1
|
Shiiya H, Ujiie H, Chiba R, Nomura S, Ohtaka K, Fujiwara-Kuroda A, Aragaki M, Takahashi K, Okada K, Kato T. Impact of pulmonary vein-first ligation during lobectomy on the postoperative survival and recurrence rates in patients with non-small cell lung cancer: a multicenter propensity score-matched study. Surg Today 2024; 54:1369-1378. [PMID: 38814330 DOI: 10.1007/s00595-024-02852-8] [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: 12/18/2023] [Accepted: 04/02/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE Surgical manipulation of the lungs increases the number of circulating tumor cells and the subsequent risk of metastasis in patients with lung cancer. This study investigated whether or not ligating the tumor-draining pulmonary vein first during lobectomy could improve the prognosis of these patients. METHODS We retrospectively evaluated patients who underwent curative lobectomy for solitary nonsmall-cell lung carcinoma between January 2012 and December 2016. We divided the patients into the vein-first group, in which all associated pulmonary veins were dissected and severed before cutting the pulmonary artery, bronchus, or pulmonary fissure, and the other procedure group. RESULTS Overall, we included 177 and 413 patients in the vein-first and other procedure groups, respectively. Propensity score matching yielded 67 pairs of patients. The 5-year overall survival (85.6% [95% confidence interval, 77.3-94.8%] vs. 69.4% [58.7-81.9%], P = 0.03%) and recurrence-free survival (73.4% [63.3-85.1%] vs. 53.5% [42.5-67.3%], P = 0.02) were significantly better in the vein-first group than in the other procedure group. The cumulative recurrence rate at 5 years post-surgery was significantly lower in the vein-first group than in the other procedure group (21.7% vs. 38.3%, P = 0.04). CONCLUSION Our study suggests that ligating the pulmonary vein first during lobectomy for lung cancer can improve the overall survival, recurrence-free survival, and cumulative recurrence rate.
Collapse
Affiliation(s)
- Haruhiko Shiiya
- Department of Thoracic Surgery, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Hideki Ujiie
- Department of Thoracic Surgery, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Ryohei Chiba
- Department of Thoracic Surgery, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Shunsuke Nomura
- Department of Thoracic Surgery, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Kazuto Ohtaka
- Department of Thoracic Surgery, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Aki Fujiwara-Kuroda
- Department of Thoracic Surgery, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Masato Aragaki
- Department of Thoracic Surgery, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Keita Takahashi
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Japan
| | - Kazufumi Okada
- Data Science Center, Promotion Unit, Institute of Health Science Innovation for Medical Care, Hokkaido University Hospital, Sapporo, Japan
| | - Tatsuya Kato
- Department of Thoracic Surgery, Hokkaido University Graduate School of Medicine, Kita 15 Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| |
Collapse
|
2
|
Zhang B, Hou Q, Zhang X, Ma Y, Yuan J, Li S, Zhao X, Sun L, Wang H, Zheng H. Anesthetic propofol inhibits ferroptosis and aggravates distant cancer metastasis via Nrf2 upregulation. Free Radic Biol Med 2023; 195:298-308. [PMID: 36586453 DOI: 10.1016/j.freeradbiomed.2022.12.092] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/21/2022] [Accepted: 12/26/2022] [Indexed: 12/29/2022]
Abstract
The impact of anesthetic management on the prognosis of patients with cancer undergoing surgery is controversial. Circulating tumor cells (CTCs) play critical roles during cancer metastasis and can be released in large quantities during surgery. The ferroptosis of CTCs is related to metastasis. Whether anesthetics affect distant metastasis by increasing the survival of CTCs is unknown. To test this hypothesis, mice were inoculated with cancer cells via tail vein injection before treatment with propofol or sevoflurane for 2 h. After 2 weeks, more metastases were observed in the propofol group compared with the sevoflurane and vehicle groups. Then, we used the ferroptosis inhibitor ferrostatin-1 to explore the effect of ferroptosis on metastasis. Similar to propofol, pretreatment with ferrostatin-1 significantly increased CTC survival in mouse lungs at 24 h and the tumor burden at 10 weeks post-inoculation. Moreover, propofol protected cancer cells from RSL3-induced ferroptosis in vitro, as evidenced by decreases in intracellular levels of reactive oxygen species (ROS), lipid peroxide, and ferroptosis markers. Further studies showed that propofol treatment upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target genes, including HO-1, NQO1, and SLC7A11. Finally, the targeted knockdown of Nrf2 abolished the anti-ferroptosis effect of propofol. Collectively, we demonstrated the risk of a specific type of anesthetic, propofol, in promoting cancer cell metastasis through Nrf2-mediated ferroptosis inhibition. These findings may guide the choice of anesthetic for surgical removal of tumors.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Qi Hou
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaoli Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yiming Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Junhu Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuai Li
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xinhua Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Li Sun
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China.
| | - Hongying Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Hui Zheng
- Department of Anesthesiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
3
|
Wankhede D, Grover S, Hofman P. Circulating Tumor Cells as a Predictive Biomarker in Resectable Lung Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2022; 14:cancers14246112. [PMID: 36551601 PMCID: PMC9776809 DOI: 10.3390/cancers14246112] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Background: In breast, prostate, and other epithelial tumors, circulating tumor cells (CTC) in peripheral blood may predict survival. Our study evaluated the prognostic significance of baseline and postoperative CTC in patients with early non-small cell lung cancer (NSCLC) through a meta-analytic approach. Methods: Prospective studies comparing survival outcomes between positive (CTC+) and negative CTC (CTC−) patients were systematically searched. Primary outcomes were overall (OS) and disease-free survival (DFS) with hazard ratio (HR) and 95% confidence interval (CI) as the effect measure. Pooled HR determined the prognostic role under a fixed-effect or random-effect model depending on heterogeneity. Results: Eighteen studies with 1321 patients were eligible. CTC+ patients were associated with an increased risk of death (HR 3.53, 95% CI 2.51−4.95; p < 0.00001) and relapse (HR 2.97, 95% CI 2.08−4.22; p < 0.00001). Subgroup analysis results were consistent in different subsets, including time points (baseline and postoperative) and sources (peripheral and pulmonary vein) of blood collection, detection methods (label-free, label-dependent, and RT-PCR), and follow-up duration. Conclusion: Our meta-analysis revealed that CTC is a promising predictive biomarker for stratifying survival outcomes in patients with early-stage NSCLC. However, future studies are required to validate these findings and standardize detection methods.
