1
|
de Priester P, Zullo L, Vasseur D, Alonso De Castro B, Marinello A, Friboulet L, Planchard D, Besse B, Aldea M. BRAF Kinase Domain Duplication as a Potential On-Target Resistance Mechanism After Dabrafenib-Trametinib Therapy in a Patient With BRAFV600E-Mutated Lung Adenocarcinoma. JCO Precis Oncol 2025; 9:e2400694. [PMID: 40239137 DOI: 10.1200/po-24-00694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/05/2025] [Accepted: 03/05/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- Pierre de Priester
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of Cancer Medicine, Henri Becquerel Cancer Center and Rouen University Hospital, Rouen-Normandie University, Rouen, France
| | - Lodovica Zullo
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Damien Vasseur
- Department of Pathology and Molecular Biology, Gustave Roussy, Villejuif, France
| | - Beatriz Alonso De Castro
- INSERM, U981 "Identification of Molecular Predictors and New Targets for Cancer Treatment," Gustave Roussy, Villejuif, France
| | | | - Luc Friboulet
- INSERM, U981 "Identification of Molecular Predictors and New Targets for Cancer Treatment," Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - David Planchard
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Mihaela Aldea
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| |
Collapse
|
2
|
Bartolomucci A, Nobrega M, Ferrier T, Dickinson K, Kaorey N, Nadeau A, Castillo A, Burnier JV. Circulating tumor DNA to monitor treatment response in solid tumors and advance precision oncology. NPJ Precis Oncol 2025; 9:84. [PMID: 40122951 PMCID: PMC11930993 DOI: 10.1038/s41698-025-00876-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 03/11/2025] [Indexed: 03/25/2025] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a dynamic biomarker in cancer, as evidenced by its increasing integration into clinical practice. Carrying tumor specific characteristics, ctDNA can be used to inform treatment selection, monitor response, and identify drug resistance. In this review, we provide a comprehensive, up-to-date summary of ctDNA in monitoring treatment response with a focus on lung, colorectal, and breast cancers, and discuss current challenges and future directions.
Collapse
Affiliation(s)
- Alexandra Bartolomucci
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Monyse Nobrega
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Nivedita Kaorey
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Alberto Castillo
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.
- Department of Pathology, McGill University, Montreal, QC, Canada.
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
3
|
Kurma K, Eslami-S Z, Alix-Panabières C, Cayrefourcq L. Liquid biopsy: paving a new avenue for cancer research. Cell Adh Migr 2024; 18:1-26. [PMID: 39219215 PMCID: PMC11370957 DOI: 10.1080/19336918.2024.2395807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The current constraints associated with cancer diagnosis and molecular profiling, which rely on invasive tissue biopsies or clinical imaging, have spurred the emergence of the liquid biopsy field. Liquid biopsy involves the extraction of circulating tumor cells (CTCs), circulating free or circulating tumor DNA (cfDNA or ctDNA), circulating cell-free RNA (cfRNA), extracellular vesicles (EVs), and tumor-educated platelets (TEPs) from bodily fluid samples. Subsequently, these components undergo molecular characterization to identify biomarkers that are critical for early cancer detection, prognosis, therapeutic assessment, and post-treatment monitoring. These innovative biosources exhibit characteristics analogous to those of the primary tumor from which they originate or interact. This review comprehensively explores the diverse technologies and methodologies employed for processing these biosources, along with their principal clinical applications.
Collapse
Affiliation(s)
- Keerthi Kurma
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| |
Collapse
|
4
|
Liu L, Soler J, Reckamp KL, Sankar K. Emerging Targets in Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:10046. [PMID: 39337530 PMCID: PMC11432526 DOI: 10.3390/ijms251810046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Lung cancer is responsible for a high burden of disease globally. Over the last two decades, the discovery of targetable oncogenic genomic alterations has revolutionized the treatment landscape for early-stage and advanced non-small cell lung cancer (NSCLC). New molecular drivers continue to emerge as promising therapeutic targets, including KRAS non-G12C, RAF/MEK, HER3, Nectin-4, folate receptor alpha, ITGB6, and PRMT5. In this review, we summarize the emerging molecular targets with a potential clinical impact in advanced NSCLC, elaborating on their clinical characteristics and specific mechanisms and molecular pathways for which targeted treatments are currently available. Additionally, we present an aggregate of ongoing clinical trials investigating the available treatment options targeting such alterations, in addition to their current recruitment status and preliminary efficacy data. These advancements may guide further research endeavors and inform future treatment strategies to improve the management of and transform outcomes for patients with advanced NSCLC.
