1
|
Stella GM, Lettieri S, Piloni D, Ferrarotti I, Perrotta F, Corsico AG, Bortolotto C. Smart Sensors and Microtechnologies in the Precision Medicine Approach against Lung Cancer. Pharmaceuticals (Basel) 2023; 16:1042. [PMID: 37513953 PMCID: PMC10385174 DOI: 10.3390/ph16071042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/23/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND AND RATIONALE The therapeutic interventions against lung cancer are currently based on a fully personalized approach to the disease with considerable improvement of patients' outcome. Alongside continuous scientific progresses and research investments, massive technologic efforts, innovative challenges, and consolidated achievements together with research investments are at the bases of the engineering and manufacturing revolution that allows a significant gain in clinical setting. AIM AND METHODS The scope of this review is thus to focus, rather than on the biologic traits, on the analysis of the precision sensors and novel generation materials, as semiconductors, which are below the clinical development of personalized diagnosis and treatment. In this perspective, a careful revision and analysis of the state of the art of the literature and experimental knowledge is presented. RESULTS Novel materials are being used in the development of personalized diagnosis and treatment for lung cancer. Among them, semiconductors are used to analyze volatile cancer compounds and allow early disease diagnosis. Moreover, they can be used to generate MEMS which have found an application in advanced imaging techniques as well as in drug delivery devices. CONCLUSIONS Overall, these issues represent critical issues only partially known and generally underestimated by the clinical community. These novel micro-technology-based biosensing devices, based on the use of molecules at atomic concentrations, are crucial for clinical innovation since they have allowed the recent significant advances in cancer biology deciphering as well as in disease detection and therapy. There is an urgent need to create a stronger dialogue between technologists, basic researchers, and clinicians to address all scientific and manufacturing efforts towards a real improvement in patients' outcome. Here, great attention is focused on their application against lung cancer, from their exploitations in translational research to their application in diagnosis and treatment development, to ensure early diagnosis and better clinical outcomes.
Collapse
Affiliation(s)
- Giulia Maria Stella
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Sara Lettieri
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Davide Piloni
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Ilaria Ferrarotti
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Fabio Perrotta
- Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", 80131 Napoli, Italy
- U.O.C. Clinica Pneumologica "L. Vanvitelli", A.O. dei Colli, Ospedale Monaldi, 80131 Napoli, Italy
| | - Angelo Guido Corsico
- Department of Internal Medicine and Medical Therapeutics, University of Pavia Medical School, 27100 Pavia, Italy
- Cardiothoracic and Vascular Department, Unit of Respiratory Diseases, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Chandra Bortolotto
- Department of Clinical-Surgical, Diagnostic and Paediatric Sciences, University of Pavia Medical School, 27100 Pavia, Italy
- Department of Diagnostic Services and Imaging, Unit of Radiology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| |
Collapse
|
2
|
Predicting oncogene mutations of lung cancer using deep learning and histopathologic features on whole-slide images. Transl Oncol 2022; 24:101494. [PMID: 35905641 PMCID: PMC9334329 DOI: 10.1016/j.tranon.2022.101494] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/29/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022] Open
Abstract
Developed a deep learning model for predicting somatic mutations of LUAD patients. Utilized LUAD subtype-related histological features to predict five major genetic mutations. Investigated multiple transfer learning scenarios to characterize morphological features. Achieved strong performance in predicting EGFR genetic mutations across multiple datasets.
Lung cancer is a leading cause of death in both men and women globally. The recent development of tumor molecular profiling has opened opportunities for targeted therapies for lung adenocarcinoma (LUAD) patients. However, the lack of access to molecular profiling or cost and turnaround time associated with it could hinder oncologists' willingness to order frequent molecular tests, limiting potential benefits from precision medicine. In this study, we developed a weakly supervised deep learning model for predicting somatic mutations of LUAD patients based on formalin-fixed paraffin-embedded (FFPE) whole-slide images (WSIs) using LUAD subtypes-related histological features and recent advances in computer vision. Our study was performed on a total of 747 hematoxylin and eosin (H&E) stained FFPE LUAD WSIs and the genetic mutation data of 232 patients who were treated at Dartmouth-Hitchcock Medical Center (DHMC). We developed our convolutional neural network-based models to analyze whole slides and predict five major genetic mutations, i.e., BRAF, EGFR, KRAS, STK11, and TP53. We additionally used 111 cases from the LUAD dataset of the CPTAC-3 study for external validation. Our model achieved an AUROC of 0.799 (95% CI: 0.686–0.904) and 0.686 (95% CI: 0.620–0.752) for predicting EGFR genetic mutations on the DHMC and CPTAC-3 test sets, respectively. Predicting TP53 genetic mutations also showed promising outcomes. Our results demonstrated that H&E stained FFPE LUAD whole slides could be utilized to predict oncogene mutations, such as EGFR, indicating that somatic mutations could present subtle morphological characteristics in histology slides, where deep learning-based feature extractors can learn such latent information.