Collapse
Affiliation(s)
- Durgesh Wankhede
- Department of Surgical Oncology, All India Institute of Medical Sciences, New Delhi 110029, India
- Correspondence:
| | - Sandeep Grover
- Center for Human Genetics, Universitatsklinikum Giessen und Marburg—Standort Marburg, 35055 Marburg, Germany
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, CHU Nice, FHU OncoAge, University Côte d’Azur, 06100 Nice, France
- Team 4, IRCAN, UMR 7284/U10181, FHU OncoAge, University Côte d’Azur, 06107 Nice, France
- Hospital-Integrated Biobank (BB-0033-00025), CHU Nice, FHU OncoAge, University Côte d’Azur, 06100 Nice, France
- European Liquid Biopsy Society, Martinistrasse 52 Building N27 Room 4.003, 20246 Hamburg, Germany
| |
Collapse
|
4
|
Duan X, Yang Z, Hao X, Zhou S, Liu Z, Zhang K, Cui Y. Early ligation of the pulmonary vein can reduce the dissemination of shed tumor cells during thoracoscopic lobectomy. J Thorac Cardiovasc Surg 2022; 164:1623-1635.e2. [DOI: 10.1016/j.jtcvs.2022.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 03/11/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
5
|
Circulating Tumor Cells and the Non-Touch Isolation Technique in Surgery for Non-Small-Cell Lung Cancer. Cancers (Basel) 2022; 14:cancers14061448. [PMID: 35326603 PMCID: PMC8946695 DOI: 10.3390/cancers14061448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/18/2022] [Accepted: 03/09/2022] [Indexed: 11/16/2022] Open
Abstract
Circulating tumor cells (CTCs) are dislodged from the primary tumor into the bloodstream, travel within the bloodstream to distant organs, and finally extravasate and proliferate as epithelial metastatic deposits. The relationship between the existence of CTCs and tumor prognosis has been demonstrated by many researchers. In surgery for malignancies, the surgical manipulation of tumors and tissues around the tumor may lead to the release of CTCs into the bloodstream. The non-touch isolation technique (NTIT) has been advocated to prevent the release of CTCs during surgery. The concept of NTIT is the prevention of intraoperative increment of CTCs from the primary tumor by the early blockade of outflow vessels, and ‘pulmonary vein (PV)-first lobectomy’ during surgery for non-small-cell lung cancer (NSCLC) corresponds to this technique. The concept of PV-first lobectomy is well known among thoracic surgeons, but evidence of its efficacy for preventing the increase of intra- and postoperative CTCs and for improving postoperative prognosis is still uncertain. Our study summarizes evidence regarding the relationship between NTIT and CTCs in NSCLC and suggests the need for further research on CTCs and CTC-detecting modalities.
Collapse
|
6
|
Qiu X, Yu H, Wu H, Hu Z, Zhou J, Lin H, Xue W, Cai W, Chen J, Yan Q, Dai W, Yang M, Tang D, Dai Y. Single-cell chromatin accessibility landscape of human umbilical cord blood in trisomy 18 syndrome. Hum Genomics 2021; 15:40. [PMID: 34193281 PMCID: PMC8246660 DOI: 10.1186/s40246-021-00338-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 05/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Trisomy 18 syndrome (Edwards syndrome, ES) is a type of aneuploidy caused by the presence of an extra chromosome 18. Aneuploidy is the leading cause of early pregnancy loss, intellectual disability, and multiple congenital anomalies. The research of trisomy 18 is progressing slowly, and the molecular characteristics of the disease mechanism and phenotype are still largely unclear. RESULTS In this study, we used the commercial Chromium platform (10× Genomics) to perform sc-ATAC-seq to measure chromatin accessibility in 11,611 single umbilical cord blood cells derived from one trisomy 18 syndrome patient and one healthy donor. We obtained 13 distinct major clusters of cells and identified them as 6 human umbilical cord blood mononuclear cell types using analysis tool. Compared with the NC group, the ES group had a lower ratio of T cells to NK cells, the ratio of monocytes/DC cell population did not change significantly, and the ratio of B cell nuclear progenitor and megakaryocyte erythroid cells was higher. The differential genes of ME-0 are enriched in Human T cell leukemia virus 1 infection pathway, and the differential peak genes of ME-1 are enriched in apopotosis pathway. We found that CCNB2 and MCM3 may be vital to the development of trisomy 18. CCNB2 and MCM3, which have been reported to be essential components of the cell cycle and chromatin. CONCLUSIONS We have identified 6 cell populations in cord blood. Disorder in megakaryocyte erythroid cells implicates trisomy 18 in perturbing fetal hematopoiesis. We identified a pathway in which the master differential regulatory pathway in the ME-0 cell population involves human T cell leukemia virus 1 infection, a pathway that is dysregulated in patients with trisomy 18 and which may increase the risk of leukemia in patients with trisomy 18. CCNB2 and MCM3 in progenitor may be vital to the development of trisomy 18. CCNB2 and MCM3, which have been reported to be essential components of the cell cycle and chromatin, may be related to chromosomal abnormalities in trisomy 18.
Collapse
Affiliation(s)
- Xiaofen Qiu
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China.,Guangxi Key Laboratory of Metabolic Diseases Research, Department of Clinical Laboratory of Guilin, No. 924 Hospital, 541002, Guilin, Guangxi, People's Republic of China.,College of Life Science, Guangxi Normal University, Guilin, Guangxi, 541004, People's Republic of China
| | - Haiyan Yu
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China
| | - Hongwei Wu
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China
| | - Zhiyang Hu
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China
| | - Jun Zhou
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China
| | - Hua Lin
- Guangxi Key Laboratory of Metabolic Diseases Research, Department of Clinical Laboratory of Guilin, No. 924 Hospital, 541002, Guilin, Guangxi, People's Republic of China
| | - Wen Xue
- Guangxi Key Laboratory of Metabolic Diseases Research, Department of Clinical Laboratory of Guilin, No. 924 Hospital, 541002, Guilin, Guangxi, People's Republic of China
| | - Wanxia Cai
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China
| | - Jiejing Chen
- Guangxi Key Laboratory of Metabolic Diseases Research, Department of Clinical Laboratory of Guilin, No. 924 Hospital, 541002, Guilin, Guangxi, People's Republic of China
| | - Qiang Yan
- Guangxi Key Laboratory of Metabolic Diseases Research, Department of Clinical Laboratory of Guilin, No. 924 Hospital, 541002, Guilin, Guangxi, People's Republic of China
| | - Weier Dai
- College of Natural Science, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ming Yang
- Guangxi Key Laboratory of Metabolic Diseases Research, Department of Clinical Laboratory of Guilin, No. 924 Hospital, 541002, Guilin, Guangxi, People's Republic of China
| | - Donge Tang
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China.
| | - Yong Dai
- Department of Clinical Medical Research Center, Guangdong Provincial Engineering Research Center of Autoimmune Disease Precision Medicine, The First Affiliated Hospital of Southern University of Science and Technology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, Guangdong, 518020, People's Republic of China. .,Guangxi Key Laboratory of Metabolic Diseases Research, Department of Clinical Laboratory of Guilin, No. 924 Hospital, 541002, Guilin, Guangxi, People's Republic of China.
