1
|
Piekarczyk P, Lechowicz U, Szopiński J, Polaczek M, Błasińska K, Modrzewska K. RT-PCR Misdiagnosis of Patient with Rare EGFR Mutation Lung Adenocarcinoma: Is NGS the Only Solution? Diagnostics (Basel) 2025; 15:842. [PMID: 40218192 PMCID: PMC11988652 DOI: 10.3390/diagnostics15070842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Background and Clinical Significance: Molecular testing plays a crucial role in lung cancer diagnosis and management. While single-gene tests (SGTs) remain an important diagnostic tool, developments in novel methods such as next generation sequencing (NGS) provide a more precise mutational profile and enable the targeted treatment of a larger scope of mutation-driven cancers. Case presentation: We present a case of a patient with a rare EGFR variant lung adenocarcinoma, who was misdiagnosed using a SGT. The initial treatment with immunotherapy was unsuccessful. Conclusions: The patient could have benefited if NGS had been performed instead of traditional real-time polymerase chain reaction (RT-PCR) and if adequate tyrosine kinase inhibitor treatment was initiated at the time of diagnosis.
Collapse
Affiliation(s)
- Piotr Piekarczyk
- 3rd Department of Lung Diseases and Oncology, National Tuberculosis and Lung Diseases Research Institute, 01-138 Warsaw, Poland
| | - Urszula Lechowicz
- Department of Genetics and Clinical Immunology, National Tuberculosis and Lung Diseases Research Institute, 01-138 Warsaw, Poland
| | - Janusz Szopiński
- 3rd Department of Lung Diseases and Oncology, National Tuberculosis and Lung Diseases Research Institute, 01-138 Warsaw, Poland
| | - Mateusz Polaczek
- 3rd Department of Lung Diseases and Oncology, National Tuberculosis and Lung Diseases Research Institute, 01-138 Warsaw, Poland
| | - Katarzyna Błasińska
- Department of Radiology, National Tuberculosis and Lung Diseases Research Institute, 01-138 Warsaw, Poland
| | - Katarzyna Modrzewska
- 3rd Department of Lung Diseases and Oncology, National Tuberculosis and Lung Diseases Research Institute, 01-138 Warsaw, Poland
| |
Collapse
|
2
|
Kim W, Cho S, Lee J, Lee J, Ji S, Sung H, Jung W, Jeon JH, Kim K, Jheon S. Mutational differences between primary cancer tissue and circulating tumor cells in early-stage non-small cell lung cancer. Transl Lung Cancer Res 2024; 13:3026-3038. [PMID: 39669998 PMCID: PMC11632435 DOI: 10.21037/tlcr-24-709] [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: 08/12/2024] [Accepted: 10/21/2024] [Indexed: 12/14/2024]
Abstract
Background Early-stage non-small cell lung cancer (NSCLC) has a high recurrence rate despite proper management, including curative surgery. Circulating tumor cells (CTCs) are believed to play a key role in the distant metastasis of lung cancer. Immunofluorescence imaging studies of CTCs have revealed that they are associated with the prognosis of NSCLC. However, the mutational profiling of CTCs from early-stage NSCLC has not been extensively explored. We hypothesized that CTCs could be detected by mutational analysis using panel sequencing and would have distinct mutations associated with distant metastasis compared to those of primary cancer tissue in early-stage NSCLC. Thus, this study examined the DNA from CTCs using targeted panel sequencing to identify mutations and compared them with mutations found in primary cancer tissue in patients with resectable early-stage NSCLC. Methods Overall, 45 patients with resectable NSCLC were prospectively enrolled from September to December 2023. Matched whole blood samples and primary cancer tissues were collected during curative surgery. Then, 405-gene targeted panel sequencing was performed on DNA from primary cancer tissues and CTCs. Results In this study, 37 patients (82%) had adenocarcinoma, and 30 (67%) were classified as having pathologic stage 1 disease. Mutated genes were detected in all (100%) and 31 patients (69%) for primary cancer tissue and CTCs from panel sequencing, respectively. The partial concordance rate of mutations between primary cancer tissue and CTCs was 17.8%, with the top 10 mutated genes differing significantly. Among primary cancer tissue samples, mutated genes differed by stage and histologic type; these findings were not observed in CTCs. CTCs predominantly displayed mutations in tumor suppressor genes, whereas primary cancer tissues exhibited mutations in both oncogenes and tumor suppressor genes. Conclusions CTCs exhibited unique mutations, showing low concordance with mutations found in primary cancer tissue. CTCs may possess specific mutations independent from those of primary cancer tissue in early-stage NSCLC.
Collapse
Affiliation(s)
- Woojung Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Joonseok Lee
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jinsu Lee
- BeyondDx Inc., Gwangmyeong, Republic of Korea
| | - Soojeong Ji
- BeyondDx Inc., Gwangmyeong, Republic of Korea
| | - Hyejin Sung
- BeyondDx Inc., Gwangmyeong, Republic of Korea
| | - Woohyun Jung
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jae Hyun Jeon
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kwhanmien Kim
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Sanghoon Jheon
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Thoracic and Cardiovascular Surgery, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
3
|
Hedblom AH, Pruneri G, Quagliata L, Costa JL, Dumanois R, Rolando C, Saunders R. Cancer patient management: Current use of next-generation sequencing in the EU TOP4. J Cancer Policy 2023; 35:100376. [PMID: 36511325 DOI: 10.1016/j.jcpo.2022.100376] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/12/2022] [Accepted: 11/25/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Next-generation sequencing (NGS) is recognised by a growing audience of medical professionals as a functional diagnostic tool in oncology. However, adoption in clinical routine proceeds haphazardly in Europe. METHODS A semi-structured interview survey was administered to 68 cancer care professionals in four EU countries between June-August and November-December 2021. Pre-screening questionnaires assessed sufficient NGS expertise, diverse geographical distribution, and professional roles. RESULTS Our findings provide a better understanding of current clinical, regulatory, and reimbursement practices for NGS in four EU countries. CONCLUSIONS Despite the impending European In-vitro Diagnostic Medical Devices Regulation (IVDR), tortuous national guidelines implementations and limping reimbursement policies are common traits across surveyed countries and produce disparity in access to advanced healthcare services amid regional distinctions. POLICY SUMMARY The evident information gap between involved parties and demand for consistent national guidelines could be filled by health economics analyses tailored to local specifics to provide factual leverage for a structured adoption of NGS testing.
