Clinical, histopathologic, and metabolic parameters were obtained. After tumor segmentation, SUV and volume-based variables, global texture, sphericity, and two novel parameters, normalized SUVpeak to centroid distance (nSCD) and normalized SUVmax to perimeter distance (nSPD), were obtained. Early recurrence (ER) and short-term mortality (STM) were used as end points. Univariate logistic regression and multivariate logistic regression with respect to ER and STM were performed. RESULTS A cohort of 173 patients was selected. ER was detected in 49/104 of patients with recurrent disease. Additionally, 100 patients died and 53 had STM. Age, pathologic lymphovascular invasion, lymph nodal infiltration, TNM stage, nSCD, and nSPD were associated with ER, although only age (aOR = 1.06, p = 0.002), pathologic lymphovascular invasion (aOR = 3.40, p = 0.022), and nSPD (aOR = 0.02, p = 0.018) were significant independent predictors of ER in multivariate analysis. Age, lymph nodal infiltration, TNM stage, nSCD, and nSPD were predictors of STM. Age (aOR = 1.05, p = 0.006), lymph nodal infiltration (aOR = 2.72, p = 0.005), and nSPD (aOR = 0.03, p = 0.022) were significantly associated with STM in multivariate analysis. Coefficient of variation (COV) and SUVmean/SUVmax ratio did not show significant predictive value with respect to ER or STM. CONCLUSION The geometric variables, nSCD and nSPD, are robust biomarkers of the poorest outcome prediction of patients with NSCLC with respect to classical PET variables. KEY POINTS • In NSCLC patients, it is crucial to find prognostic parameters since TNM system alone cannot explain the variation in lung cancer survival. • Age, lymphovascular invasion, lymph nodal infiltration, and metabolic geometrical parameters were useful as prognostic parameters. • The displacement grade of the highest point of metabolic activity towards the periphery assessed by geometric variables obtained from [18F]FDG PET/CT was a robust biomarker of the poorest outcome prediction of patients with NSCLC.
Collapse
Affiliation(s)
| | - Ana Maria García Vicente
- Department of Nuclear Medicine, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Jesús J Bosque
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Mariano Amo-Salas
- Department of Mathematics, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | - Julián Pérez-Beteta
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | | | - Víctor M Pérez-García
- Department of Mathematics, Mathematical Oncology Laboratory (MOLAB), Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | | |
Collapse
|
4
|
Sinha A, Zou Y, Patel AS, Yoo S, Jiang F, Sato T, Kong R, Watanabe H, Zhu J, Massion PP, Borczuk AC, Powell CA. Early-Stage Lung Adenocarcinoma MDM2 Genomic Amplification Predicts Clinical Outcome and Response to Targeted Therapy. Cancers (Basel) 2022; 14:cancers14030708. [PMID: 35158979 PMCID: PMC8833784 DOI: 10.3390/cancers14030708] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/18/2022] [Accepted: 01/28/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Invasive subtypes of lung adenocarcinoma (LUAD) show MDM2 amplification that is associated with poor survival. Mouse double minute 2 (MDM2) is frequently amplified in lung adenocarcinoma (LUAD) and is a negative regulator of p53, which binds to p53 and regulates its activity and stability. Genomic amplification and overexpression of MDM2, together with genetic alterations in p53, leads to genomic and genetic heterogeneity in LUAD that represents a therapeutic target. In vitro assays in a panel of LUAD cell lines showed that tumor cell response to MDM2-targeted therapy is associated with MDM2 amplification. Abstract Lung cancer is the most common cause of cancer-related deaths in both men and women, accounting for one-quarter of total cancer-related mortality globally. Lung adenocarcinoma is the major subtype of non-small cell lung cancer (NSCLC) and accounts for around 40% of lung cancer cases. Lung adenocarcinoma is a highly heterogeneous disease and patients often display variable histopathological morphology, genetic alterations, and genomic aberrations. Recent advances in transcriptomic and genetic profiling of lung adenocarcinoma by investigators, including our group, has provided better stratification of this heterogeneous disease, which can facilitate devising better treatment strategies suitable for targeted patient cohorts. In a recent study we have shown gene expression profiling identified novel clustering of early stage LUAD patients and correlated with tumor invasiveness and patient survival. In this study, we focused on copy number alterations in LUAD patients. SNP array data identified amplification at chromosome 12q15 on MDM2 locus and protein overexpression in a subclass of LUAD patients with an invasive subtype of the disease. High copy number amplification and protein expression in this subclass correlated with poor overall survival. We hypothesized that MDM2 copy number and overexpression predict response to MDM2-targeted therapy. In vitro functional data on a panel of LUAD cells showed that MDM2-targeted therapy effectively suppresses cell proliferation, migration, and invasion in cells with MDM2 amplification/overexpression but not in cells without MDM2 amplification, independent of p53 status. To determine the key signaling mechanisms, we used RNA sequencing (RNA seq) to examine the response to therapy in MDM2-amplified/overexpressing p53 mutant and wild-type LUAD cells. RNA seq data shows that in MDM2-amplified/overexpression with p53 wild-type condition, the E2F → PEG10 → MMPs pathway is operative, while in p53 mutant genetic background, MDM2-targeted therapy abrogates tumor progression in LUAD cells by suppressing epithelial to mesenchymal transition (EMT) signaling. Our study provides a potentially clinically relevant strategy of selecting LUAD patients for MDM2-targeted therapy that may provide for increased response rates and, thus, better survival.