| |
Collapse
|
7
|
Rushton AJ, Nteliopoulos G, Shaw JA, Coombes RC. A Review of Circulating Tumour Cell Enrichment Technologies. Cancers (Basel) 2021; 13:cancers13050970. [PMID: 33652649 PMCID: PMC7956528 DOI: 10.3390/cancers13050970] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Circulating tumour cells (CTCs) are cancer cells shed into the bloodstream from tumours and their analysis can provide important insights into cancer detection and monitoring, with the potential to direct personalised therapies for the patient. These CTCs are rare in the blood, which makes their detection and enrichment challenging and to date, only one technology (the CellSearch) has gained FDA approval for determining the prognosis of patients with advanced breast, prostate and colorectal cancers. Here, we review the wide range of enrichment technologies available to isolate CTCs from other blood components and highlight the important characteristics that new technologies should possess for routine clinical use. Abstract Circulating tumour cells (CTCs) are the precursor cells for the formation of metastatic disease. With a simple blood draw, liquid biopsies enable the non-invasive sampling of CTCs from the blood, which have the potential to provide important insights into cancer detection and monitoring. Since gaining FDA approval in 2004, the CellSearch system has been used to determine the prognosis of patients with metastatic breast, prostate and colorectal cancers. This utilises the cell surface marker Epithelial Cell Adhesion Molecule (EpCAM), to enrich CTCs, and many other technologies have adopted this approach. More recently, the role of mesenchymal-like CTCs in metastasis formation has come to light. It has been suggested that these cells are more aggressive metastatic precursors than their epithelial counterparts; however, mesenchymal CTCs remain undetected by EpCAM-based enrichment methods. This has prompted the development of a variety of ‘label free’ enrichment technologies, which exploit the unique physical properties of CTCs (such as size and deformability) compared to other blood components. Here, we review a wide range of both immunocapture and label free CTC enrichment technologies, summarising the most significant advantages and disadvantages of each. We also highlight the important characteristics that technologies should possess for routine clinical use, since future developments could have important clinical implications, with the potential to direct personalised therapies for patients with cancer.
Collapse
Affiliation(s)
- Amelia J. Rushton
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
- Correspondence:
| | - Georgios Nteliopoulos
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
| | - Jacqueline A. Shaw
- Leicester Cancer Research Centre, University of Leicester, Leicester LE2 7LX, UK;
| | - R. Charles Coombes
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London W12 0NN, UK; (G.N.); (R.C.C.)
| |
Collapse
|
8
|
Huang CS, Chien HC, Chen CK, Yeh YC, Hsu PK, Chen HS, Hsieh CC, Hsu HS, Huang BS, Shih CC. Significance of preoperative biopsy in radiological solid-dominant clinical stage I non-small-cell lung cancer. Interact Cardiovasc Thorac Surg 2020; 32:537-545. [PMID: 33332546 DOI: 10.1093/icvts/ivaa297] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/21/2020] [Accepted: 10/27/2020] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES The present study aimed to clarify the association between preoperative biopsy and surgical outcomes in clinical stage I non-small-cell lung cancer (NSCLC) with different proportions of ground-glass opacity (GGO). METHODS Data on patients who underwent pulmonary resection for NSCLC from 2006 to 2016 were drawn from a prospective registered database and analysed retrospectively. Patient characteristics collected included tumour size, location and staging, surgical approach, consolidation-tumour ratio, histopathology and the presence or absence of preoperative biopsy to identify the independent prognostic factors of disease-free survival (DFS) and cancer-specific survival. A 1:1 propensity score matching was conducted between the preoperative biopsy and reference groups based on their baseline characteristics measured before the decision for preoperative biopsy. RESULTS A total of 1427 patients were collected to achieve an overall 5-year DFS as 84.5% (median follow-up: 67.3 months), stratified to be 99.5% in the GGO-dominant group (n = 430) and 78.2% in the solid-dominant group (n = 997). Only 2 patients (0.5%) in the GGO-dominant group experienced tumour recurrence. For solid-dominant tumours matched with propensity scores (279 in preoperative biopsy vs 279 in reference group), the independent predictors of DFS included preoperative biopsy, sublobar resection, pathological staging and angiolymphatic invasion. Preoperative biopsy was a predictor of cancer-specific survival in univariable analysis but was not in multivariable analysis. Significant differences were also found between matched groups in those with late-delay surgery, but not in patients receiving preoperative biopsy with early-delay surgery (≤21 days). CONCLUSION Preoperative biopsy may worsen surgical outcomes in patients with clinical stage I, solid-dominant NSCLC.
Collapse
Affiliation(s)
- Chien-Sheng Huang
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Hung-Che Chien
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Ku Chen
- Department of Radiology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Chen Yeh
- Department of Pathology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Po-Kuei Hsu
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hui-Shan Chen
- Department of Health Care Administration, Chang Jung Christian University, Tainan City, Taiwan
| | - Chih-Cheng Hsieh
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Han-Shui Hsu
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Biing-Shiun Huang
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chun-Che Shih
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei, Taiwan.,Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
9
|
Ito K, Shimada J, Shimomura M, Terauchi K, Nishimura M, Yanada M, Iwasaki Y, Ueshima Y, Kato D, Suzuki H, Inoue M. Safety and reliability of computed tomography-guided lipiodol marking for undetectable pulmonary lesions. Interact Cardiovasc Thorac Surg 2020; 30:546-551. [PMID: 31899511 DOI: 10.1093/icvts/ivz304] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/20/2019] [Accepted: 11/24/2019] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES This study aimed to evaluate the safety and reliability of percutaneous computed tomography (CT)-guided lipiodol marking for undetectable pulmonary lesions before video-assisted thoracic surgery (VATS). METHODS We retrospectively analysed the cases of CT-guided lipiodol marking followed by VATS in 9 institutes from May 2006 to March 2018. Lipiodol (0.2-0.5 ml) was percutaneously injected closely adjacent to undetectable pulmonary lesions with computed-tomography guidance. Lipiodol spots were identified using C-arm-shaped fluoroscopy during VATS. We grasped the lipiodol spots, including the target lesions, with ring-shaped forceps and resected them. RESULTS Of 1182 lesions, 1181 (99.9%) were successfully marked. In 1 case, the injected lipiodol diffused, and no spot was created. Of the 1181 lesions, 1179 (99.8%) were successfully resected with intraoperative fluoroscopy. Two lipiodol spots were not detected because of the lipiodol distribution during the division of pleural adhesions. The mean lesion size was 9.1 mm (range 1-48 mm). The mean distance from the pleural surface was 10.2 mm (range 0-43 mm). Lipiodol marking-induced pneumothorax occurred in 495 (57.1%) of 867 cases. Of these, chest drainage was required in 59 patients (6.8%). The other complications were 19 (2.2%) cases of bloody sputum, 3 (0.35%) cases of intravascular air, 1 (0.12%) case of pneumonia and 1 (0.12%) case of cerebral infarction. There were no lipiodol marking-induced deaths or sequelae. CONCLUSIONS Preoperative CT-guided lipiodol marking followed by VATS resection was shown to be a safe and reliable procedure with a high success rate and acceptably low severe complication rate.