Collapse
Affiliation(s)
- Louisa Liu
- Samuel-Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Joshua Soler
- Riverside School of Medicine, University of California, Riverside, CA 92521, USA
| | - Karen L Reckamp
- Samuel-Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kamya Sankar
- Samuel-Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
5
|
Nokin MJ, Darbo E, Richard E, San José S, de Hita S, Prouzet-Mauleon V, Turcq B, Gerardelli L, Crake R, Velasco V, Koopmansch B, Lambert F, Xue JY, Sang B, Horne J, Ziemons E, Villanueva A, Blomme A, Herfs M, Cataldo D, Calvayrac O, Porporato P, Nadal E, Lito P, Jänne PA, Ricciuti B, Awad MM, Ambrogio C, Santamaría D. In vivo vulnerabilities to GPX4 and HDAC inhibitors in drug-persistent versus drug-resistant BRAF V600E lung adenocarcinoma. Cell Rep Med 2024; 5:101663. [PMID: 39094577 PMCID: PMC11384943 DOI: 10.1016/j.xcrm.2024.101663] [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: 07/03/2023] [Revised: 05/22/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024]
Abstract
The current targeted therapy for BRAFV600E-mutant lung cancer consists of a dual blockade of RAF/MEK kinases often combining dabrafenib/trametinib (D/T). This regimen extends survival when compared to single-agent treatments, but disease progression is unavoidable. By using whole-genome CRISPR screening and RNA sequencing, we characterize the vulnerabilities of both persister and D/T-resistant cellular models. Oxidative stress together with concomitant induction of antioxidant responses is boosted by D/T treatment. However, the nature of the oxidative damage, the choice of redox detoxification systems, and the resulting therapeutic vulnerabilities display stage-specific differences. Persister cells suffer from lipid peroxidation and are sensitive to ferroptosis upon GPX4 inhibition in vivo. Biomarkers of lipid peroxidation are detected in clinical samples following D/T treatment. Acquired alterations leading to mitogen-activated protein kinase (MAPK) reactivation enhance cystine transport to boost GPX4-independent antioxidant responses. Similarly to BRAFV600E-mutant melanoma, histone deacetylase (HDAC) inhibitors decrease D/T-resistant cell viability and extend therapeutic response in vivo.
Collapse
Affiliation(s)
- Marie-Julie Nokin
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France; Laboratory of Biology of Tumor and Development (LBTD), GIGA-Cancer, University of Liege, 4000 Liege, Belgium.
| | - Elodie Darbo
- Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France
| | - Elodie Richard
- Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France
| | - Sonia San José
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France; Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain
| | - Sergio de Hita
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France; Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France; Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain
| | - Valérie Prouzet-Mauleon
- Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France; CRISP'edit, TBMCore, University of Bordeaux, CNRS UAR 3427, INSERM US05, 33000 Bordeaux, France
| | - Béatrice Turcq
- Bordeaux Institute of Oncology (BRIC), INSERM U1312, University of Bordeaux, 33000 Bordeaux, France; CRISP'edit, TBMCore, University of Bordeaux, CNRS UAR 3427, INSERM US05, 33000 Bordeaux, France
| | - Laura Gerardelli
- Laboratory of Biology of Tumor and Development (LBTD), GIGA-Cancer, University of Liege, 4000 Liege, Belgium
| | - Rebekah Crake
- Laboratory of Biology of Tumor and Development (LBTD), GIGA-Cancer, University of Liege, 4000 Liege, Belgium
| | - Valérie Velasco
- Department of Biopathology, Institut Bergonié, 33076 Bordeaux, France
| | - Benjamin Koopmansch
- Department of Human Genetics, University Hospital Center of Liege, 4000 Liege, Belgium
| | - Frederic Lambert
- Department of Human Genetics, University Hospital Center of Liege, 4000 Liege, Belgium
| | - Jenny Y Xue
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ben Sang
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Julie Horne
- Laboratory of Pharmaceutical Analytical Chemistry, CIRM, University of Liege, 4000 Liege, Belgium
| | - Eric Ziemons
- Laboratory of Pharmaceutical Analytical Chemistry, CIRM, University of Liege, 4000 Liege, Belgium
| | - Alberto Villanueva
- Chemoresistance and Predictive Factors Group, Program Against Cancer Therapeutic Resistance (ProCURE), Catalan Institute of Oncology (ICO); Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet del Llobregat, 08908 Barcelona, Spain
| | - Arnaud Blomme
- Laboratory of Cancer Signaling, GIGA-Stem Cells, University of Liege, 4000 Liege, Belgium
| | - Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer, University of Liege, 4000 Liege, Belgium
| | - Didier Cataldo
- Laboratory of Biology of Tumor and Development (LBTD), GIGA-Cancer, University of Liege, 4000 Liege, Belgium
| | - Olivier Calvayrac
- Cancer Research Centre of Toulouse, INSERM UMR1037, CNRS UMR5071, 31100 Toulouse, France
| | - Paolo Porporato
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy
| | - Ernest Nadal
- Molecular Mechanisms of Cancer Program, Department of Medical Oncology, Catalan Institute of Oncology (ICO), Preclinical and Experimental Research in Thoracic Tumors (PReTT) Group, Oncobell Program, IDIBELL, L'Hospitalet del Llobregat, 08908 Barcelona, Spain
| | - Piro Lito
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Pasi A Jänne
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Biagio Ricciuti
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Mark M Awad
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, 10126 Torino, Italy.
| | - David Santamaría
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France; Centro de Investigación del Cáncer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain.
