1
|
Tan N, Li Y, Ying J, Chen W. Histological transformation in lung adenocarcinoma: Insights of mechanisms and therapeutic windows. J Transl Int Med 2024; 12:452-465. [PMID: 39513032 PMCID: PMC11538883 DOI: 10.1515/jtim-2024-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2024] Open
Abstract
Histological transformation from lung adenocarcinoma (ADC) to small cell lung carcinoma (SCLC), large cell neuroendocrine carcinoma (LCNEC), squamous cell carcinoma (SCC), and sarcomatoid carcinoma (PSC) after targeted therapies is recognized as a mechanism of resistance in ADC treatments. Patients with transformed lung cancer typically experience a poor prognosis and short survival time. However, effective treatment options for these patients are currently lacking. Therefore, understanding the mechanisms underlying histological transformation is crucial for the development of effective therapies. Hypotheses including intratumoral heterogeneity, cancer stem cells, and alteration of suppressor genes have been proposed to explain the mechanism of histological transformation. In this review, we provide a comprehensive overview of the known molecular features and signaling pathways of transformed tumors, and summarized potential therapies based on previous findings.
Collapse
Affiliation(s)
- Nuopei Tan
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Chen
- Office of Cancer Screening, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
2
|
Tan L, Brown C, Mersiades A, Lee CK, John T, Kao S, Newnham G, O'Byrne K, Parakh S, Bray V, Jasas K, Yip S, Wong SQ, Ftouni S, Guinto J, Chandrashekar S, Clarke S, Pavlakis N, Stockler MR, Dawson SJ, Solomon BJ. A Phase II trial of alternating osimertinib and gefitinib therapy in advanced EGFR-T790M positive non-small cell lung cancer: OSCILLATE. Nat Commun 2024; 15:1823. [PMID: 38418463 PMCID: PMC10902357 DOI: 10.1038/s41467-024-46008-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 02/07/2024] [Indexed: 03/01/2024] Open
Abstract
In this phase II, single arm trial (ACTRN12617000720314), we investigate if alternating osimertinib and gefitinib would delay the development of resistance to osimertinib in advanced, non-small cell lung cancer (NSCLC) with the epidermal growth factor receptor (EGFR) T790M mutation (n = 47) by modulating selective pressure on resistant clones. The primary endpoint is progression free-survival (PFS) rate at 12 months, and secondary endpoints include: feasibility of alternating therapy, overall response rate (ORR), overall survival (OS), and safety. The 12-month PFS rate is 38% (95% CI 27.5-55), not meeting the pre-specified primary endpoint. Serial circulating tumor DNA (ctDNA) analysis reveals decrease and clearance of the original activating EGFR and EGFR-T790M mutations which are prognostic of clinical outcomes. In 73% of participants, loss of T790M ctDNA is observed at progression and no participants have evidence of the EGFR C797S resistance mutation following the alternating regimen. These findings highlight the challenges of treatment strategies designed to modulate clonal evolution and the clinical importance of resistance mechanisms beyond suppression of selected genetic mutations in driving therapeutic escape to highly potent targeted therapies.
Collapse
Affiliation(s)
- Lavinia Tan
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Chris Brown
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Antony Mersiades
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Chee Khoon Lee
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- St George Hospital, Sydney, NSW, Australia
| | - Thomas John
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Steven Kao
- Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | | | | | - Sagun Parakh
- Austin Hospital, Olivia Newton John Cancer and Wellness and Research Centre, Melbourne, VIC, Australia
| | | | - Kevin Jasas
- Sir Charles Gairdner Hospital, Perth, WA, Australia
| | - Sonia Yip
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Stephen Q Wong
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Sarah Ftouni
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - Jerick Guinto
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | | | - Stephen Clarke
- Royal North Shore Hospital, Sydney, NSW, Australia
- University of Sydney, Sydney, NSW, Australia
| | - Nick Pavlakis
- Royal North Shore Hospital, Sydney, NSW, Australia
- University of Sydney, Sydney, NSW, Australia
| | - Martin R Stockler
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
- Centre for Cancer Research, The University of Melbourne, Melbourne, VIC, Australia.
| | - Benjamin J Solomon
- Peter MacCallum Cancer Centre, Melbourne, Vic, Australia.
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
3
|
Santonja A, Cooper WN, Eldridge MD, Edwards PAW, Morris JA, Edwards AR, Zhao H, Heider K, Couturier D, Vijayaraghavan A, Mennea P, Ditter E, Smith CG, Boursnell C, Manzano García R, Rueda OM, Beddowes E, Biggs H, Sammut S, Rosenfeld N, Caldas C, Abraham JE, Gale D. Comparison of tumor-informed and tumor-naïve sequencing assays for ctDNA detection in breast cancer. EMBO Mol Med 2023; 15:e16505. [PMID: 37161793 PMCID: PMC10245040 DOI: 10.15252/emmm.202216505] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 05/11/2023] Open
Abstract
Analysis of circulating tumor DNA (ctDNA) to monitor cancer dynamics and detect minimal residual disease has been an area of increasing interest. Multiple methods have been proposed but few studies have compared the performance of different approaches. Here, we compare detection of ctDNA in serial plasma samples from patients with breast cancer using different tumor-informed and tumor-naïve assays designed to detect structural variants (SVs), single nucleotide variants (SNVs), and/or somatic copy-number aberrations, by multiplex PCR, hybrid capture, and different depths of whole-genome sequencing. Our results demonstrate that the ctDNA dynamics and allele fractions (AFs) were highly concordant when analyzing the same patient samples using different assays. Tumor-informed assays showed the highest sensitivity for detection of ctDNA at low concentrations. Hybrid capture sequencing targeting between 1,347 and 7,491 tumor-identified mutations at high depth was the most sensitive assay, detecting ctDNA down to an AF of 0.00024% (2.4 parts per million, ppm). Multiplex PCR targeting 21-47 tumor-identified SVs per patient detected ctDNA down to 0.00047% AF (4.7 ppm) and has potential as a clinical assay.
Collapse
Affiliation(s)
- Angela Santonja
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Wendy N Cooper
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Matthew D Eldridge
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Paul A W Edwards
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
- Department of PathologyUniversity of CambridgeCambridgeUK
| | - James A Morris
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Abigail R Edwards
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
| | - Hui Zhao
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Katrin Heider
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Dominique‐Laurent Couturier
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
- MRC Biostatistics UnitUniversity of CambridgeCambridgeUK
| | - Aadhitthya Vijayaraghavan
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Paulius Mennea
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Emma‐Jane Ditter
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Christopher G Smith
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Chris Boursnell
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Raquel Manzano García
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Oscar M Rueda
- MRC Biostatistics UnitUniversity of CambridgeCambridgeUK
| | - Emma Beddowes
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Heather Biggs
- Department of OncologyUniversity of CambridgeCambridgeUK
- Precision Breast Cancer Institute, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's HospitalCambridgeUK
| | - Stephen‐John Sammut
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
- Department of OncologyUniversity of CambridgeCambridgeUK
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
- Department of OncologyUniversity of CambridgeCambridgeUK
- Precision Breast Cancer Institute, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's HospitalCambridgeUK
| | - Jean E Abraham
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
- Department of OncologyUniversity of CambridgeCambridgeUK
- Precision Breast Cancer Institute, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke's HospitalCambridgeUK
| | - Davina Gale
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing CentreCambridgeUK
- Cancer Research UK Cambridge Centre, Cancer Research UK Cambridge Institute, Li Ka Shing CentreCambridgeUK
| |
Collapse
|
4
|
Laface C, Maselli FM, Santoro AN, Iaia ML, Ambrogio F, Laterza M, Guarini C, De Santis P, Perrone M, Fedele P. The Resistance to EGFR-TKIs in Non-Small Cell Lung Cancer: From Molecular Mechanisms to Clinical Application of New Therapeutic Strategies. Pharmaceutics 2023; 15:1604. [PMID: 37376053 DOI: 10.3390/pharmaceutics15061604] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/13/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Almost 17% of Western patients affected by non-small cell lung cancer (NSCLC) have an activating epidermal growth factor receptor (EGFR) gene mutation. Del19 and L858R are the most-common ones; they are positive predictive factors for EGFR tyrosine kinase inhibitors (TKIs). Currently, osimertinib, a third-generation TKI, is the standard first-line therapy for advanced NSCLC patients with common EGFR mutations. This drug is also administered as a second-line treatment for those patients with the T790M EGFR mutation and previously treated with first- (erlotinib, gefitinib) or second- (afatinib) generation TKIs. However, despite the high clinical efficacy, the prognosis remains severe due to intrinsic or acquired resistance to EGRF-TKIs. Various mechanisms of resistance have been reported including the activation of other signalling pathways, the development of secondary mutations, the alteration of the downstream pathways, and phenotypic transformation. However, further data are needed to achieve the goal of overcoming resistance to EGFR-TKIs, hence the necessity of discovering novel genetic targets and developing new-generation drugs. This review aimed to deepen the knowledge of intrinsic and acquired molecular mechanisms of resistance to EGFR-TKIs and the development of new therapeutic strategies to overcome TKIs' resistance.
Collapse
Affiliation(s)
- Carmelo Laface
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | | | | | - Maria Laura Iaia
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Francesca Ambrogio
- Section of Dermatology, Department of Biomedical Science and Human Oncology, University of Bari, 70124 Bari, Italy
| | - Marigia Laterza
- Division of Cardiac Surgery, University of Bari, 70124 Bari, Italy
| | - Chiara Guarini
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Pierluigi De Santis
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Martina Perrone
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| | - Palma Fedele
- Medical Oncology, Dario Camberlingo Hospital, 72021 Francavilla Fontana, Italy
| |
Collapse
|
5
|
Pezzuto F, Hofman V, Bontoux C, Fortarezza F, Lunardi F, Calabrese F, Hofman P. The significance of co-mutations in EGFR-mutated non-small cell lung cancer: Optimizing the efficacy of targeted therapies? Lung Cancer 2023; 181:107249. [PMID: 37244040 DOI: 10.1016/j.lungcan.2023.107249] [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: 02/27/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/29/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most common cause of cancer death worldwide. In non-squamous NSCLC, the identification of oncogenic drivers and the development of target-specific molecules led to remarkable progress in therapeutic strategies and overall survival over the last decade. Nevertheless, responses are limited by systematically acquired mechanisms of resistance early on after starting a targeted therapy. Moreover, mounting evidence has demonstrated that each oncogenic-driven cluster is actually heterogeneous in terms of molecular features, clinical behaviour, and sensitivity to targeted therapy. In this review, we aimed to examine the prognostic and predictive significance of oncogene-driven co-mutations, focusing mainly on EGFR and TP53. A narrative review was performed by searching MEDLINE databases for English articles published over the last decade (from January 2012 until November 2022). The bibliographies of key references were manually reviewed to select those eligible for the topic. The genetic landscape of EGFR-mutated NSCLC is more complicated than what is known so far. In particular, the occurrence of TP53 co-mutations stratify patients carrying EGFR mutations in terms of treatment response. The study provides a deeper understanding of the mechanisms underlying the variability of the genetic landscape of EGFR-mutated NSCLC and summarizes notably the clinical importance of TP53 co-mutations for an open avenue to more properly addressing the clinical decision-making in the near future.
Collapse
Affiliation(s)
- Federica Pezzuto
- University Côte d'Azur, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, BB-0033-00025, Pasteur Hospital, 30 voie romaine, 06001 Nice, France; Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Via A. Gabelli 61, 35121 Padova, Italy.
| | - Véronique Hofman
- University Côte d'Azur, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, BB-0033-00025, Pasteur Hospital, 30 voie romaine, 06001 Nice, France
| | - Christophe Bontoux
- University Côte d'Azur, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, BB-0033-00025, Pasteur Hospital, 30 voie romaine, 06001 Nice, France
| | - Francesco Fortarezza
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Via A. Gabelli 61, 35121 Padova, Italy
| | - Francesca Lunardi
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Via A. Gabelli 61, 35121 Padova, Italy
| | - Fiorella Calabrese
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova Medical School, Via A. Gabelli 61, 35121 Padova, Italy
| | - Paul Hofman
- University Côte d'Azur, Laboratory of Clinical and Experimental Pathology, FHU OncoAge, BB-0033-00025, Pasteur Hospital, 30 voie romaine, 06001 Nice, France.
| |
Collapse
|
6
|
Ferrara MG, Belluomini L, Smimmo A, Sposito M, Avancini A, Giannarelli D, Milella M, Pilotto S, Bria E. Meta-analysis of the prognostic impact of TP53 co-mutations in EGFR-mutant advanced non-small-cell lung cancer treated with tyrosine kinase inhibitors. Crit Rev Oncol Hematol 2023; 184:103929. [PMID: 36773668 DOI: 10.1016/j.critrevonc.2023.103929] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/24/2023] [Accepted: 01/24/2023] [Indexed: 02/12/2023] Open
Abstract
PURPOSE To assess the prognostic impact of TP53 mutations in EGFR-mutant advanced NSCLC patients treated with TKIs. METHODS Studies exploring the clinical outcomes of EGFR mutant/TP53 wild-type versus EGFR/TP53 co-mutant patients treated with TKIs were selected. Data were cumulated by adopting a fixed and random-effect model. RESULTS Overall, 29 trials were eligible. The PFS analysis showed that TP53 co-mutant group has shorter PFS versus EGFR mutant/TP53 wild-type group (HR = 1.67, 95% CI 1.51-1.83, heterogeneity I2 =20%, p = 0.18). Patients affected by EGFR/TP53 co-mutant NSCLC have a higher chance of shorter OS versus EGFR mutant/TP53 wild type (HR= 1.89, 95% CI 1.67-2.14, heterogeneity I2 = 21%; p = 0.19). The subgroup analysis showed no significant difference between first-second versus third-generation TKIs in both PFS and OS (p = 0.31, p = 0.08). CONCLUSIONS TP53 mutations represent a clinically relevant mechanism of resistance to EGFR-TKIs, regardless of their generation. A personalized therapeutical approach should be explored in dedicated clinical trials.
Collapse
Affiliation(s)
- Miriam Grazia Ferrara
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy; Medical Oncology, Università Cattolica del Sacro Cuore, Roma, Italy.
| | - Lorenzo Belluomini
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy.
| | - Annafrancesca Smimmo
- Biostatistical Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| | - Marco Sposito
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy.
| | - Alice Avancini
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy; Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy.
| | - Diana Giannarelli
- Biostatistical Unit, Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy.
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy.
| | - Sara Pilotto
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy.