Collapse
Affiliation(s)
- Abhilasha Sinha
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Yong Zou
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (Y.Z.); (P.P.M.)
| | - Ayushi S. Patel
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | | | - Feng Jiang
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Takashi Sato
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo 160-8582, Japan
- Department of Respiratory Medicine, Kitasato University School of Medicine, Sagamihara 252-0374, Japan
| | - Ranran Kong
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Thoracic Surgery, The Second Affiliated Hospital of Medical School, Xi’an Jiaotong University, Xi’an 710004, China
| | - Hideo Watanabe
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jun Zhu
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Sema4, 333 Ludlow St., Stamford, CT 06902, USA;
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Icahn Institute for Data Science and Genomic Technology, New York, NY 10029, USA
| | - Pierre P. Massion
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA; (Y.Z.); (P.P.M.)
| | - Alain C. Borczuk
- Department of Pathology, Weill Cornell Medicine, New York, NY 10021, USA;
| | - Charles A. Powell
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; (A.S.); (A.S.P.); (F.J.); (T.S.); (R.K.); (H.W.)
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Correspondence: ; Tel.: +1-212-241-5656
| |
Collapse
|
5
|
Iijima Y, Ishikawa M, Iwai S, Motono N, Yamagishi S, Koizumi K, Uramoto H. Is extrathoracic metastasis screening necessary for clinical stage IA non-small cell lung cancer? Sci Prog 2022; 105:368504221085152. [PMID: 35275504 PMCID: PMC10358501 DOI: 10.1177/00368504221085152] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Detecting distant metastases when staging lung cancer is critical to avoid unnecessary surgery and provide appropriate multidisciplinary treatment. However, it is controversial as to whether staging studies should be performed routinely for patients with early-stage lung cancer who have no evidence of distant metastasis. Thus, this study aimed to examine the need for extrathoracic metastasis screening in patients with clinical stage IA non-small cell lung cancer, understand the association between extrathoracic metastasis and other clinical features, and evaluate the diagnostic efficiency of imaging screening for preoperative extrathoracic metastasis in patients with early-stage lung cancer. METHODS From 2010 to 2019, 510 patients diagnosed with clinical T1N0 lung cancer, excluding contralateral lung metastases, pleural dissemination, malignant pleural effusion, and malignant pericardial effusion, were treated for primary lung cancer. Patients were divided into two groups, and their clinicopathological characteristics were investigated. RESULTS Five patients (1.0%) had extrathoracic metastases. The histological types were adenocarcinoma in three of the cases, and squamous cell carcinoma and large cell neuroendocrine carcinoma in the other two cases. The T factor was T1b in one case and T1c in four cases. Four patients had solid tumors and one had a solid predominant tumor with an average tumor diameter of 23.0 ± 2.9 mm. The size of solid tumors with extrathoracic metastases was larger than their counterparts. CONCLUSION When evaluating stage IA non-small cell lung cancer with a solid component diameter < 22 mm, or clinical T1mi-1bN0 in computed tomography evaluation, screening for preoperative extrathoracic metastasis may be omitted.