Collapse
|
3
|
Bauml JM, Li BT, Velcheti V, Govindan R, Curioni-Fontecedro A, Dooms C, Takahashi T, Duda AW, Odegaard JI, Cruz-Guilloty F, Jin L, Zhang Y, Anderson A, Skoulidis F. Clinical validation of Guardant360 CDx as a blood-based companion diagnostic for sotorasib. Lung Cancer 2022; 166:270-278. [PMID: 34838325 PMCID: PMC10325630 DOI: 10.1016/j.lungcan.2021.10.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/17/2021] [Accepted: 10/14/2021] [Indexed: 01/22/2023]
Abstract
OBJECTIVES Effective therapy for non-small-cell lung cancer (NSCLC) depends on morphological and genomic classification, with comprehensive screening for guideline-recommended biomarkers critical to guide treatment. Companion diagnostics, which provide robust genotyping results, represent an important component of personalized oncology. We evaluated the clinical validity of Guardant360 CDx as a companion diagnostic for sotorasib for detection of KRAS p.G12C, an important oncogenic NSCLC driver mutation. MATERIALS AND METHODS KRAS p.G12C was tested in NSCLC patients from CodeBreaK100 (NCT03600833) in pretreatment plasma samples using Guardant360 CDx liquid biopsy and archival tissue samples using therascreen® KRAS RGQ polymerase chain reaction (PCR) kit tissue testing. Matched tissue and plasma samples were procured from other clinical trials or commercial vendors, and results were compared. Demographics and clinical characteristics and objective response rate (ORR) were evaluated. RESULTS Of 126 CodeBreaK patients, 112 (88.9%) were tested for KRASp.G12C mutations with Guardant360 CDx. Among 189 patients in the extended analysis cohort, the positive and negative percent agreement (95% CI) for Guardant360 CDx plasma testing relative to therascreen® KRAS RGQ PCR kit tissue testing were 0.71 (0.62, 0.79) and 1.00 (0.95, 1.00), respectively; overall percent agreement (95% CI) was 0.82 (0.76, 0.87). TP53 co-mutations were the most common regardless of KRAS p.G12C status (KRAS p.G12C-positive, 53.4%; KRAS p.G12C-negative, 45.5%). STK11 was co-mutated in 26.1% of KRAS p.G12C-positive samples. The ORR was similar among patients selected by plasma and tissue testing. CONCLUSION Comprehensive genotyping for all therapeutic targets including KRAS p.G12C is critical for management of NSCLC. Liquid biopsy using Guardant360 CDx has clinical validity for identification of patients with KRASp.G12C-mutant NSCLC and, augmented by tissue testing methodologies as outlined on the approved product label, will identify patients for treatment with sotorasib.
Collapse
Affiliation(s)
| | - Bob T Li
- Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Vamsidhar Velcheti
- NYU Langone - Laura and Isaac Perlmutter Cancer Center, New York, NY, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Abstract
This overview of the molecular pathology of lung cancer includes a review of the most salient molecular alterations of the genome, transcriptome, and the epigenome. The insights provided by the growing use of next-generation sequencing (NGS) in lung cancer will be discussed, and interrelated concepts such as intertumor heterogeneity, intratumor heterogeneity, tumor mutational burden, and the advent of liquid biopsy will be explored. Moreover, this work describes how the evolving field of molecular pathology refines the understanding of different histologic phenotypes of non-small-cell lung cancer (NSCLC) and the underlying biology of small-cell lung cancer. This review will provide an appreciation for how ongoing scientific findings and technologic advances in molecular pathology are crucial for development of biomarkers, therapeutic agents, clinical trials, and ultimately improved patient care.