Collapse
Affiliation(s)
- Kazuhiro Ito
- Division of Thoracic Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Thoracic Surgery, Kyoto Yamashiro Medical Center, Kizugawa, Japan
| | - Junichi Shimada
- Division of Thoracic Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masanori Shimomura
- Division of Thoracic Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan.,Department of Thoracic Surgery, Ayabe City Hospital, Ayabe, Japan
| | | | | | - Masashi Yanada
- Department of Thoracic Surgery, Japanese Red Cross Kyoto Daini Hospital, Kyoto, Japan
| | - Yasushi Iwasaki
- Department of Thoracic Surgery, Kyoto Chubu Medical Center, Nantan, Japan
| | - Yasuo Ueshima
- Department of Thoracic Surgery, Japanese Red Cross Kyoto Daiichi Hospital, Kyoto, Japan
| | - Daishiro Kato
- Division of Thoracic Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hirofumi Suzuki
- Department of Thoracic Surgery, Osaka General Hospital of West Japan Railway Company, Osaka, Japan
| | - Masayoshi Inoue
- Division of Thoracic Surgery, Department of Surgery, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
10
|
Sawabata N, Nakamura T, Kawaguchi T, Watanabe T, Ouji NS, Ito T, Taniguchi S. Circulating tumor cells detected only after surgery for non-small cell lung cancer: is it a predictor of recurrence? J Thorac Dis 2020; 12:4623-4632. [PMID: 33145035 PMCID: PMC7578482 DOI: 10.21037/jtd-20-1636] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background Surgical manipulation of a tumor can lead to shedding of tumor cells that can enter the circulation and lead to metastasis. The present study evaluated the clinical relevance of circulating tumor cells (CTCs) that were identified immediately after non-small cell lung cancer resection in patients without preoperative CTCs, and whether postoperative CTC detection was associated with recurrence. Methods Immediate preoperative testing for CTCs was performed for 147 patients with pulmonary nodules. This study included 81 lung cancer patients (55.1%) with negative preoperative results for CTCs and who completed postoperative testing for CTCs. The clinical relevance of postoperative CTC detection was evaluated based on the clinicopathological characteristics and recurrence patterns. Results Among the eligible patients, the postoperative CTC results were none detected in 58 patients (71.6%, “Group N”), only a single CTC detected in 6 patients (7.4%, “Group S”), and CTC clusters detected in 17 patients (21.0%, “Group C”). The presence of postoperative CTCs was associated with tumor vessel invasion, lymph duct invasion, and pleural invasion. Distant metastasis was very common in cases with postoperatively detected CTC clusters. The 2-year recurrence-free survival rates were 94.6% for Group N, 62.5% for Group S, and 52.9% for Group C (P<0.01). Multivariate analysis revealed that recurrence was independently related to the postoperative detection of single CTCs and CTC clusters. Conclusions In cases without preoperative CTCs, we postoperatively detected CTCs and the postoperative CTC results were an independent predictor of recurrence.
Collapse
Affiliation(s)
- Noriyoshi Sawabata
- Respiratory Disease Center, Hoshigaoka Medical Center, Hirakata City, Osaka, Japan.,Department of Thoracic and Cardio-Vascular Surgery, Nara Medical University, Shijo-cho Kashihara City, Nara, Japan
| | - Toshitaka Nakamura
- Respiratory Disease Center, Hoshigaoka Medical Center, Hirakata City, Osaka, Japan
| | - Takeshi Kawaguchi
- Department of Thoracic and Cardio-Vascular Surgery, Nara Medical University, Shijo-cho Kashihara City, Nara, Japan
| | - Takashi Watanabe
- Department of Thoracic and Cardio-Vascular Surgery, Nara Medical University, Shijo-cho Kashihara City, Nara, Japan
| | - Noriko Sageshima Ouji
- Department of Immunology, Nara Medical University, Shijo-cho Kashihara City, Nara, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Shijo-cho Kashihara City, Nara, Japan
| | - Shigeki Taniguchi
- Department of Thoracic and Cardio-Vascular Surgery, Nara Medical University, Shijo-cho Kashihara City, Nara, Japan
| |
Collapse
|
11
|
Isozaki A, Mikami H, Hiramatsu K, Sakuma S, Kasai Y, Iino T, Yamano T, Yasumoto A, Oguchi Y, Suzuki N, Shirasaki Y, Endo T, Ito T, Hiraki K, Yamada M, Matsusaka S, Hayakawa T, Fukuzawa H, Yatomi Y, Arai F, Di Carlo D, Nakagawa A, Hoshino Y, Hosokawa Y, Uemura S, Sugimura T, Ozeki Y, Nitta N, Goda K. A practical guide to intelligent image-activated cell sorting. Nat Protoc 2019; 14:2370-2415. [PMID: 31278398 DOI: 10.1038/s41596-019-0183-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Accepted: 04/18/2019] [Indexed: 02/08/2023]
Abstract
Intelligent image-activated cell sorting (iIACS) is a machine-intelligence technology that performs real-time intelligent image-based sorting of single cells with high throughput. iIACS extends beyond the capabilities of fluorescence-activated cell sorting (FACS) from fluorescence intensity profiles of cells to multidimensional images, thereby enabling high-content sorting of cells or cell clusters with unique spatial chemical and morphological traits. Therefore, iIACS serves as an integral part of holistic single-cell analysis by enabling direct links between population-level analysis (flow cytometry), cell-level analysis (microscopy), and gene-level analysis (sequencing). Specifically, iIACS is based on a seamless integration of high-throughput cell microscopy (e.g., multicolor fluorescence imaging, bright-field imaging), cell focusing, cell sorting, and deep learning on a hybrid software-hardware data management infrastructure, enabling real-time automated operation for data acquisition, data processing, intelligent decision making, and actuation. Here, we provide a practical guide to iIACS that describes how to design, build, characterize, and use an iIACS machine. The guide includes the consideration of several important design parameters, such as throughput, sensitivity, dynamic range, image quality, sort purity, and sort yield; the development and integration of optical, microfluidic, electrical, computational, and mechanical components; and the characterization and practical usage of the integrated system. Assuming that all components are readily available, a team of several researchers experienced in optics, electronics, digital signal processing, microfluidics, mechatronics, and flow cytometry can complete this protocol in ~3 months.