Collapse
Affiliation(s)
- Yoshihito Iijima
- Department of Thoracic Surgery, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Masahito Ishikawa
- Department of Thoracic Surgery, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Shun Iwai
- Department of Thoracic Surgery, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Nozomu Motono
- Department of Thoracic Surgery, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| | - Shigeki Yamagishi
- Department of Thoracic Surgery, Aidu Chuo Hospital, Aizuwakamatsu, Fukushima, Japan
| | - Kiyoshi Koizumi
- Department of Thoracic Surgery, Aidu Chuo Hospital, Aizuwakamatsu, Fukushima, Japan
| | - Hidetaka Uramoto
- Department of Thoracic Surgery, Kanazawa Medical University, Kahoku-gun, Ishikawa, Japan
| |
Collapse
|
6
|
Cheng JT, Yao YH, Gao YE, Zhang SL, Chen HJ, Wang Z, Yan HH, Zhou Q, Tu HY, Zhang XC, Su J, Xie Z, Lizaso A, Chen SY, Lin X, Xiang JX, Wu YL, Yang JJ. Integrated histological and molecular analyses of rebiopsy samples at osimertinib progression improve post-progression survivals: A single-center retrospective study. Lung Cancer 2020; 150:97-106. [PMID: 33126092 DOI: 10.1016/j.lungcan.2020.10.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 09/30/2020] [Accepted: 10/11/2020] [Indexed: 01/10/2023]
Abstract
BACKGROUND This single-center retrospective cohort study sought to investigate the impact of rebiopsy analysis after osimertinib progression in improving the survival outcomes. METHODS Eighty-nine patients with EGFR T790M-positive advanced NSCLC who received second- or further-line osimertinib between January 2017 and July 2019 were included in this study. The co-primary study endpoints were post-progression progression-free survival (pPFS), defined as the time from osimertinib progression until progression from further-line treatment, and post-progression overall survival (pOS), defined as the time from osimertinib progression until death or the last follow-up date. RESULTS Pairwise analysis revealed that receiving targeted therapy as further-line treatment after osimertinib progression did not statistically improve the pPFS (P = 0.285) or the pOS (P = 0.903) compared to chemotherapy. However, patients who submitted rebiopsy samples at osimertinib progression for histological and molecular analyses, particularly those who had actionable markers and received highly matched therapy, had significantly longer pPFS and pOS as compared to those who received low-level matched therapy (pPFS = 10.0 m vs. 4.1 m, P = 0.005; pOS = 19.4 m vs. 10.0 m, P = 0.023), unmatched therapy (pPFS = 10.0 m vs. 4.7 m, P = 0.009; pOS = 19.4 m vs. 7.0 m, P = 0.001), and those without rebiopsy data (Rebiopsy vs Non-rebiopsy; pPFS = 6.1 m vs. 3.3 m, P = 0.014; pOS = 11.7 m vs. 6.8 m, P = 0.011). CONCLUSION Our real-world cohort study demonstrates that integrated histological and molecular analyses of rebiopsy specimens after osimertinib progression could provide more opportunities for individualized treatments to improve the post-progression survival of patients with advanced NSCLC. Our findings provide clinical evidence that supports the inclusion of NGS-based analysis of rebiopsy specimens as standard-of-care after osimertinib progression and warrants further prospective evaluation.
Collapse
Affiliation(s)
- Jiang-Tao Cheng
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China; Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yi-Hui Yao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China; Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Yu-Er Gao
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China; Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Shi-Ling Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China; Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hua-Jun Chen
- Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhen Wang
- Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hong-Hong Yan
- Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Qing Zhou
- Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Hai-Yan Tu
- Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Xu-Chao Zhang
- Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Jian Su
- Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Zhi Xie
- Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | | | | | - Xuan Lin
- Burning Rock Biotech, Guangzhou, 510300, China
| | | | - Yi-Long Wu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China; Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| | - Jin-Ji Yang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China; Cancer Center, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, 510080, China.