| |
Collapse
|
6
|
Imyanitov EN, Mitiushkina NV, Kuligina ES, Tiurin VI, Venina AR. Pathways and targeting avenues of BRAF in non-small cell lung cancer. Expert Opin Ther Targets 2024; 28:613-622. [PMID: 38941191 DOI: 10.1080/14728222.2024.2374742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 06/27/2024] [Indexed: 06/30/2024]
Abstract
INTRODUCTION BRAF is a serine-threonine kinase implicated in the regulation of MAPK signaling cascade. BRAF mutation-driven activation occurs in approximately 2-4% of treatment-naive non-small cell carcinomas (NSCLCs). BRAF upregulation is also often observed in tumors with acquired resistance to receptor tyrosine kinase inhibitors (TKIs). AREAS COVERED This review describes the spectrum of BRAF mutations and their functional roles, discusses treatment options available for BRAF p.V600 and non-V600 mutated NSCLCs, and identifies some gaps in the current knowledge. EXPERT OPINION Administration of combined BRAF/MEK inhibitors usually produces significant, although often a short-term, benefit to NSCLC patients with BRAF V600 (class 1) mutations. There are no established treatments for BRAF class 2 (L597, K601, G464, G469A/V/R/S, fusions, etc.) and class 3 (D594, G596, G466, etc.) mutants, which account for up to two-thirds of BRAF-driven NSCLCs. Many important issues related to the use of immune therapy for the management of BRAF-mutated NSCLC deserve further investigation. The rare occurrence of BRAF mutations in NSCLC is compensated by high overall incidence of lung cancer disease; therefore, clinical studies on BRAF-associated NSCLC are feasible.
Collapse
Affiliation(s)
- Evgeny N Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, St.-Petersburg, Russia
| | - Natalia V Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Ekatherina Sh Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Vladislav I Tiurin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| | - Aigul R Venina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, St.-Petersburg, Russia
| |
Collapse
|
7
|
Planchard D, Sanborn RE, Negrao MV, Vaishnavi A, Smit EF. BRAF V600E-mutant metastatic NSCLC: disease overview and treatment landscape. NPJ Precis Oncol 2024; 8:90. [PMID: 38627602 PMCID: PMC11021522 DOI: 10.1038/s41698-024-00552-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/15/2024] [Indexed: 04/19/2024] Open
Abstract
In this review, we cover the current understanding of BRAF mutations and associated clinical characteristics in patients with metastatic NSCLC, approved and emerging treatment options, BRAF sequencing approaches, and unmet needs. The BRAFV600E mutation confers constitutive activity of the MAPK pathway, leading to enhanced growth, proliferation, and survival of tumor cells. Testing for BRAF mutations enables patients to be treated with therapies that directly target BRAFV600E and the MAPK pathway, but BRAF testing lags behind other oncogene testing in metastatic NSCLC. Additional therapies targeting BRAFV600E mutations provide options for patients with metastatic NSCLC. Emerging therapies and combinations under investigation could potentially overcome issues of resistance and target non-V600E mutations. Therefore, because targeted therapies with enhanced efficacy are on the horizon, being able to identify BRAF mutations in metastatic NSCLC may become even more important.
Collapse
Affiliation(s)
- David Planchard
- Thoracic Cancer Group, Department of Medical Oncology, Gustave Roussy, Villejuif, France.
| | - Rachel E Sanborn
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, USA
| | - Marcelo V Negrao
- Department of Thoracic/Head and Neck Medical Oncology, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Aria Vaishnavi
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Egbert F Smit
- Department of Pulmonary Disease, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
8
|
Chen CP, Lin SF, Yeh CN, Huang WK, Pan YR, Hsiao YT, Lo CH, Wu CE. Synergistic effects of the combination of trametinib and alpelisib in anaplastic thyroid cancer with BRAF and PI3KCA co-mutations. Heliyon 2024; 10:e29055. [PMID: 38576565 PMCID: PMC10990975 DOI: 10.1016/j.heliyon.2024.e29055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
Background Anaplastic thyroid cancer (ATC), a rare and aggressive malignancy with a poor prognosis, has shown promise with the approved dabrafenib/trametinib combination for BRAFV600E mutation. Co-occurring PI3KCA mutations, identified as negative prognostic factors in lung cancer with BRAFV600E mutation, emphasize the need to target both pathways. Exploring trametinib and alpelisib combination becomes crucial for ATC. Methods A patient-derived xenograft (PDX) and primary cell line were obtained from an ATC patient with BRAF and PI3KCA co-mutation. Individual testing of targeted therapies against BRAF, MEK, and PI3KCA was followed by a combination treatment. Synergistic effects were evaluated using the combination index. Immunoblotting assessed the efficacy, with validation performed using a PDX model. Results In this study, the ATC0802 cell line and PDX were established from a refractory ATC patient. NGS revealed BRAF and PI3KCA co-mutations pre- and post-dabrafenib/trametinib treatment. Trametinib/alpelisib combination showed synergy, suppressing both pERK and pAKT levels, unlike monotherapies or BRAF knockdown. The combination induced apoptosis and, in the PDX model, demonstrated superior tumor growth inhibition compared to monotherapies. Conclusions The combination of trametinib and alpelisib showed promise as a strategy for treating ATC with co-mutations in BRAF and PI3KCA, both in vitro and in vivo. This combination offers insights into overcoming resistance to BRAF-targeted treatments in ATC with mutations in BRAF and PI3KCA.