Collapse
Affiliation(s)
| | - Giancarlo Pruneri
- Fondazione IRCCS, Istituto Nazionale Tumori, Milan, Italy; School of Medicine, University of Milan, Milan, Italy
| | | | | | | | | | | |
Collapse
|
4
|
Zhang S, Wang W, Xu C, Zhang Y, Cai X, Wang Q, Song Z, Li Z, Yu J, Zhong W, Wang Z, Liu J, Liu A, Li W, Zhan P, Liu H, Lv T, Miao L, Min L, Lin G, Huang L, Yuan J, Jiang Z, Pu X, Rao C, Lv D, Yu Z, Li X, Tang C, Zhou C, Zhang J, Guo H, Chu Q, Meng R, Liu X, Wu J, Zhou J, Zhu Z, Pan W, Dong X, Pang F, Wang K, Yao C, Lin G, Li S, Yang Z, Luo J, Jia H, Nie X, Wang L, Zhu Y, Hu X, Xie Y, Lin X, Cai J, Xia Y, Feng H, Wang L, Du Y, Yao W, Shi X, Niu X, Yuan D, Yao Y, Kang J, Zhang J, Zhang C, Gao W, Huang J, Zhang Y, Sun P, Wang H, Ye M, Wang D, Wang Z, Wan B, Lv D, Yu G, Shi L, Xia Y, Gao F, Zhang X, Xu T, Zhou W, Wang H, Liu Z, Yang N, Wu L, Wang Q, Wang G, Hong Z, Wang J, Fang M, Fang Y, Zhang Y, Song Y, Ma S, Fang W, Lu Y. Chinese expert consensus on the diagnosis and treatment of HER2-altered non-small cell lung cancer. Thorac Cancer 2022; 14:91-104. [PMID: 36444143 PMCID: PMC9807451 DOI: 10.1111/1759-7714.14743] [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: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) possesses tyrosine kinase activity and participates in cell growth, differentiation and migration, and survival. Its alterations, mainly including mutations, amplifications, and overexpression are associated with poor prognosis and are one of the major drivers in non-small cell lung cancer (NSCLC). Several clinical trials had been investigating on the treatments of HER2-altered NSCLC, including conventional chemotherapy, programmed death 1 (PD-1) inhibitors, tyrosine kinase inhibitors (TKIs) and antibody-drug conjugates (ADCs), however, the results were either disappointing or encouraging, but inconsistent. Trastuzumab deruxtecan (T-DXd) was recently approved by the Food and Drug Administration as the first targeted agent for treating HER2-mutant NSCLC. Effective screening of patients is the key to the clinical application of HER2-targeted agents such as TKIs and ADCs. Various testing methods are nowadays available, including polymerase chain reaction (PCR), next-generation sequencing (NGS), fluorescence in situ hybridization (FISH), immunohistochemistry (IHC), etc. Each method has its pros and cons and should be reasonably assigned to appropriate patients for diagnosis and guiding treatment decisions. To help standardize the clinical workflow, our expert group reached a consensus on the clinical management of HER2-altered NSCLC, focusing on the diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Shirong Zhang
- Translational Medicine Research Center, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Cancer CenterZhejiang University School of MedicineHangzhouChina
| | - Wenxian Wang
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Chunwei Xu
- Institute of Cancer and Basic Medicine (ICBM)Chinese Academy of SciencesHangzhouChina,Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Yongchang Zhang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Xiuyu Cai
- Department of VIP Inpatient, Sun Yet‐Sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Qian Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingChina
| | - Zhengbo Song
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Ziming Li
- Department of Shanghai Lung Cancer Center, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Jinpu Yu
- Department of Cancer Molecular Diagnostics CoreTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesSchool of MedicineGuangzhouChina
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jingjing Liu
- Department of Thoracic CancerJilin Cancer HospitalChangchunChina
| | - Anwen Liu
- Department of OncologySecond Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Wen Li
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Cancer CenterZhejiang UniversityHangzhouChina
| | - Ping Zhan
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Hongbing Liu
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Tangfeng Lv
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Liyun Miao
- Department of Respiratory Medicine, Affiliated Drum Tower HospitalMedical School of Nanjing UniversityNanjingChina
| | - Lingfeng Min
- Department of Respiratory Medicine, Clinical Medical School of Yangzhou UniversitySubei People's Hospital of Jiangsu ProvinceYangzhouChina
| | - Gen Lin
- Department of Medical OncologyFujian Medical University Cancer Hospital and Fujian Cancer HospitalFuzhouChina
| | - Long Huang
- Department of OncologySecond Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Jingping Yuan
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhansheng Jiang
- Department of Integrative OncologyTianjin Medical University Cancer Institute and HospitalTianjinChina
| | - Xingxiang Pu
- Department of Medical Oncology, Lung Cancer and Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Chuangzhou Rao
- Department of Radiotherapy and Chemotherapy, Hwamei HospitalUniversity of Chinese Academy of SciencesNingboChina
| | - Dongqing Lv
- Department of Pulmonary MedicineTaizhou Hospital of Wenzhou Medical UniversityTaizhouChina
| | - Zongyang Yu
- Department of Respiratory Medicine, the 900th Hospital of the Joint Logistics Team (the Former Fuzhou General Hospital)Fujian Medical UniversityFuzhouChina
| | - Xiaoyan Li
- Department of Oncology, Beijing Tiantan HospitalCapital Medical UniversityBeijingChina
| | - Chuanhao Tang
- Department of Medical OncologyPeking University International HospitalBeijingChina
| | - Chengzhi Zhou
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease; Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University (The First Affiliated Hospital of Guangzhou Medical University)GuangzhouChina
| | - Junping Zhang
- Department of Thoracic Oncology, Shanxi Academy of Medical SciencesShanxi Bethune HospitalTaiyuanChina
| | - Hui Guo
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Rui Meng
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Xuewen Liu
- Department of Oncology, the Third Xiangya HospitalCentral South UniversityChangshaChina
| | - Jingxun Wu
- Department of Medical Oncology, the First Affiliated Hospital of MedicineXiamen UniversityXiamenChina
| | - Jin Zhou
- Department of Medical Oncology, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, School of MedicineUniversity of Electronic Science and TechnologyChengduChina
| | - Zhengfei Zhu
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Weiwei Pan
- Department of Cell Biology, College of MedicineJiaxing UniversityJiaxingChina
| | - Xiaowei Dong
- Department of MedicalShanghai OrigiMed Co., LtdShanghaiChina
| | - Fei Pang
- Department of MedicalShanghai OrigiMed Co., LtdShanghaiChina
| | - Kai Wang
- Department of MedicalShanghai OrigiMed Co., LtdShanghaiChina
| | - Chao Yao
- Department of MedicalShanghai OrigiMed Co., LtdShanghaiChina
| | - Guomin Lin
- Department of MedicalShanghai OrigiMed Co., LtdShanghaiChina
| | - Site Li
- Department of IVD Medical Marketing3D MedicinesShanghaiChina
| | - Zhi Yang
- Department of IVD Medical Marketing3D MedicinesShanghaiChina
| | | | - Hongtao Jia
- Department of MedicalAiyi TechnologyBeijingChina
| | - Xiuqing Nie
- Department of Clinical TrialNovocodex BiopharmaceuticShaoxingChina
| | - Liping Wang
- Department of Oncology, Baotou Cancer HospitalBaotouChina
| | - Youcai Zhu
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun HospitalThe Third Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Xiao Hu