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.
| |
Collapse
|
7
|
Li YZ, Kong SN, Liu YP, Yang Y, Zhang HM. Can Liquid Biopsy Based on ctDNA/cfDNA Replace Tissue Biopsy for the Precision Treatment of EGFR-Mutated NSCLC? J Clin Med 2023; 12:jcm12041438. [PMID: 36835972 PMCID: PMC9966257 DOI: 10.3390/jcm12041438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/16/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
More and more clinical trials have explored the role of liquid biopsy in the diagnosis and treatment of EGFR-mutated NSCLC. In certain circumstances, liquid biopsy has unique advantages and offers a new way to detect therapeutic targets, analyze drug resistance mechanisms in advanced patients, and monitor MRD in patients with operable NSCLC. Although its potential cannot be ignored, more evidence is needed to support the transition from the research stage to clinical application. We reviewed the latest progress in research on the efficacy and resistance mechanisms of targeted therapy for advanced NSCLC patients with plasma ctDNA EGFR mutation and the evaluation of MRD based on ctDNA detection in perioperative and follow-up monitoring.
Collapse
|
8
|
Miura S, Koh Y, Azuma K, Yoshioka H, Koyama K, Teraoka S, Ishii H, Kibata K, Ozawa Y, Tokito T, Oyanagi J, Shimokawa T, Kurata T, Yamamoto N, Tanaka H. Afatinib plus osimertinib in the treatment of osimertinib-resistant non-small cell lung carcinoma: a phase I clinical trial. BMC Cancer 2023; 23:6. [PMID: 36597021 PMCID: PMC9808978 DOI: 10.1186/s12885-022-10467-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Conquering acquired resistance to osimertinib remains a major challenge in treating patients with epidermal growth factor receptor (EGFR) mutation-positive non-small-cell lung cancer (NSCLC). Thus, we aimed to determine the safety and efficacy of combination treatment with osimertinib and afatinib for patients with acquired resistance to osimertinib. METHODS This open-label phase I study was a feasibility study of the combination of afatinib and osimertinib for patients with advanced EGFR-positive NSCLC who had progressive disease after receiving osimertinib. The primary endpoint was to determine the maximum tolerated dose (MTD). We enrolled patients who received afatinib at three different dose levels (level 1, 20 mg; level 2, 30 mg; level 3, 40 mg) combined with osimertinib at a standard dose of 80 mg once per day. RESULTS Thirteen patients were enrolled in this study. The MTD was defined as 30 mg afatinib when combined with daily oral administration of osimertinib (80 mg). The most frequent adverse events were diarrhea (76.9%), anemia (76.9%), and rash (69.2%). Considering the toxicity profiles during all treatment periods, the recommended oral dose of afatinib was determined as 20 mg daily, with an osimertinib dose of 80 mg. For all evaluable patients (n = 12), the response rate was 7.7% and the disease-control rate was 46.2%. CONCLUSION Combination therapy with osimertinib and afatinib was tolerable; however, the synergistic effect of afatinib with osimertinib may be limited in osimertinib-resistant patients. TRIAL REGISTRATION Japan Registry of Clinical Trials ID: jRCTs051180008, registered date: 08/11/2018.
Collapse
Affiliation(s)
- Satoru Miura
- grid.416203.20000 0004 0377 8969Department of Internal Medicine, Niigata Cancer Center Hospital, 2-15-3 Kawagishicho, Chuo-ku, Niigata, Niigata 951-8566 Japan
| | - Yasuhiro Koh
- grid.412857.d0000 0004 1763 1087Internal Medicine III, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan ,grid.412857.d0000 0004 1763 1087Center for Biomedical Sciences, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan
| | - Koichi Azuma
- grid.410781.b0000 0001 0706 0776Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine 67 Asahi-machi, Kurume, 830-0011 Japan
| | - Hiroshige Yoshioka
- grid.410783.90000 0001 2172 5041Department of Thoracic Oncology, Kansai Medical University Hospital, 3-1, Shinmachi 2 Chome, Hirakata, Osaka, 573-1191 Japan
| | - Kenichi Koyama
- grid.416203.20000 0004 0377 8969Department of Internal Medicine, Niigata Cancer Center Hospital, 2-15-3 Kawagishicho, Chuo-ku, Niigata, Niigata 951-8566 Japan
| | - Shunsuke Teraoka
- grid.412857.d0000 0004 1763 1087Internal Medicine III, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan
| | - Hidenobu Ishii
- grid.410781.b0000 0001 0706 0776Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine 67 Asahi-machi, Kurume, 830-0011 Japan
| | - Kayoko Kibata
- grid.410783.90000 0001 2172 5041Department of Thoracic Oncology, Kansai Medical University Hospital, 3-1, Shinmachi 2 Chome, Hirakata, Osaka, 573-1191 Japan
| | - Yuichi Ozawa
- grid.412857.d0000 0004 1763 1087Internal Medicine III, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan
| | - Takaaki Tokito
- grid.410781.b0000 0001 0706 0776Division of Respirology, Neurology, and Rheumatology, Department of Internal Medicine, Kurume University School of Medicine 67 Asahi-machi, Kurume, 830-0011 Japan
| | - Jun Oyanagi
- grid.412857.d0000 0004 1763 1087Internal Medicine III, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan ,grid.412857.d0000 0004 1763 1087Center for Biomedical Sciences, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan
| | - Toshio Shimokawa
- grid.412857.d0000 0004 1763 1087Clinical Study Support Center, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan
| | - Takayasu Kurata
- grid.410783.90000 0001 2172 5041Department of Thoracic Oncology, Kansai Medical University Hospital, 3-1, Shinmachi 2 Chome, Hirakata, Osaka, 573-1191 Japan
| | - Nobuyuki Yamamoto
- grid.412857.d0000 0004 1763 1087Internal Medicine III, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan ,grid.412857.d0000 0004 1763 1087Center for Biomedical Sciences, Wakayama Medical University, 811-1, Kimiidera, Wakayama, 641-8509 Japan
| | - Hiroshi Tanaka
- grid.416203.20000 0004 0377 8969Department of Internal Medicine, Niigata Cancer Center Hospital, 2-15-3 Kawagishicho, Chuo-ku, Niigata, Niigata 951-8566 Japan
| |
Collapse
|
9
|
Yang D, Xu F, Li Y, Lai X, Xian B, Yu P, Chen R, Li Z, Ma D. Assessment of durable chemoimmunotherapy response via circulating tumor DNA in advanced esophageal squamous cell carcinoma. Thorac Cancer 2022; 13:2786-2791. [PMID: 35997004 PMCID: PMC9527169 DOI: 10.1111/1759-7714.14610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/26/2022] [Accepted: 07/31/2022] [Indexed: 11/28/2022] Open
Abstract
Immune checkpoint inhibitor (ICI)‐based therapies have shown promising advances for the first‐line treatment of advanced or metastatic esophageal cancer (EC). However, few studies concerning the identification of patients who achieve durable response from ICIs have been previously reported. In the present study, pre‐ and on‐treatment plasma circulating tumor DNA (ctDNA) were analyzed in 10 patients with advanced esophageal squamous cell cancer (ESCC) receiving first‐line chemoimmunotherapy. Patients with decreased molecular tumor burden index (mTBI) >7% experienced longer progression‐free survival (PFS) and durable clinical benefit (DCB, PFS ≥ 6 months). In addition, five patients showed stable disease at first scan, all three patients with decreased mTBI > 7% achieved DCB, while two cases with decreased mTBI ≤ 7% experienced non‐DCB. Our results demonstrate that ctDNA monitor might help identify which ESCC patients respond to chemoimmunotherapy.
Collapse
Affiliation(s)
- Dongyang Yang
- Secondary division of Medical Oncology, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Fei Xu
- Secondary division of Medical Oncology, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ying Li
- Secondary division of Medical Oncology, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaorong Lai
- Secondary division of Medical Oncology, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Bohong Xian
- Secondary division of Medical Oncology, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Pengli Yu
- Department of Medicine, Geneplus-Beijing Institute, Beijing, China
| | - Rongrong Chen
- Department of Medicine, Geneplus-Beijing Institute, Beijing, China
| | - Zijun Li
- Department of Gastroenterology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dong Ma
- Secondary division of Medical Oncology, Cancer Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
10
|
Vaquero J, Pavy A, Gonzalez-Sanchez E, Meredith M, Arbelaiz A, Fouassier L. Genetic alterations shaping tumor response to anti-EGFR therapies. Drug Resist Updat 2022; 64:100863. [DOI: 10.1016/j.drup.2022.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
11
|
Vokes NI, Chambers E, Nguyen T, Coolidge A, Lydon CA, Le X, Sholl L, Heymach JV, Nishino M, Van Allen EM, Jänne PA. Concurrent TP53 Mutations Facilitate Resistance Evolution in EGFR-Mutant Lung Adenocarcinoma. J Thorac Oncol 2022; 17:779-792. [PMID: 35331964 PMCID: PMC10478031 DOI: 10.1016/j.jtho.2022.02.011] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 11/26/2022]
Abstract
INTRODUCTION Patients with EGFR-mutant NSCLC experience variable duration of benefit on EGFR tyrosine kinase inhibitors. The effect of concurrent genomic alterations on outcome has been incompletely described. METHODS In this retrospective study, targeted next-generation sequencing data were collected from patients with EGFR-mutant lung cancer treated at the Dana-Farber Cancer Institute. Clinical data were collected and correlated with somatic mutation data. Associations between TP53 mutation status, genomic features, and mutational processes were analyzed. RESULTS A total of 269 patients were identified for inclusion in the cohort. Among 185 response-assessable patients with pretreatment specimens, TP53 alterations were the most common event associated with decreased first-line progression-free survival and decreased overall survival, along with DNMT3A, KEAP1, and ASXL1 alterations. Reduced progression-free survival on later-line osimertinib in 33 patients was associated with MET, APC, and ERBB4 alterations. Further investigation of the effect of TP53 alterations revealed an association with worse outcomes even in patients with good initial radiographic response, and faster acquisition of T790M and other resistance mechanisms. TP53-mutated tumors had higher mutational burdens and increased mutagenesis with exposure to therapy and tobacco. Cell cycle alterations were not independently predictive, but portended worse OS in conjunction with TP53 alterations. CONCLUSIONS TP53 alterations associate with faster resistance evolution independent of mechanism in EGFR-mutant NSCLC and may cooperate with other genomic events to mediate acquisition of resistance mutations to EGFR tyrosine kinase inhibitors.
Collapse
Affiliation(s)
- Natalie I Vokes
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Emily Chambers
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tom Nguyen
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Alexis Coolidge
- Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Christine A Lydon
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Xiuning Le
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - John V Heymach
- Department of Thoracic and Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Mizuki Nishino
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts; Department of Imaging, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Eliezer M Van Allen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Broad Institute of Harvard and Massachusetts Institute of Technology, Boston, Massachusetts
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
12
|
Liang X, Lin A, Wang Q, Zhang J, Luo P. Cell plasticity in patients with NSCLC: The controversial origins of transformed SCLC. Biomed Pharmacother 2022; 149:112909. [DOI: 10.1016/j.biopha.2022.112909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 11/02/2022] Open
|
13
|
Favaro PF, Stewart SD, McDonald BR, Cawley J, Contente-Cuomo T, Wong S, Hendricks WPD, Trent JM, Khanna C, Murtaza M. Feasibility of circulating tumor DNA analysis in dogs with naturally occurring malignant and benign splenic lesions. Sci Rep 2022; 12:6337. [PMID: 35428782 PMCID: PMC9012871 DOI: 10.1038/s41598-022-09716-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 03/28/2022] [Indexed: 11/27/2022] Open
Abstract
Comparative studies of naturally occurring canine cancers have provided new insight into many areas of cancer research. Development and validation of circulating tumor DNA (ctDNA) analysis in pet dogs can help address diagnostic needs in veterinary as well as human oncology. Dogs have high incidence of naturally occurring spontaneous cancers, demonstrate molecular heterogeneity and clonal evolution during therapy, allow serial sampling of blood from the same individuals during the course of disease progression, and have relatively compressed intervals for disease progression amenable to longitudinal studies. Here, we present a feasibility study of ctDNA analysis performed in 48 dogs including healthy dogs and dogs with either benign splenic lesions or malignant splenic tumors (hemangiosarcoma) using shallow whole genome sequencing (sWGS) of cell-free DNA. To enable detection and quantification of ctDNA using sWGS, we adapted two informatic approaches and compared their performance for the canine genome. At the time of initial clinical presentation, mean ctDNA fraction in dogs with malignant splenic tumors was 11.2%, significantly higher than dogs with benign lesions (3.2%; p = 0.001). ctDNA fraction was 14.3% and 9.0% in dogs with metastatic and localized disease, respectively (p = 0.227). In dogs treated with surgical resection of malignant tumors, mean ctDNA fraction decreased from 11.0% prior to resection to 7.9% post-resection (p = 0.047 for comparison of paired samples). Our results demonstrate that ctDNA analysis is feasible in dogs with hemangiosarcoma using a cost-effective approach such as sWGS. Additional studies are needed to validate these findings, and determine the role of ctDNA to assess burden of disease and treatment response in dogs with cancer.
Collapse
Affiliation(s)
- Patricia Filippsen Favaro
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
- Department of Surgery and Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Samuel D Stewart
- Ethos Veterinary Health, Woburn, MA, USA
- Ethos Discovery, San Diego, CA, USA
| | - Bradon R McDonald
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
- Department of Surgery and Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Jacob Cawley
- Ethos Veterinary Health, Woburn, MA, USA
- Ethos Discovery, San Diego, CA, USA
| | | | - Shukmei Wong
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | | | - Jeffrey M Trent
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA
| | - Chand Khanna
- Ethos Veterinary Health, Woburn, MA, USA.
- Ethos Discovery, San Diego, CA, USA.
| | - Muhammed Murtaza
- Translational Genomics Research Institute (TGen), Phoenix, AZ, USA.