Collapse
Affiliation(s)
- Chiao-Ping Chen
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Shu-Fu Lin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, New Taipei Municipal TuCheng Hospital (Built and Operated by Chang Gung Medical Foundation), College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chun-Nan Yeh
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of General Surgery, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yi-Ru Pan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- Department of General Surgery, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Tien Hsiao
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chih-Hong Lo
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Chiao-En Wu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Liver Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
9
|
Mezquita L, Oulhen M, Aberlenc A, Deloger M, Aldea M, Honore A, Lecluse Y, Howarth K, Friboulet L, Besse B, Planchard D, Farace F. Resistance to BRAF inhibition explored through single circulating tumour cell molecular profiling in BRAF-mutant non-small-cell lung cancer. Br J Cancer 2024; 130:682-693. [PMID: 38177660 PMCID: PMC10876548 DOI: 10.1038/s41416-023-02535-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 11/24/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Resistance mechanisms to combination therapy with dabrafenib plus trametinib remain poorly understood in patients with BRAFV600E-mutant advanced non-small-cell lung cancer (NSCLC). We examined resistance to BRAF inhibition by single CTC sequencing in BRAFV600E-mutant NSCLC. METHODS CTCs and cfDNA were examined in seven BRAFV600E-mutant NSCLC patients at failure to treatment. Matched tumour tissue was available for four patients. Single CTCs were isolated by fluorescence-activated cell sorting following enrichment and immunofluorescence (Hoechst 33342/CD45/pan-cytokeratins) and sequenced for mutation and copy number-alteration (CNA) analyses. RESULTS BRAFV600E was found in 4/4 tumour biopsies and 5/7 cfDNA samples. CTC mutations were mostly found in MAPK-independent pathways and only 1/26 CTCs were BRAFV600E mutated. CTC profiles encompassed the majority of matched tumour biopsy CNAs but 72.5% to 84.5% of CTC CNAs were exclusive to CTCs. Extensive diversity, involving MAPK, MAPK-related, cell cycle, DNA repair and immune response pathways, was observed in CTCs and missed by analyses on tumour biopsies and cfDNA. Driver alterations in clinically relevant genes were recurrent in CTCs. CONCLUSIONS Resistance was not driven by BRAFV600E-mutant CTCs. Extensive tumour genomic heterogeneity was found in CTCs compared to tumour biopsies and cfDNA at failure to BRAF inhibition, in BRAFV600E-mutant NSCLC, including relevant alterations that may represent potential treatment opportunities.
Collapse
Affiliation(s)
- Laura Mezquita
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, F-94805, Villejuif, France
- Medical Oncology Department, Hospital Clinic of Barcelona, Laboratory of Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, Barcelona, Spain
| | - Marianne Oulhen
- Gustave Roussy, Université Paris-Saclay, "Rare Circulating Cells" Translational Platform, CNRS UMS3655-INSERM US23 AMMICA, F-94805, Villejuif, France
- INSERM, U981 "Identification of Molecular Predictors and new Targets for Cancer Treatment", F-94805, Villejuif, France
| | - Agathe Aberlenc
- Gustave Roussy, Université Paris-Saclay, "Rare Circulating Cells" Translational Platform, CNRS UMS3655-INSERM US23 AMMICA, F-94805, Villejuif, France
- INSERM, U981 "Identification of Molecular Predictors and new Targets for Cancer Treatment", F-94805, Villejuif, France
| | - Marc Deloger
- Gustave Roussy, Université Paris-Saclay, Bioinformatics Platform, CNRS UMS3655-INSERM US23 AMMICA, F-94805, Villejuif, France
| | - Mihaela Aldea
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, F-94805, Villejuif, France
| | - Aurélie Honore
- Gustave Roussy, Université Paris-Saclay, Genomic Platform, CNRS UMS3655-INSERM US23 AMMICA, F-94805, Villejuif, France
| | - Yann Lecluse
- Gustave Roussy, Université Paris-Saclay, "Flow cytometry and Imaging" Platform, CNRS UMS3655-INSERM US23AMMICA, F-94805, Villejuif, France
| | | | - Luc Friboulet
- INSERM, U981 "Identification of Molecular Predictors and new Targets for Cancer Treatment", F-94805, Villejuif, France
| | - Benjamin Besse
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, F-94805, Villejuif, France
| | - David Planchard
- Gustave Roussy, Université Paris-Saclay, Department of Medicine, F-94805, Villejuif, France
| | - Françoise Farace
- Gustave Roussy, Université Paris-Saclay, "Rare Circulating Cells" Translational Platform, CNRS UMS3655-INSERM US23 AMMICA, F-94805, Villejuif, France.
- INSERM, U981 "Identification of Molecular Predictors and new Targets for Cancer Treatment", F-94805, Villejuif, France.
| |
Collapse
|
10
|
Parisi C, Tagliamento M, Belcaid L, Aldea M, Bayle A, Remon-Masip J, Italiano A, Planchard D, Besse B, Barlesi F. Circulating tumor DNA in clinical trials for solid tumors: Challenges and current applications. THE JOURNAL OF LIQUID BIOPSY 2023; 1:100007. [PMID: 40027283 PMCID: PMC11863815 DOI: 10.1016/j.jlb.2023.100007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/27/2023] [Accepted: 08/27/2023] [Indexed: 03/05/2025]
Abstract
Tumor derived biomarkers including circulating tumor DNA (ctDNA) and/or circulating tumors cells (CTCs) may be detected and quantified through liquid biopsy (LB). ctDNA analysis through LB is a validated tool for monitoring response to systemic treatment and detecting molecular mechanisms of resistance at the time of progression of advanced stage malignancies. Several applications of ctDNA have been investigated in the diagnostic phase of cancer or in the post-curative treatment surveillance phase (e.g., minimal residual disease assessment after neoadjuvant or adjuvant therapy). Recently, the improvement of ctDNA technology and its implementation have affected early phase trials design, with significant changes in the inclusion and randomization phases. Implementation of LB has resulted in large-scale development of academic programs aimed at exploiting all the potential applications of ctDNA, such as patients extended molecular screening, molecular oriented treatment decision making, monitoring of anti-cancer treatments response. In this rapid evolving field, the challenge is no longer the technique, but the evaluation of the results and the interpretation of their impact on diagnosis, prognosis, or therapeutic decision. Leading research cancer centers may favor education for scientific community, by capturing data on this evolving technology and sharing knowledge. In this review we summarize the main applications and challenges of ctDNA genotyping in clinical trials, with special focus on ongoing studies. We finally describe the most important next generation academic and industry-sponsored programs addressing early cancer detection and prevention in high-risk populations through ctDNA genotyping.