Collapse
Affiliation(s)
- Akihiro Isozaki
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Hideharu Mikami
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | | | - Shinya Sakuma
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya, Japan
| | - Yusuke Kasai
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya, Japan
| | - Takanori Iino
- Department of Electrical Engineering and Information Systems, The University of Tokyo, Tokyo, Japan
| | - Takashi Yamano
- Laboratory of Applied Molecular Microbiology, Kyoto University, Kyoto, Japan
| | - Atsushi Yasumoto
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Yusuke Oguchi
- Department of Biological Sciences, The University of Tokyo, Tokyo, Japan
| | - Nobutake Suzuki
- Department of Biological Sciences, The University of Tokyo, Tokyo, Japan
| | | | | | - Takuro Ito
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Japan Science and Technology Agency, Saitama, Japan
| | - Kei Hiraki
- Department of Chemistry, The University of Tokyo, Tokyo, Japan
| | - Makoto Yamada
- Department of Intelligence Science and Technology, Graduate School of Informatics, Kyoto University, Kyoto, Japan
| | - Satoshi Matsusaka
- Clinical Research and Regional Innovation, Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Takeshi Hayakawa
- Department of Precision Mechanics, Chuo University, Tokyo, Japan
| | - Hideya Fukuzawa
- Laboratory of Applied Molecular Microbiology, Kyoto University, Kyoto, Japan
| | - Yutaka Yatomi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Fumihito Arai
- Department of Micro-Nano Mechanical Science and Engineering, Nagoya University, Nagoya, Japan
| | - Dino Di Carlo
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, USA.,Department of Mechanical Engineering, University of California, Los Angeles, Los Angeles, CA, USA.,California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, USA
| | - Atsuhiro Nakagawa
- Department of Neurosurgery, Graduate School of Medicine, Tohoku University, Sendai, Japan
| | - Yu Hoshino
- Department of Chemical Engineering, Kyushu University, Fukuoka, Japan
| | - Yoichiroh Hosokawa
- Division of Materials Science, Graduate School of Science and Technology, Nara Institute of Science and Technology, Ikoma, Japan
| | - Sotaro Uemura
- Department of Biological Sciences, The University of Tokyo, Tokyo, Japan
| | - Takeaki Sugimura
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Japan Science and Technology Agency, Saitama, Japan
| | - Yasuyuki Ozeki
- Department of Electrical Engineering and Information Systems, The University of Tokyo, Tokyo, Japan
| | - Nao Nitta
- Department of Chemistry, The University of Tokyo, Tokyo, Japan.,Japan Science and Technology Agency, Saitama, Japan
| | - Keisuke Goda
- Department of Chemistry, The University of Tokyo, Tokyo, Japan. .,Japan Science and Technology Agency, Saitama, Japan. .,Department of Electrical Engineering, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
12
|
Nelep C, Eberhardt J. Automated rare single cell picking with the ALS cellcelector™. Cytometry A 2018; 93:1267-1270. [PMID: 30184320 PMCID: PMC6586056 DOI: 10.1002/cyto.a.23568] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/19/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023]
Abstract
Molecular analysis of rare single cells like circulating tumor cells (CTCs) from whole blood patient samples bears multiple challenges. One of those challenges is the efficient and ideally loss-free isolation of CTCs over contaminating white and red blood cells. While there is a multitude of commercial and non-commercial systems available for the enrichment of CTCs their cell output does not deliver the purity most molecular analysis methods require. Here we describe the ALS CellCelector™ which can solve this challenge allowing the retrieval of 100% pure single CTCs from blood processed by different upstream enrichment techniques. It is a multifunctional, extremely flexible system for automated screening of cell culture plates, Petri dishes, and microscope slides. Fixed or live single cells or multicellular clusters detected during screening can be picked out of those plates automatically. The complete scan and picking process is fully documented hence allowing highest standardization and reproducibility of all processes. Use of CellCelector allowed the isolation of pure single tumor cells or clusters from liquid biopsies of breast, prostate, ovarian, colorectal, lung, and brain cancers for their subsequent molecular analysis. © 2018 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
|
13
|
Hashimoto M, Tanaka F, Yoneda K, Takuwa T, Matsumoto S, Okumura Y, Kondo N, Tsujimura T, Nakano T, Hasegawa S. Positive correlation between postoperative tumor recurrence and changes in circulating tumor cell counts in pulmonary venous blood (pvCTC) during surgical manipulation in non-small cell lung cancer. J Thorac Dis 2018; 10:298-306. [PMID: 29600060 DOI: 10.21037/jtd.2017.12.56] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background In non-small cell lung cancer (NSCLC), circulating tumor cells (CTC) are shed and circulate to the peripheral blood through the pulmonary vein. Previously, CTC count in pulmonary venous blood (pvCTC) was shown to significantly increase after surgical manipulation. Therefore, we assessed the correlation between the changes in the pvCTC count (ΔpvCTC) and clinical outcomes. Methods Consecutive patients with peripheral-type, NSCLC, who underwent lobectomy or bi-lobectomy through open thoracotomy, were enrolled prospectively. Before and after lobectomy, 2.5 mL of blood was drawn from the associated lobar pulmonary vein (PV), and was served for the quantitative evaluation of CTC using the CellSearch® system. The cut-off point of ΔpvCTC was determined according to clinical outcomes and ΔpvCTC using receiver operation characteristic (ROC) curve. Then the correlation between ΔpvCTC and clinical outcomes was evaluated by Kaplan-Meier analyses and log-rank test. In addition, the correlation between ΔpvCTC and perioperative variables was assessed. Results A total of 30 patients were enrolled, tumor recurrence occurred in 11 patients over a median follow-up of 64.4 months. Of these, 7 patients had distant metastasis and 4 had local recurrence. The median ΔpvCTC was 49 cells/2.5 mL, and pvCTC-count was increased during surgical manipulation in 24 patients (80%). We divided patients into two groups based on ΔpvCTC with the cut-off value as 119 cells/2.5 mL according to ROC curve. Significant shorter time to distant metastasis (TDM) (P=0.0123) was observed in high ΔpvCTC group (ΔpvCTC ≥119 cells/2.5 mL) than low ΔpvCTC group (ΔpvCTC <119 cells/ 2.5mL). Neither disease-free survival (DFS) nor overall survival (OS) was significantly correlated with ΔpvCTC. Conclusions Increasing pvCTC count during surgical manipulation was significantly correlated with postoperative distant metastasis in completely resected NSCLC patients. Significant shorter TDM was observed in patient with high ΔpvCTC group.
Collapse
Affiliation(s)
- Masaki Hashimoto
- Department of Thoracic Surgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Fumihiro Tanaka
- Second Department of Surgery (Chest Surgery), University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Kazue Yoneda
- Second Department of Surgery (Chest Surgery), University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Teruhisa Takuwa
- Department of Thoracic Surgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Seiji Matsumoto
- Department of Thoracic Surgery, Hyogo College of Medicine, Nishinomiya, Japan
| | | | - Nobuyuki Kondo
- Department of Thoracic Surgery, Hyogo College of Medicine, Nishinomiya, Japan
| | - Tohru Tsujimura
- Department of Pathology, Hyogo College of Medicine, Nishinomiya, Japan
| | - Takashi Nakano
- Center for Respiratory Medicine, Otemae Hospital, Osaka, Japan
| | - Seiki Hasegawa
- Department of Thoracic Surgery, Hyogo College of Medicine, Nishinomiya, Japan
| |
Collapse
|
14
|
Liu HE, Triboulet M, Zia A, Vuppalapaty M, Kidess-Sigal E, Coller J, Natu VS, Shokoohi V, Che J, Renier C, Chan NH, Hanft VR, Jeffrey SS, Sollier-Christen E. Workflow optimization of whole genome amplification and targeted panel sequencing for CTC mutation detection. NPJ Genom Med 2017; 2:34. [PMID: 29263843 PMCID: PMC5677973 DOI: 10.1038/s41525-017-0034-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 08/29/2017] [Accepted: 09/11/2017] [Indexed: 12/30/2022] Open
Abstract
Genomic characterization of circulating tumor cells (CTCs) may prove useful as a surrogate for conventional tissue biopsies. This is particularly important as studies have shown different mutational profiles between CTCs and ctDNA in some tumor subtypes. However, isolating rare CTCs from whole blood has significant hurdles. Very limited DNA quantities often can't meet NGS requirements without whole genome amplification (WGA). Moreover, white blood cells (WBC) germline contamination may confound CTC somatic mutation analyses. Thus, a good CTC enrichment platform with an efficient WGA and NGS workflow are needed. Here, Vortex label-free CTC enrichment platform was used to capture CTCs. DNA extraction was optimized, WGA evaluated and targeted NGS tested. We used metastatic colorectal cancer (CRC) as the clinical target, HCT116 as the corresponding cell line, GenomePlex® and REPLI-g as the WGA methods, GeneRead DNAseq Human CRC Panel as the 38 gene panel. The workflow was further validated on metastatic CRC patient samples, assaying both tumor and CTCs. WBCs from the same patients were included to eliminate germline contaminations. The described workflow performed well on samples with sufficient DNA, but showed bias for rare cells with limited DNA input. REPLI-g provided an unbiased amplification on fresh rare cells, enabling an accurate variant calling using the targeted NGS. Somatic variants were detected in patient CTCs and not found in age matched healthy donors. This demonstrates the feasibility of a simple workflow for clinically relevant monitoring of tumor genetics in real time and over the course of a patient's therapy using CTCs.