- Department of Surgery and Center for Human Genomics and Precision Medicine, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
14
|
Hayashi H, Yonesaka K, Nakamura A, Fujimoto D, Azuma K, Sakata S, Tachihara M, Ikeda S, Yokoyama T, Hataji O, Yano Y, Hirano K, Daga H, Okada H, Chiba Y, Sakai K, Nishio K, Yamamoto N, Nakagawa K. Alternating Therapy with Osimertinib and Afatinib for Treatment-Naive Patients with EGFR-Mutated Advanced Non–Small Cell Lung Cancer: A Single-Group, Open-Label Phase 2 Trial (WJOG10818L). Lung Cancer 2022; 168:38-45. [DOI: 10.1016/j.lungcan.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/01/2022] [Accepted: 04/09/2022] [Indexed: 12/16/2022]
|
15
|
Nakano K, Koh Y, Yamamichi G, Yumiba S, Tomiyama E, Matsushita M, Hayashi Y, Wang C, Ishizuya Y, Yamamoto Y, Kato T, Hatano K, Kawashima A, Ujike T, Fujita K, Kiyotani K, Katayama K, Yamaguchi R, Imoto S, Imamura R, Nonomura N, Uemura M. Perioperative Circulating Tumor DNA Enables Identification of Patients with Poor Prognosis in Upper Tract Urothelial Carcinoma. Cancer Sci 2022; 113:1830-1842. [PMID: 35293110 PMCID: PMC9128184 DOI: 10.1111/cas.15334] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 02/15/2022] [Accepted: 02/26/2022] [Indexed: 11/30/2022] Open
Abstract
Perioperative systemic chemotherapy improves the prognosis of upper tract urothelial carcinoma (UTUC). The first objective of this study is to verify whether perioperative circulating tumor DNA (ctDNA) analysis using a pan-cancer gene panel and next-generation sequencing can identify patients with poor prognosis who require perioperative chemotherapy. Secondly, we will investigate whether ctDNA is useful for minimal residual disease (MRD) detection and treatment monitoring in UTUC. This study included fifty patients with untreated UTUC, including 43 cases of localized UTUC. We performed targeted ultradeep sequencing of plasma cfDNA and buffy coat DNA and whole-exome sequencing of cancer tissues, allowing exclusion of possible false positives. We attempted to stratify the prognosis according to the perioperative ctDNA levels in patients with localized UTUC. In patients with metastatic UTUC, ctDNA was evaluated before, during, and after systemic treatment. Twenty-three (46%) of 50 patients with untreated UTUC were ctDNA-positive, and 17 (40%) of 43 patients with localized UTUC were ctDNA-positive. Of the detected TP53 mutations, 19% were false-positive due to clonal hematopoiesis of indeterminate potential (CHIP). Among preoperative risk factors, only the preoperative ctDNA fraction>2% was a significant and independent risk factor associated with worse recurrence-free survival (RFS). Furthermore, the existence of ctDNA early point after the operation was significantly associated with worse RFS, suggesting the presence of MRD. ctDNA also showed potential as a real-time marker for systemic therapy in patients with metastatic UTUC. Detection of ctDNA may indicate potential metastasis and guide decisions of perioperative chemotherapy.
Collapse
Affiliation(s)
- Kosuke Nakano
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoko Koh
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Gaku Yamamichi
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Satoru Yumiba
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eisuke Tomiyama
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Makoto Matsushita
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yujiro Hayashi
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Cong Wang
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yu Ishizuya
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshiyuki Yamamoto
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Taigo Kato
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Koji Hatano
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Atsunari Kawashima
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takeshi Ujike
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazutoshi Fujita
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kazuma Kiyotani
- Project for Immunogenomics, Cancer Precision Medicine Center, Japanese Foundation for Cancer Research, Tokyo, 135-8550, Japan
| | - Kotoe Katayama
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo
| | - Rui Yamaguchi
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo.,Division of Cancer Systems Biology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa-ku, Nagoya, Aichi, Japan.,Division of Cancer Informatics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Aichi, Japan
| | - Seiya Imoto
- Division of Health Medical Intelligence, Human Genome Center, The Institute of Medical Science, The University of Tokyo
| | - Ryoichi Imamura
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Norio Nonomura
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Motohide Uemura
- Department of Urology, Osaka University Graduate School of Medicine, Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
16
|
Small cell lung cancer transformation: From pathogenesis to treatment. Semin Cancer Biol 2022; 86:595-606. [PMID: 35276343 DOI: 10.1016/j.semcancer.2022.03.006] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 02/05/2023]
Abstract
Small cell lung cancer (SCLC) is a type of neuroendocrine tumor with high malignancy and poor prognosis. Besides the de novo SCLC, there is transformed SCLC, which has similar characteristics of pathological morphology, molecular characteristics, clinical manifestations and drug sensitivity. However, de novo SCLC and transformed SCLC have different pathogenesis and tumor microenvironment. SCLC transformation is one of the mechanisms of resistance to chemotherapy, immunotherapy, and targeted therapy in NSCLC. Two hypotheses have been used to explain the pathogenesis of SCLC transformation. Although SCLC transformation is not common in clinical practice, it has been repeatedly identified in many small patient series and case reports. It usually occurs in epidermal growth factor receptor (EGFR) mutant lung adenocarcinoma after treatment with tyrosine kinase inhibitors (TKIs). SCLC transformation can also occur in anaplastic lymphoma kinase (ALK)-positive lung cancer after treatment with ALK inhibitors and in wild-type EGFR or ALK NSCLC treated with immunotherapy. Chemotherapy was previously used to treat transformed SCLC, yet it is associated with an unsatisfactory prognosis. We comprehensively review the advancements in transformed SCLC, including clinical and pathological characteristics, and the potential effective treatment after SCLC transformation, aiming to give a better understanding of transformed SCLC and provide support for clinical uses.
Collapse
|
17
|
Xu J, Pu Y, Lin R, Xiao S, Fu Y, Wang T. PEAC: An Ultrasensitive and Cost-Effective MRD Detection System in Non-small Cell Lung Cancer Using Plasma Specimen. Front Med (Lausanne) 2022; 9:822200. [PMID: 35308511 PMCID: PMC8928926 DOI: 10.3389/fmed.2022.822200] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Circulating tumor DNA (ctDNA), a tumor-derived fraction of cell-free DNA (cfDNA), has emerged as a promising marker in targeted therapy, immunotherapy, and minimal residual disease (MRD) monitoring in postsurgical patients. However, ctDNA level in early-stage cancers and postsurgical patients is very low, which posed many technical challenges to improve the detection rate and sensitivity, especially in the clinical practice of MRD detection. These challenges usually include insufficient DNA input amount, limit of detection (LOD), and high experimental costs. To resolve these challenges, we developed an ultrasensitive ctDNA MRD detection system in this study, namely PErsonalized Analysis of Cancer (PEAC), to simultaneously detect up to 37 mutations, which account for 70–80% non-small cell lung cancer (NSCLC) driver mutations from low plasma sample volume and enables LOD of 0.01% at a single-site level. We demonstrated the high performance achieved by PEAC on both cfDNA reference standards and clinical plasma samples from three NSCLC patient cohorts. For cfDNA reference standards, PEAC achieved a specificity of 99% and a sensitivity of 87% for the mutations at 0.01% allele fraction. In the second cohort, PEAC showed 100% concordance rate between ddPCR and Next-generation sequencing (NGS) among 29 samples. In the third cohort, 22 of 59 patients received EGFR TKI treatment. Among them, three in four patients identified low level actionable gene mutations only by PEAC had partial responses after targeted therapy, demonstrating high ctDNA detection ability of PEAC. Overall, the developed PEAC system can detect the majority of NSCLC driver mutations using 8–10 ml plasma samples, and has the advantages of high detection sensitivity and lower costs compared with the existing technologies such as ddPCR and NGS. These advantages make the PEAC system quite appropriate for ctDNA and MRD detection in early-stage NSCLC and postsurgical recurrence monitoring.
Collapse
Affiliation(s)
- Jianping Xu
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Peking Union Medical College, Beijing, China
- *Correspondence: Jianping Xu
| | - Yue Pu
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Rui Lin
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Shanshan Xiao
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Yingxue Fu
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Tao Wang
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
- Tao Wang
| |
Collapse
|
18
|
Breadner DA, Vincent MD, Correa R, Black M, Warner A, Sanatani M, Bhat V, Morris C, Jones G, Allan A, Palma DA, Raphael J. Exploitation of treatment induced tumor lysis to enhance the sensitivity of ctDNA analysis: A first-in-human pilot study. Lung Cancer 2022; 165:145-151. [PMID: 35124411 DOI: 10.1016/j.lungcan.2022.01.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/30/2021] [Accepted: 01/19/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Blood-based liquid biopsies examining circulating tumour DNA (ctDNA) have increasing applications in non-small cell lung cancer (NSCLC). Limitations in sensitivity remain a barrier to ctDNA replacing tissue-based testing. We hypothesized that testing immediately after starting treatment would yield an increased abundance of ctDNA in plasma because of tumor lysis, allowing for the detection of genetic alterations that were occult in baseline testing. METHODS Three prospective cohorts of patients with stage III/IV NSCLC were enrolled. Cohort 1 (C1) contained patients starting platinum doublet chemoradiation (n = 10) and cohort 2 (C2) initiating platinum doublet cytotoxic chemotherapy ± immunotherapy (n = 10). Cohort 3 (C3) contained patients receiving palliative radiation. Two baseline samples were collected. In C1 and C2, subsequent samples were collected 3, 6, 24 and 48 h post initiation of chemotherapy. Patients in C3 had samples collected immediately prior to the next three radiotherapy fractions. Samples were analyzed for ctDNA using the 36-gene amplicon-based NGS Inivata InVisionFirst®-Lung assay. RESULTS A total of 40 patients were enrolled. Detectable ctDNA was present at baseline in 32 patients (80%), 4 additional patients (50%) had detectable ctDNA in post-treatment samples. Seven patients with detectable ctDNA at baseline (23%) had new genetic alterations detected in post-treatment samples. Mutant molecule numbers increased with treatment in 24 of 31 (77%) pts with detectable ctDNA. ctDNA levels peaked a median of 7 h (IQR:2-26 h) after the initiation of chemotherapy and a median of 2 days (IQR:1-3 days) after radiation was commenced. CONCLUSION ctDNA levels increase in the hours to days after starting treatment. ctDNA testing in the acute post-treatment phase can yield results that were not evident in pre-treatment testing. Application of this principle could improve ctDNA utility as an alternate to tissue-based testing and improve sensitivity for the detection of treatment-resistant clones.(NCT03986463).
Collapse
Affiliation(s)
- Daniel A Breadner
- Division of Medical Oncology, London Regional Cancer Program, 800 Commissioners Road East, N6A5W9 London, Ontario, Canada.
| | - Mark D Vincent
- Division of Medical Oncology, London Regional Cancer Program, 800 Commissioners Road East, N6A5W9 London, Ontario, Canada
| | - Rohann Correa
- Division of Radiation Oncology, London Regional Cancer Program, 800 Commissioners Road East, N6A5W9 London, Ontario, Canada
| | - Morgan Black
- Division of Medical Oncology, London Regional Cancer Program, 800 Commissioners Road East, N6A5W9 London, Ontario, Canada
| | - Andrew Warner
- Division of Radiation Oncology, London Regional Cancer Program, 800 Commissioners Road East, N6A5W9 London, Ontario, Canada
| | - Michael Sanatani
- Division of Medical Oncology, London Regional Cancer Program, 800 Commissioners Road East, N6A5W9 London, Ontario, Canada
| | - Vasudeva Bhat
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, 1151 Richmond St, N6A 5C1 London, Ontario, Canada
| | - Clive Morris
- Inivata Inc., 7020 Kit Creek Road, Suite 140, Research Triangle Park, 27560, NC, United States
| | - Greg Jones
- Inivata Inc., 7020 Kit Creek Road, Suite 140, Research Triangle Park, 27560, NC, United States
| | - Alison Allan
- Department of Anatomy and Cell Biology, Schulich School of Medicine and Dentistry, 1151 Richmond St, N6A 5C1 London, Ontario, Canada
| | - David A Palma
- Division of Radiation Oncology, London Regional Cancer Program, 800 Commissioners Road East, N6A5W9 London, Ontario, Canada
| | - Jacques Raphael
- Division of Medical Oncology, London Regional Cancer Program, 800 Commissioners Road East, N6A5W9 London, Ontario, Canada
| |
Collapse
|
19
|
Kolesar J, Peh S, Thomas L, Baburaj G, Mukherjee N, Kantamneni R, Lewis S, Pai A, Udupa KS, Kumar An N, Rangnekar VM, Rao M. Integration of liquid biopsy and pharmacogenomics for precision therapy of EGFR mutant and resistant lung cancers. Mol Cancer 2022; 21:61. [PMID: 35209919 PMCID: PMC8867675 DOI: 10.1186/s12943-022-01534-8] [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: 11/30/2021] [Accepted: 02/07/2022] [Indexed: 11/22/2022] Open
Abstract
The advent of molecular profiling has revolutionized the treatment of lung cancer by comprehensively delineating the genomic landscape of the epidermal growth factor receptor (EGFR) gene. Drug resistance caused by EGFR mutations and genetic polymorphisms of drug metabolizing enzymes and transporters impedes effective treatment of EGFR mutant and resistant lung cancer. This review appraises current literature, opportunities, and challenges associated with liquid biopsy and pharmacogenomic (PGx) testing as precision therapy tools in the management of EGFR mutant and resistant lung cancers. Liquid biopsy could play a potential role in selection of precise tyrosine kinase inhibitor (TKI) therapies during different phases of lung cancer treatment. This selection will be based on the driver EGFR mutational status, as well as monitoring the development of potential EGFR mutations arising during or after TKIs treatment, since some of these new mutations may be druggable targets for alternative TKIs. Several studies have identified the utility of liquid biopsy in the identification of EGFR driver and acquired resistance with good sensitivities for various blood-based biomarkers. With a plethora of sequencing technologies and platforms available currently, further evaluations using randomized controlled trials (RCTs) in multicentric, multiethnic and larger patient cohorts could enable optimization of liquid-based assays for the detection of EGFR mutations, and support testing of CYP450 enzymes and drug transporter polymorphisms to guide precise dosing of EGFR TKIs.