Collapse
Affiliation(s)
- James J Saller
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Theresa A Boyle
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| |
Collapse
|
5
|
Dang ATH, Tran VU, Tran TT, Thi Pham HA, Le DT, Nguyen L, Nguyen NV, Thi Nguyen TH, Nguyen CV, Le HT, Thi Nguyen ML, Le VT, Nguyen PH, Vo BT, Thi Dao HT, Nguyen LT, Van Nguyen TC, Bui QTN, Nguyen LH, Nguyen NH, Thi Nguyen QT, Le TX, Do TTT, Dinh KT, Do HN, Phan MD, Nguyen HN, Tran LS, Giang H. Actionable Mutation Profiles of Non-Small Cell Lung Cancer patients from Vietnamese population. Sci Rep 2020; 10:2707. [PMID: 32066856 PMCID: PMC7026432 DOI: 10.1038/s41598-020-59744-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
Comprehensive profiling of actionable mutations in non-small cell lung cancer (NSCLC) is vital to guide targeted therapy, thereby improving the survival rate of patients. Despite the high incidence and mortality rate of NSCLC in Vietnam, the actionable mutation profiles of Vietnamese patients have not been thoroughly examined. Here, we employed massively parallel sequencing to identify alterations in major driver genes (EGFR, KRAS, NRAS, BRAF, ALK and ROS1) in 350 Vietnamese NSCLC patients. We showed that the Vietnamese NSCLC patients exhibited mutations most frequently in EGFR (35.4%) and KRAS (22.6%), followed by ALK (6.6%), ROS1 (3.1%), BRAF (2.3%) and NRAS (0.6%). Interestingly, the cohort of Vietnamese patients with advanced adenocarcinoma had higher prevalence of EGFR mutations than the Caucasian MSK-IMPACT cohort. Compared to the East Asian cohort, it had lower EGFR but higher KRAS mutation prevalence. We found that KRAS mutations were more commonly detected in male patients while EGFR mutations was more frequently found in female. Moreover, younger patients (<61 years) had higher genetic rearrangements in ALK or ROS1. In conclusions, our study revealed mutation profiles of 6 driver genes in the largest cohort of NSCLC patients in Vietnam to date, highlighting significant differences in mutation prevalence to other cohorts.
Collapse
Affiliation(s)
- Anh-Thu Huynh Dang
- University of Medicine and Pharmacy at Ho Chi Minh city, Ho Chi Minh city, Vietnam
| | | | | | | | | | - Lam Nguyen
- Pham Ngoc Thach Hospital, Ho Chi Minh city, Vietnam
| | | | | | | | - Ha Thu Le
- Ha Noi Oncology hospital, Ha Noi, Vietnam
| | | | | | - Phuc Huu Nguyen
- University of Medicine and Pharmacy at Ho Chi Minh city, Ho Chi Minh city, Vietnam
| | | | | | | | | | | | | | | | | | - Truong Xuan Le
- University of Medicine and Pharmacy at Ho Chi Minh city, Ho Chi Minh city, Vietnam
| | - Thanh-Thuy Thi Do
- University of Medicine and Pharmacy at Ho Chi Minh city, Ho Chi Minh city, Vietnam
| | | | | | - Minh-Duy Phan
- Gene Solutions, Ho Chi Minh city, Vietnam
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Hoai-Nghia Nguyen
- University of Medicine and Pharmacy at Ho Chi Minh city, Ho Chi Minh city, Vietnam.
| | - Le Son Tran
- Gene Solutions, Ho Chi Minh city, Vietnam.
- Institute of Molecular and Cellular Biology, Astar, Singapore.
| | - Hoa Giang
- Gene Solutions, Ho Chi Minh city, Vietnam.
- Medical Genetics Institute, Ho Chi Minh City, Vietnam.
| |
Collapse
|
6
|
Garg S, Grenier S, Misyura M, Sukhai MA, Thomas M, Kamel-Reid S, Stockley T. Assessing the Diagnostic Yield of Targeted Next-Generation Sequencing for Melanoma and Gastrointestinal Tumors. J Mol Diagn 2020; 22:467-475. [PMID: 32036084 DOI: 10.1016/j.jmoldx.2019.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 11/19/2019] [Accepted: 12/20/2019] [Indexed: 12/21/2022] Open
Abstract
A common rationale in molecular diagnostic laboratories is that implementation of next-generation sequencing (NGS) enables simultaneous multigene testing, allowing increased information benefit compared with non-NGS assays. However, minimal published data exist to support this justification. The current study compared clinical diagnostic yield of TruSight Tumor 26 Sequencing Panel (TST26) in melanoma, colorectal (CRC), and gastrointestinal stromal (GIST) tumors with non-NGS assays. A total of 1041 formalin-fixed, paraffin-embedded tumors, of melanoma, CRC, and GIST, were profiled. NGS results were compared with non-NGS single-gene or single-variant assays with respect to variant output and diagnostic yield. A total of 79% melanoma and 94% CRC tumors were variant positive by panel testing. TST26 panel improved serine/threonine-protein kinase B-raf (BRAF) variant detection in melanoma compared with single-variant BRAF Val600Glu/Lys (V600E/K) routine tests by 24% and detected variants in genes other than BRAF, NRAS, and KIT, which could impact patient management in 20% additional cases. NGS enhanced diagnostic yield in CRC by 36% when compared with routine single-gene assays. In contrast, no added benefit of NGS-based testing for GIST tumors was observed. TST26 panel either missed or inaccurately called complex insertion/deletion variants in KIT exon 11, which were accurately identified by non-NGS methods. Findings of this study demonstrate the differential impact of cancer site and variant type on diagnostic test information yield from NGS assays.