Collapse
Affiliation(s)
| | - Melanie Triboulet
- Department of Surgery, Stanford University School of Medicine, Stanford, CA USA
| | - Amin Zia
- Stanford Center for Genomics and Personalized Medicine, Stanford University, Stanford, CA USA
| | | | - Evelyn Kidess-Sigal
- Department of Surgery, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Hepatology and Gastroenterology, Charité University Hospital, Berlin, Germany
| | - John Coller
- Stanford Functional Genomics Facility, Stanford University, Stanford, CA USA
| | - Vanita S. Natu
- Stanford Functional Genomics Facility, Stanford University, Stanford, CA USA
| | - Vida Shokoohi
- Stanford Functional Genomics Facility, Stanford University, Stanford, CA USA
| | - James Che
- Vortex Biosciences, Inc., Menlo Park, CA USA
| | | | - Natalie H. Chan
- Department of Surgery, Stanford University School of Medicine, Stanford, CA USA
| | - Violet R. Hanft
- Department of Surgery, Stanford University School of Medicine, Stanford, CA USA
| | - Stefanie S. Jeffrey
- Department of Surgery, Stanford University School of Medicine, Stanford, CA USA
| | | |
Collapse
|
15
|
A Prospective Study of Loose Tissue Fragments in Non-Small Cell Lung Cancer Resection Specimens: An Alternative View to "Spread Through Air Spaces". Am J Surg Pathol 2017. [PMID: 28622180 DOI: 10.1097/pas.0000000000000889] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The World Health Organization Classification of Lung Tumors considers "Spread Through Air Spaces" a form of invasion in lung adenocarcinoma. The recently described spread of free-floating cell clusters during lung specimen sectioning, otherwise known as "Spread Through A Knife Surface," represents an ex vivo artifact. The purpose of this study was to prospectively investigate the presence and frequency of these free-floating tumor cell clusters in surgically resected lung cancer specimens and their possible relation to gross examination procedures. A prospective, multi-institutional study of non-small cell lung cancer resection specimen was undertaken. At prosection the first cut was made with a clean knife; the second cut was made in a parallel plane to the first. Four tissue blocks were taken from upper and lower parts of first and second cuts. Hematoxylin and eosin-stained slides were examined for displaced benign and/or malignant tissue fragments. Forty-four resection specimens were studied. The mean number of tumor clusters for blocks 1 to 4 was 0.36, 1.44, 1.86, and 1.95, respectively, and for benign fragments was 0.11, 0.11, 0.13, and 0.25, respectively. Almost all cell clusters were intra-alveolar. Comparison of tumor cell clusters in block 1 with blocks 2 to 4 was significant with P-values (Friedman test for repeated measures 0.03) 0.031, 0.02, and 0.05, respectively. Overall 93% of the loose tissue fragments could be explained by mechanical forces associated with tissue handling. While the 2015 World Health Organization Classification of Lung Tumors recognizes Spread Through Air Spaces as a form of lung cancer invasion, such is debatable and in many instances likely represents mechanical artifact, including dissemination along the prosecting knife blade.
Collapse
|
16
|
Matsutani N, Sawabata N, Yamaguchi M, Woo T, Kudo Y, Kawase A, Shiono S, Iinuma H, Morita S, Kawamura M. Does lung cancer surgery cause circulating tumor cells?-A multicenter, prospective study. J Thorac Dis 2017; 9:2419-2426. [PMID: 28932547 DOI: 10.21037/jtd.2017.07.33] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND Cancer relapse is caused by residual isolated tumor cells (ITCs) remaining in the body after surgery. It is speculated that surgical manipulation may cause circulating tumor cells (CTCs) which are the origin of ITCs in the body. The occurrence of CTCs in surgical patients with non-small cell lung cancer (NSCLC) has been shown in retrospective observation, but not prospectively, thus we conducted a multicenter prospective study regarding the occurrence of CTCs by surgical manipulation. METHODS Patients with T1b-2N0M0 lung cancer were studied. Blood samples were collected from the peripheral artery in the operating room at both pre- and post-lobectomy to extract CTCs by a size selection method. The CTCs detection rate, pathological findings, and background of each patient were studied. RESULTS The histological diagnosis of 29 patients were adenocarcinoma in 24 patients, squamous cell carcinoma in 3 patients, and other types in 2 patients. The number of pre-CTCs positive patients was 13 and the number of post CTCs positive patients was 17. Among the 16 patients who were pre-CTCs negative, 4 patients showed post CTCs positive, while all pre-CTCs positive patients remained post-CTCs positive. CONCLUSIONS The likelihood of CTC dislodgement by surgical manipulation is indicated based on the result that CTCs were detected after lung cancer surgery, where there were no cases of pre-CTCs positive and post CTCs negative.
Collapse
Affiliation(s)
- Noriyuki Matsutani
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | | | - Masafumi Yamaguchi
- Department of General Thoracic Oncology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Tetsukan Woo
- Department of Thoracic Surgery, Saiseikai Yokohama City South Hospital, Kanagawa, Japan
| | - Yujin Kudo
- Department of Thoracic Surgery, Tokyo Medical University, Tokyo, Japan
| | - Akikazu Kawase
- First Department of Surgery, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Satoshi Shiono
- Department of Thoracic Surgery, Yamagata Prefectural Central Hospital, Yamagata, Japan
| | - Hisae Iinuma
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| | - Shigeki Morita
- Department of Diagnostic Pathology, University Hospital, Teikyo University School of Medicine, Tokyo, Japan
| | - Masafumi Kawamura
- Department of Surgery, Teikyo University School of Medicine, Tokyo, Japan
| |
Collapse
|
17
|
Mascalchi M, Maddau C, Sali L, Bertelli E, Salvianti F, Zuccherelli S, Matucci M, Borgheresi A, Raspanti C, Lanzetta M, Falchini M, Mazza E, Vella A, Luconi M, Pinzani P, Pazzagli M. Circulating tumor cells and microemboli can differentiate malignant and benign pulmonary lesions. J Cancer 2017; 8:2223-2230. [PMID: 28819424 PMCID: PMC5560139 DOI: 10.7150/jca.18418] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 02/26/2017] [Indexed: 01/02/2023] Open
Abstract
The presence of circulating tumor cells (CTC) or microemboli (CTM) in the peripheral blood can theoretically anticipate malignancy of solid lesions in a variety of organs. We aimed to preliminarily assess this capability in patients with pulmonary lesions of suspected malignant nature. We used a cell-size filtration method (ScreenCell) and cytomorphometric criteria to detect CTC/CTM in a 3 mL sample of peripheral blood that was taken just before diagnostic percutaneous CT-guided fine needle aspiration (FNA) or core biopsy of the suspicious lung lesion. At least one CTC/CTM was found in 47 of 67 (70%) patients with final diagnoses of lung malignancy and in none of 8 patients with benign pulmonary nodules. In particular they were detected in 38 (69%) of 55 primary lung cancers and in 9 (75%) of 12 lung metastases from extra-pulmonary cancers. Sensitivity of CTC/CTM presence for malignancy was 70.1% (95%CI: 56.9-83.1%), specificity 100%, positive predictive value 100% and negative predictive value 28.6% (95%CI: 11.9-45.3%). Remarkably, the presence of CTC/CTM anticipated the diagnosis of primary lung cancer in 3 of 5 patients with non-diagnostic or inconclusive results of FNA or core biopsy, whereas CTC/CTM were not observed in 1 patient with sarcoidosis and 1 with amarthocondroma. These results suggest that presently, due to the low sensitivity, the search of CTC/CTM cannot replace CT guided percutaneous FNA or core biopsy in the diagnostic work-up of patients with suspicious malignant lung lesions. However, the high specificity may as yet indicate a role in cases with non-diagnostic or inconclusive FNA or core biopsy results that warrants to be further investigated.