- Zhejiang Key Laboratory of Radiation OncologyCancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital)HangzhouChina
| | - Yanru Xie
- Department of OncologyLishui Municipal Central HospitalLishuiChina
| | - Xinqing Lin
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease; Guangzhou Institute of Respiratory HealthThe First Affiliated Hospital of Guangzhou Medical University (The First Affiliated Hospital of Guangzhou Medical University)GuangzhouChina
| | - Jing Cai
- Department of OncologySecond Affiliated Hospital of Nanchang UniversityNanchangChina
| | - Yang Xia
- Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Cancer CenterZhejiang UniversityHangzhouChina
| | - Huijing Feng
- Department of Thoracic Oncology, Shanxi Academy of Medical SciencesShanxi Bethune HospitalTaiyuanChina
| | - Lin Wang
- Department of Pathology, Shanxi Academy of Medical SciencesShanxi Bethune HospitalTaiyuanChina
| | - Yingying Du
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
| | - Wang Yao
- Department of Interventional Oncology, The First Affiliated HospitalSun Yat‐sen UniversityGuangzhouChina
| | - Xuefei Shi
- Department of Respiratory Medicine, Huzhou HospitalZhejiang University School of MedicineHuzhouChina
| | - Xiaomin Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest HospitalShanghai Jiao Tong UniversityShanghaiChina
| | - Dongmei Yuan
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Yanwen Yao
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Jing Kang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesSchool of MedicineGuangzhouChina
| | - Jiatao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesSchool of MedicineGuangzhouChina
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital, Guangdong Academy of Medical SciencesSchool of MedicineGuangzhouChina
| | - Wenbin Gao
- Department of OncologyThe Third Affiliated Hospital of Shenzhen UniversityShenzhenChina
| | - Jianhui Huang
- Department of OncologyLishui Municipal Central HospitalLishuiChina
| | - Yinbin Zhang
- Department of Oncology, the Second Affiliated Hospital of Medical CollegeXi'an Jiaotong UniversityXi'anChina
| | - Pingli Sun
- Department of PathologyThe Second Hospital of Jilin UniversityChangchunChina
| | - Hong Wang
- Senior Department of OncologyThe 5th Medical Center of PLA General HospitalBeijingChina
| | - Mingxiang Ye
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Dong Wang
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Zhaofeng Wang
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Bing Wan
- Department of Respiratory MedicineThe Affiliated Jiangning Hospital of Nanjing Medical UniversityNanjingChina
| | - Donglai Lv
- Department of Clinical OncologyThe 901 Hospital of Joint Logistics Support Force of People Liberation ArmyHefeiChina
| | - Genhua Yu
- Department of Radiation OncologyZhebei Mingzhou HospitalHuzhouChina
| | - Lin Shi
- Department of Respiratory Medicine, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yuanli Xia
- Department of Thoracic Disease Diagnosis and Treatment Center, Zhejiang Rongjun HospitalThe Third Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Feng Gao
- Department of Thoracic SurgeryThe Fourth Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Xiaochen Zhang
- Department of Oncology, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Tao Xu
- Department of Pulmonary and Critical Care MedicineThe Affiliated Hospital of Qingdao UniversityQingdaoChina
| | - Wei Zhou
- Institute of Cancer and Basic Medicine (ICBM)Chinese Academy of SciencesHangzhouChina
| | - Haixia Wang
- Institute of Cancer and Basic Medicine (ICBM)Chinese Academy of SciencesHangzhouChina
| | - Zhefeng Liu
- Senior Department of OncologyThe 5th Medical Center of PLA General HospitalBeijingChina
| | - Nong Yang
- Department of Medical Oncology, Lung Cancer and Gastrointestinal Unit, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Lin Wu
- Department of Medical Oncology, Lung Cancer and Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| | - Qiming Wang
- Department of Internal Medicine, The Affiliated Cancer Hospital of Zhengzhou UniversityHenan Cancer HospitalZhengzhouChina
| | - Guansong Wang
- Institute of Respiratory Diseases, Xinqiao HospitalThird Military Medical UniversityChongqingChina
| | - Zhuan Hong
- Department of Medical Oncology, Jiangsu Cancer HospitalNanjing Medical University Affiliated Cancer HospitalNanjingChina
| | - Jiandong Wang
- Department of Pathology, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Meiyu Fang
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw HospitalZhejiang UniversityHangzhouChina
| | - Yiping Zhang
- Department of ChemotherapyChinese Academy of Sciences University Cancer Hospital (Zhejiang Cancer Hospital)HangzhouChina
| | - Yong Song
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingChina
| | - Shenglin Ma
- Department of Oncology, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou Cancer Hospital, Cancer CenterZhejiang University School of MedicineHangzhouChina
| | - Wenfeng Fang
- Department of Medical Oncology, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Yuanzhi Lu
- Department of Clinical PathologyThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| |
Collapse
|
5
|
Sobahy TM, Tashkandi G, Bahussain D, Al-Harbi R. Clinically actionable cancer somatic variants (CACSV): a tumor interpreted dataset for analytical workflows. BMC Med Genomics 2022; 15:95. [PMID: 35468810 PMCID: PMC9036759 DOI: 10.1186/s12920-022-01235-7] [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: 01/20/2021] [Accepted: 04/12/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The recent development and enormous application of parallel sequencing technology in oncology has produced immense amounts of cell-specific genetic information. However, publicly available cell-specific genetic variants are not explained by well-established guidelines. Additionally, cell-specific variants interpretation and classification has remained a challenging task and lacks standardization. The Association for Molecular Pathology (AMP), the American Society of Clinical Oncology (ASCO), and the College of American Pathologists (CAP) published the first consensus guidelines for cell-specific variants cataloging and clinical annotations. METHODS AMP-ASCO-CAP recommended sources and information were downloaded and used as follows: relative knowledge in oncology clinical practice guidelines; approved, investigative or preclinical drugs; supporting literature and each gene-tumor site correlation. All information was homogenized into a single knowledgebase. Finally, we incorporated the consensus recommendations into a new computational method. RESULTS A subset of cancer genetic variants was manually curated to benchmark our method and well-known computational algorithms. We applied the new method on freely available tumor-specific databases to produce a clinically actionable cancer somatic variants (CACSV) dataset in an easy-to-integrate format for most clinical analytical workflows. The research also showed the current challenges and limitations of using different classification systems or computational methods. CONCLUSION CACSV is a step toward cell-specific genetic variants standardized interpretation as it is readily adaptable by most clinical laboratory pipelines for somatic variants clinical annotations. CACSV is freely accessible at ( https://github.com/tsobahytm/CACSV/tree/main/dataset ).