| |
Collapse
|
7
|
Qu CH, Li T, Tang ZP, Zhu XR, Han JY, Tian H. Platelet Count is Associated with the Rate of Lymph Node Metastasis in Lung Adenocarcinoma. Cancer Manag Res 2020; 12:9765-9774. [PMID: 33116836 PMCID: PMC7548228 DOI: 10.2147/cmar.s273328] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 09/08/2020] [Indexed: 12/14/2022] Open
Abstract
Purpose Emerging studies have revealed that platelets are involved in tumor metastasis in lung adenocarcinoma (ADC). The solid pathological subtype of lung ADC is associated with metastasis, recurrence, and poor prognosis. However, there is no study exploring the relationship between platelets and different lung pathological subtypes. Patients and Methods The association between platelet counts and lymph node metastasis was analyzed in 852 patients with lung ADC who underwent surgery and lymph node dissection. Multivariate logistic analysis was conducted to identify the risk factors of lymph node metastasis. Then, lymph node metastasis and other factors were analyzed to determine their correlation with platelet count and histological subtype. Results We found that the platelet count was associated with lymph node metastasis (P = 0.01) in multivariable analysis, independent of tumor size, predominant subtype, visceral pleural invasion, and microvessel invasion. In patients with a platelet count ≥300 × 109/L, the rate of lymph node metastasis was 38.5%, almost twice as high as that in patients with a platelet count <300 × 109/L (23.2%). Additionally, elevated platelet counts, even those within the normal range, were significantly associated with a higher rate of lymph node metastasis. The mean platelet count in patients with solid-predominant histology (269.70 ± 69.38 × 109/L) was significantly higher than that in patients with other histologies (P < 0.001). Conclusion Elevated platelet counts are significantly associated with a higher rate of lymph node metastasis, even if the platelet counts are within the reference range. Platelet counts were significantly higher in patients with solid-predominant histology than in patients with other histologies. In addition, VEGF-C may play an important role in lymphatic metastasis in patients with lung ADC. We hypothesize that antiplatelet therapy may reduce lymph node metastasis in lung ADC patients.
Collapse
Affiliation(s)
- Cheng-Hao Qu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China.,Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Tong Li
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China.,Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Zhan-Peng Tang
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China.,Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Xi-Rui Zhu
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China.,Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Jing-Yi Han
- Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China.,Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China
| | - Hui Tian
- Department of Thoracic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, People's Republic of China
| |
Collapse
|
8
|
Moussa AM, Ziv E, Solomon SB, Camacho JC. Microwave Ablation in Primary Lung Malignancies. Semin Intervent Radiol 2019; 36:326-333. [PMID: 31680724 DOI: 10.1055/s-0039-1700567] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide. Eighty-five percent of cases correspond to non-small cell lung cancer (NSCLC) and pivotal nonsurgical options for early-stage disease include percutaneous ablation and stereotactic body radiation therapy (SBRT). Microwave Ablation (MWA) is a locoregional treatment option that has many advantages over radiofrequency ablation and has been able to overcome the limitations of this technique in the treatment of early-stage NSCLC. In this review article, we highlight the current evidence supporting the use of MWA in patients with early-stage NSCLC and discuss the technical considerations of the procedure, including optimal patient selection and planning strategies, as well as the potential complications and reported outcomes. Finally, we mention future trends involving ablation in NSCLC, including its role in combination with SBRT in central tumors, management of post-SBRT local recurrence, and its potential as an adjuvant treatment option for patients with resistance to systemic therapy or in combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- Amgad M Moussa
- Department of Interventional Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Etay Ziv
- Department of Interventional Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Stephen B Solomon
- Department of Interventional Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Juan C Camacho
- Department of Interventional Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
9
|
Qian J, Xu J, Wang S, Qian F, Yang W, Zhang B, Zhang Y, Nie W, Lou Y, Lu J, Zhang X, Zhang W, Chu T, Zhong H, Fang W, Zhao H, Han B. Adjuvant Chemotherapy Candidates in Stage I Lung Adenocarcinomas Following Complete Lobectomy. Ann Surg Oncol 2019; 26:2392-2400. [PMID: 31011907 DOI: 10.1245/s10434-019-07366-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND This study aimed to explore adjuvant chemotherapy (ACT) candidates based on a recurrence risk-scoring model in completely lobectomized stage I patients with lung adenocarcinoma (LAD). METHODS A retrospective study was performed on 4606 patients (non-ACT group: n = 3514; ACT group: n = 1092) who underwent complete lobectomy for LAD at Shanghai Chest Hospital from 2008 to 2014. The nomogram predicting recurrence-free survival (RFS) was developed in the non-ACT group using Cox proportional hazards regression. The nomogram-based risk score was calculated in the entire cohort. Differences of RFS between the non-ACT and ACT groups were compared as stratified by the risk score. The score cut-off points were determined using the X-tile software. RESULTS Six independent predictors, including age, sex, tumor size, pathological subtype, visceral pleural invasion (VPI), and lymphovascular invasion (LVI) were associated with RFS. The nomogram more accurately predicted RFS than the 8th TNM staging {C-index: 0.784 [95% confidence interval (CI) 0.756-0.812] vs. 0.719 (95% CI 0.689-0.749), p = 0.0017}. A significant RFS difference was observed among the low-, intermediate- and high-risk groups (p < 0.0001), as divided by the optimal cut-points of risk score (203 and 244). ACT did not improve RFS for patients at intermediate-risk, or was even detrimental for low-risk patients; however, improved RFS was observed in ACT receivers at high-risk (p = 0.0416). ACT candidates with a risk score ≥ 245 constituted 2.6% of stage I patients. CONCLUSIONS The nomogram provided an individual prediction of RFS for stage I LAD following lobectomy. High-risk patients (score ≥ 245) may benefit from postoperative ACT.