Collapse
Affiliation(s)
- Claudia Parisi
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
- Department of Medical and Surgical Sciences and Translational Medicine, St Andrea University Hospital, Sapienza University, Rome, Italy
| | - Marco Tagliamento
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy
| | - Laila Belcaid
- Department of Oncology, Copenaghen University Hospital, Rigshospitalet, Denmark
| | - Mihaela Aldea
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Arnaud Bayle
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
| | | | - Antoine Italiano
- Drug Development Department (DITEP), Gustave Roussy, Villejuif, France
- Medical Oncology Department, Institute Bergonié, Bordeaux, France
| | - David Planchard
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| | - Fabrice Barlesi
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Paris-Saclay University, Orsay, France
| |
Collapse
|
11
|
Puri M, Gawri K, Dawar R. Therapeutic strategies for BRAF mutation in non-small cell lung cancer: a review. Front Oncol 2023; 13:1141876. [PMID: 37645429 PMCID: PMC10461310 DOI: 10.3389/fonc.2023.1141876] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 07/24/2023] [Indexed: 08/31/2023] Open
Abstract
Lung cancer is the leading cause of cancer related deaths. Among the two broad types of lung cancer, non-small cell lung cancer accounts for 85% of the cases. The study of the genetic alteration has facilitated the development of targeted therapeutic interventions. Some of the molecular alterations which are important targets for drug therapy include Kirsten rat sarcoma (KRAS), Epidermal Growth Factor Receptor (EGFR), V-RAF murine sarcoma viral oncogene homolog B (BRAF), anaplastic lymphoma kinase gene (ALK). In the setting of extensive on-going clinical trials, it is imperative to periodically review the advancements and the newer drug therapies being available. Among all mutations, BRAF mutation is common with incidence being 8% overall and 1.5 - 4% in NSCLC. Here, we have summarized the BRAF mutation types and reviewed the various drug therapy available - for both V600 and nonV600 group; the mechanism of resistance to BRAF inhibitors and strategies to overcome it; the significance of comprehensive profiling of concurrent mutations, and the role of immune checkpoint inhibitor in BRAF mutated NSCLC. We have also included the currently ongoing clinical trials and recent advancements including combination therapy that would play a role in improving the overall survival and outcome of NSCLC.
Collapse
Affiliation(s)
- Megha Puri
- Department of Internal Medicine, Saint Peter’s University Hospital, New Brunswick, NJ, United States
| | - Kunal Gawri
- Department of Pulmonary, Critical Care and Sleep Medicine, University of Buffalo, Buffalo, NY, United States
| | - Richa Dawar
- Sylvester Comprehensive Cancer Center, University of Miami Health System, Miami, FL, United States
| |
Collapse
|
12
|
Nigro MC, Marchese PV, Deiana C, Casadio C, Galvani L, Di Federico A, De Giglio A. Clinical Utility and Application of Liquid Biopsy Genotyping in Lung Cancer: A Comprehensive Review. LUNG CANCER (AUCKLAND, N.Z.) 2023; 14:11-25. [PMID: 36762267 PMCID: PMC9904307 DOI: 10.2147/lctt.s388047] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 01/26/2023] [Indexed: 02/05/2023]
Abstract
Precision medicine has revolutionized the therapeutic management of cancer patients with a major impact on non-small cell lung cancer (NSCLC), particularly lung adenocarcinoma, where advances have been remarkable. Tissue biopsy, required for tumor molecular testing, has significant limitations due to the difficulty of the biopsy site or the inadequacy of the histological specimen. In this context, liquid biopsy, consisting of the analysis of tumor-released materials circulating in body fluids, such as blood, is increasingly emerging as a valuable and non-invasive biomarker for detecting circulating tumor DNA (ctDNA) carrying molecular tumor signatures. In advanced/metastatic NSCLC, liquid biopsy drives target therapy by monitoring response to treatment and identifying eventual genomic mechanisms of resistance. In addition, recent data have shown a significant ability to detect minimal residual disease in early-stage lung cancer, underlying the potential application of liquid biopsy in the adjuvant setting, in early detection of recurrence, and also in the screening field. In this article, we present a review of the currently available data about the utility and application of liquid biopsy in lung cancer, with a particular focus on the approach to different techniques of analysis for liquid biopsy and a comparison with tissue samples as well as the potential practical uses in early and advanced/metastatic NSCLC.