Collapse
Affiliation(s)
- Turki M. Sobahy
- grid.415310.20000 0001 2191 4301King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Research Center, Jeddah, 21499 Kingdom of Saudi Arabia
| | - Ghassan Tashkandi
- grid.415310.20000 0001 2191 4301King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Research Center, Jeddah, 21499 Kingdom of Saudi Arabia
| | - Donya Bahussain
- grid.415310.20000 0001 2191 4301King Faisal Specialist Hospital & Research Center-Jeddah (KFSHRC-J), Research Center, Jeddah, 21499 Kingdom of Saudi Arabia
| | - Raneem Al-Harbi
- grid.412125.10000 0001 0619 1117Genetic Medicine Department, College of Medicine, King Abdulaziz University (KAU), Jeddah, 7393 Kingdom of Saudi Arabia
| |
Collapse
|
6
|
Consensus for HER2 alterations testing in non-small-cell lung cancer. ESMO Open 2022; 7:100395. [PMID: 35149428 PMCID: PMC8844658 DOI: 10.1016/j.esmoop.2022.100395] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/27/2021] [Accepted: 01/06/2022] [Indexed: 02/08/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2) is a transmembrane glycoprotein receptor with intracellular tyrosine kinase activity. Its alterations, including mutation, amplification and overexpression, could result in oncogenic potential and have been detected in many cancers such as non-small-cell lung cancer (NSCLC). Such alterations are, in general, considered markers of poor prognosis. Anti-HER2 antibody-drug conjugates, e.g. trastuzumab deruxtecan (T-DXd, DS-8201) and disitamab vedotin (RC48), were recently approved for HER2-positive breast and gastric cancers. Meanwhile, several HER2-targeted drugs, such as T-DXd, neratinib, afatinib, poziotinib and pyrotinib, have been evaluated in patients with advanced NSCLC, with several of them demonstrating clinical benefit. Therefore, identifying HER2 alterations is pivotal for NSCLC patients to benefit from these targeted therapies. Recent guidelines on HER2 testing were developed for breast and gastric cancer, however, and have not been fully established for NSCLC. The expert group here reached a consensus on HER2 alteration testing in NSCLC with the focus on clinicopathologic characteristics, therapies, detection methods and diagnostic criteria for HER2-altered NSCLC patients. We hope this consensus could improve the clinical management of NSCLC patients with HER2 alterations. Human epidermal growth factor receptor 2 (HER2) alterations lead to poor prognosis in non-small-cell lung cancer (NSCLC). Identifying HER2 alterations is pivotal to guide the anti-HER2-targeted therapies in NSCLC. The requirements for HER2 mutation, amplification or expression testing are distinct in NSCLC. This consensus fills the gap in the criteria for HER2 alteration testing in NSCLC.
Collapse
|
7
|
Masciale V, Banchelli F, Grisendi G, D’Amico R, Maiorana A, Stefani A, Morandi U, Stella F, Dominici M, Aramini B. OUP accepted manuscript. Stem Cells Transl Med 2022; 11:239-247. [PMID: 35356974 PMCID: PMC8968653 DOI: 10.1093/stcltm/szab029] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 11/26/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose Lung cancer relapse may be associated with the presence of a small population of cancer stem cells (CSCs) with unlimited proliferative potential. Our study assessed the relationship between CSCs and the relapse rate in patients harboring adenocarcinoma (ADL) and squamous cell carcinoma of the lung (SCCL). Experimental design This is an observational prospective cohort study (NCT04634630) assessing the influence of CSC frequency on relapse rate after major lung resection in 35 patients harboring early (I-II) (n = 21) and locally advanced (IIIA) (n = 14) ADL and SCCL. There was a 2-year enrollment period followed by a 1-year follow-up period. Surgical tumor specimens were processed, and CSCs were quantified by cytofluorimetric analysis. Results Cancer stem cells were expressed in all patients with a median of 3.1% of the primary cell culture. Primary analysis showed no influence of CSC frequency on the risk of relapse (hazard ratio [HR] = 1.05, 95% confidence interval [CI] = 0.85-1.30). At secondary analysis, patients with locally advanced disease with higher CSC frequency had an increased risk of relapse (HR = 1.26, 95% CI = 1.14-1.39), whereas this was not observed in early-stage patients (HR = 0.90, 95% CI = 0.65-1.25). Conclusion No association was found between CSC and relapse rates after major lung resection in patients harboring ACL and SCCL. However, in locally advanced-stage patients, a positive correlation was observed between CSC frequency and risk of relapse. These results indicate a need for further molecular investigations into the prognostic role of CSCs at different lung cancer stages. Clinical Trial Registration NCT04634630.
Collapse
Affiliation(s)
- Valentina Masciale
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Federico Banchelli
- Center of Medical Statistic, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Roberto D’Amico
- Center of Medical Statistic, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonino Maiorana
- Institute of Pathology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Alessandro Stefani
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Uliano Morandi
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Franco Stella
- Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine—DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni—L. Pierantoni Hospital, Forlì, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine—DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni—L. Pierantoni Hospital, Forlì, Italy
- Corresponding author: Beatrice Aramini, Division of Thoracic Surgery, Department of Experimental, Diagnostic and Specialty Medicine - DIMES of the Alma Mater Studiorum, University of Bologna, G.B. Morgagni - L. Pierantoni Hospital, 34 Carlo Forlanini Street, 47121 Forlì, Italy Forlì, Italy.
| |
Collapse
|
8
|
Oversoe SK, Sorensen BS, Tabaksblat EM, Gronbaek H, Kelsen J. Cell-Free DNA and Clinical Characteristics in Patients with Small Intestinal or Pancreatic Neuroendocrine Tumors. Neuroendocrinology 2022; 112:43-50. [PMID: 33461190 DOI: 10.1159/000514457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 01/15/2021] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Neuroendocrine tumors (NETs) are rare and characterized by a heterogeneous clinical course and an unmet need for better prognostic markers. Plasma cell-free DNA (cfDNA) has prognostic value in other malignancies but is not previously investigated in NETs. We studied cfDNA levels in patients with mainly low-grade small intestinal NET -(siNET) or pancreatic NET (pNET) and evaluated the prognostic potential of cfDNA. MATERIALS AND METHODS We included 70 NET patients, siNET (n = 50) and pNET (n = 20). Plasma cfDNA levels were determined by droplet digital PCR for the beta-2-microglobulin gene every 6 months during a period of 3 years, including in a subgroup of 19 patients during peptide receptor radionuclide therapy (PRRT) therapy. RESULTS cfDNA levels were higher in both siNET and pNET compared to a previously established healthy cohort (p < 0.0001). -cfDNA levels did not predict overall survival (crude hazard ratio [HR] 0.95 [0.57-1.58], p = 0.837, adjusted for smoking status HR 0.77 [0.51-1.17], p = 0.22). The impact of cfDNA level on progression-free survival showed different trends in siNET and pNET. There was no effect of PRRT treatment on cfDNA levels and no difference in cfDNA levels between patients with and without progressive disease after PRRT (ANOVA p = 0.66). cfDNA levels were significantly higher in never-smokers and previous smokers than in current smokers (p = 0.029). DISCUSSION/CONCLUSION cfDNA levels are higher in NET patients than in healthy controls; however, there was no association with prognosis, and cfDNA levels were unaffected by PRRT. Our observations suggest that cfDNA levels are not associated with the disease course in low-grade NET in contrast to other malignancies.