Collapse
Affiliation(s)
- Jie Qian
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jianlin Xu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shuyuan Wang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Fangfei Qian
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wenjia Yang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yanwei Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Nie
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqing Lou
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Lu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xueyan Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Zhang
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Tianqing Chu
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hua Zhong
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wentao Fang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Heng Zhao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Baohui Han
- Department of Pulmonary, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
10
|
Chen T, Luo J, Gu H, Gu Y, Huang J, Luo Q, Yang Y. Should minimally invasive lung adenocarcinoma be transferred from stage IA1 to stage 0 in future updates of the TNM staging system? J Thorac Dis 2018; 10:6247-6253. [PMID: 30622797 DOI: 10.21037/jtd.2018.10.78] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Background The 8th International Association Study of Lung Cancer (IASLC) TNM classification staging project for lung cancer has classified patients with adenocarcinoma in situ (AIS) into stage 0, while patients with a minimally invasive adenocarcinoma (MIA) were classified into stage IA1. However, MIA patients, similar to AIS patients, have an approximately a 100% 5-year survival. This study aimed to investigate if MIA could be transferred from stage IA to stage 0 in the next TNM staging system. Methods We retrospectively reviewed 1,524 consecutive patients with a pathologically confirmed AIS, MIA and an invasive adenocarcinoma (IADC) in stage IA1. Disease-free survival (DFS) and overall survival (OS) were analyzed to evaluate survival difference between these three groups. Results There were 412 AIS, 675 MIA and 437 IADC patients in stage IA1. No statistically significant differences for DFS and OS (P=0.109) were seen between the AIS and MIA groups. Patients of the IADC group had significantly worse DFS (P=0.003) but the OS rate (P=0.941) was insignificant when compared with the MIA patients. Similar survival results were seen when comparisons were made between the IADC and AIS/MIA groups. The IADC group had a worse DFS (P=0.001) rate but no OS (P=0.380) difference with the AIS/MIA groups. Conclusions Patients with AIS and MIA had similar post-surgical survival rates. We propose that MIA may potentially be transferred from IA1 to stage 0 in the future.
Collapse
Affiliation(s)
- Tianxiang Chen
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China.,School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Jizhuang Luo
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Haiyong Gu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yu Gu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jia Huang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qingquan Luo
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yunhai Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
11
|
Xi JJ, Yin JC, Wang L, Lu CL, Wang Q, Jiang W. A surveillance method-oriented detection of post-operative spatial-temporal recurrence for non-small cell lung cancer. J Thorac Dis 2018; 10:6107-6117. [PMID: 30622782 DOI: 10.21037/jtd.2018.10.32] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background This study evaluated spatial-temporal recurrence patterns after curative resection for non-small cell lung cancer (NSCLC) to clarify and recommend appropriate post-operative surveillance. Methods A total of 2,486 consecutive patients between January 2005 and December 2012 with NSCLC (stage I-IIIA) who underwent definitive surgical resection were retrospectively analyzed. We used a hazard rate curve to evaluate event dynamics. Disease-free survival (DFS) was evaluated by the Kaplan-Meier method. Univariate and multivariate analyses with Cox proportional hazards regression identified risk factors that predicted DFS. Results The median follow-up was 50.1 months. Recurrences were diagnosed in 852 (34.3%) patients. Four hundred eighty-nine events first occurred in the chest, 177 in the brain, 117 in the bone, and 71 in the abdomen. Of all recurrences, 78.5% occurred in the first 3 years. Univariate and multivariate analyses identified the age at diagnosis (P<0.001), histology (P=0.023), tumor size (P<0.001), pathologic N stage (P<0.001), and grade (P=0.043) as independent risk factors for intra-thoracic recurrences. Histology (P<0.001), tumor size (P<0.001), surgical method (P=0.021), pathologic N stage (P<0.001), and grade (P=0.005) were independent to predict extra-thoracic recurrences. The hazard rate curve displayed an initial surge of time to any treatment failure during 12 months after surgery. Based on sub-group analysis, both intra- and extra-recurrences increased with stage and brain recurrences in stage IIIA occurred earlier than stage II. Hazard rate curve of brain recurrences in squamous cell carcinoma showed a moderate peak during 9-15 months. Hazard rate curves of brain and bone recurrences in adenocarcinoma displayed clear peaks at 9-27 and 15-30 months, respectively. Conclusions Intra- and extra-thoracic recurrences correlate with different clinicopathological factors. Brain MRI and bone ECT were recommended for selected patients in particular time to early detect extra-thoracic recurrences.