Collapse
Affiliation(s)
- Swati Garg
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario
| | - Sylvie Grenier
- Division of Genome Diagnostics, Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Maksym Misyura
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario
| | - Mahadeo A Sukhai
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario
| | - Mariam Thomas
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario
| | - Suzanne Kamel-Reid
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario; Division of Genome Diagnostics, Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Tracy Stockley
- Advanced Molecular Diagnostics Laboratory, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario; Division of Genome Diagnostics, Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Tran LS, Pham HAT, Tran VU, Tran TT, Dang ATH, Le DT, Nguyen SL, Nguyen NV, Nguyen TV, Vo BT, Dao HTT, Nguyen NH, Tran TH, Nguyen CV, Pham PC, Dang-Mai AT, Dinh-Nguyen TK, Phan VH, Do TTT, Truong Dinh K, Do HN, Phan MD, Giang H, Nguyen HN. Ultra-deep massively parallel sequencing with unique molecular identifier tagging achieves comparable performance to droplet digital PCR for detection and quantification of circulating tumor DNA from lung cancer patients. PLoS One 2019; 14:e0226193. [PMID: 31841547 PMCID: PMC6913927 DOI: 10.1371/journal.pone.0226193] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 11/21/2019] [Indexed: 12/24/2022] Open
Abstract
The identification and quantification of actionable mutations are of critical importance for effective genotype-directed therapies, prognosis and drug response monitoring in patients with non-small-cell lung cancer (NSCLC). Although tumor tissue biopsy remains the gold standard for diagnosis of NSCLC, the analysis of circulating tumor DNA (ctDNA) in plasma, known as liquid biopsy, has recently emerged as an alternative and noninvasive approach for exploring tumor genetic constitution. In this study, we developed a protocol for liquid biopsy using ultra-deep massively parallel sequencing (MPS) with unique molecular identifier tagging and evaluated its performance for the identification and quantification of tumor-derived mutations from plasma of patients with advanced NSCLC. Paired plasma and tumor tissue samples were used to evaluate mutation profiles detected by ultra-deep MPS, which showed 87.5% concordance. Cross-platform comparison with droplet digital PCR demonstrated comparable detection performance (91.4% concordance, Cohen's kappa coefficient of 0.85 with 95% CI = 0.72-0.97) and great reliability in quantification of mutation allele frequency (Intraclass correlation coefficient of 0.96 with 95% CI = 0.90-0.98). Our results highlight the potential application of liquid biopsy using ultra-deep MPS as a routine assay in clinical practice for both detection and quantification of actionable mutation landscape in NSCLC patients.
Collapse
Affiliation(s)
- Le Son Tran
- Gene Solutions, Ho Chi Minh, Vietnam
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | - Hong-Anh Thi Pham
- Gene Solutions, Ho Chi Minh, Vietnam
- Graduate program of Genetics, Ho Chi Minh city University of Science, Ho Chi Minh city, Vietnam
| | - Vu-Uyen Tran
- Gene Solutions, Ho Chi Minh, Vietnam
- Graduate program of Genetics, Ho Chi Minh city University of Science, Ho Chi Minh city, Vietnam
| | - Thanh-Truong Tran
- Gene Solutions, Ho Chi Minh, Vietnam
- Graduate program of Genetics, Ho Chi Minh city University of Science, Ho Chi Minh city, Vietnam
| | | | | | | | | | | | - Binh Thanh Vo
- Gene Solutions, Ho Chi Minh, Vietnam
- Graduate program of Genetics, Ho Chi Minh city University of Science, Ho Chi Minh city, Vietnam
| | - Hong-Thuy Thi Dao
- Gene Solutions, Ho Chi Minh, Vietnam
- Graduate program of Genetics, Ho Chi Minh city University of Science, Ho Chi Minh city, Vietnam
| | | | - Tam Huu Tran
- Center for Standardization and QC in Medical Lab of Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | | | | | | | | | - Van Hieu Phan
- Center for Forensic Science, Ho Chi Minh City, Vietnam
| | | | | | | | - Minh-Duy Phan
- Gene Solutions, Ho Chi Minh, Vietnam
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Hoa Giang
- Gene Solutions, Ho Chi Minh, Vietnam
- Medical Genetics Institute, Ho Chi Minh City, Vietnam
| | | |
Collapse
|
8
|