Collapse
Affiliation(s)
- Stine Karlsen Oversoe
- Department of Hepatology and Gastroenterology, Denmark, Aarhus University Hospital, Aarhus, Denmark
- Department of Internal Medicine, Randers Regional Hospital, Randers, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | | | - Henning Gronbaek
- Department of Hepatology and Gastroenterology, Denmark, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Kelsen
- Department of Hepatology and Gastroenterology, Denmark, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
9
|
Fernandes MGO, Cruz-Martins N, Machado JC, Costa JL, Hespanhol V. The value of cell-free circulating tumour DNA profiling in advanced non-small cell lung cancer (NSCLC) management. Cancer Cell Int 2021; 21:675. [PMID: 34915883 PMCID: PMC8680243 DOI: 10.1186/s12935-021-02382-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 11/30/2021] [Indexed: 01/04/2023] Open
Abstract
AbstractLiquid biopsy (LB) has boosted a remarkable change in the management of cancer patients by contributing to tumour genomic profiling. Plasma circulating cell-free tumour DNA (ctDNA) is the most widely searched tumour-related element for clinical application. Specifically, for patients with lung cancer, LB has revealed valuable to detect the diversity of targetable genomic alterations and to detect and monitor the emergence of resistance mechanisms. Furthermore, its non-invasive nature helps to overcome the difficulty in obtaining tissue samples, offering a comprehensive view about tumour diversity. However, the use of the LB to support diagnostic and therapeutic decisions still needs further clarification. In this sense, this review aims to provide a critical view of the clinical importance of plasma ctDNA analysis, the most widely applied LB, and its limitations while anticipating concepts that will intersect the present and future of LB in non-small cell lung cancer patients.
Graphical Abstract
Collapse
|
10
|
Zheng Y, Vioix H, Liu FX, Singh B, Sharma S, Sharda D. Diagnostic and economic value of biomarker testing for targetable mutations in non-small-cell lung cancer: a literature review. Future Oncol 2021; 18:505-518. [PMID: 34865516 DOI: 10.2217/fon-2021-1040] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
We aimed to assess the diagnostic and economic value of next-generation sequencing (NGS) versus single-gene testing, and of liquid biopsy (LBx) versus tissue biopsy (TBx) in non-small-cell lung cancer biomarker testing through literature review. Embase and MEDLINE were searched to identify relevant studies (n = 43) from 2015 to 2020 in adults with advanced non-small-cell lung cancer. For NGS versus single-gene testing, concordance was 70-99% and sensitivity was 86-100%. For LBx versus TBx, specificity was 43-100% and sensitivity was ≥60%. Turnaround times were longer for NGS versus single-gene testing (but not vs sequential testing) and faster for LBx versus TBx. NGS was cost-effective, and LBx reduced US per-patient costs. NGS versus single-gene testing and LBx versus TBx were concordant. NGS and LBx may be cost-effective for initial screening.
Collapse
Affiliation(s)
- Ying Zheng
- US Health Economics and Outcomes Research, EMD Serono, Inc., An affiliate of Merck KGaA, Rockland, MA 02370, USA
| | - Helene Vioix
- Global Evidence & Value Development, Merck Healthcare KGaA, Darmstadt, Germany
| | - Frank X Liu
- US Health Economics and Outcomes Research, EMD Serono, Inc., An affiliate of Merck KGaA, Rockland, MA 02370, USA
| | | | - Sakshi Sharma
- HEOR, Parexel, Access Consulting, Mohali, Punjab, India
| | - Deepti Sharda
- HEOR, Parexel, Access Consulting, Mohali, Punjab, India
| |
Collapse
|
11
|
Torres GF, Bonilla CE, Buitrago G, Arrieta O, Malapelle U, Rolfo C, Cardona AF. How clinically useful is comprehensive genomic profiling for patients with non-small cell lung cancer? A systematic review. Crit Rev Oncol Hematol 2021; 166:103459. [PMID: 34461270 DOI: 10.1016/j.critrevonc.2021.103459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 08/02/2021] [Accepted: 08/22/2021] [Indexed: 12/18/2022] Open
Abstract
Given the lack of a gold standard, the clinical usefulness of Comprehensive Genomic Profiling (CGP) has not been established. This systematic review aimed to evaluate evidence about the clinical benefit of CGP for patients with Non-small cell lung carcinoma (NSCLC). All controlled studies that evaluated the ability of CGP to detect actionable targets (ATs) reported increases in the number of samples with ATs. The frequency of ATs detected in uncontrolled case series ranged from 0.7 % for RET mutations to 45 % for EGFR mutations. The studies that evaluated therapies targeted to EGFR, ALK, ROS-1, MET, and RET mutations documented significant improvement in clinical outcomes. This review suggests that CGP tests may be clinically helpful for treating patients with NSCLC. Although current evidence is associated with a high risk of bias, the significant impact of NSCLC on individuals and society may justify the routine use of CGP testing for this disease.
Collapse
Affiliation(s)
| | - Carlos Eduardo Bonilla
- Clinical Oncology Department, Instituto Nacional de Cancerología - INC, Bogotá, Colombia
| | - Giancarlo Buitrago
- Instituto de Investigaciones Clínicas, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Oscar Arrieta
- Thoracic Oncology Unit, National Cancer Institute (INCan), México City, Mexico
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Center, Mount Sinai Hospital System & Icahn School of Medicine, Mount Sinai, New York, NY, USA
| | - Andrés F Cardona
- Foundation for Clinical and Applied Cancer Research (FICMAC), Bogotá, Colombia; Molecular Oncology and Biology Systems Research Group (Fox-G/ONCOLGroup), Universidad el Bosque, Bogotá, Colombia; Clinical and Traslational Oncology Group, Clínica del Country, Bogotá, Colombia
| |
Collapse
|
12
|
Dameri M, Ferrando L, Cirmena G, Vernieri C, Pruneri G, Ballestrero A, Zoppoli G. Multi-Gene Testing Overview with a Clinical Perspective in Metastatic Triple-Negative Breast Cancer. Int J Mol Sci 2021; 22:7154. [PMID: 34281208 PMCID: PMC8268401 DOI: 10.3390/ijms22137154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
Next-generation sequencing (NGS) is the technology of choice for the routine screening of tumor samples in clinical practice. In this setting, the targeted sequencing of a restricted number of clinically relevant genes represents the most practical option when looking for genetic variants associated with cancer, as well as for the choice of targeted treatments. In this review, we analyze available NGS platforms and clinical applications of multi-gene testing in breast cancer, with a focus on metastatic triple-negative breast cancer (mTNBC). We make an overview of the clinical utility of multi-gene testing in mTNBC, and then, as immunotherapy is emerging as a possible targeted therapy for mTNBC, we also briefly report on the results of the latest clinical trials involving immune checkpoint inhibitors (ICIs) and TNBC, where NGS could play a role for the potential predictive utility of homologous recombination repair deficiency (HRD) and tumor mutational burden (TMB).