Collapse
Affiliation(s)
- Jun-Jie Xi
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Jia-Cheng Yin
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Lin Wang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Chun-Lai Lu
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| | - Wei Jiang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai 200032, China
| |
Collapse
|
12
|
Luo J, Ma K, Shi Y, Chen Z, Zhao M, Huang Y, Wang S, Xi J, Zhan C, Xu S, Wang Q. Genetic analyses of differences between solid and nonsolid predominant lung adenocarcinomas. Thorac Cancer 2018; 9:1656-1663. [PMID: 30276966 PMCID: PMC6275839 DOI: 10.1111/1759-7714.12876] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Solid predominant lung adenocarcinomas (LUAD) have distinct histopathological and clinical characteristics compared with nonsolid subtypes. A comprehensive comparison of altered genes found in solid and nonsolid subtypes has not previously been performed. In this study, we analyzed differences in gene expression, genetic mutations, and DNA methylation to better understand the risk factors for these two subtypes of LUAD. METHODS Differentially expressed genes (DEGs) and differentially mutated genes (DMGs) were analyzed from RNA-seq data downloaded from The Cancer Genome Atlas (TCGA) and Broad Institute database. To understand the functional significance of molecular changes, we examined the DEGs and DMGs with Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway analysis. RESULTS A total of 184 patients in the TCGA cohort and 140 patients in the Broad Institute cohort were included in this study. We identified 75 DEGs, of which 15 were upregulated and 56 downregulated in the solid group relative to the nonsolid group. The DEGs were mainly involved in the regulation of water and fluid transport. We discovered 38 significantly differentially expressed genes that overlapped in the two groups. The DMGs were mainly enriched for pathways involved in cell-cell adhesion, cell adhesion, biological adhesion, and hemophilic cell adhesion. We additionally discovered nine significantly methylated genes between solid and nonsolid LUAD. CONCLUSIONS Our study identified distinct DEGs, DMGs, and methylation genes for solid and nonsolid LUAD subtypes. These findings improve our understanding of the different carcinogenesis mechanisms in LUAD and will help to develop new therapeutic strategies.
Collapse
Affiliation(s)
- Jizhuang Luo
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ke Ma
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yu Shi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zongwei Chen
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengnan Zhao
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiwei Huang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shuai Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Junjie Xi
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Zhan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Songtao Xu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qun Wang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Abstract
PURPOSE OF REVIEW After 'curative' resection, many patients are still at risk for further lung cancer, either as a recurrence or a new metachronous primary. In theory, close follow-up should improve survival by catching relapse early - but in reality, many experts feel that surveillance for recurrence is of uncertain value. In this article, we explore the reasons behind the controversy, what the current guidelines recommend, and what future solutions are in development that may ultimately resolve this debate. RECENT FINDINGS Although postoperative surveillance for a new lung cancer may impart a survival advantage, this benefit does not appear to extend to the phenomenon of recurrence. Nevertheless, close radiographic follow-up after curative resection is still recommended by most professional societies, with more frequent scanning in the first 2 years, and then annual screening thereafter. Given the radiation risk, however, low-dose and minimal-dose computed tomography options are under investigation, as well as timing scans around expected peaks of recurrence rather than a set schedule. SUMMARY Applying the same surveillance algorithm to all lung cancer patients after curative resection may not be cost-effective or reasonable, especially if there is no demonstrable mortality benefit. Therefore, future research should focus on finding safer nonradiographic screening options, such as blood or breath biomarkers, or developing nomograms for predicting which patients will relapse and require closer follow-up. Ultimately, however, better tools for surveillance may be moot until we develop better treatment options for lung cancer recurrence.