Applying comprehensive histologic assessment and genetic testing to synchronous multifocal lung adenocarcinomas and further survival analysis. Chin Med J (Engl) 2019; 132:227-231. [PMID: 30614868 PMCID: PMC6365266 DOI: 10.1097/cm9.0000000000000055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
9
|
Tulchinsky E, Demidov O, Kriajevska M, Barlev NA, Imyanitov E. EMT: A mechanism for escape from EGFR-targeted therapy in lung cancer. Biochim Biophys Acta Rev Cancer 2018; 1871:29-39. [PMID: 30419315 DOI: 10.1016/j.bbcan.2018.10.003] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 10/07/2018] [Accepted: 10/21/2018] [Indexed: 02/08/2023]
Abstract
Epithelial mesenchymal transition (EMT) is a reversible developmental genetic programme of transdifferentiation of polarised epithelial cells to mesenchymal cells. In cancer, EMT is an important factor of tumour cell plasticity and has received increasing attention for its role in the resistance to conventional and targeted therapies. In this paper we provide an overview of EMT in human malignancies, and discuss contribution of EMT to the development of the resistance to Epidermal Growth Factor Receptor (EGFR)-targeted therapies in non-small cell lung cancer (NSCLC). Patients with the tumours bearing specific mutations in EGFR have a good clinical response to selective EGFR inhibitors, but the resistance inevitably develops. Several mechanisms responsible for the resistance include secondary mutations in the EGFR gene, genetic or non-mutational activation of alternative survival pathways, transdifferentiation of NSCLC to the small cell lung cancer histotype, or formation of resistant tumours with mesenchymal characteristics. Mechanistically, application of an EGFR inhibitor does not kill all cancer cells; some cells survive the exposure to a drug, and undergo genetic evolution towards resistance. Here, we present a theory that these quiescent or slow-proliferating drug-tolerant cell populations, or so-called "persisters", are generated via EMT pathways. We review the EMT-activated mechanisms of cell survival in NSCLC, which include activation of ABC transporters and EMT-associated receptor tyrosine kinase AXL, immune evasion, and epigenetic reprogramming. We propose that therapeutic inhibition of these pathways would eliminate pools of persister cells and prevent or delay cancer recurrence when applied in combination with the agents targeting EGFR.
Collapse
Affiliation(s)
- Eugene Tulchinsky
- Leicester Cancer Research Centre, Leicester University, UK; Moscow Institute of Physics and Technology, Dolgoprudny, Moscow, region, 117303, Russia.
| | - Oleg Demidov
- Instutute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia
| | | | - Nickolai A Barlev
- Moscow Institute of Physics and Technology, Dolgoprudny, Moscow, region, 117303, Russia; Instutute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia
| | | |
Collapse
|
10
|
Yan B, Hu Y, Ma T, Wang Y. IDH1 mutation promotes lung cancer cell proliferation through methylation of Fibulin-5. Open Biol 2018; 8:rsob.180086. [PMID: 30305430 PMCID: PMC6223204 DOI: 10.1098/rsob.180086] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/27/2018] [Indexed: 12/21/2022] Open
Abstract
Mutation in isocitrate dehydrogenase (IDH) leads to an aberrant function of the enzyme, leading to the production of hydroxyglutarate, as well as changes in cellular metabolism, DNA methylation and histone modification. Previous studies uncovered mutations in IDH1 in several malignancies, with the most frequent mutation being IDH1 R132H. It has been demonstrated that IDH1 expression is induced in non-small-cell lung cancer (NSCLC). However, the contribution of IDH1 mutation in the malignant transformation and development of NSCLC is unclear. In our study, we show that IDH1 R132H enhanced the migration and proliferation of NSCLC cells. Moreover, IDH1 R132H was a crucial modulator of 2-hydroxyglutarate, whose production from cells with IDH1 mutation promoted the binding of DNA-methyltransferase 1 (DNMT1) to the Fibulin-5 promoter, leading to its methylation. As a result, Fibulin-5 silencing in cells with IDH1 mutation enhanced the migration and proliferation of NSCLC cells. We show that the IDH1 mutation was present in tissues sampled from patients with NSCLC, which was reversely linked to Fibulin-5 expression. In this study, we suggest an innovative model for IDH1 R132H/Fibulin-5 pathway, which could throw light upon the activity of IDH1 R132H in NSCLC.