Collapse
Affiliation(s)
- Jill Kolesar
- Department of Pharmacy Practice & Science, University of Kentucky, Lexington, KY, 40536, USA
| | - Spencer Peh
- Department of Pharmacy Practice & Science, University of Kentucky, Lexington, KY, 40536, USA
| | - Levin Thomas
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Gayathri Baburaj
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Nayonika Mukherjee
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Raveena Kantamneni
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Shirley Lewis
- Department of Radiotherapy and Oncology, Kasturba Medical College, Manipal Comprehensive Cancer Care Centre, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Ananth Pai
- Department of Medical Oncology, Kasturba Medical College, Manipal Comprehensive Cancer Care Centre, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Karthik S Udupa
- Department of Medical Oncology, Kasturba Medical College, Manipal Comprehensive Cancer Care Centre, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Naveena Kumar An
- Department of Surgical Oncology, Kasturba Medical College, Manipal Comprehensive Cancer Care Centre, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Vivek M Rangnekar
- Markey Cancer Centre and Department of Radiation Medicine, University of Kentucky, Lexington, KY, 40536, USA
| | - Mahadev Rao
- Department of Pharmacy Practice, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
20
|
Canale M, Andrikou K, Priano I, Cravero P, Pasini L, Urbini M, Delmonte A, Crinò L, Bronte G, Ulivi P. The Role of TP53 Mutations in EGFR-Mutated Non-Small-Cell Lung Cancer: Clinical Significance and Implications for Therapy. Cancers (Basel) 2022; 14:cancers14051143. [PMID: 35267450 PMCID: PMC8909869 DOI: 10.3390/cancers14051143] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/29/2022] [Accepted: 02/16/2022] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Non-Small-Cell Lung Cancer (NSCLC) is the primary cause of cancer-related death worldwide. Patients carrying Epidermal Growth Factor Receptor (EGFR) mutations usually benefit from targeted therapy treatment. Nonetheless, primary or acquired resistance mechanisms lead to treatment discontinuation and disease progression. Tumor protein 53 (TP53) mutations are the most common mutations in NSCLC, and several reports highlighted a role for these mutations in influencing prognosis and responsiveness to EGFR targeted therapy. In this review, we discuss the emerging data about the role of TP53 in predicting EGFR mutated NSCLC patients’ prognosis and responsiveness to targeted therapy. Abstract Non-Small-Cell Lung Cancer (NSCLC) is the primary cause of cancer-related death worldwide. Oncogene-addicted patients usually benefit from targeted therapy, but primary and acquired resistance mechanisms inevitably occur. Tumor protein 53 (TP53) gene is the most frequently mutated gene in cancer, including NSCLC. TP53 mutations are able to induce carcinogenesis, tumor development and resistance to therapy, influencing patient prognosis and responsiveness to therapy. TP53 mutants present in different forms, suggesting that different gene alterations confer specific acquired protein functions. In recent years, many associations between different TP53 mutations and responses to Epidermal Growth Factor Receptor (EGFR) targeted therapy in NSCLC patients have been found. In this review, we discuss the current landscape concerning the role of TP53 mutants to guide primary and acquired resistance to Tyrosine-Kinase Inhibitors (TKIs) EGFR-directed, investigating the possible mechanisms of TP53 mutants within the cellular compartments. We also discuss the role of the TP53 mutations in predicting the response to targeted therapy with EGFR-TKIs, as a possible biomarker to guide patient stratification for treatment.
Collapse
Affiliation(s)
- Matteo Canale
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (M.U.); (P.U.)
| | - Kalliopi Andrikou
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (K.A.); (I.P.); (A.D.); (L.C.); (G.B.)
| | - Ilaria Priano
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (K.A.); (I.P.); (A.D.); (L.C.); (G.B.)
| | - Paola Cravero
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (K.A.); (I.P.); (A.D.); (L.C.); (G.B.)
- Correspondence: (P.C.); (L.P.)
| | - Luigi Pasini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (M.U.); (P.U.)
- Correspondence: (P.C.); (L.P.)
| | - Milena Urbini
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (M.U.); (P.U.)
| | - Angelo Delmonte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (K.A.); (I.P.); (A.D.); (L.C.); (G.B.)
| | - Lucio Crinò
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (K.A.); (I.P.); (A.D.); (L.C.); (G.B.)
| | - Giuseppe Bronte
- Department of Medical Oncology, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (K.A.); (I.P.); (A.D.); (L.C.); (G.B.)
| | - Paola Ulivi
- Biosciences Laboratory, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, 47014 Meldola, Italy; (M.C.); (M.U.); (P.U.)
| |
Collapse
|
21
|
Li H, Ma ZL, Li B, Pan YJ, Xiang JQ, Zhang YW, Sun YH, Hou T, Lizaso A, Chen Y, Li X, Hu H. Potential utility of longitudinal somatic mutation and methylation profiling for predicting molecular residual disease in postoperative non-small cell lung cancer patients. Cancer Med 2021; 10:8377-8386. [PMID: 34664796 PMCID: PMC8633238 DOI: 10.1002/cam4.4339] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 06/19/2021] [Accepted: 09/06/2021] [Indexed: 11/08/2022] Open
Abstract
GROWING EFFORTS ARE BEING INVESTED IN INVESTIGATING VARIOUS MOLECULAR APPROACHES TO DETECT MINIMAL RESIDUAL DISEASE (MRD) AND PREDICT DISEASE RECURRENCE. IN OUR STUDY, WE INVESTIGATED THE UTILITY OF PARALLEL LONGITUDINAL ANALYSIS OF MUTATION AND DNA METHYLATION PROFILES FOR PREDICTING MRD IN POSTOPERATIVE NON-SMALL-CELL LUNG CANCER (NSCLC) PATIENTS. TUMOR TISSUES AND LONGITUDINAL BLOOD SAMPLES WERE OBTAINED FROM 65 PATIENTS WITH RESECTED STAGE IA-IIIB NSCLC. SOMATIC MUTATION AND DNA METHYLATION PROFILING WERE PERFORMED USING ULTRA-DEEP TARGETED SEQUENCING AND TARGETED BISULFITE SEQUENCING, RESPECTIVELY. DYNAMIC CHANGES IN PLASMA-BASED MUTATION AND TUMOR-INFORMED METHYLATION PROFILES, REFLECTED AS MRD SCORE, WERE OBSERVED FROM BEFORE SURGERY (BASELINE) TO POSTOPERATIVE FOLLOW-UP, REFLECTING THE DECREASE IN TUMOR BURDEN OF THE PATIENTS WITH RESECTED NSCLC. MUTATIONS WERE DETECTED FROM PLASMA SAMPLES IN 63% OF THE PATIENTS AT BASELINE, WHICH SIGNIFICANTLY REDUCED TO 23-25% DURING POST-OPERATIVE FOLLOW-UPS. MRD SCORE POSITIVE RATE WAS 95.7% AT BASELINE, WHICH REDUCED TO 74% AT THE FIRST AND 70% AT THE SECOND FOLLOW-UP. AMONG THE 5 RELAPSED PATIENTS WITH PARALLEL LONGITUDINAL ANALYSIS OF MUTATION AND METHYLATION PROFILE, ELEVATED MRD SCORE WAS OBSERVED AT FOLLOW-UP BETWEEN 0.5-7 MONTHS PRIOR TO RADIOLOGIC RECURRENCE FOR ALL 5 PATIENTS. OF THEM, 4 PATIENTS ALSO HAD CONCOMITANT INCREASE IN ALLELIC FRACTION OF MUTATIONS IN AT LEAST 1 FOLLOW-UP TIME POINT, BUT ONE PATIENT HAD NO MUTATION DETECTED THROUGHOUT ALL FOLLOW-UPS. OUR RESULTS DEMONSTRATE THAT LONGITUDINAL PROFILING OF MUTATION AND DNA METHYLATION MAY HAVE POTENTIAL FOR DETECTING MRD AND PREDICTING RECURRENCE IN POSTOPERATIVE NSCLC PATIENTS.
Collapse
Affiliation(s)
- Hang Li
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China.,Institute of Thoracic Oncology, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ze-Lin Ma
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China.,Institute of Thoracic Oncology, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Bin Li
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China.,Institute of Thoracic Oncology, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yun-Jian Pan
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China.,Institute of Thoracic Oncology, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jia-Qing Xiang
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China.,Institute of Thoracic Oncology, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ya-Wei Zhang
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China.,Institute of Thoracic Oncology, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi-Hua Sun
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China.,Institute of Thoracic Oncology, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ting Hou
- Burning Rock Biotech, Guangzhou, China
| | | | - Yan Chen
- Burning Rock Biotech, Guangzhou, China
| | - Xi Li
- Burning Rock Biotech, Guangzhou, China
| | - Hong Hu
- Department of Thoracic Surgery and State Key Laboratory of Genetic Engineering, Fudan University Shanghai Cancer Center, Shanghai, China.,Institute of Thoracic Oncology, Fudan University, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Zhang M, Chen J. [Advances in Clinical Application of Liquid Biopsy in Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2021; 24:723-728. [PMID: 34696544 PMCID: PMC8560984 DOI: 10.3779/j.issn.1009-3419.2021.102.33] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Lung cancer, with the highest incidence in China, is the leading cause of death in cancer patients. Of these, about 85% are patients with non-small cell lung cancer (NSCLC). Therefore, the diagnosis and treatment of patients with lung cancer have always been a top priority nowadays. Fluid biopsy has many advantages, such as safety, convenience, repeatability, low trauma and so on, which are not available in traditional invasive biopsy. In recent years, with the rapid progress of molecular biological detection technology, fluid biopsy, as a new technology, has become the focus of attention. What's more, it contributes to the development of precision treatment and individualized treatment of lung cancer. Liquid biopsy mainly detects circulating tumor DNA (ctDNA), circulating tumor cells (CTCs) and exosomes in peripheral blood. We will make an introduce to the detection and clinical applications of ctDNA, CTCs and exocrine in this article, in order that it can provide insights into future clinical treatment for NSCLC.
.
Collapse
Affiliation(s)
- Min Zhang
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian 116000, China
| | - Jun Chen
- Department of Oncology, The Second Hospital of Dalian Medical University, Dalian 116000, China
| |
Collapse
|
23
|
Reita D, Pabst L, Pencreach E, Guérin E, Dano L, Rimelen V, Voegeli AC, Vallat L, Mascaux C, Beau-Faller M. Molecular Mechanism of EGFR-TKI Resistance in EGFR-Mutated Non-Small Cell Lung Cancer: Application to Biological Diagnostic and Monitoring. Cancers (Basel) 2021; 13:4926. [PMID: 34638411 PMCID: PMC8507869 DOI: 10.3390/cancers13194926] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/20/2021] [Accepted: 09/23/2021] [Indexed: 12/21/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is the most common cancer in the world. Activating epidermal growth factor receptor (EGFR) gene mutations are a positive predictive factor for EGFR tyrosine kinase inhibitors (TKIs). For common EGFR mutations (Del19, L858R), the standard first-line treatment is actually third-generation TKI, osimertinib. In the case of first-line treatment by first (erlotinib, gefitinib)- or second-generation (afatinib) TKIs, osimertinib is approved in second-line treatment for patients with T790M EGFR mutation. Despite the excellent disease control results with EGFR TKIs, acquired resistance inevitably occurs and remains a biological challenge. This leads to the discovery of novel biomarkers and possible drug targets, which vary among the generation/line of EGFR TKIs. Besides EGFR second/third mutations, alternative mechanisms could be involved, such as gene amplification or gene fusion, which could be detected by different molecular techniques on different types of biological samples. Histological transformation is another mechanism of resistance with some biological predictive factors that needs tumor biopsy. The place of liquid biopsy also depends on the generation/line of EGFR TKIs and should be a good candidate for molecular monitoring. This article is based on the literature and proposes actual and future directions in clinical and translational research.
Collapse
Affiliation(s)
- Damien Reita
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospital, CEDEX, 67098 Strasbourg, France; (D.R.); (E.P.); (E.G.); (L.D.); (V.R.); (A.-C.V.); (L.V.)
- Bio-imagery and Pathology (LBP), UMR CNRS 7021, Strasbourg University, 67400 Illkirch-Graffenstaden, France
| | - Lucile Pabst
- Department of Pneumology, Strasbourg University Hospital, CEDEX, 67091 Strasbourg, France; (L.P.); (C.M.)
| | - Erwan Pencreach
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospital, CEDEX, 67098 Strasbourg, France; (D.R.); (E.P.); (E.G.); (L.D.); (V.R.); (A.-C.V.); (L.V.)
- INSERM U1113, IRFAC, Strasbourg University, 67000 Strasbourg, France
| | - Eric Guérin
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospital, CEDEX, 67098 Strasbourg, France; (D.R.); (E.P.); (E.G.); (L.D.); (V.R.); (A.-C.V.); (L.V.)
- INSERM U1113, IRFAC, Strasbourg University, 67000 Strasbourg, France
| | - Laurent Dano
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospital, CEDEX, 67098 Strasbourg, France; (D.R.); (E.P.); (E.G.); (L.D.); (V.R.); (A.-C.V.); (L.V.)
| | - Valérie Rimelen
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospital, CEDEX, 67098 Strasbourg, France; (D.R.); (E.P.); (E.G.); (L.D.); (V.R.); (A.-C.V.); (L.V.)
| | - Anne-Claire Voegeli
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospital, CEDEX, 67098 Strasbourg, France; (D.R.); (E.P.); (E.G.); (L.D.); (V.R.); (A.-C.V.); (L.V.)
| | - Laurent Vallat
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospital, CEDEX, 67098 Strasbourg, France; (D.R.); (E.P.); (E.G.); (L.D.); (V.R.); (A.-C.V.); (L.V.)
| | - Céline Mascaux
- Department of Pneumology, Strasbourg University Hospital, CEDEX, 67091 Strasbourg, France; (L.P.); (C.M.)
- INSERM U1113, IRFAC, Strasbourg University, 67000 Strasbourg, France
| | - Michèle Beau-Faller
- Department of Biochemistry and Molecular Biology, Strasbourg University Hospital, CEDEX, 67098 Strasbourg, France; (D.R.); (E.P.); (E.G.); (L.D.); (V.R.); (A.-C.V.); (L.V.)
- INSERM U1113, IRFAC, Strasbourg University, 67000 Strasbourg, France
| |
Collapse
|
24
|
Romero A, Serna-Blasco R, Calvo V, Provencio M. Use of Liquid Biopsy in the Care of Patients with Non-Small Cell Lung Cancer. Curr Treat Options Oncol 2021; 22:86. [PMID: 34424428 DOI: 10.1007/s11864-021-00882-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2021] [Indexed: 12/17/2022]
Abstract
OPINION STATEMENT Recent technological advances have enabled the development of liquid biopsy-based approaches, which have revolutionized the diagnostic world. The analysis of circulating tumor DNA (ctDNA) has several clinical applications. First, ctDNA genotyping is becoming widely used for non-invasive biomarker testing. Of note, in lung cancer patients in whom biopsies are difficult to obtain, ctDNA has led to significant improvement in the diagnosis and identification of therapeutic targets. In addition, ctDNA quantification over the course of the disease can be useful for tumor response to treatment monitoring and for early detection of resistance mutations. ctDNA levels per se are also of prognostic significance and could be used to tailor treatments. Finally, improvements in assay sensitivity are facilitating the development of liquid biopsy-based tests for the detection of ctDNA at very low allele frequencies (AFs), which can be used for the measurement of minimal residual disease and ultimately for the development of strategies (by complementing imaging techniques) aimed to improve the efficiency of lung cancer screening programs.