Collapse
Affiliation(s)
- Maria Concetta Nigro
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, University of Bologna, Bologna, 40138, Italy
| | - Paola Valeria Marchese
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, University of Bologna, Bologna, 40138, Italy,Correspondence: Paola Valeria Marchese, Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, University of Bologna, Via Albertoni 15, Bologna, 40138, Italy, Email
| | - Chiara Deiana
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, University of Bologna, Bologna, 40138, Italy
| | - Chiara Casadio
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, University of Bologna, Bologna, 40138, Italy
| | - Linda Galvani
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, University of Bologna, Bologna, 40138, Italy
| | - Alessandro Di Federico
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, University of Bologna, Bologna, 40138, Italy
| | - Andrea De Giglio
- Department of Experimental, Diagnostic and Specialty Medicine, S.Orsola-Malpighi University Hospital, University of Bologna, Bologna, 40138, Italy,Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, 40138, Italy
| |
Collapse
|
13
|
Guaitoli G, Zullo L, Tiseo M, Dankner M, Rose AAN, Facchinetti F. Non-small-cell lung cancer: how to manage BRAF-mutated disease. Drugs Context 2023; 12:dic-2022-11-3. [PMID: 37168877 PMCID: PMC10166262 DOI: 10.7573/dic.2022-11-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 03/17/2023] [Indexed: 05/13/2023] Open
Abstract
BRAF mutations are reported in about 3-5% of non-small-cell lung cancer (NSCLC), almost exclusively in adenocarcinoma histology, and are classified into three different classes. The segmentation of BRAF mutations into V600 (class 1) and non-V600 (classes 2 and 3) relies on their biological characteristics and is of interest for predicting the therapeutic benefit of targeted therapies and immunotherapy. Given the relative rarity of this molecular subset of disease, evidence supporting treatment choices is limited. This review aims to offer a comprehensive update about available therapeutic options for patients with NSCLC harbouring BRAF mutations to guide the physician in the choice of treatment strategies. We collected the most relevant available data, from single-arm phase II studies and retrospective analyses conducted in advanced NSCLC, regarding the efficacy of BRAF and MEK inhibitors in both V600 and non-V600 BRAF mutations. We included case reports and smaller experiences that could provide information on specific alterations. With respect to immunotherapy, we reviewed retrospective evidence on immune-checkpoint inhibitors in this molecular subset, whereas data about chemo-immunotherapy in this molecular subgroup are lacking. Moreover, we included the available, though limited, retrospective evidence of immunotherapy as consolidation after chemo-radiation for unresectable stage III BRAF-mutant NSCLC, and an overview of ongoing clinical trials in the peri-operative setting that could open new perspectives in the future.
Collapse
Affiliation(s)
- Giorgia Guaitoli
- Université Paris-Saclay, Gustave Roussy, INSERM U981, Villejuif, France
- PhD Program Clinical & Experimental Medicine, University of Modena & Reggio Emilia, Modena, Italy
| | - Lodovica Zullo
- Department of Experimental Medicine (DIMES), University of Genova, Genova, Italy
- Department of Cancer Medicine, Gustave Roussy Cancer Campus, Villejuif, France
| | - Marcello Tiseo
- Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Matthew Dankner
- Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montréal, Québec, Canada
- Faculty of Medicine, McGill University, Montréal, Québec, Canada
| | - April AN Rose
- Lady Davis Institute, Segal Cancer Centre, Jewish General Hospital, McGill University, Montréal, Québec, Canada
- Department of Oncology, McGill University, Montréal, Québec, Canada
| | - Francesco Facchinetti
- Université Paris-Saclay, Gustave Roussy, INSERM U981, Villejuif, France
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
14
|
Buszka K, Ntzifa A, Owecka B, Kamińska P, Kolecka-Bednarczyk A, Zabel M, Nowicki M, Lianidou E, Budna-Tukan J. Liquid Biopsy Analysis as a Tool for TKI-Based Treatment in Non-Small Cell Lung Cancer. Cells 2022; 11:2871. [PMID: 36139444 PMCID: PMC9497234 DOI: 10.3390/cells11182871] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/07/2022] [Accepted: 09/09/2022] [Indexed: 12/03/2022] Open
Abstract
The treatment of non-small cell lung cancer (NSCLC) has recently evolved with the introduction of targeted therapy based on the use of tyrosine kinase inhibitors (TKIs) in patients with certain gene alterations, including EGFR, ALK, ROS1, BRAF, and MET genes. Molecular targeted therapy based on TKIs has improved clinical outcomes in a large number of NSCLC patients with advanced disease, enabling significantly longer progression-free survival (PFS). Liquid biopsy is an increasingly popular diagnostic tool for treating TKI-based NSCLC. The studies presented in this article show that detection and analysis based on liquid biopsy elements such as circulating tumor cells (CTCs), cell-free DNA (cfDNA), exosomes, and/or tumor-educated platelets (TEPs) can contribute to the appropriate selection and monitoring of targeted therapy in NSCLC patients as complementary to invasive tissue biopsy. The detection of these elements, combined with their molecular analysis (using, e.g., digital PCR (dPCR), next generation sequencing (NGS), shallow whole genome sequencing (sWGS)), enables the detection of mutations, which are required for the TKI treatment. Despite such promising results obtained by many research teams, it is still necessary to carry out prospective studies on a larger group of patients in order to validate these methods before their application in clinical practice.