Collapse
Affiliation(s)
- Mario Mascalchi
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Cristina Maddau
- Oncological Prevention Laboratory, Institute for Cancer Research and Prevention (ISPO), Florence, Italy
| | - Lapo Sali
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Elena Bertelli
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Francesca Salvianti
- Clinical Biochemistry Unit, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Stefania Zuccherelli
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Marzia Matucci
- Oncological Prevention Laboratory, Institute for Cancer Research and Prevention (ISPO), Florence, Italy
| | - Alessandra Borgheresi
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Claudio Raspanti
- Interventional Radiology Unit, Careggi Hospital, Florence, Italy
| | - Monica Lanzetta
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Massimo Falchini
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Ernesto Mazza
- Interventional Radiology Unit, Careggi Hospital, Florence, Italy
| | - Alessandra Vella
- Nuclear Medicine Unit, Le Scotte University Hospital, Siena, Italy
| | - Michaela Luconi
- Endocrinology Unit, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Pamela Pinzani
- Clinical Biochemistry Unit, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| | - Mario Pazzagli
- Clinical Biochemistry Unit, "Mario Serio" Department of Biomedical, Experimental and Clinical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
18
|
Chudasama D, Burnside N, Beeson J, Karteris E, Rice A, Anikin V. Perioperative detection of circulating tumour cells in patients with lung cancer. Oncol Lett 2017; 14:1281-1286. [PMID: 28789342 PMCID: PMC5529936 DOI: 10.3892/ol.2017.6366] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 11/30/2016] [Indexed: 11/24/2022] Open
Abstract
Lung cancer is a leading cause of mortality and despite surgical resection a proportion of patients may develop metastatic spread. The detection of circulating tumour cells (CTCs) may allow for improved prediction of metastatic spread and survival. The current study evaluates the efficacy of the ScreenCell® filtration device, to capture, isolate and propagate CTCs in patients with primary lung cancer. Prior to assessment of CTCs, the present study detected cancer cells in a proof-of-principle- experiment using A549 human lung carcinoma cells as a model. Ten patients (five males and five females) with pathologically diagnosed primary non-small cell lung cancer undergoing surgical resection, had their blood tested for CTCs. Samples were taken from a peripheral vessel at the baseline, from the pulmonary vein draining the lobe containing the tumour immediately prior to division, a further central sample was taken following completion of the resection, and a final peripheral sample was taken three days post-resection. A significant increase in CTCs was observed from baseline levels following lung manipulation. No association was able to be made between increased levels of circulating tumour cells and survival or the development of metastatic deposits. Manipulation of the lung during surgical resection for non-small cell lung carcinoma results in a temporarily increased level of CTCs; however, no clinical impact for this increase was observed. Overall, the study suggests the ScreenCell® device has the potential to be used as a CTC isolation tool, following further work, adaptations and improvements to the technology and validation of results.
Collapse
Affiliation(s)
- Dimple Chudasama
- Department of Cardiothoracic Surgery, Harefield Hospital, Royal Brompton and Harefield Trust, Harefield, Middlesex UB9 6JH, UK.,Department of Life Sciences, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
| | - Nathan Burnside
- Department of Cardiothoracic Surgery, Harefield Hospital, Royal Brompton and Harefield Trust, Harefield, Middlesex UB9 6JH, UK
| | - Julie Beeson
- Department of Cardiothoracic Surgery, Harefield Hospital, Royal Brompton and Harefield Trust, Harefield, Middlesex UB9 6JH, UK
| | - Emmanouil Karteris
- Department of Life Sciences, Brunel University London, Uxbridge, Middlesex UB8 3PH, UK
| | - Alexandra Rice
- Department of Pathology, Harefield Hospital, Royal Brompton and Harefield Trust, Harefield, Middlesex UB9 6JH, UK
| | - Vladimir Anikin
- Department of Cardiothoracic Surgery, Harefield Hospital, Royal Brompton and Harefield Trust, Harefield, Middlesex UB9 6JH, UK
| |
Collapse
|
19
|
Perioperative circulating tumor cells in surgical patients with non-small cell lung cancer: does surgical manipulation dislodge cancer cells thus allowing them to pass into the peripheral blood? Surg Today 2016; 46:1402-1409. [PMID: 26951195 DOI: 10.1007/s00595-016-1318-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 02/16/2016] [Indexed: 02/05/2023]
Abstract
PURPOSE We herein evaluated the status of circulating tumor cells (CTC) dislodged from the tumor during surgery in patients who underwent pulmonary resection for non-small cell lung cancer (NSCLC) to assess the clinical implications. METHODS Tumor cells in the peripheral arterial blood before surgery (Before) and immediately after lung resection (After) and in the blood from the pulmonary vein of the resected lung were detected using a size selective method. The clinicopathological characteristics and the prognosis were then analyzed according to the CTC status: no tumor cells detected (N), single tumor cell or total number less than 4 cells (S), and existence of clustered cells (C). RESULTS According to the CTC status, the patients were classified into the following three groups: Before-C and After-C, Group I (n = 6); Before-S or N and After-C, Group II (n = 9); and Before-S or N and After-S or N, Group III (n = 8). Group III showed a high rate of p-stage IA, smaller tumor size, lower CEA level, lower SUVmax level, and a higher relapse-free survival rate than the other groups. CONCLUSIONS CTCs were detected in patients after undergoing lung resection, some of which may have been dislodged by the surgical procedure. The presence of clustered CTCs after the operation indicated an unfavorable outcome.