Collapse
Affiliation(s)
- Bingdi Yan
- Department of Pneumology, The 2nd Hospital Affiliated to Jilin University, Changchun, China
| | - Yanbing Hu
- Department of Ultrasound, The 2nd Hospital Affiliated to Jilin University, Changchun, China
| | - Tiangang Ma
- Department of Pneumology, The 2nd Hospital Affiliated to Jilin University, Changchun, China
| | - Yanjun Wang
- Nursing Department, The 2nd Hospital Affiliated to Jilin University, Changchun, China
| |
Collapse
|
11
|
Toth LN, de Abreu FB, Tafe LJ. Non–small cell lung cancers with isocitrate dehydrogenase 1 or 2 ( IDH1/2 ) mutations. Hum Pathol 2018; 78:138-143. [DOI: 10.1016/j.humpath.2018.04.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 04/07/2018] [Accepted: 04/20/2018] [Indexed: 02/06/2023]
|
12
|
Soca-Chafre G, Hernández-Pedro N, Aviles-Salas A, Versón CA, Sánchez KC, Cardona AF, Avila-Moreno F, Barrios-Bernal P, Flores-Estrada D, Arrieta O. Targeted next generation sequencing identified a high frequency genetic mutated profile in wood smoke exposure-related lung adenocarcinoma patients. Oncotarget 2018; 9:30499-30512. [PMID: 30093964 PMCID: PMC6078143 DOI: 10.18632/oncotarget.25369] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 04/21/2018] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Wood smoke exposure (WSE) has been associated with an increased risk of lung cancer development. WSE has been related with high frequency of EGFR mutations and low frequency of KRAS mutations. The aim of this study was to evaluate large scale genomic alterations in lung adenocarcinomas associated with WSE using targeted next generation sequencing. METHODS DNA multi-targeted sequencing was performed in 42 fresh-frozen samples of advanced lung adenocarcinomas. The TruSeQ Cancer Panel (Illumina) was used for genomic library construction and sequencing assays. RESULTS WSE rate was higher in women (p=0.037) and non-smokers (p=0.001). WSE correlated with mutations in the genes SMARCB1 (p=0.002), Ataxia telangiectasia mutated (p=0.004), Kinase Insert Domain Receptor (p=0.006), and were borderline significant in RET and EGFR exon. Genomic alterations significantly co-occurred in the tumor suppressor gene ATM with the following genes: SMARCB1, EGFR exon 7, RET and KDR. Clinical factors associated with poor prognosis were ECOG ≥ 2 (p= 0.014), mutations in KDR (p= 0.004) and APC genes (p < 0.001). CONCLUSIONS Lung adenocarcinoma patients with WSE showed a distinctive mutated profile for the SMARCB1, ATM, EGFR exon 7, RET and KDR genes. ECOG status and KDR gene mutations were significantly associated with poor prognosis.
Collapse
Affiliation(s)
- Giovanny Soca-Chafre
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN) México City, México
| | - Norma Hernández-Pedro
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN) México City, México
| | | | - Carmen Alaez Versón
- Translational Genomics Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
| | - Karol Carrillo Sánchez
- Translational Genomics Laboratory, Instituto Nacional de Medicina Genómica (INMEGEN), México City, México
| | - Andrés F. Cardona
- Clinical and Translational Oncology Group, Institute of Oncology, Clínica del Country, Bogotá, Colombia
| | - Federico Avila-Moreno
- Universidad Nacional Autónoma de México (UNAM), Facultad de Estudios Superiores (FES) Iztacala, Biomedicine Research Unit, Cancer Epigenomics and Lung Diseases Laboratory 12, México State, México
- National Institute of Respiratory Diseases (INER) “Ismael Cosío Villegas”, Research Unit, México City, México
| | - Pedro Barrios-Bernal
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN) México City, México
| | | | - Oscar Arrieta
- Personalized Medicine Laboratory, Instituto Nacional de Cancerología (INCAN) México City, México
- Thoracic Oncology Clinic, INCAN, México City, México
| |
Collapse
|
13
|
High-throughput detection of clinically targetable alterations using next-generation sequencing. Oncotarget 2018; 8:40345-40358. [PMID: 28404952 PMCID: PMC5522202 DOI: 10.18632/oncotarget.15875] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 01/23/2017] [Indexed: 12/17/2022] Open
Abstract
Next-generation sequencing (NGS) has revolutionized the therapeutic care of patients by allowing high-throughput and parallel sequencing of large numbers of genes in a single run. However, most of available commercialized cancer panels target a large number of mutations that do not have direct therapeutic implications and that are not fully adapted to low quality formalin-fixed, paraffin-embedded (FFPE) samples. Here, we designed an amplicon-based NGS panel assay of 16 currently actionable genes according to the most recent recommendations of the French National Cancer Institute (NCI). We developed a panel of short amplicons (<150 bp) using dual-strand library preparation. The clinical validation of this panel was performed on well-characterized controls and 140 routine diagnostic samples, including highly degraded and cross-linked genomic DNA extracted from FFPE tumor samples. All mutations were detected with elevated inter-laboratory and inter-run reproducibility. Importantly, we could detect clinically actionable alterations in FFPE samples with variant allele frequencies as low as 1%. In addition, the overall molecular diagnosis rate was increased from 40.7% with conventional techniques to 59.2% with our NGS panel, including 41 novel actionable alterations normally not explored by conventional techniques. Taken together, we believe that this new actionable target panel represents a relevant, highly scalable and robust tool that is easy to implement and is fully adapted to daily clinical practice in hospital and academic laboratories.