Collapse
|
14
|
Chen T, Luo J, Gu H, Gu Y, Huang Q, Wang Y, Zheng J, Yang Y, Chen H. Impact of Solid Minor Histologic Subtype in Postsurgical Prognosis of Stage I Lung Adenocarcinoma. Ann Thorac Surg 2017; 105:302-308. [PMID: 29162222 DOI: 10.1016/j.athoracsur.2017.08.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 07/26/2017] [Accepted: 08/07/2017] [Indexed: 12/23/2022]
Abstract
BACKGROUND Few studies focus on the outcome and effect of a postsurgical treatment strategy for early stage patients with minor solid components pattern. This study investigated the prognosis and the adjuvant chemotherapy benefit among stage I invasive lung adenocarcinoma patients with minor solid components pattern according to the eighth TNM staging classification. METHODS A total of 3,308 lung adenocarcinoma patients with mixed histologic components was divided into three groups: solid predominant, solid minor, and solid absent pattern. Disease-free survival and overall survival were analyzed to evaluate survival difference among patients in the different groups using the Kaplan-Meier approach and multivariable Cox models. RESULTS Both solid predominant and solid minor groups showed significantly worse disease-free survival (p < 0.001) and overall survival (p < 0.001) compared with the solid absent group. There were no significant disease-free survival (hazard ratio [HR] 1.41, 95% confidence interval [CI]: 0.87 to 2.30, p = 0.161) or overall survival (HR 1.60, 95% CI: 0.83 to 3.09, p = 0.159) difference between the former two groups. For patients in stage IB, adjuvant chemotherapy improves disease-free survival (HR 0.33, 95% CI: 0.11 to 1.02, p = 0.044) but not overall survival (HR 0.61, 95% CI: 0.21 to 1.77, p = 0.360) in the solid predominant group. No adjuvant chemotherapy benefits for disease-free survival (HR 1.04, 95% CI: 0.49 to 2.22; p = 0.922) and overall survival (HR 0.49, 95% CI: 0.13 to 1.90; p = 0.291) were seen for the solid minor group. CONCLUSIONS Solid minor components predict a significantly worse prognosis compared with the solid absent pattern. However, adjuvant chemotherapy may be unhelpful to improve outcomes for stage IB patients with solid minor components after surgery.
Collapse
Affiliation(s)
- Tianxiang Chen
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jizhuang Luo
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Haiyong Gu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Gu
- Department of Radiation Oncology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Qingyuan Huang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yiyang Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiajie Zheng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yunhai Yang
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Haiquan Chen
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China; Department of Thoracic Surgery, Shanghai Cancer Center, Fudan University, Shanghai, China.
| |
Collapse
|
15
|
Gao Q, Li P, Jiang X, Zhan Z, Yan Q, Zhang B, Huang C. Worse disease-free, tumor-specific, and overall survival in surgically-resected lung adenocarcinoma patients with ALK rearrangement. Oncotarget 2017; 8:86066-86081. [PMID: 29156778 PMCID: PMC5689668 DOI: 10.18632/oncotarget.20973] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 08/26/2017] [Indexed: 12/12/2022] Open
Abstract
Introduction This study determined the prevalence of anaplastic lymphoma kinase (ALK) rearrangement, and identified the associations of ALK rearrangement with clinicopathologic characteristics and treatment outcomes in patients with surgically-resected stage I-III lung adenocarcinoma. Methods A total of 534 surgically-resected lung adenocarcinoma patients were studied. The prevalence of ALK protein over-expression was determined by a fully-automated immunochemistry assay (with mouse monoclonal Ventana D5F3 antibody), and the associations of ALK rearrangement with clinicopathologic characteristics and treatment outcomes were analyzed. Results Forty-two (7.9%) of the 534 lung adenocarcinoma patients were ALK IHC-positive. ALK rearrangement was significantly associated with younger age (P = 0.011), high T-stage (P = 0.025), high pathologic stage (P = 0.002), solid predominant adenocarcinoma with mucin production (P = 0.006), invasive mucinous adenocarcinoma (P = 0.009), and receipt of adjuvant therapy after surgery (P = 0.036), but no significant associations were found between the ALK rearrangement and sex or smoking status. ALK IHC-positivity was significantly associated with a shorter disease-free survival, tumor-specific survival, and overall survival (P = 0.001, 0.026, and 0.007, respectively). Multivariate analysis showed that ALK IHC-positivity was an adverse prognostic factor for disease-free survival (HR, 1.80; 95% CI 1.18-2.77; P = 0.007), tumor-specific survival (HR, 2.59; 95% CI 1.35-4.97; P = 0.004), and overall survival (HR, 1.92; 95% CI 1.07-3.44; P = 0.030). Conclusion The clinical characteristics of patients with ALK-positive lung adenocarcinoma were similar to those of EGFR-mutated patients. ALK rearrangement was an adverse prognostic factor in surgically-resected lung adenocarcinoma patients.