Collapse
|
20
|
Mascalchi M, Falchini M, Maddau C, Salvianti F, Nistri M, Bertelli E, Sali L, Zuccherelli S, Vella A, Matucci M, Voltolini L, Pegna AL, Luconi M, Pinzani P, Pazzagli M. Prevalence and number of circulating tumour cells and microemboli at diagnosis of advanced NSCLC. J Cancer Res Clin Oncol 2016; 142:195-200. [PMID: 26210156 DOI: 10.1007/s00432-015-2021-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Accepted: 07/14/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE Timing and magnitude of blood release of circulating tumour cells (CTC) and circulating tumour microemboli (CTM) from primary solid cancers are uncertain. We investigated prevalence and number of CTC and CTM at diagnosis of advanced non-small cell lung cancer (NSCLC). METHODS Twenty-eight consecutive patients with suspected stage III-IV lung cancer gave consent to provide 15 mL of peripheral blood soon before diagnostic CT-guided fine-needle aspiration biopsy (FNAB). CTC and CTM (clusters of ≥3 CTC) were isolated by cell size filtration (ScreenCell), identified and counted by cytopathologists using morphometric criteria and (in 6 cases) immunostained for vimentin. RESULTS FNAB demonstrated NSCLC in 26 cases. At least one CTC/3 mL blood (mean 6.8 ± 3.7) was detected in 17 (65 %) and one CTM (mean 4.5 ± 3.3) in 15 (58 %) of 26 NSCLC cases. No correlation between number of CTC or CTM and tumour type or stage was observed. Neoplastic cells from both FNA and CTC/CTM were positive for vimentin but heterogeneously. CONCLUSIONS CTC can be detected in two-thirds and CTM in more than half of patients with advanced NSCLC at diagnosis. Reasons underlying lack of CTC and CTM in some advanced lung cancers deserve further investigations.
Collapse
Affiliation(s)
- Mario Mascalchi
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy.
| | - Massimo Falchini
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Cristina Maddau
- Oncological Prevention Laboratory, Cancer Prevention and Research Institute (ISPO), Florence, Italy
| | - Francesca Salvianti
- Clinical Biochemistry Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Marco Nistri
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Elena Bertelli
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Lapo Sali
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Stefania Zuccherelli
- Diagnostic and Interventional Radiology Units, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | - Alessandra Vella
- Nuclear Medicine Unit, Le Scotte University Hospital, Siena, Italy
| | - Marzia Matucci
- Oncological Prevention Laboratory, Cancer Prevention and Research Institute (ISPO), Florence, Italy
| | - Luca Voltolini
- Division of Thoracic Surgery, Careggi Hospital, Florence, Italy
| | | | - Michaela Luconi
- Endocrinology Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Pamela Pinzani
- Clinical Biochemistry Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| | - Mario Pazzagli
- Clinical Biochemistry Unit, "Mario Serio" Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy
| |
Collapse
|
21
|
Sawabata N, Funaki S, Shintani Y, Okumura M. Lung excision of non-small-cell lung cancer leaves cancer cells in residual lobe: cytological detection using pulmonary vein blood. Interact Cardiovasc Thorac Surg 2015; 22:131-5. [PMID: 26538102 DOI: 10.1093/icvts/ivv301] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Accepted: 09/21/2015] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES Lung excision to treat non-small-cell lung cancer (NSCLC) is associated with a worse prognosis when compared with a lobectomy. Cancer relapse may be caused by tumour cells remaining in the residual lobe, the possibility of dislodged cancer cells in the residual lobe is assessed using pulmonary vein blood (PVB) from the resected lung. METHODS Twenty-eight patients with pathological stage I NSCLC who underwent lung excision followed by a lobectomy were evaluated according to the status of isolated tumour cells (ITCs) (origin of circulating tumour cells) in PVB from the resected lobe. Survival was also assessed according to the status of ITCs. RESULTS The rate of ITC presence was 60.7% and depended on margin distance/tumour size (M/T) with a threshold of 1.0-30.8% (4/13) in M/T greater than or equal to 1.0 and 86.7% (13/15) in M/T smaller than 1.0 (P = 0.001). PVB-ITC status was no ITCs (N) in 11 (39.3%), only singular cells (S) in 13 (50.0%) and clustered cells (C) in 4 (14.3%). In addition, the survival status of patients with clustered cells was exclusively wrong. CONCLUSION After pulmonary excision for lung cancer, tumour cells remain in the residual lobe and the morphology of which may indicate recurrence.
Collapse
Affiliation(s)
- Noriyoshi Sawabata
- Japan Community Healthcare Organization, Hoshigaoka Medical Center, Hirakata, Japan Division of Health Science, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Soichiro Funaki
- Division of General Thoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yasushi Shintani
- Division of General Thoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Meinosin Okumura
- Division of General Thoracic Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
22
|
Reddy RM, Murlidhar V, Zhao L, Grabauskiene S, Zhang Z, Ramnath N, Lin J, Chang AC, Carrott P, Lynch W, Orringer MB, Beer DG, Nagrath S. Pulmonary venous blood sampling significantly increases the yield of circulating tumor cells in early-stage lung cancer. J Thorac Cardiovasc Surg 2015; 151:852-858. [PMID: 26614417 DOI: 10.1016/j.jtcvs.2015.09.126] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 08/19/2015] [Accepted: 09/26/2015] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To identify circulating tumor cells (CTCs) in the blood of patients with early-stage lung cancer and to show that sampling pulmonary vein (PV) blood using microfluidic chip technology will yield significantly more CTCs. Improving early detection of lung cancer is critical to improving lung cancer survival. Reproducible detection of CTCs is limited currently in early stage tumors. METHODS Patients undergoing pulmonary resection had PV blood drawn before resection. Peripheral blood was sampled at preoperative, intraoperative, and postoperative times. Samples were analyzed on microfluidic chips using antibody-based capture. RESULTS A total of 32 patients with primary lung cancer were evaluated. Twenty patients had 1 or more CTCs detected in at least 1 sample (62.5%). The mean number of CTCs from peripheral vein sources at the preoperative, intraoperative, and postoperative time points was 1.3, 1.9, and 0.6 respectively. The average number of CTCs in the PV was 340.0 (range, 0.0-5422.50; P > .01). When PV CTCs were present, the number of CTCs was correlated with pathological tumor size (P = .0236). The number of PV CTCs was not correlated with any other clinical feature (eg, smoking status, preoperative or postoperative stage). Furthermore, the number of PV CTCs was significantly higher when preoperative bronchoscopic biopsy was performed, compared with computed tomography-guided biopsy (P = .0311). Seven patients had evidence of CTC clusters, or microemboli. CONCLUSIONS With a single vein draining the entire tumor basin, lung cancers are unique, allowing the high-yield isolation of CTCs from the PV. This method may facilitate future studies to improve the detection and analysis of early-stage lung CTCs.
Collapse
Affiliation(s)
- Rishindra M Reddy
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan.
| | - Vasudha Murlidhar
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Svetlana Grabauskiene
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Zhuo Zhang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| | - Nithya Ramnath
- Division of Hematology/Oncology, Department of Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jules Lin
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Andrew C Chang
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Phillip Carrott
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - William Lynch
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Mark B Orringer
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - David G Beer
- Section of Thoracic Surgery, Department of Surgery, University of Michigan, Ann Arbor, Michigan
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|