Collapse
Affiliation(s)
- Martina Dameri
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (M.D.); (L.F.); (G.C.); (A.B.)
| | - Lorenzo Ferrando
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (M.D.); (L.F.); (G.C.); (A.B.)
| | - Gabriella Cirmena
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (M.D.); (L.F.); (G.C.); (A.B.)
| | - Claudio Vernieri
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
- IFOM, The FIRC Institute of Molecular Oncology, 20139 Milan, Italy
| | - Giancarlo Pruneri
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy;
- School of Medicine, University of Milan, 20122 Milan, Italy
| | - Alberto Ballestrero
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (M.D.); (L.F.); (G.C.); (A.B.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Gabriele Zoppoli
- Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy; (M.D.); (L.F.); (G.C.); (A.B.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy
| |
Collapse
|
13
|
Clinical Application of Next-Generation Sequencing of Plasma Cell-Free DNA for Genotyping Untreated Advanced Non-Small Cell Lung Cancer. Cancers (Basel) 2021; 13:cancers13112707. [PMID: 34070940 PMCID: PMC8199488 DOI: 10.3390/cancers13112707] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Plasma ctDNA is a material source for molecular analysis particularly useful when tissue is not available or sufficient. NGS-based plasma genotyping should be integrated into the clinical workup of newly diagnosed advanced NSCLC. Abstract Background: Analysis of circulating tumor DNA (ctDNA) has remarkable potential as a non-invasive lung cancer molecular diagnostic method. This prospective study addressed the clinical value of a targeted-gene amplicon-based plasma next-generation sequencing (NGS) assay to detect actionable mutations in ctDNA in patients with newly diagnosed advanced lung adenocarcinoma. Methods: ctDNA test performance and concordance with tissue NGS were determined, and the correlation between ctDNA findings, clinical features, and clinical outcomes was evaluated in 115 patients with paired plasma and tissue samples. Results: Targeted-gene NGS-based ctDNA and NGS-based tissue analysis detected 54 and 63 genomic alterations, respectively; 11 patients presented co-mutations, totalizing 66 hotspot mutations detected, 51 on both tissue and plasma, 12 exclusively on tissue, and 3 exclusively on plasma. NGS-based ctDNA revealed a diagnostic performance with 81.0% sensitivity, 95.3% specificity, 94.4% PPV, 83.6% NPV, test accuracy of 88.2%, and Cohen’s Kappa 0.764. PFS and OS assessed by both assays did not significantly differ. Detection of ctDNA alterations was statistically associated with metastatic disease (p = 0.013), extra-thoracic metastasis (p = 0.004) and the number of organs involved (p = 0.010). Conclusions: This study highlights the potential use of ctDNA for mutation detection in newly diagnosed NSCLC patients due to its high accuracy and correlation with clinical outcomes.
Collapse
|
14
|
Asada K, Kaneko S, Takasawa K, Machino H, Takahashi S, Shinkai N, Shimoyama R, Komatsu M, Hamamoto R. Integrated Analysis of Whole Genome and Epigenome Data Using Machine Learning Technology: Toward the Establishment of Precision Oncology. Front Oncol 2021; 11:666937. [PMID: 34055633 PMCID: PMC8149908 DOI: 10.3389/fonc.2021.666937] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 04/26/2021] [Indexed: 12/17/2022] Open
Abstract
With the completion of the International Human Genome Project, we have entered what is known as the post-genome era, and efforts to apply genomic information to medicine have become more active. In particular, with the announcement of the Precision Medicine Initiative by U.S. President Barack Obama in his State of the Union address at the beginning of 2015, "precision medicine," which aims to divide patients and potential patients into subgroups with respect to disease susceptibility, has become the focus of worldwide attention. The field of oncology is also actively adopting the precision oncology approach, which is based on molecular profiling, such as genomic information, to select the appropriate treatment. However, the current precision oncology is dominated by a method called targeted-gene panel (TGP), which uses next-generation sequencing (NGS) to analyze a limited number of specific cancer-related genes and suggest optimal treatments, but this method causes the problem that the number of patients who benefit from it is limited. In order to steadily develop precision oncology, it is necessary to integrate and analyze more detailed omics data, such as whole genome data and epigenome data. On the other hand, with the advancement of analysis technologies such as NGS, the amount of data obtained by omics analysis has become enormous, and artificial intelligence (AI) technologies, mainly machine learning (ML) technologies, are being actively used to make more efficient and accurate predictions. In this review, we will focus on whole genome sequencing (WGS) analysis and epigenome analysis, introduce the latest results of omics analysis using ML technologies for the development of precision oncology, and discuss the future prospects.
Collapse
Affiliation(s)
- Ken Asada
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Syuzo Kaneko
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Ken Takasawa
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Hidenori Machino
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Satoshi Takahashi
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Norio Shinkai
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ryo Shimoyama
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Masaaki Komatsu
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Ryuji Hamamoto
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
15
|
Zhang T, Xu Z, Liu G, Jiang B, de Bock GH, Groen HJM, Vliegenthart R, Xie X. Simultaneous Identification of EGFR,KRAS,ERBB2, and TP53 Mutations in Patients with Non-Small Cell Lung Cancer by Machine Learning-Derived Three-Dimensional Radiomics. Cancers (Basel) 2021; 13:1814. [PMID: 33920322 PMCID: PMC8070114 DOI: 10.3390/cancers13081814] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/25/2022] Open
Abstract
PURPOSE To develop a machine learning-derived radiomics approach to simultaneously discriminate epidermal growth factor receptor (EGFR), Kirsten rat sarcoma viral oncogene (KRAS), Erb-B2 receptor tyrosine kinase 2 (ERBB2), and tumor protein 53 (TP53) genetic mutations in patients with non-small cell lung cancer (NSCLC). METHODS This study included consecutive patients from April 2018 to June 2020 who had histologically confirmed NSCLC, and underwent pre-surgical contrast-enhanced CT and post-surgical next-generation sequencing (NGS) tests to determine the presence of EGFR, KRAS, ERBB2, and TP53 mutations. A dedicated radiomics analysis package extracted 1672 radiomic features in three dimensions. Discriminative models were established using the least absolute shrinkage and selection operator to determine the presence of EGFR, KRAS, ERBB2, and TP53 mutations, based on radiomic features and relevant clinical factors. RESULTS In 134 patients (63.6 ± 8.9 years), the 20 most relevant radiomic features (13 for KRAS) to mutations were selected to construct models. The areas under the curve (AUCs) of the combined model (radiomic features and relevant clinical factors) for discriminating EGFR, KRAS, ERBB2, and TP53 mutations were 0.78 (95% CI: 0.70-0.86), 0.81 (0.69-0.93), 0.87 (0.78-0.95), and 0.84 (0.78-0.91), respectively. In particular, the specificity to exclude EGFR mutations was 0.96 (0.87-0.99). The sensitivity to determine KRAS, ERBB2, and TP53 mutations ranged from 0.82 (0.69-90) to 0.92 (0.62-0.99). CONCLUSIONS Machine learning-derived 3D radiomics can simultaneously discriminate the presence of EGFR, KRAS, ERBB2, and TP53 mutations in patients with NSCLC. This noninvasive and low-cost approach may be helpful in screening patients before invasive sampling and NGS testing.