Collapse
Affiliation(s)
- Karolina Buszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Aliki Ntzifa
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Barbara Owecka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Paula Kamińska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, 60-812 Poznan, Poland
| | - Agata Kolecka-Bednarczyk
- Department of Immunology, Chair of Pathomorphology and Clinical Immunology, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Maciej Zabel
- Division of Anatomy and Histology, University of Zielona Góra, 65-046 Zielona Góra, Poland
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| | - Evi Lianidou
- Analysis of Circulating Tumor Cells Lab, Lab of Analytical Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15771 Athens, Greece
| | - Joanna Budna-Tukan
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland
| |
Collapse
|
15
|
Pesta M, Shetti D, Kulda V, Knizkova T, Houfkova K, Bagheri MS, Svaton M, Polivka J. Applications of Liquid Biopsies in Non-Small-Cell Lung Cancer. Diagnostics (Basel) 2022; 12:1799. [PMID: 35892510 PMCID: PMC9330570 DOI: 10.3390/diagnostics12081799] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 11/17/2022] Open
Abstract
The concept of liquid biopsy as an analysis tool for non-solid tissue carried out for the purpose of providing information about solid tumors was introduced approximately 20 years ago. Additional to the detection of circulating tumor cells (CTCs), the liquid biopsy approach quickly included the analysis of circulating tumor DNA (ctDNA) and other tumor-derived markers such as circulating cell-free RNA or extracellular vesicles. Liquid biopsy is a non-invasive technique for detecting multiple cancer-associated biomarkers that is easy to obtain and can reflect the characteristics of the entire tumor mass. Currently, ctDNA is the key component of the liquid biopsy approach from the point of view of the prognosis assessment, prediction, and monitoring of the treatment of non-small-cell lung cancer (NSCLC) patients. ctDNA in NSCLC patients carries variants or rearrangements that drive carcinogenesis, such as those in EGFR, KRAS, ALK, or ROS1. Due to advances in pharmacology, these variants are the subject of targeted therapy. Therefore, the detection of these variants has gained attention in clinical medicine. Recently, methods based on qPCR (ddPCR, BEAMing) and next-generation sequencing (NGS) are the most effective approaches for ctDNA analysis. This review addresses various aspects of the use of liquid biopsy with an emphasis on ctDNA as a biomarker in NSCLC patients.
Collapse
Affiliation(s)
- Martin Pesta
- Department of Biology, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00 Plzen, Czech Republic; (D.S.); (T.K.); (K.H.)
| | - Dattatrya Shetti
- Department of Biology, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00 Plzen, Czech Republic; (D.S.); (T.K.); (K.H.)
| | - Vlastimil Kulda
- Department of Medical Chemistry and Biochemistry, Faculty of Medicine in Pilsen, Charles University, Karlovarska 48, 301 66 Plzen, Czech Republic;
| | - Tereza Knizkova
- Department of Biology, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00 Plzen, Czech Republic; (D.S.); (T.K.); (K.H.)
| | - Katerina Houfkova
- Department of Biology, Faculty of Medicine in Pilsen, Charles University, Alej Svobody 1655/76, 323 00 Plzen, Czech Republic; (D.S.); (T.K.); (K.H.)
| | - Mahyar Sharif Bagheri
- Department of Histology, Faculty of Medicine in Pilsen, Charles University, Karlovarska 48, 301 66 Plzen, Czech Republic; (M.S.B.); (J.P.)
| | - Martin Svaton
- Department of Pneumology and Phthisiology, Faculty of Medicine in Pilsen, Charles University, University Hospital in Pilsen, E. Benese 13, 301 00 Plzen, Czech Republic;
| | - Jiri Polivka
- Department of Histology, Faculty of Medicine in Pilsen, Charles University, Karlovarska 48, 301 66 Plzen, Czech Republic; (M.S.B.); (J.P.)
| |
Collapse
|
16
|
Hu D, Guo E, Yang B, Qin X, Fu Y, Fan J, Zhuang X, Yao Q, Lu F, Li W, Xiao R, Wu X, Yang X, Wang Z, Liu C, You L, Zang R, Zhou Q, Zhao W, Chen G, Sun C. Mutation profiles in circulating cell‐free
DNA
predict acquired resistance to Olaparib in high‐grade serous ovarian carcinoma. Cancer Sci 2022; 113:2849-2861. [PMID: 35661486 PMCID: PMC9357630 DOI: 10.1111/cas.15456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/23/2022] [Accepted: 05/29/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Dianxing Hu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Ensong Guo
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Bin Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xu Qin
- Department of Stomatology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Yu Fu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Junpeng Fan
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xucui Zhuang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Qianqian Yao
- Department of Medical Science Shanghai AccuraGen Biotechnology Co., Ltd Shanghai China
| | - Funian Lu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Wenting Li
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Rourou Xiao
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xue Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Xiaohang Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Zizhuo Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chen Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Lixin You
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Rongyu Zang
- Department of Gynecologic Oncology, Zhongshan Hospital Fudan University Shanghai China
| | - Qi Zhou
- Department of Gynecology Oncology Chongqing University Cancer Hospital Chongqing China
| | - Weidong Zhao
- Department of Gynecologic Oncology Anhui Provincial Cancer Hospital Hefei China
| | - Gang Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
17
|
Riudavets M, Cascetta P, Planchard D. Targeting BRAF-mutant non-small cell lung cancer: current status and future directions. Lung Cancer 2022; 169:102-114. [DOI: 10.1016/j.lungcan.2022.05.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/24/2022] [Indexed: 10/18/2022]