Collapse
Affiliation(s)
- Atocha Romero
- Molecular Oncology Laboratory, Biomedical Sciences Research Institute, Puerta de Hierro-Majadahonda University Hospital, Madrid, Spain.
| | - Roberto Serna-Blasco
- Molecular Oncology Laboratory, Biomedical Sciences Research Institute, Puerta de Hierro-Majadahonda University Hospital, Madrid, Spain
| | - Virginia Calvo
- Medical Oncology Department, Hospital Puerta de Hierro-Majadahonda University Hospital, Calle Joaquín Rodrigo, 1, 28222, Madrid, Majadahonda, Spain
| | - Mariano Provencio
- Medical Oncology Department, Hospital Puerta de Hierro-Majadahonda University Hospital, Calle Joaquín Rodrigo, 1, 28222, Madrid, Majadahonda, Spain.
| |
Collapse
|
25
|
Di Capua D, Bracken-Clarke D, Ronan K, Baird AM, Finn S. The Liquid Biopsy for Lung Cancer: State of the Art, Limitations and Future Developments. Cancers (Basel) 2021; 13:cancers13163923. [PMID: 34439082 PMCID: PMC8391249 DOI: 10.3390/cancers13163923] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary During the development and progression of lung tumors, processes such as necrosis and vascular invasion shed tumor cells or cellular components into various fluid compartments. Liquid biopsies consist of obtaining a bodily fluid, typically peripheral blood, in order to isolate and investigate these shed tumor constituents. Circulating tumor cells (CTCs) are one such constituent, which can be isolated from blood and can act as a diagnostic aid and provide valuable prognostic information. Liquid-based biopsies may also have a potential future role in lung cancer screening. Circulating tumor DNA (ctDNA) is found in small quantities in blood and, with the recent development of sensitive molecular and sequencing technologies, can be used to directly detect actionable genetic alterations or monitor for resistance mutations and guide clinical management. While potential benefits of liquid biopsies are promising, they are not without limitations. In this review, we summarize the current state and limitations of CTCs and ctDNA and possible future directions. Abstract Lung cancer is a leading cause of cancer-related deaths, contributing to 18.4% of cancer deaths globally. Treatment of non-small cell lung carcinoma has seen rapid progression with targeted therapies tailored to specific genetic drivers. However, identifying genetic alterations can be difficult due to lack of tissue, inaccessible tumors and the risk of complications for the patient with serial tissue sampling. The liquid biopsy provides a minimally invasive method which can obtain circulating biomarkers shed from the tumor and could be a safer alternative to tissue biopsy. While tissue biopsy remains the gold standard, liquid biopsies could be very beneficial where serial sampling is required, such as monitoring disease progression or development of resistance mutations to current targeted therapies. Liquid biopsies also have a potential role in identifying patients at risk of relapse post treatment and as a component of future lung cancer screening protocols. Rapid developments have led to multiple platforms for isolating circulating tumor cells (CTCs) and detecting circulating tumor DNA (ctDNA); however, standardization is lacking, especially in lung carcinoma. Additionally, clonal hematopoiesis of uncertain clinical significance must be taken into consideration in genetic sequencing, as it introduces the potential for false positives. Various biomarkers have been investigated in liquid biopsies; however, in this review, we will concentrate on the current use of ctDNA and CTCs, focusing on the clinical relevance, current and possible future applications and limitations of each.
Collapse
Affiliation(s)
- Daniel Di Capua
- Department of Histopathology, St. James’s Hospital, D08NHY1 Dublin, Ireland;
| | - Dara Bracken-Clarke
- Department of Medical Oncology, St. James’ Hospital, D08NHY1 Dublin, Ireland;
| | - Karine Ronan
- Faculty of Medicine, University College Dublin, D04V1W8 Dublin, Ireland;
| | - Anne-Marie Baird
- School of Medicine, Trinity Translational Medicine Institute, Trinity College, D02PN40 Dublin, Ireland;
| | - Stephen Finn
- Department of Histopathology, St. James’s Hospital, D08NHY1 Dublin, Ireland;
- Correspondence:
| |
Collapse
|
26
|
Su Z, Chen M, Ding R, Shui L, Zhao Q, Luo W. Long non‑coding RNA HCG11 suppresses the malignant phenotype of non‑small cell lung cancer cells by targeting a miR‑875/SATB2 axis. Mol Med Rep 2021; 24:552. [PMID: 34080031 PMCID: PMC8188752 DOI: 10.3892/mmr.2021.12191] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 02/04/2021] [Indexed: 02/07/2023] Open
Abstract
Long non‑coding RNAs (lncRNAs) are involved in the development and progression of a variety of diseases. However, the role of the lncRNA HLA complex group 11 (HCG11) in non‑small cell lung cancer (NSCLC) remains unclear. The present study showed that the expression levels of HCG11 were reduced in tumor tissues compared with adjacent normal tissues, and similar results were obtained in experiments using lung cancer cell lines. Additionally, patients with high HCG11 expression had an increased survival rate compared with patients with low HCG11 expression. Further studies have shown that overexpression of HCG11 inhibited NSCLC cell proliferation in vitro and in vivo. Interestingly, it was observed that HCG11 expression was negatively associated with the expression levels of oncogenic microRNA‑875 (miR‑875) in patient specimens. Specifically, HCG11 served as a sponge of miR‑875. Notably, it was determined that special AT‑rich sequence‑binding protein 2 (SATB2) was a direct target gene of miR‑875, and overexpression of miR‑875 largely abrogated the effects of HCG11 in NSCLC cells. In conclusion, HCG11 was shown to suppress the malignant properties of NSCLC cells by targeting a miR‑875/SATB2 axis, and may therefore be a promising target for the treatment of NSCLC.
Collapse
Affiliation(s)
- Zhou Su
- Department of Oncology, Sichuan Mianyang 404 Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Mi Chen
- Department of Oncology, The Third Hospital of Mianyang/Sichuan Mental Health Center, Mianyang, Sichuan 621000, P.R. China
| | - Ruilin Ding
- Institute of Drug Clinical Trial/GCP Center, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Lian Shui
- Department of Oncology, Sichuan Mianyang 404 Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Qingmei Zhao
- Department of Oncology, Sichuan Mianyang 404 Hospital, Mianyang, Sichuan 621000, P.R. China
| | - Wenjuan Luo
- Department of Oncology, Sichuan Mianyang 404 Hospital, Mianyang, Sichuan 621000, P.R. China
| |
Collapse
|
27
|
Sheng J, Wang H, Liu X, Deng Y, Yu Y, Xu P, Shou J, Pan H, Li H, Zhou X, Han W, Sun T, Pan H, Fang Y. Deep Sequencing of T-Cell Receptors for Monitoring Peripheral CD8 + T Cells in Chinese Advanced Non-Small-Cell Lung Cancer Patients Treated With the Anti-PD-L1 Antibody. Front Mol Biosci 2021; 8:679130. [PMID: 34307450 PMCID: PMC8299707 DOI: 10.3389/fmolb.2021.679130] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022] Open
Abstract
Background: Atezolizumab, a high-affinity engineered human anti-PD-L1 antibody, has produced a clinical benefit for patients with advanced non-small-cell lung cancer (NSCLC). However, associated with T-cell regulation, the immunomodulatory effect of PD-L1 blockade and its biomarker in peripheral immunity remains elusive. Methods: In a prospective cohort with 12 Chinese advanced NSCLC patients who received atezolizumab 1,200 mg every 3 weeks as a second-line treatment, blood samples were obtained before and 6 weeks after atezolizumab initiation, and when disease progression was confirmed. Patients were classified into a response or progression group according to response evaluation criteria in solid tumors (RECIST) 1.1. Fresh peripheral blood mononuclear cells (PBMCs) from patients were stained with antihuman CD3, CD8, and PD-1 antibodies for flow cytometry analysis. T-cell receptor (TCR)-β chains of CD8+ T cells were analyzed by next-generation sequencing (NGS) at the deep level. Diversity, clonality, and similarity of TCR have been calculated before and after treatment in both groups. Results: Clonal expansion with high PD-1 expression was detected in all patients' peripheral CD8+ T cells before the treatment of atezolizumab. Unlike the progression group, the diversity of TCR repertoire and singletons in the TCRβ pool increased over time with atezolizumab administration, and the TCR repertoire dynamically changes in the response group. The percentage of CD8+ PD-1high terminal exhausted T cells declined in the response group after the PD-L1 blockade. Two patterns of TCR changes among patients who received PD-L1-targeted immunotherapy were observed. Conclusions: Deep sequencing of the T-cell receptors confirmed the existence of CD8+ PD-1high T cells with an exhaustion phenotype in Chinese NSCLC patients. Our study demonstrated that efficient anti-PD-L1 therapy could reshape the TCR repertoire for antitumor patients. Furthermore, singleton frequency may help us select patients who are sensitive to anti-PD-L1 immunotherapy.
Collapse
Affiliation(s)
- Jin Sheng
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Huadi Wang
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Xiao Liu
- Department of Human Genetics, University of Chicago, Chicago, IL, United States
| | - Yunyun Deng
- Hangzhou ImmuQuad Biotechnologies, LLC, Hangzhou, China
| | - Yingying Yu
- Hangzhou ImmuQuad Biotechnologies, LLC, Hangzhou, China
| | - Pengfei Xu
- Hangzhou ImmuQuad Biotechnologies, LLC, Hangzhou, China
| | - Jiawei Shou
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Hong Pan
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Hongsen Li
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Xiaoyun Zhou
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Weidong Han
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China.,Laboratory of Cancer Biology, Institute of Clinical Science, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Tao Sun
- Hangzhou ImmuQuad Biotechnologies, LLC, Hangzhou, China.,Zhejiang-California International Nano-Systems Institute, Zhejiang University, Hangzhou, China
| | - Hongming Pan
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| | - Yong Fang
- Department of Medical Oncology, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
28
|
Underhill HR. Leveraging the Fragment Length of Circulating Tumour DNA to Improve Molecular Profiling of Solid Tumour Malignancies with Next-Generation Sequencing: A Pathway to Advanced Non-invasive Diagnostics in Precision Oncology? Mol Diagn Ther 2021; 25:389-408. [PMID: 34018157 PMCID: PMC8249304 DOI: 10.1007/s40291-021-00534-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 12/20/2022]
Abstract
Circulating cell-free DNA (ccfDNA) has emerged as a promising diagnostic tool in oncology. Identification of tumour-derived ccfDNA (i.e. circulating tumour DNA [ctDNA]) provides non-invasive access to a malignancy’s molecular landscape to diagnose, inform therapeutic strategies, and monitor treatment efficacy. Current applications of ccfDNA to detect somatic mutations, however, have been largely constrained to tumour-informed searches and identification of common mutations because of the interaction between ctDNA signal and next-generation sequencing (NGS) noise. Specifically, the low allele frequency of ctDNA associated with non-metastatic and early-stage lesions may be indistinguishable from artifacts that accrue during sample preparation and NGS. Thus, using ccfDNA to achieve non-invasive and personalized molecular profiling to optimize individual patient care is a highly sought goal that remains limited in clinical practice. There is growing evidence, however, that further advances in the field of ccfDNA diagnostics may be achieved by improving detection of somatic mutations through leveraging the inherently shorter fragment lengths of ctDNA compared to non-neoplastic ccfDNA. Here, the origins and rationale for seeking to improve the mutation-based detection of ctDNA by using ccfDNA size profiling are reviewed. Subsequently, in vitro and in silico methods to enrich for a target ccfDNA fragment length are detailed to identify current practices and provide perspective into the potential of using ccfDNA size profiling to impact clinical applications in oncology.
Collapse
Affiliation(s)
- Hunter R Underhill
- Division of Medical Genetics, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA. .,Department of Radiology, University of Utah, Salt Lake City, UT, USA. .,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
29
|
Pizzutilo EG, Pedrani M, Amatu A, Ruggieri L, Lauricella C, Veronese SM, Signorelli D, Cerea G, Giannetta L, Siena S, Sartore-Bianchi A. Liquid Biopsy for Small Cell Lung Cancer either De Novo or Transformed: Systematic Review of Different Applications and Meta-Analysis. Cancers (Basel) 2021; 13:2265. [PMID: 34066817 PMCID: PMC8125928 DOI: 10.3390/cancers13092265] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 05/02/2021] [Accepted: 05/03/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The potential added value of liquid biopsy (LB) is not well determined in the case of small cell lung cancer (SCLC), an aggressive tumor that can occur either de novo or from the histologic transformation of non-small cell lung cancer (NSCLC). METHODS A systematic review of studies adopting LB in patients with SCLC have been performed to assess the clinical utility of circulating tumor DNA (ctDNA) or circulating tumor cells (CTCs). RESULTS After a screening of 728 records, 62 studies (32 evaluating CTCs, 27 ctDNA, and 3 both) met predetermined eligibility criteria. Only four studies evaluated LB in the diagnostic setting for SCLC, while its prognostic significance was evaluated in 38 studies and prominently supported by both ctDNA and CTCs. A meta-analysis of 11 studies as for CTCs enumeration showed an HR for overall survival of 2.63 (1.71-4.05), with a potential publication bias. The feasibility of tumor genomic profiling and the predictive role of LB in terms of response/resistance to chemotherapy was assessed in 11 and 24 studies, respectively, with greater consistency for those regarding ctDNA. Intriguingly, several case reports suggest that LB can indirectly capture the transition to SCLC in NSCLC treated with EGFR tyrosine kinase inhibitors. CONCLUSIONS While dedicated trials are needed, LB holds potential clinical roles in both de novo and transformed SCLC. CtDNA analysis appears the most valuable and practicable tool for both disease monitoring and genomic profiling.
Collapse
Affiliation(s)
- Elio Gregory Pizzutilo
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Martino Pedrani
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
| | - Lorenzo Ruggieri
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Calogero Lauricella
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
| | - Silvio Marco Veronese
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
| | - Diego Signorelli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
| | - Giulio Cerea
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
| | - Laura Giannetta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy; (E.G.P.); (M.P.); (A.A.); (L.R.); (C.L.); (S.M.V.); (D.S.); (G.C.); (L.G.); (S.S.)
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, 20122 Milan, Italy
| |
Collapse
|
30
|
Januszyk P, Januszyk K, Wierzbik-Strońska M, Boroń D, Grabarek B. Analysis of the Differences in the Expression of mRNAs and miRNAs Associated with Drug Resistance in Endometrial Cancer Cells Treated with Salinomycin. Curr Pharm Biotechnol 2021; 22:541-548. [DOI: 10.2174/1389201021666200629151008] [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: 02/04/2020] [Revised: 03/26/2020] [Accepted: 05/29/2020] [Indexed: 01/03/2023]
Abstract
Background:
It is important to understand the molecular mechanisms involved in cancer
drug resistance and to study the activity of new drugs, e.g. salinomycin.
Objective:
The purpose of the study was to analyze changes in the expression of genes associated with drug resistance in the
Ishikawa endometrial cancer cell line when treated with salinomycin. In addition, changes in the level of miRNA potentially
regulating these mRNAs were evaluated.
Materials and Methods:
Endometrial cancer cells were treated with 1 μM of salinomycin for 12, 24
and 48 hours periods. Untreated cells were a control culture. The molecular analysis consists of mRNA
and miRNA microarray analysis and the RTqPCR technique.