Collapse
Affiliation(s)
- Tiening Zhang
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China;
| | - Zhihan Xu
- Siemens Healthineers Ltd., Zhouzhu Rd.278, Shanghai 200120, China;
| | - Guixue Liu
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China; (G.L.); (B.J.)
| | - Beibei Jiang
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China; (G.L.); (B.J.)
| | - Geertruida H. de Bock
- Department of Epidemiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ Groningen, The Netherlands;
| | - Harry J. M. Groen
- Department of Lung Diseases, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands;
| | - Rozemarijn Vliegenthart
- Department of Radiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9700 RB Groningen, The Netherlands;
| | - Xueqian Xie
- Department of Radiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Haining Rd.100, Shanghai 200080, China; (G.L.); (B.J.)
| |
Collapse
|
16
|
Dealing with NSCLC EGFR mutation testing and treatment: A comprehensive review with an Italian real-world perspective. Crit Rev Oncol Hematol 2021; 160:103300. [PMID: 33744362 DOI: 10.1016/j.critrevonc.2021.103300] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 01/12/2023] Open
Abstract
Since their discovery, relevant efforts have been made to optimize the detection approaches to EGFR mutations as well as the clinical management of EGFR-mutated NSCLC. The recent shift from single gene testing to novel comprehensive detection platforms along with the development of new generation tyrosine kinase inhibitors, targeting both common and uncommon EGFR-mutations, is leading to a progressive increase in the number of patients who may benefit from targeted approaches, with subsequent impact on their long-term survival and quality of life. However, a prompt and adequate implementation of the most recent diagnostic and treatment advances in the routine practice often remains critical to be specifically addressed. In this review we provide a complete and updated overview of the different detection platforms and therapeutic options currently available for the clinical management of advanced EGFR-positive NSCLC, summarizing scientific evidence and describing molecular testing as well as treatment practice in the real-word scenario.
Collapse
|
17
|
Belardinilli F, Pernazza A, Mahdavian Y, Cerbelli B, Bassi M, Gradilone A, Coppa A, Pignataro MG, Anile M, Venuta F, Della Rocca C, Giannini G, d'Amati G. A multidisciplinary approach for the differential diagnosis between multiple primary lung adenocarcinomas and intrapulmonary metastases. Pathol Res Pract 2021; 220:153387. [PMID: 33647865 DOI: 10.1016/j.prp.2021.153387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/13/2021] [Indexed: 11/26/2022]
Abstract
PURPOSE The distinction between multiple primary lung cancers (MPLCs) and intrapulmonary metastases has a significant impact on tumor staging and therapeutic choices. Several criteria have been proposed to solve this diagnostic issue, but a definitive consensus is still missing. We tested the efficacy of a combined clinical, histopathological and molecular ("real world") approach for the correct classification of multiple lung tumors in a selected cohort of patients. METHODS 24 multiple lung tumors with a diagnosis of adenocarcinoma from 10 patients were retrospectively reviewed. Radiological, pathological and clinical information, including follow-up, were integrated with molecular profiling via a routine multigene panel sequencing. RESULTS Comprehensive histologic assessment revealed readily distinguishable histologic patterns between multiple tumors suggesting unrelated lesions in 7 cases, in agreement with clinical, radiological and molecular data, thus leading to final diagnosis of MPLCs. In the remaining 3 cases, the differential diagnosis between MPLCs and intrapulmonary metastases was challenging, since the histologic features of the lesions were similar or identical. The final interpretation (2 MPLCs and 1 most likely intrapulmonary metastases) was reached thanks to the integration of all available data, and was confirmed by follow-up. CONCLUSIONS A multidisciplinary approach including a routinely affordable multigene panel sequencing is a useful tool to discriminate MPLCs from intrapulmonary metastases in multiple lung nodules sharing the adenocarcinoma histotype.
Collapse
Affiliation(s)
- Francesca Belardinilli
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy
| | - Angelina Pernazza
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Latina, 04100, Italy
| | - Yasaman Mahdavian
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy
| | - Bruna Cerbelli
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Latina, 04100, Italy
| | - Massimiliano Bassi
- Department of General and Specialist Surgery "P. Stefanini" Sapienza University, Rome, 00161, Italy
| | - Angela Gradilone
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy
| | | | - Maria Gemma Pignataro
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University, Rome, 00161, Italy
| | - Marco Anile
- Department of General and Specialist Surgery "P. Stefanini" Sapienza University, Rome, 00161, Italy
| | - Federico Venuta
- Department of General and Specialist Surgery "P. Stefanini" Sapienza University, Rome, 00161, Italy
| | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University, Latina, 04100, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161, Rome, Italy.
| | - Giulia d'Amati
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University, Rome, 00161, Italy.
| |
Collapse
|
18
|
How the COVID-19 Pandemic Impacted Oncological Molecular Diagnosis: A Picture from a National Reference Center for Molecular Pathology. BIOMED RESEARCH INTERNATIONAL 2020; 2020:8397053. [PMID: 33029526 PMCID: PMC7537698 DOI: 10.1155/2020/8397053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 09/17/2020] [Indexed: 01/06/2023]
Abstract
Introduction The Portuguese healthcare system had to adapt at short notice to the COVID-19 pandemic. We implemented workflow changes to our molecular pathology laboratory, a national reference center, to maximize safety and productivity. We assess the impact this situation had on our caseload and what conclusions can be drawn about the wider impact of the pandemic in oncological therapy in Portugal. Material and Methods. We reviewed our database for all oncological molecular tests requested between March and April of 2019 and 2020. For each case, we recorded age, sex, region of the country, requesting institution, sample type, testing method, and turnaround time (TAT). A comparison between years was made. Results The total number of tests decreased from 421 in 2019 to 319 in 2020 (p = 0.0027). The greatest reduction was in clinical trial-related cases. Routine cases were similar between years (267 vs. 256). TAT was higher in 2019 (mean 15 days vs. 12.3 days; p = 0.0003). Medium- to large-sized public hospitals in the north of the country were mostly responsible for the reduction in cases (p = 0.0153). Conclusions Case reduction was observed at hospitals that have mostly been involved in the treatment of COVID-19 and in the north of the country, the region worst-hit by the pandemic. Similar to other studies, our TAT decreased, even with a similar number of routine cases. Thus, we conclude that it is possible to successfully adapt the workflow of a molecular pathology laboratory to new safety standards without losing efficiency.