Collapse
|
14
|
Abstract
The identification of certain genomic alterations (EGFR, ALK, ROS1, BRAF) or immunological markers (PD-L1) in tissues or cells has led to targeted treatment for patients presenting with late stage or metastatic lung cancer. These biomarkers can be detected by immunohistochemistry (IHC) and/or by molecular biology (MB) techniques. These approaches are often complementary but depending on, the quantity and quality of the biological material, the urgency to get the results, the access to technological platforms, the financial resources and the expertise of the team, the choice of the approach can be questioned. The possibility of detecting simultaneously several molecular targets, and of analyzing the degree of tumor mutation burden and of the micro-satellite instability, as well as the recent requirement to quantify the expression of PD-L1 in tumor cells, has led to case by case development of algorithms and international recommendations, which depend on the quality and quantity of biological samples. This review will highlight the different predictive biomarkers detected by IHC for treatment of lung cancer as well as the present advantages and limitations of this approach. A number of perspectives will be considered.
Collapse
|
15
|
Illei PB, Belchis D, Tseng LH, Nguyen D, De Marchi F, Haley L, Riel S, Beierl K, Zheng G, Brahmer JR, Askin FB, Gocke CD, Eshleman JR, Forde PM, Lin MT. Clinical mutational profiling of 1006 lung cancers by next generation sequencing. Oncotarget 2017; 8:96684-96696. [PMID: 29228562 PMCID: PMC5722514 DOI: 10.18632/oncotarget.18042] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/10/2017] [Indexed: 12/15/2022] Open
Abstract
Analysis of lung adenocarcinomas for actionable mutations has become standard of care. Here, we report our experience using next generation sequencing (NGS) to examine AKT1, BRAF, EGFR, ERBB2, KRAS, NRAS, and PIK3CA genes in 1006 non-small cell lung cancers in a clinical diagnostic setting. NGS demonstrated high sensitivity. Among 760 mutations detected, the variant allele frequency (VAF) was 2-5% in 33 (4.3%) mutations and 2-10% in 101 (13%) mutations. A single bioinformatics pipeline using Torrent Variant Caller, however, missed a variety of EGFR mutations. Mutations were detected in KRAS (36% of tumors), EGFR (19%) including 8 (0.8%) within the extracellular domain (4 at codons 108 and 4 at codon 289), BRAF (6.3%), and PIK3CA (3.7%). With a broader reportable range, exon 19 deletion and p.L858R accounted for only 36% and 26% of EGFR mutations and p.V600E accounted for only 24% of BRAF mutations. NGS provided accurate sequencing of complex mutations seen in 19% of EGFR exon 19 deletion mutations. Doublet (compound) EGFR mutations were observed in 29 (16%) of 187 EGFR-mutated tumors, including 69% with two non-p.L858R missense mutations and 24% with p.L858 and non-p.L858R missense mutations. Concordant VAFs suggests doublet EGFR mutations were present in a dominant clone and cooperated in oncogenesis. Mutants with predicted impaired kinase, observed in 25% of BRAF-mutated tumors, were associated with a higher incidence of concomitant activating KRAS mutations. NGS demonstrates high analytic sensitivity, broad reportable range, quantitative VAF measurement, single molecule sequencing to resolve complex deletion mutations, and simultaneous detection of concomitant mutations.
Collapse
Affiliation(s)
- Peter B. Illei
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Deborah Belchis
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Li-Hui Tseng
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Doreen Nguyen
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Federico De Marchi
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Division of Hematology and Bone Marrow Transplantation, University of Udine Hospital, Udine, Italy
| | - Lisa Haley
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Stacy Riel
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Katie Beierl
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Gang Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Julie R. Brahmer
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Frederic B. Askin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Christopher D. Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - James R. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Patrick M. Forde
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
16
|
Lin L, Zhuang W, Wang W, Xu C, Chen R, Guan Y, Yi X, Shao Y, Tai Y, Chen Y, Wu M, Wei J, Chen G. Genetic mutations in lung enteric adenocarcinoma identified using next-generation sequencing. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2017; 10:9583-9590. [PMID: 31966835 PMCID: PMC6966000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 08/15/2017] [Indexed: 06/10/2023]
Abstract
Primary lung enteric adenocarcinoma is a rare type of invasive lung carcinoma. Its morphological and immunohistochemical characteristics are similar to those of metastatic colorectal carcinoma, but there is no associated primary colorectal carcinoma. The purpose of this study is to identify mutations by assessing the genetic profile of lung enteric adenocarcinoma with next-generation sequencing (NGS). This study included 11 lung enteric adenocarcinoma patients (5 males and 6 females) from three different centers who received treatment between Feb 2013 and Dec 2016. Immunohistochemical analysis failed to reveal any markers that differentiated this carcinoma from primary gastrointestinal adenocarcinoma. NGS analysis identified ALK/ROS1 primary point mutations in 5 patients (71.42%, 5/7) and MSH2/MSH6 point mutations in 3 patients (42.86%, 3/7). There was no case with drive genes changed, such as EGFR mutation, ALK rearrangement, ROS1 rearrangement, RET rearrangement, MET amplification or 14 exon skipping mutation. The median overall survival of the 11 lung enteric adenocarcinoma patients was 9.0 months. Further, subgroup analysis showed that the median OS of patients with ALK/ROS1 primary point mutations was 6.5 months and that of patients with MSH2/MSH6 primary point mutations was 26.0 months. These two mutations were the most frequent features, but this carcinoma generally showed genetic heterogeneity. Even though we have revealed some hitherto unidentified genetic mutations associated with lung enteric adenocarcinoma, the findings are preliminary and further investigations on more patients will be required to validate our findings.