Results:
The following was observed about the number of mRNAs differentiating the cell culture exposed
to the drug compared to a control culture: H-12 vs. C - 9 mRNAs, H_24 vs. C - 6 mRNAs, and
H_48 vs. C - 1 mRNA. It was noted that 4 of the 9 differentiating mRNAs were characteristic for 12
hours of exposure to salinomycin and they correspond to the following genes: TUFT1, ABCB1,
MTMR11, and MX2. After 24 hours, 2 mRNAs were characteristic for this time of incubation cells
with salinomycin: TUFT1 and MYD88 and after 48 hours, SLC30A5 could also be observed.
Discussion:
The highest differences in expression were indicated for TUFT1, MTMR11, and SLC30A5.
The highest influence probability was determined between TUFT1 and hsa- miR-3188 (FC + 2.48),
MTMR11and has-miR-16 (FC -1.74), and between SLC30A5 and hsa-miR-30d (FC -2.01).
Conclusions:
Salinomycin induces changes in the activity of mRNA and miRNA participating in drug
resistance; however, the observed changes in character are the expected result of anti-cancer treatment.
Collapse
Affiliation(s)
- Piotr Januszyk
- Department of Gynecology and Obstetrics with Gynecologic Oncology, Ludwik Rydygier Memorial Specialized Hospital, Krakow, Poland
| | - Krzysztof Januszyk
- Faculty of Health Science, Public Higher Medical Professional School in Opole, Poland
| | - Magdalena Wierzbik-Strońska
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine, University of Technology in Katowice, Zabrze, Poland
| | - Dariusz Boroń
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine, University of Technology in Katowice, Zabrze, Poland
| | - Beniamin Grabarek
- Department of Histology, Cytophysiology and Embryology, Faculty of Medicine, University of Technology in Katowice, Zabrze, Poland
| |
Collapse
|
31
|
Chen J, Alduais Y, Chen B. Therapeutic and Systemic Adverse Events of Immune Checkpoint Inhibitors Targeting the PD-1/PD-L1 axis for Clinical Management of NSCLC. Cell Transplant 2021; 30:9636897211041587. [PMID: 34606729 PMCID: PMC8493325 DOI: 10.1177/09636897211041587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Non-small-cell lung cancer takes up the majority of lung carcinoma-caused deaths. It is reported that targeting PD-1/PD-L1, a well-known immune evasion checkpoint, can eradicate tumor. Checkpoint inhibitors, such as monoclonal antibodies, are actively employed in cancer treatment. Thus, this review aimed to assess the therapeutic and toxic effects of PD-1/PD-L1 inhibitors in treatment of NSCLC. So far, 6 monoclonal antibodies blocking PD-1/PD-L1 interaction are identified and used in clinical trials and randomized controlled trials for NSCLC therapy. These antibody-based therapies for NSCLC were collected by using search engine PubMed, and articles about the assessment of adverse events were collected by using Google search. Route of administration and dosage are critical parameters for efficient immunotherapy. Although antibodies can improve overall survival and are expected to be target-specific, they can cause systemic adverse effects in the host. Targeting certain biomarkers can limit the toxicity of adverse effects of the antibody-mediated therapy. Clinical experts with knowledge of adverse effects (AEs) of checkpoint inhibitors can help manage and reduce mortalities associated with antibody-based therapy of NSCLC.
Collapse
Affiliation(s)
- Jing Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009, Jiangsu, China
| | - Yaser Alduais
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009, Jiangsu, China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009, Jiangsu, China
| |
Collapse
|
32
|
Esposito Abate R, Frezzetti D, Maiello MR, Gallo M, Camerlingo R, De Luca A, De Cecio R, Morabito A, Normanno N. Next Generation Sequencing-Based Profiling of Cell Free DNA in Patients with Advanced Non-Small Cell Lung Cancer: Advantages and Pitfalls. Cancers (Basel) 2020; 12:E3804. [PMID: 33348595 PMCID: PMC7766403 DOI: 10.3390/cancers12123804] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Lung cancer (LC) is the main cause of death for cancer worldwide and non-small cell lung cancer (NSCLC) represents the most common histology. The discovery of genomic alterations in driver genes that offer the possibility of therapeutic intervention has completely changed the approach to the diagnosis and therapy of advanced NSCLC patients, and tumor molecular profiling has become mandatory for the choice of the most appropriate therapeutic strategy. However, in approximately 30% of NSCLC patients tumor tissue is inadequate for biomarker analysis. The development of highly sensitive next generation sequencing (NGS) technologies for the analysis of circulating cell-free DNA (cfDNA) is emerging as a valuable alternative to assess tumor molecular landscape in case of tissue unavailability. Additionally, cfDNA NGS testing can better recapitulate NSCLC heterogeneity as compared with tissue testing. In this review we describe the main advantages and limits of using NGS-based cfDNA analysis to guide the therapeutic decision-making process in advanced NSCLC patients, to monitor the response to therapy and to identify mechanisms of resistance early. Therefore, we provide evidence that the implementation of cfDNA NGS testing in clinical research and in the clinical practice can significantly improve precision medicine approaches in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Daniela Frezzetti
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Monica Rosaria Maiello
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Marianna Gallo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Rosa Camerlingo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Rossella De Cecio
- Department of Pathology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy;
| | - Alessandro Morabito
- Department of Thoracic Medical Oncology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy;
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| |
Collapse
|
33
|
Iacovino M, Ciaramella V, Paragliola F, Suarato G, Busiello G, Sparano F. Use of liquid biopsy in monitoring therapeutic resistance in EGFR oncogene addicted NSCLC. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:391-400. [PMID: 36046387 PMCID: PMC9400718 DOI: 10.37349/etat.2020.00024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 11/10/2020] [Indexed: 11/19/2022] Open
Abstract
Liquid biopsy has emerged as a minimally invasive alternative to tumor tissue analysis for the management of lung cancer patients, especially for epidermal growth factor receptor (EGFR) oncogene addicted tumor. In these patients, despite the clear benefits of tyrosine kinase inhibitors therapy, the development of acquired resistance and progressive disease is inevitable in most cases and liquid biopsy is important for molecular characterization at resistance and, being non-invasive, may be useful for disease monitoring. In this review, the authors will focus on the applications of liquid biopsy in EGFR-mutated non small cells lung cancer at diagnosis, during treatment and at progression, describing available data and possible future scenarios.
Collapse
Affiliation(s)
- Marialucia Iacovino
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Vincenza Ciaramella
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Fernando Paragliola
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Gabriella Suarato
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Gesualdina Busiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| | - Francesca Sparano
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania Luigi Vanvitelli, 80131 Naples, Italy
| |
Collapse
|
34
|
Duan J, Xu J, Wang Z, Bai H, Cheng Y, An T, Gao H, Wang K, Zhou Q, Hu Y, Song Y, Ding C, Peng F, Liang L, Hu Y, Huang C, Zhou C, Shi Y, Han J, Wang D, Tian Y, Yang Z, Zhang L, Chuai S, Ye J, Zhu G, Zhao J, Wu YL, Wang J. Refined Stratification Based on Baseline Concomitant Mutations and Longitudinal Circulating Tumor DNA Monitoring in Advanced EGFR-Mutant Lung Adenocarcinoma Under Gefitinib Treatment. J Thorac Oncol 2020; 15:1857-1870. [PMID: 32916309 DOI: 10.1016/j.jtho.2020.08.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/16/2020] [Accepted: 08/04/2020] [Indexed: 02/05/2023]
Abstract
INTRODUCTION The optimal treatment for EGFR-mutant lung adenocarcinoma (LUAD) remains challenging because of intratumor heterogeneity. We aimed to explore a refined stratification model based on the integrated analysis of circulating tumor DNA (ctDNA) tracking. METHODS ctDNA was prospectively collected at baseline and at every 8 weeks in patients with advanced treatment-naive EGFR-mutant LUAD under gefitinib treatment enrolled in a phase 2 trial and analyzed using next-generation sequencing of a 168-gene panel. RESULTS Three subgroups categorized by baseline comutations-EGFR-sensitizing mutations (59, 32.8%), EGFR-sensitizing mutations with tumor suppressor mutations (97, 53.9%), and EGFR-sensitizing mutations with other driver mutations (24, 13.3%)-exhibited distinct progression-free survival (13.2 [11.3-15.2] versus 9.3 [7.6-10.5] versus 4.0 [2.4-9.3] months) and overall survival (32.0 [29.2-41.5] versus 21.7 [19.3-27.0] versus 15.5 [10.5-33.7] months, respectively), providing evidence for initial stratification. A total of 63.7% of the patients achieved week 8 ctDNA clearance, with significant difference noted among the three subgroups (74.5% versus 64.0% versus 29.4%, respectively, p = 0.004, Fisher's exact test). Patients without week 8 ctDNA clearance had worse progression-free survival (clearance versus nonclearance 11.2 [9.9-13.2] versus 7.4 [5.6-9.6] months, p = 0.016, Cox regression], especially in the second subgroup [5.8 (5.6-11.5) months], suggesting the necessity of adaptive stratification during treatment. During follow-up, 56.0% and 20.8% of the patients eventually harbored p.T790M and non-p.T790M mutations, respectively, with a significant difference in non-p.T790M mutations among the three subgroups (7.5% versus 15.7% versus 80.0%, respectively, p < 0.001, Fisher's exact test), giving clues to postline treatment. CONCLUSIONS The patients with baseline comutations and ctDNA nonclearance at first visit might require combined therapy because of the limited survival benefit of EGFR tyrosine kinase inhibitor monotherapy. We proposed a refined stratification mode for the whole-course management of EGFR-mutant LUAD.
Collapse
Affiliation(s)
- Jianchun Duan
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - JiaChen Xu
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Hua Bai
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Ying Cheng
- Division of Thoracic Oncology, Jilin Cancer Hospital, Jilin Province Cancer Hospital, Changchun, People's Republic of China
| | - Tongtong An
- Department of Thoracic Medical Oncology, Peking University School of Oncology, Beijing Cancer Hospital and Institute, Beijing, People's Republic of China
| | - Hongjun Gao
- The 307th Hospital of Chinese People's Liberation Army, Beijing, People's Republic of China
| | - Kai Wang
- Department of Respiratory Medicine, Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Yanping Hu
- Department of Oncology, Hubei Cancer Hospital, Hubei, People's Republic of China
| | - Yong Song
- Nanjing General Hospital of Nanjing Military Command and PLA Bayi Hospital, Nanjing, People's Republic of China
| | - Cuimin Ding
- Department of Respiratory Medicine, The Fourth Hospital of Hebei Medical University, Hebei, People's Republic of China
| | - Feng Peng
- Department of Thoracic Oncology, Cancer Center, West China Hospital, Medical School, Sichuan University, Chengdu, People's Republic of China
| | - Li Liang
- Peking University Third Hospital, Beijing, People's Republic of China
| | - Yi Hu
- Chinese People's Liberation Army General Hospital, Beijing, People's Republic of China
| | - Cheng Huang
- Department of Medical Oncology, Fujian Provincial Cancer Hospital, Teaching Hospital of Fujian University of Traditional Chinese Medicine, Teaching Hospital of Fujian Medical University, Fuzhou, People's Republic of China
| | - Caicun Zhou
- Shanghai Pulmonary Hospital, Tongji University, Shanghai, People's Republic of China
| | - Yuankai Shi
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jiefei Han
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Di Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yanhua Tian
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhenlin Yang
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Li Zhang
- Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Shaokun Chuai
- Burning Rock Biotech, Guangdong, People's Republic of China
| | - Junyi Ye
- Burning Rock Biotech, Guangdong, People's Republic of China
| | - Guanshan Zhu
- Amoy Diagnostics, Xiamen, People's Republic of China
| | - Junhui Zhao
- Department of Medical Oncology, Affiliated Hospital of Qinghai University, Qinghai, People's Republic of China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital and Guangdong Academy of Medical Sciences, Guangzhou, People's Republic of China
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China.
| |
Collapse
|
35
|
Heidrich I, Ačkar L, Mossahebi Mohammadi P, Pantel K. Liquid biopsies: Potential and challenges. Int J Cancer 2020; 148:528-545. [PMID: 32683679 DOI: 10.1002/ijc.33217] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/09/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022]
Abstract
The analysis of tumor cells or tumor cell products obtained from blood or other body fluids ("liquid biopsy" [LB]) provides a broad range of opportunities in the field of oncology. Clinical application areas include early detection of cancer or tumor recurrence, individual risk assessment and therapy monitoring. LB allows to portray the entire disease as tumor cells or tumor cell products are released from all metastatic or primary tumor sites, providing comprehensive and real-time information on tumor cell evolution, therapeutic targets and mechanisms of resistance to therapy. Here, we focus on the most prominent LB markers, circulating tumor cells (CTCs) and circulating tumor-derived DNA (ctDNA), in the blood of patients with breast, prostate, lung and colorectal cancer, as the four most frequent tumor types in Europe. After a brief introduction of key technologies used to detect CTCs and ctDNA, we discuss recent clinical studies on these biomarkers for early detection and prognostication of cancer as well as prediction and monitoring of cancer therapies. We also point out current methodological and biological limitations that still hamper the implementation of LB into clinical practice.
Collapse
Affiliation(s)
- Isabel Heidrich
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lucija Ačkar
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Parinaz Mossahebi Mohammadi
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, Center of Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
36
|
Assessment of Pre-Analytical Sample Handling Conditions for Comprehensive Liquid Biopsy Analysis. J Mol Diagn 2020; 22:1070-1086. [PMID: 32497717 DOI: 10.1016/j.jmoldx.2020.05.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Revised: 05/05/2020] [Accepted: 05/19/2020] [Indexed: 12/25/2022] Open
Abstract
Liquid biopsies as a minimally invasive approach have the potential to revolutionize molecular diagnostics. Yet, although protocols for sample handling and the isolation of circulating tumor DNA (ctDNA) are numerous, comprehensive guidelines for diagnostics and research considering all aspects of real-life multicenter clinical studies are currently not available. These include limitations in sample volume, transport, and blood collection tubes. We tested the impact of commonly used (EDTA and heparin) and specialized blood collection tubes and storage conditions on the yield and purity of cell-free DNA for the application in down-stream analysis. Moreover, we evaluated the feasibility of a combined workflow for ctDNA and tumor cell genomic testing and parallel flow cytometric analysis of leukocytes. For genomic analyses, EDTA tubes showed good results if stored for a maximum of 4 hours at room temperature or for up to 24 hours when stored at 4°C. Spike-in experiments revealed that EDTA tubes in combination with density gradient centrifugation allowed the parallel isolation of ctDNA, leukocytes, and low amounts of tumor cells (0.1%) and their immunophenotyping by flow cytometry and down-stream genomic analysis by whole genome sequencing. In conclusion, adhering to time and temperature limits allows the use of routine EDTA blood samples for liquid biopsy analyses. We further provide a workflow enabling the parallel analysis of cell-free and cellular features for disease monitoring and for clonal evolution studies.