Collapse
Affiliation(s)
- Qiongqiong Gao
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Cancer Institute & Hospital, Tianjin 300060, P.R. China
| | - Pupu Li
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Cancer Institute & Hospital, Tianjin 300060, P.R. China
| | - Xiangli Jiang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Cancer Institute & Hospital, Tianjin 300060, P.R. China
| | - Zhongli Zhan
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Department of Pathology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin 300060, P.R. China
| | - Qingna Yan
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Department of Pathology, Tianjin Lung Cancer Center, Tianjin Cancer Institute & Hospital, Tianjin 300060, P.R. China
| | - Bo Zhang
- Department of Ultrasound Diagnosis, Second Hospital of Tianjin Medical University, Tianjin 300060, P.R. China
| | - Chun Huang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, P.R. China.,Tianjin's Clinical Research Center for Cancer, Tianjin 300060, P.R. China.,Department of Thoracic Oncology, Tianjin Cancer Institute & Hospital, Tianjin 300060, P.R. China
| |
Collapse
|
16
|
Chen T, Luo J, Wang R, Gu H, Gu Y, Huang Q, Wang Y, Zheng J, Gu C, Pan X, Yang J, Yang Y, Zhao H. Visceral pleural invasion predict a poor survival among lung adenocarcinoma patients with tumor size ≤ 3cm. Oncotarget 2017; 8:66576-66583. [PMID: 29029538 PMCID: PMC5630438 DOI: 10.18632/oncotarget.16476] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/14/2017] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION The impact of visceral pleural invasion (VPI) on survival remains controversial for patients with early stage non-small cell lung cancer (NSCLC). This study investigated the survival status of VPI among patients with lymph node-negative lung invasive adenocarcinoma smaller than 3cm. METHODS We retrospectively reviewed 2537 consecutive patients with pathologic stage I lung invasive adenocarcinoma. All patients had received lobectomy and system lymph nodes resection. Patients were classified into 4 groups according to tumor size and visceral pleural invasion status. Disease-free survival (DFS) and overall survival (OS) were analyzed to evaluate survival difference between these groups. RESULTS 548 patients with VPI while 1989 patients without VPI were included in this study. For patients with tumor size ≤2cm, patients with VPI had significant worse DFS (HR,4.85; 95% CI, 2.98-7.91; p = .000) and OS(HR,3.52; 95% CI, 1.59-7.78; p = .002) compared with non-VPI group. For patients with tumor size between 2-3cm, patients with VPI had significant worse DFS (HR, 1.72; 95% CI, 1.16-2.55; p = .006) but no significant OS (HR, 1.31; 95% CI, 0.76-2.24; p = .330) compared with non-VPI group. For patients with VPI, there were no survival difference between tumor size 2-3cm group and ≤2cm group for both DFS(HR,1.02; 95% CI, 0.65-1.61; p = .939) and OS(HR,1.45; 95% CI, 0.71-2.97; p = .315). CONCLUSIONS VPI could predict a poor survival even for node-negative invasive lung adenocarcinoma patients with tumor size less than 3cm.
Collapse
Affiliation(s)
- Tianxiang Chen
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jizhuang Luo
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Rui Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Haiyong Gu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yu Gu
- Department of Radiation Oncology, Shanghai Cancer Hospital, Fudan University, Shanghai, China
| | - Qingyuan Huang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yiyang Wang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jiajie Zheng
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Chang Gu
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xufeng Pan
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jun Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yunhai Yang
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Heng Zhao
- Department of Thoracic Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
17
|
Mao R, Chen C, Fei K. The Clinical Implications and Thoughts on Different Patterns in Resected Lung Adenocarcinoma. J Thorac Oncol 2017; 12:e24-e25. [PMID: 28215725 DOI: 10.1016/j.jtho.2016.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Accepted: 12/04/2016] [Indexed: 01/15/2023]
Affiliation(s)
- Rui Mao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| | - Ke Fei
- Department of Thoracic Surgery, East China Sanatorium, Wuxi, People's Republic of China
| |
Collapse
|