Collapse
|
19
|
Dlamini Z, Francies FZ, Hull R, Marima R. Artificial intelligence (AI) and big data in cancer and precision oncology. Comput Struct Biotechnol J 2020; 18:2300-2311. [PMID: 32994889 PMCID: PMC7490765 DOI: 10.1016/j.csbj.2020.08.019] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 08/21/2020] [Accepted: 08/21/2020] [Indexed: 02/07/2023] Open
Abstract
Artificial intelligence (AI) and machine learning have significantly influenced many facets of the healthcare sector. Advancement in technology has paved the way for analysis of big datasets in a cost- and time-effective manner. Clinical oncology and research are reaping the benefits of AI. The burden of cancer is a global phenomenon. Efforts to reduce mortality rates requires early diagnosis for effective therapeutic interventions. However, metastatic and recurrent cancers evolve and acquire drug resistance. It is imperative to detect novel biomarkers that induce drug resistance and identify therapeutic targets to enhance treatment regimes. The introduction of the next generation sequencing (NGS) platforms address these demands, has revolutionised the future of precision oncology. NGS offers several clinical applications that are important for risk predictor, early detection of disease, diagnosis by sequencing and medical imaging, accurate prognosis, biomarker identification and identification of therapeutic targets for novel drug discovery. NGS generates large datasets that demand specialised bioinformatics resources to analyse the data that is relevant and clinically significant. Through these applications of AI, cancer diagnostics and prognostic prediction are enhanced with NGS and medical imaging that delivers high resolution images. Regardless of the improvements in technology, AI has some challenges and limitations, and the clinical application of NGS remains to be validated. By continuing to enhance the progression of innovation and technology, the future of AI and precision oncology show great promise.
Collapse
Affiliation(s)
- Zodwa Dlamini
- SAMRC/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Extramural Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Faculty of Health Sciences, Hatfield 0028, South Africa
| | - Flavia Zita Francies
- SAMRC/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Extramural Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Faculty of Health Sciences, Hatfield 0028, South Africa
| | - Rodney Hull
- SAMRC/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Extramural Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Faculty of Health Sciences, Hatfield 0028, South Africa
| | - Rahaba Marima
- SAMRC/UP Precision Prevention & Novel Drug Targets for HIV-Associated Cancers (PPNDTHAC) Extramural Unit, Pan African Cancer Research Institute (PACRI), University of Pretoria, Faculty of Health Sciences, Hatfield 0028, South Africa
| |
Collapse
|
20
|
Luo R, Song J, Xiao X, Xie Z, Zhao Z, Zhang W, Miao S, Tang Y, Ran L. Identifying CpG methylation signature as a promising biomarker for recurrence and immunotherapy in non-small-cell lung carcinoma. Aging (Albany NY) 2020; 12:14649-14676. [PMID: 32723974 PMCID: PMC7425482 DOI: 10.18632/aging.103517] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 06/04/2020] [Indexed: 12/15/2022]
Abstract
Epigenetic alterations are crucial to oncogenesis and regulation of gene expression in non-small-cell lung carcinoma (NSCLC). DNA methylation (DNAm) biomarkers may provide molecular-level prediction of relapse risk in cancer. Identification of optimal treatment is warranted for improving clinical management of NSCLC patients. Using machine learning algorithm we identified 4 recurrence predictive CpG methylation markers (cg00253681/ART4, cg00111503/KCNK9, cg02715629/FAM83A, cg03282991/C6orf10) and constructed a risk score model that potently predicted recurrence-free survival and prognosis for patients with NSCLC (P = 0.0002). Integrating genomic, transcriptomic, proteomic and clinical data, the DNAm-based risk score was observed to significantly associate with clinical stage, cell proliferation markers, somatic alterations, tumor mutation burden (TMB) as well as DNA damage response (DDR) genes, and potentially predict the efficacy of immunotherapy. In general, our identified DNAm signature shows a significant correlation to TMB and DDR pathways, and serves as an effective biomarker for predicting NSCLC recurrence and response to immunotherapy. These findings demonstrate the utility of 4-DNAm-marker panel in the prognosis, treatment decision-making and evaluation of therapeutic responses for NSCLC.
Collapse
Affiliation(s)
- Ruihan Luo
- Department of Bioinformatics, The Basic Medical School of Chongqing Medical University, Chongqing, China
| | - Jing Song
- Department of Bioinformatics, The Basic Medical School of Chongqing Medical University, Chongqing, China
| | - Xiao Xiao
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengbo Xie
- Information Center Department, Chongqing Medical University, Chongqing, China
| | - Zhiyuan Zhao
- Information Center Department, Chongqing Medical University, Chongqing, China
| | - Wanfeng Zhang
- Department of Bioinformatics, The Basic Medical School of Chongqing Medical University, Chongqing, China
| | - Shiqi Miao
- Department of Bioinformatics, The Basic Medical School of Chongqing Medical University, Chongqing, China
| | - Yongyao Tang
- Molecular and Tumor Research Center, Chongqing Medical University, Chongqing, China
| | - Longke Ran
- Department of Bioinformatics, The Basic Medical School of Chongqing Medical University, Chongqing, China
| |
Collapse
|
21
|
Wadowska K, Bil-Lula I, Trembecki Ł, Śliwińska-Mossoń M. Genetic Markers in Lung Cancer Diagnosis: A Review. Int J Mol Sci 2020; 21:4569. [PMID: 32604993 PMCID: PMC7369725 DOI: 10.3390/ijms21134569] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/19/2020] [Accepted: 06/25/2020] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is the most often diagnosed cancer in the world and the most frequent cause of cancer death. The prognosis for lung cancer is relatively poor and 75% of patients are diagnosed at its advanced stage. The currently used diagnostic tools are not sensitive enough and do not enable diagnosis at the early stage of the disease. Therefore, searching for new methods of early and accurate diagnosis of lung cancer is crucial for its effective treatment. Lung cancer is the result of multistage carcinogenesis with gradually increasing genetic and epigenetic changes. Screening for the characteristic genetic markers could enable the diagnosis of lung cancer at its early stage. The aim of this review was the summarization of both the preclinical and clinical approaches in the genetic diagnostics of lung cancer. The advancement of molecular strategies and analytic platforms makes it possible to analyze the genome changes leading to cancer development-i.e., the potential biomarkers of lung cancer. In the reviewed studies, the diagnostic values of microsatellite changes, DNA hypermethylation, and p53 and KRAS gene mutations, as well as microRNAs expression, have been analyzed as potential genetic markers. It seems that microRNAs and their expression profiles have the greatest diagnostic potential value in lung cancer diagnosis, but their quantification requires standardization.
Collapse
Affiliation(s)
- Katarzyna Wadowska
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (I.B.-L.)
| | - Iwona Bil-Lula
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (I.B.-L.)
| | - Łukasz Trembecki
- Department of Radiation Oncology, Lower Silesian Oncology Center, 53-413 Wroclaw, Poland;
- Department of Oncology, Faculty of Medicine, Wroclaw Medical University, 53-413 Wroclaw, Poland
| | - Mariola Śliwińska-Mossoń
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Haematology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (I.B.-L.)
| |
Collapse
|
22
|
Tsao MS, Yatabe Y. Old Soldiers Never Die: Is There Still a Role for Immunohistochemistry in the Era of Next-Generation Sequencing Panel Testing? J Thorac Oncol 2020; 14:2035-2038. [PMID: 31757371 DOI: 10.1016/j.jtho.2019.09.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Ming Sound Tsao
- Laboratory Medicine Program, Department of Pathology, University Health Network, Princess Margaret Cancer Centre and Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center, Tokyo, Japan
| |
Collapse
|