Collapse
|
37
|
Del Re M, Addeo A, Passaro A, Petrini I, van Schaik RH, Danesi R. Circulating tumor DNA and the future of EGFR-mutant lung cancer treatment. Pharmacogenomics 2020; 20:1255-1257. [PMID: 31829835 DOI: 10.2217/pgs-2019-0150] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Affiliation(s)
- Marzia Del Re
- Department of Clinical & Experimental Medicine, Unit of Clinical Pharmacology & Pharmacogenetics, University Hospital of Pisa, Pisa, Italy
| | - Alfredo Addeo
- Department of Oncology, University Hospital Geneva, Geneva, Switzerland
| | - Antonio Passaro
- Division of Thoracic Oncology, IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Iacopo Petrini
- Department of Translational Research & New Technologies in Surgery & Medicine, General Pathology, University of Pisa, Pisa, Italy
| | - Ron Hn van Schaik
- Department of Clinical Chemistry, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Romano Danesi
- Department of Clinical & Experimental Medicine, Unit of Clinical Pharmacology & Pharmacogenetics, University Hospital of Pisa, Pisa, Italy
| |
Collapse
|
38
|
Deep sequencing of circulating tumor DNA detects molecular residual disease and predicts recurrence in gastric cancer. Cell Death Dis 2020; 11:346. [PMID: 32393783 PMCID: PMC7214415 DOI: 10.1038/s41419-020-2531-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/29/2022]
Abstract
Identifying locoregional gastric cancer patients who are at high risk for relapse after resection could facilitate early intervention. By detecting molecular residual disease (MRD), circulating tumor DNA (ctDNA) has been shown to predict post-operative relapse in several cancers. Here, we aim to evaluate MRD detection by ctDNA and its association with clinical outcome in resected gastric cancer. This prospective cohort study enrolled 46 patients with stage I–III gastric cancer that underwent resection with curative intent. Sixty resected tumor samples and 296 plasma samples were obtained for targeted deep sequencing and longitudinal ctDNA profiling. ctDNA detection was correlated with clinicopathologic features and post-operative disease-free (DFS) and overall survival (OS). ctDNA was detected in 45% of treatment-naïve plasma samples. Primary tumor extent (T stage) was independently associated with pre-operative ctDNA positivity (p = 0.006). All patients with detectable ctDNA in the immediate post-operative period eventually experienced recurrence. ctDNA positivity at any time during longitudinal post-operative follow-up was associated with worse DFS and OS (HR = 14.78, 95%CI, 7.991–61.29, p < 0.0001 and HR = 7.664, 95% CI, 2.916–21.06, p = 0.002, respectively), and preceded radiographic recurrence by a median of 6 months. In locoregional gastric cancer patients treated with curative intent, these results indicate that ctDNA-detected MRD identifies patients at high risk for recurrence and can facilitate novel treatment intensification studies in the adjuvant setting to improve survival.
Collapse
|
39
|
Gini B, Thomas N, Blakely CM. Impact of concurrent genomic alterations in epidermal growth factor receptor ( EGFR)-mutated lung cancer. J Thorac Dis 2020; 12:2883-2895. [PMID: 32642201 PMCID: PMC7330397 DOI: 10.21037/jtd.2020.03.78] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Comprehensive characterization of the genomic landscape of epidermal growth factor receptor (EGFR)-mutated lung cancers have identified patterns of secondary mutations beyond the primary oncogenic EGFR mutation. These include concurrent pathogenic alterations affecting p53 (60–65%), RTKs (5–10%), PIK3CA/KRAS (3–23%), Wnt (5–10%), and cell cycle (7–25%) pathways as well as transcription factors such as MYC and NKX2-1 (10–15%). The majority of these co-occurring alterations were detected or enriched in samples collected from patients at resistance to tyrosine kinase inhibitor (TKI) treatment, indicating a potential functional role in driving resistance to therapy. Of note, these co-occurring tumor genomic alterations are not necessarily mutually exclusive, and evidence suggests that multiple clonal and sub-clonal cancer cell populations can co-exist and contribute to EGFR TKI resistance. Computational tools aimed to classify, track and predict the evolution of cancer clonal populations during therapy are being investigated in pre-clinical models to guide the selection of combination therapy switching strategies that may delay the development of treatment resistance. Here we review the most frequently identified tumor genomic alterations that co-occur with mutated EGFR and the evidence that these alterations effect responsiveness to EGFR TKI treatment.
Collapse
Affiliation(s)
- Beatrice Gini
- Department of Medicine, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Nicholas Thomas
- Department of Medicine, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| | - Collin M Blakely
- Department of Medicine, University of California, San Francisco, California, USA.,Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, California, USA
| |
Collapse
|
40
|
Qin K, Hou H, Liang Y, Zhang X. Prognostic value of TP53 concurrent mutations for EGFR- TKIs and ALK-TKIs based targeted therapy in advanced non-small cell lung cancer: a meta-analysis. BMC Cancer 2020; 20:328. [PMID: 32299384 PMCID: PMC7164297 DOI: 10.1186/s12885-020-06805-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 03/30/2020] [Indexed: 12/26/2022] Open
Abstract
Background The prognostic significance of TP53 concurrent mutations in patients with epidermal growth factor receptor (EGFR)- or anaplastic lymphoma kinase (ALK)- mutated advanced non–small-cell lung cancer (NSCLC) who received EGFR-tyrosine kinase inhibitors (TKIs) or ALK-TKIs based targeted therapy remains controversial. Therefore, the present meta-analysis was performed to investigate the association between TP53 concurrent mutations and prognosis of patients with advanced NSCLC undergoing EGFR-TKIs or ALK-TKIs treatments. Methods Eligible studies were identified by searching the online databases PubMed, Embase, Medline, The Cochrane library and Web of Science. Hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated to clarify the correlation between TP53 mutation status and prognosis of patients. This meta-analysis was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Results In total, 15 studies with 1342 patients were included for final analysis. Overall, concurrent TP53 mutation was associated with unfavorable progression-free survival (PFS) (HR = 1.88, 95%CI: 1.59–2.23, p < 0.001, I2 = 0.0%, P = 0.792) and overall survival (OS) (HR = 1.92, 95%CI: 1.55–2.38, p < 0.001, I2 = 0.0%, P = 0.515). Subgroup analysis based on type of targeted therapy (EGFR-TKIs or ALK-TKIs, pathological type of cancer (adenocarcinoma only or all NSCLC subtypes) and line of treatment (first-line only or all lines) all showed that TP53 mutations was associated with shorter survivals of patients with EGFR-TKIs or ALK-TKIs treatments. Particularly, in patients with first-line EGFR-TKIs treatment, significantly poorer prognosis was observed in patients with TP53 concurrent mutations (pooled HR for PFS: 1.69, 95% CI 1.25–2.27, P < 0.001, I2 = 0.0%, P = 0.473; pooled HR for OS: 1.94, 95% CI 1.36–2.76, P < 0.001, I2 = 0.0%, P = 0.484). Begg’s funnel plots and Egger’s tests indicated no significant publication bias in this study. Conclusions This meta-analysis indicated that concurrent TP53 mutations was a negative prognostic factor and associated with poorer outcomes of patients with EGFR-TKIs or ALK-TKIs treatments in advanced NSCLC. In addition, our study provided evidence that TP53 mutations might be involved in primary resistance to EGFR-TKIs treatments in patients with sensitive EGFR mutations in advanced NSCLC.
Collapse
Affiliation(s)
- Kang Qin
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266005, Shandong Province, China
| | - Helei Hou
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266005, Shandong Province, China
| | - Yu Liang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266005, Shandong Province, China
| | - Xiaochun Zhang
- Department of Medical Oncology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, 266005, Shandong Province, China.
| |
Collapse
|
41
|
Parikh AR, Mojtahed A, Schneider JL, Kanter K, Van Seventer EE, Fetter IJ, Thabet A, Fish MG, Teshome B, Fosbenner K, Nadres B, Shahzade HA, Allen JN, Blaszkowsky LS, Ryan DP, Giantonio B, Goyal L, Nipp RD, Roeland E, Weekes CD, Wo JY, Zhu AX, Dias-Santagata D, Iafrate AJ, Lennerz JK, Hong TS, Siravegna G, Horick N, Clark JW, Corcoran RB. Serial ctDNA Monitoring to Predict Response to Systemic Therapy in Metastatic Gastrointestinal Cancers. Clin Cancer Res 2020; 26:1877-1885. [PMID: 31941831 PMCID: PMC7165022 DOI: 10.1158/1078-0432.ccr-19-3467] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/09/2019] [Accepted: 01/10/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE ctDNA offers a promising, noninvasive approach to monitor therapeutic efficacy in real-time. We explored whether the quantitative percent change in ctDNA early after therapy initiation can predict treatment response and progression-free survival (PFS) in patients with metastatic gastrointestinal cancer. EXPERIMENTAL DESIGN A total of 138 patients with metastatic gastrointestinal cancers and tumor profiling by next-generation sequencing had serial blood draws pretreatment and at scheduled intervals during therapy. ctDNA was assessed using individualized droplet digital PCR measuring the mutant allele fraction in plasma of mutations identified in tumor biopsies. ctDNA changes were correlated with tumor markers and radiographic response. RESULTS A total of 138 patients enrolled. A total of 101 patients were evaluable for ctDNA and 68 for tumor markers at 4 weeks. Percent change of ctDNA by 4 weeks predicted partial response (PR, P < 0.0001) and clinical benefit [CB: PR and stable disease (SD), P < 0.0001]. ctDNA decreased by 98% (median) and >30% for all PR patients. ctDNA change at 8 weeks, but not 2 weeks, also predicted CB (P < 0.0001). Four-week change in tumor markers also predicted response (P = 0.0026) and CB (P = 0.022). However, at a clinically relevant specificity threshold of 90%, 4-week ctDNA change more effectively predicted CB versus tumor markers, with a sensitivity of 60% versus 24%, respectively (P = 0.0109). Patients whose 4-week ctDNA decreased beyond this threshold (≥30% decrease) had a median PFS of 175 days versus 59.5 days (HR, 3.29; 95% CI, 1.55-7.00; P < 0.0001). CONCLUSIONS Serial ctDNA monitoring may provide early indication of response to systemic therapy in patients with metastatic gastrointestinal cancer prior to radiographic assessments and may outperform standard tumor markers, warranting further evaluation.
Collapse
Affiliation(s)
- Aparna R Parikh
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Amikasra Mojtahed
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jaime L Schneider
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Katie Kanter
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Emily E Van Seventer
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Isobel J Fetter
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ashraf Thabet
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Madeleine G Fish
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bezaye Teshome
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Kathryn Fosbenner
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Brandon Nadres
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Heather A Shahzade
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jill N Allen
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lawrence S Blaszkowsky
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David P Ryan
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Bruce Giantonio
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Lipika Goyal
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ryan D Nipp
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Eric Roeland
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Colin D Weekes
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Andrew X Zhu
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - A John Iafrate
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jochen K Lennerz
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Giulia Siravegna
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nora Horick
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey W Clark
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ryan B Corcoran
- Cancer Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
42
|
Circulating tumour DNA: A new biomarker to monitor resistance in NSCLC patients treated with EGFR-TKIs. Biochim Biophys Acta Rev Cancer 2020; 1873:188363. [PMID: 32275933 DOI: 10.1016/j.bbcan.2020.188363] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/03/2020] [Accepted: 04/03/2020] [Indexed: 12/24/2022]
Abstract
Targeted molecular therapies have markedly improved the therapeutic management of lung cancer, while the discovery of epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) has revolutionized the treatment of non-small cell lung cancer (NSCLC). However, the clinical benefit of targeted therapies is limited by the eventual emergence of resistance. Identifying and monitoring the underlying mechanism of EGFR-TKI resistance could lead to more precise therapy and advances in treatment. Presently, tissue biopsy remains the gold standard for genotyping but it is limited by sampling bias, lack of available tissue, and potential complications. Analysis of circulating tumour DNA (ctDNA) may overcome the current limitations of tissue biopsies and provide a comprehensive landscape of the resistance mechanisms in a minimally invasive manner. Well-developed, analytically valid detection technologies are prerequisites for integrating ctDNA detection into clinical cancer management. Here, we provide an overview of available methodologies for ctDNA detection and we also discuss the potential clinical applications of ctDNA to monitor the resistance mechanisms.
Collapse
|
43
|
Towards the overcoming of anticancer drug resistance mediated by p53 mutations. Drug Resist Updat 2020; 49:100671. [DOI: 10.1016/j.drup.2019.100671] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 12/21/2022]
|
44
|
Rath B, Plangger A, Hamilton G. Non-small cell lung cancer-small cell lung cancer transformation as mechanism of resistance to tyrosine kinase inhibitors in lung cancer. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2020; 3:171-178. [PMID: 35582610 PMCID: PMC9090586 DOI: 10.20517/cdr.2019.85] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 01/13/2020] [Accepted: 02/18/2020] [Indexed: 12/11/2022]
Abstract
Mutated or rearranged driver kinases in non-small cell lung cancer (NSCLC) cells are clinically amenable to treatment with tyrosine kinase inhibitors (TKIs) resulting in prolonged survival and significant benefit compared to cytotoxic chemotherapy. The most frequent genomic alterations are observed for epidermal growth factor receptor and anaplastic lymphoma kinase, which can be blocked by a range of specific TKIs in sequence. In clinics, resistance to TKIs emerges after approximately one year and comprises secondary mutations of the kinases (on-target) or alternative pathways circumventing the original kinase (off-target) alterations. A special feature of NSCLC is the occurrence of histological transformation to small cell lung cancer (SCLC) in up to 14% of cases, which, in general, is accompanied by resistance to the original TKIs. SCLC transformed tumors may be treated with the classical platinum/etoposide regimen but thus far there are no definitive guidelines. Four transformed pleural SCLC lines in our lab indicate the presence of a gradual NSCLC-SCLC shift with overlapping drug sensitivities. In conclusion, the treatment of NSCLC-SCLC transformed cancer cells would need a better chemosensitivity assessment using functional genomics to guide further therapy.