Collapse
Affiliation(s)
- Li Lin
- Department of Oncology, Peking University International HospitalBeijing 102206, People’s Republic of China
| | - Wu Zhuang
- Department of Medical Thoracic Oncology, Fujian Cancer Hospital, Fujian Medical University Cancer HospitalFujian 350014, Fuzhou, People’s Republic of China
| | - Wenxian Wang
- Department of Chemotherapy, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, People’s Republic of China
| | - Chunwei Xu
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer HospitalFujian 350014, Fuzhou, People’s Republic of China
| | - Rongrong Chen
- Geneplus-BeijingBeijing 102200, People’s Republic of China
| | - Yanfang Guan
- Geneplus-BeijingBeijing 102200, People’s Republic of China
| | - Xin Yi
- Geneplus-BeijingBeijing 102200, People’s Republic of China
| | - Yun Shao
- Department of Pathology, Affiliated Hospital Cancer Center, Academy of Military Medical SciencesBeijing 100071, People’s Republic of China
| | - Yanhong Tai
- Department of Pathology, Affiliated Hospital Cancer Center, Academy of Military Medical SciencesBeijing 100071, People’s Republic of China
| | - Yanping Chen
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer HospitalFujian 350014, Fuzhou, People’s Republic of China
| | - Meijuan Wu
- Department of Pathology, Zhejiang Cancer HospitalHangzhou 310022, Zhejiang, People’s Republic of China
| | - Jianguo Wei
- Department of Pathology, Shaoxing People’s HospitalShaoxing 312000, Zhejiang, People’s Republic of China
| | - Gang Chen
- Department of Pathology, Fujian Cancer Hospital, Fujian Medical University Cancer HospitalFujian 350014, Fuzhou, People’s Republic of China
| |
Collapse
|
17
|
Park J, Yoo HM, Jang W, Shin S, Kim M, Kim Y, Lee SW, Kim JG. Distribution of somatic mutations of cancer-related genes according to microsatellite instability status in Korean gastric cancer. Medicine (Baltimore) 2017; 96:e7224. [PMID: 28640116 PMCID: PMC5484224 DOI: 10.1097/md.0000000000007224] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
In studies of the molecular basis of gastric cancer (GC), microsatellite instability (MSI) is one of the key factors. Somatic mutations found in GC are expected to contribute to MSI-high (H) tumorigenesis. We estimated somatic mutation distribution according to MSI status in 52 matched pair GC samples using the Ion Torrent Ion S5 XL with the AmpliSeq Cancer Hotspot panel.Seventy-five (9.8%) somatic variants consisting of 34 hotspot mutations and 41 other likely pathogenic variants were identified in 34 GC samples. The TP53 mutations was most common (35%, 26/75), followed by EGFR (8%, 6/75), HNF1A (8%, 6/75), PIK3CA (8%, 6/75), and ERBB2 (5%, 4/75). To determine MSI status, 52 matched pair samples were estimated using 15 MSI markers. Thirty-nine MS stable (S), 5 MSI-low (L), and 8 MSI-H were classified. GCs with MSI-H tended to have more variants significantly compared with GCs with MS stable (MSS) and MSI-L (standardized J-T statistic = 3.161 for number of variants; P = .002). The mean number of all variants and hotspot mutations per tumor samples only in GCs with MSI-H were 3.9 (range, 1-6) and 1.1 (range, 0-3), respectively. Whereas, the mean number of all variants and hotspot mutations per tumor samples only in GCs with MSS/MSI-L were 1 (0-5)/0.8 (0-1) and 0.5 (0-3)/0.8 (0-1), respectively.In conclusion, GC with MSI-H harbored more mutations in genes that act as a tumor suppressor or oncogene compared to GC with MSS/MSI-L. This finding suggests that the accumulation of MSIs contributes to the genetic diversity and complexities of GC. In addition, targeted NGS approach allows for detection of common and also rare clinically actionable mutations and profiles of comutations in multiple patients simultaneously. Because GC shows distinctive patterns related to ethnics, further studies pertaining to different racial/ethnic groups or cancer types may reinforce our investigations.
Collapse
Affiliation(s)
| | - Han Mo Yoo
- Division of Gastrointestinal Surgery, Department of Surgery
| | | | | | | | | | - Seung-Woo Lee
- Division of Gastroenterology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Jeong Goo Kim
- Division of Gastrointestinal Surgery, Department of Surgery
| |
Collapse
|