Collapse
Affiliation(s)
- Barbara Rath
- Department of Vascular Surgery, Medical University of Vienna, Vienna A-1090, Austria
| | - Adelina Plangger
- Department of Vascular Surgery, Medical University of Vienna, Vienna A-1090, Austria
| | - Gerhard Hamilton
- Department of Vascular Surgery, Medical University of Vienna, Vienna A-1090, Austria
| |
Collapse
|
45
|
Tuaeva NO, Falzone L, Porozov YB, Nosyrev AE, Trukhan VM, Kovatsi L, Spandidos DA, Drakoulis N, Kalogeraki A, Mamoulakis C, Tzanakakis G, Libra M, Tsatsakis A. Translational Application of Circulating DNA in Oncology: Review of the Last Decades Achievements. Cells 2019; 8:E1251. [PMID: 31615102 PMCID: PMC6829588 DOI: 10.3390/cells8101251] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Revised: 09/30/2019] [Accepted: 10/12/2019] [Indexed: 02/06/2023] Open
Abstract
In recent years, the introduction of new molecular techniques in experimental and clinical settings has allowed researchers and clinicians to propose circulating-tumor DNA (ctDNA) analysis and liquid biopsy as novel promising strategies for the early diagnosis of cancer and for the definition of patients' prognosis. It was widely demonstrated that through the non-invasive analysis of ctDNA, it is possible to identify and characterize the mutational status of tumors while avoiding invasive diagnostic strategies. Although a number of studies on ctDNA in patients' samples significantly contributed to the improvement of oncology practice, some investigations generated conflicting data about the diagnostic and prognostic significance of ctDNA. Hence, to highlight the relevant achievements obtained so far in this field, a clearer description of the current methodologies used, as well as the obtained results, are strongly needed. On these bases, this review discusses the most relevant studies on ctDNA analysis in cancer, as well as the future directions and applications of liquid biopsy. In particular, special attention was paid to the early diagnosis of primary cancer, to the diagnosis of tumors with an unknown primary location, and finally to the prognosis of cancer patients. Furthermore, the current limitations of ctDNA-based approaches and possible strategies to overcome these limitations are presented.
Collapse
Affiliation(s)
- Natalia O Tuaeva
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
| | - Luca Falzone
- Department of Biomedical and Biotechnlogical Sciences, University of Catania, 95123 Catania, Italy.
- Epidemiology Unit, IRCCS Istituto Nazionale Tumori "Fondazione G. Pascale", 80131 Naples, Italy.
| | - Yuri B Porozov
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
- ITMO University, Saint Petersburg 197101, Russia.
| | - Alexander E Nosyrev
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
| | - Vladimir M Trukhan
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
| | - Leda Kovatsi
- Laboratory of Forensic Medicine and Toxicology, School of Medicine, Aristotle University of Thessaloniki, 54248 Thessaloniki, Greece.
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece.
| | - Nikolaos Drakoulis
- Research Group of Clinical Pharmacology and Pharmacogenomics, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, 15771 Zografou, Greece.
| | - Alexandra Kalogeraki
- Department of Pathology-Cytopathology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece.
| | - Charalampos Mamoulakis
- Department of Urology, University General Hospital of Heraklion, University of Crete, Medical School, Heraklion, 70013 Crete, Greece.
| | - George Tzanakakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, Heraklion, 70013 Crete, Greece.
| | - Massimo Libra
- Department of Biomedical and Biotechnlogical Sciences, University of Catania, 95123 Catania, Italy.
- Research Center for Prevention, Diagnosis and Treatment of Cancer, University of Catania, 95123 Catania, Italy.
| | - Aristides Tsatsakis
- I.M. Sechenov First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia.
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, Heraklion, 71003 Crete, Greece.
| |
Collapse
|
46
|
van der Pol Y, Mouliere F. Toward the Early Detection of Cancer by Decoding the Epigenetic and Environmental Fingerprints of Cell-Free DNA. Cancer Cell 2019; 36:350-368. [PMID: 31614115 DOI: 10.1016/j.ccell.2019.09.003] [Citation(s) in RCA: 179] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/18/2019] [Accepted: 09/06/2019] [Indexed: 12/15/2022]
Abstract
Widespread adaptation of liquid biopsy for the early detection of cancer has yet to reach clinical utility. Circulating tumor DNA is commonly detected though the presence of genetic alterations, but only a minor fraction of tumor-derived cell-free DNA (cfDNA) fragments exhibit mutations. The cellular processes occurring in cancer development mark the chromatin. These epigenetic marks are reflected by modifications in the cfDNA methylation, fragment size, and structure. In this review, we describe how going beyond DNA sequence information alone, by analyzing cfDNA epigenetic and immune signatures, boosts the potential of liquid biopsy for the early detection of cancer.
Collapse
Affiliation(s)
- Ymke van der Pol
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Florent Mouliere
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Pathology, Cancer Center Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
47
|
Villatoro S, Mayo-de-Las-Casas C, Jordana-Ariza N, Viteri-Ramírez S, Garzón-Ibañez M, Moya-Horno I, García-Peláez B, González-Cao M, Malapelle U, Balada-Bel A, Martínez-Bueno A, Campos R, Reguart N, Majem M, Blanco R, Blasco A, Catalán MJ, González X, Troncone G, Karachaliou N, Rosell R, Molina-Vila MA. Prospective detection of mutations in cerebrospinal fluid, pleural effusion, and ascites of advanced cancer patients to guide treatment decisions. Mol Oncol 2019; 13:2633-2645. [PMID: 31529604 PMCID: PMC6887582 DOI: 10.1002/1878-0261.12574] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 09/06/2019] [Accepted: 09/13/2019] [Indexed: 12/18/2022] Open
Abstract
Many advanced cases of cancer show central nervous system, pleural, or peritoneal involvement. In this study, we prospectively analyzed if cerebrospinal fluid (CSF), pleural effusion (PE), and/or ascites (ASC) can be used to detect driver mutations and guide treatment decisions. We collected 42 CSF, PE, and ASC samples from advanced non‐small‐cell lung cancer and melanoma patients. Cell‐free DNA (cfDNA) was purified and driver mutations analyzed and quantified by PNA‐Q‐PCR or next‐generation sequencing. All 42 fluid samples were evaluable; clinically relevant mutations were detected in 41 (97.6%). Twenty‐three fluids had paired blood samples, 22 were mutation positive in fluid but only 14 in blood, and the abundance of the mutant alleles was significantly higher in fluids. Of the 34 fluids obtained at progression to different therapies, EGFR resistance mutations were detected in nine and ALK acquired mutations in two. The results of testing of CSF, PE, and ASC were used to guide treatment decisions, such as initiation of osimertinib treatment or selection of specific ALK tyrosine–kinase inhibitors. In conclusion, fluids close to metastatic sites are superior to blood for the detection of relevant mutations and can offer valuable clinical information, particularly in patients progressing to targeted therapies.
Collapse
Affiliation(s)
- Sergio Villatoro
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Clara Mayo-de-Las-Casas
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Núria Jordana-Ariza
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | | | - Mónica Garzón-Ibañez
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Irene Moya-Horno
- Dr Rosell Oncology Institute, General Hospital of Catalonia, Sant Cugat del Vallés, Spain
| | - Beatriz García-Peláez
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - María González-Cao
- Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Italy
| | - Ariadna Balada-Bel
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | | | - Raquel Campos
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Noemí Reguart
- Department of Medical Oncology, Hospital Clínic, Barcelona, Spain
| | - Margarita Majem
- Medical Oncology Service, Hospital de Sant Pau, Barcelona, Spain
| | - Remei Blanco
- Medical Oncology Service, Hospital de Terrassa, Spain
| | - Ana Blasco
- Medical Oncology Department, Hospital General Universitario de Valencia, Spain
| | - María J Catalán
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| | - Xavier González
- Dr Rosell Oncology Institute, General Hospital of Catalonia, Sant Cugat del Vallés, Spain
| | | | - Niki Karachaliou
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain.,Dr Rosell Oncology Institute, University Hospital Sagrat Cor, Barcelona, Spain
| | - Rafael Rosell
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain.,Dr Rosell Oncology Institute, Quirón Dexeus University Hospital, Barcelona, Spain.,Cancer Biology and Precision Medicine Program, Catalan Institute of Oncology, Germans Trias i Pujol Health Sciences Institute and Hospital, Badalona, Spain
| | - Miguel A Molina-Vila
- Laboratory of Oncology, Pangaea Oncology, Quirón Dexeus University Hospital, Barcelona, Spain
| |
Collapse
|
48
|
Reversal of EGFR inhibitors' resistance by co-delivering EGFR and integrin αvβ3 inhibitors with nanoparticles in non-small cell lung cancer. Biosci Rep 2019; 39:BSR20181259. [PMID: 31316001 PMCID: PMC6712436 DOI: 10.1042/bsr20181259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 06/30/2019] [Accepted: 07/10/2019] [Indexed: 12/15/2022] Open
Abstract
Purpose: Tumor cells, with drug resistance, are associated with failed treatment and poor prognosis. Our aim was to explore potential strategy to overcome the epidermal growth factor receptor (EGFR) inhibitors resistance in non-small cell lung cancer (NSCLC).Materials and methods: Flow cytometry was used to examine and sort cells. Using MTT assay, we detected the cell viability under different conditions. Using RT-qPCR and Western blot, we determined the targeted gene expression in mRNA and protein levels. The morphology of the prepared nanoparticles was pictured by transmission electron microscopy. We also performed immunohistochemistry (IHC) and immunofluorescence (IF) to detect the proteins expression. Subcutaneous cancer models in nude mice were constructed to evaluate the anti-cancer effects in vivo Results: Here, we observed enhanced expression of integrin αvβ3 in tumor tissues from EGFR inhibitors resistant patients. Also, integrin αvβ3-positive NSCLC cells revealed significant EGFR inhibitors resistance, resulting from the activation of Galectin-3/KRAS/RalB/TBK1/NF-κB signaling pathway. Co-encapsulating integrin αvβ3 inhibitor and EGFR inhibitor further improved the drug delivery system, leading to superior anti-cancer effects and reduced systemic toxicity.Conclusion: Our results demonstrated that co-encapsulation of erlotinib and cilengitide by MPEG-PLA (Erlo+Cilen/PP) nanoparticles revealed enhanced tumor suppression along with reduced organ damages, providing an innovative approach for NSCLC treatment.
Collapse
|
49
|
Del Re M, Crucitta S, Gianfilippo G, Passaro A, Petrini I, Restante G, Michelucci A, Fogli S, de Marinis F, Porta C, Chella A, Danesi R. Understanding the Mechanisms of Resistance in EGFR-Positive NSCLC: From Tissue to Liquid Biopsy to Guide Treatment Strategy. Int J Mol Sci 2019; 20:ijms20163951. [PMID: 31416192 PMCID: PMC6720634 DOI: 10.3390/ijms20163951] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 08/07/2019] [Accepted: 08/09/2019] [Indexed: 02/06/2023] Open
Abstract
Liquid biopsy has emerged as an alternative source of nucleic acids for the management of Epidermal Growth Factor Receptor (EGFR)-mutant non-Small Cell Lung Cancer (NSCLC). The use of circulating cell-free DNA (cfDNA) has been recently introduced in clinical practice, resulting in the improvement of the identification of druggable EGFR mutations for the diagnosis and monitoring of response to targeted therapy. EGFR-dependent (T790M and C797S mutations) and independent (Mesenchymal Epithelial Transition [MET] gene amplification, Kirsten Rat Sarcoma [KRAS], Phosphatidyl-Inositol 4,5-bisphosphate 3-Kinase Catalytic subunit Alpha isoform [PI3KCA], and RAF murine sarcoma viral oncogene homolog B1 [BRAF] gene mutations) mechanisms of resistance to EGFR tyrosine kinase inhibitors (TKIs) have been evaluated in plasma samples from NSCLC patients using highly sensitive methods (i.e., digital droplet PCR, Next Generation Sequencing), allowing for the switch to other therapies. Therefore, liquid biopsy is a non-invasive method able to detect the molecular dynamic changes that occur under the pressure of treatment, and to capture tumor heterogeneity more efficiently than is allowed by tissue biopsy. This review addresses how liquid biopsy may be used to guide the choice of treatment strategy in EGFR-mutant NSCLC.
Collapse
Affiliation(s)
- Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy.
| | - Stefania Crucitta
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Giulia Gianfilippo
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology, 20141 Milano, Italy
| | - Iacopo Petrini
- General Pathology, Department of Translational Research & New Technologies in Surgery and Medicine, University of Pisa, 56126 Pisa, Italy
| | - Giuliana Restante
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Angela Michelucci
- Unit of Molecular Genetics, Department of Laboratory Medicine, University Hospital, 56126 Pisa, Italy
| | - Stefano Fogli
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Filippo de Marinis
- Division of Thoracic Oncology, European Institute of Oncology, 20141 Milano, Italy
| | - Camillo Porta
- Department of Internal Medicine, University of Pavia, 27100 Pavia, Italy
- Division of Translational Oncology, I.R.C.C.S. Istituti Clinici Scientifici Maugeri, 27100 Pavia, Italy
| | - Antonio Chella
- Unit of Respiratory Medicine, Department of Critical Area and Surgical, Medical and Molecular Pathology, University Hospital, 56126 Pisa, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
50
|
Identification and monitoring of somatic mutations in circulating cell-free tumor DNA in lung cancer patients. Lung Cancer 2019; 134:225-232. [PMID: 31319985 DOI: 10.1016/j.lungcan.2019.06.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 06/04/2019] [Accepted: 06/11/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVES Circulating cell-free tumor DNA (ctDNA) isolated from the peripheral blood of non-small-cell lung cancer (NSCLC) patients provides biomarkers for both therapeutic target selection, particularly when direct tumor biopsy is unfeasible, and also for drug resistance monitoring. This study evaluates the reliability and feasibility of ctDNA analysis in an in-house clinical molecular diagnostic workflow. MATERIALS AND METHODS Mutation profiling by both standard methods and Next-Generation sequencing (NGS) was carried out and compared on 2 independent lung cancer patient cohorts. Cohort 1 consisted of 50 EGFR-mutated NSCLC patients, established on tumour biopsy, for whom ctDNA was collected at disease progression after TKI-inhibitor treatment and could be used to monitor drug resistance. Cohort 2 consisted of 50 newly diagnosed lung cancer patients for whom tumour biopsy was not possible and only ctDNA was available, providing the possibility of biomarker identification. RESULTS ctDNA analysis of Cohort 1 verified the persistence of the tumour-detected EGFR activating mutation at disease progression by both standard and NGS methods, in 84% and 92% of the cases respectively. The T790M EGFR resistance mutation was identified in 71% of the ctDNA EGFR mutated samples providing vital information for their disease management. In newly diagnosed Cohort 2 patients, EGFR activating mutations were detected in 16% of the patients by both standard and NGS analysis of ctDNA in peripheral blood, providing indication to targeted-therapy otherwise unavailable for this group of patients. CONCLUSION The presented study investigated lung cancer ctDNA analysis, comparing conventional methods versus NGS sequencing, and demonstrated the successful use of plasma ctDNA as a template for targeted NGS tumor gene panel in an in-house routine clinical practice. More importantly, these data underline the advantages of the clinical application of ctDNA NGS analysis for identification of therapeutic targets, real-time monitoring of therapy, and resistance mechanisms in lung cancer patients.
Collapse
|