|
18
|
Riudavets M, Lamberts V, Vasseur D, Auclin E, Aldea M, Jovelet C, Naltet C, Lavaud P, Gazzah A, Aboubakar F, Dorta M, Remon J, Rouleau E, Ngocamus M, Nicotra C, Lacroix L, Besse B, Mezquita L, Planchard D. Clinical utility and outcomes impact of crystal digital PCR of sensitizing and resistance EGFR mutations in patients with advanced non-small cell lung cancer. Clin Lung Cancer 2022; 23:e377-e383. [DOI: 10.1016/j.cllc.2022.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/05/2022] [Accepted: 05/02/2022] [Indexed: 11/03/2022]
|
19
|
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: 8] [Impact Index Per Article: 2.7] [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
|
20
|
Tabbò F, Pisano C, Mazieres J, Mezquita L, Nadal E, Planchard D, Pradines A, Santamaria D, Swalduz A, Ambrogio C, Novello S, Ortiz-Cuaran S. How far we have come targeting BRAF-mutant non-small cell lung cancer (NSCLC). Cancer Treat Rev 2022; 103:102335. [DOI: 10.1016/j.ctrv.2021.102335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 12/19/2021] [Accepted: 12/27/2021] [Indexed: 12/27/2022]
|
21
|
Ueda K, Yamada T, Ohta R, Matsuda A, Sonoda H, Kuriyama S, Takahashi G, Iwai T, Takeda K, Miyasaka T, Shinji S, Chika N, Ishida H, Yoshida H. BRAF V600E mutations in right-side colon cancer: Heterogeneity detected by liquid biopsy. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2022; 48:1375-1383. [DOI: 10.1016/j.ejso.2022.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/30/2021] [Accepted: 01/17/2022] [Indexed: 10/19/2022]
|
22
|
Takam Kamga P, Swalduz A, Costantini A, Julié C, Emile JF, Pérol M, Avrillon V, Ortiz-Cuaran S, de Saintigny P, Leprieur EG. High Circulating Sonic Hedgehog Protein Is Associated With Poor Outcome in EGFR-Mutated Advanced NSCLC Treated With Tyrosine Kinase Inhibitors. Front Oncol 2022; 11:747692. [PMID: 34970481 PMCID: PMC8712335 DOI: 10.3389/fonc.2021.747692] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 11/22/2021] [Indexed: 12/24/2022] Open
Abstract
Introduction Growing preclinical evidence has suggested that the Sonic hedgehog (Shh) pathway is involved in resistance to tyrosine kinase inhibitor (TKI) therapy for EGFR-mutated (EGFRm) non-small cell lung cancer (NSCLC). However, little is known concerning the prognostic value of this pathway in this context. Materials and Methods We investigated the relationship between plasma levels of Shh and EGFRm NSCLC patients’ outcome with EGFR TKIs. We included 74 consecutive patients from two institutions with EGFRm advanced NSCLC treated by EGFR TKI as first-line therapy. Plasma samples were collected longitudinally for each patient and were analyzed for the expression of Shh using an ELISA assay. The activation of the Shh–Gli1 pathway was assessed through immunohistochemistry (IHC) of Gli1 and RT-qPCR analysis of the transcripts of Gli1 target genes in 14 available tumor biopsies collected at diagnosis (baseline). Results Among the 74 patients, only 61 had baseline (diagnosis) plasma samples, while only 49 patients had plasma samples at the first evaluation. Shh protein was detectable in all samples at diagnosis (n = 61, mean = 1,041.2 ± 252.5 pg/ml). Among the 14 available tumor biopsies, nuclear expression of Gli1 was observed in 57.1% (8/14) of patients’ biopsies. Shh was significantly (p < 0.05) enriched in youth (age < 68), male, nonsmokers, patients with a PS > 1, and patients presenting more than 2 metastatic sites and L858R mutation. Higher levels of Shh correlated with poor objective response to TKI, shorter progression-free survival (PFS), and T790M-independent mechanism of resistance. In addition, the rise of plasma Shh levels along the treatment was associated with the emergence of drug resistance in patients presenting an initial good therapy response. Conclusion These data support that higher levels of plasma Shh at diagnosis and increased levels of Shh along the course of the disease are related to the emergence of TKI resistance and poor outcome for EGFR-TKI therapy, suggesting that Shh levels could stand both as a prognostic and as a resistance biomarker for the management of EGFR-mutated NSCLC patients treated with EGFR-TKI.
Collapse
Affiliation(s)
- Paul Takam Kamga
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France
| | - Aurélie Swalduz
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Adrien Costantini
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France.,Department of Respiratory Diseases and Thoracic Oncology, APHP-Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Catherine Julié
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France.,Department of Pathology, APHP-Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Jean-François Emile
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France.,Department of Pathology, APHP-Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Maurice Pérol
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Virginie Avrillon
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Sandra Ortiz-Cuaran
- Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Pierre de Saintigny
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France.,Univ Lyon, Université Claude Bernard Lyon 1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Etienne Giroux Leprieur
- Université Paris-Saclay, UVSQ, EA 4340 BECCOH, Boulogne-Billancourt, France.,Department of Respiratory Diseases and Thoracic Oncology, APHP-Hopital Ambroise Pare, Boulogne-Billancourt, France
| |
Collapse
|
23
|
Pathak N, Chitikela S, Malik PS. Recent advances in lung cancer genomics: Application in targeted therapy. ADVANCES IN GENETICS 2021; 108:201-275. [PMID: 34844713 DOI: 10.1016/bs.adgen.2021.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Genomic characterization of lung cancer has not only improved our understanding of disease biology and carcinogenesis but also revealed several therapeutic opportunities. Targeting tumor dependencies on specific genomic alterations (oncogene addiction) has accelerated the therapeutic developments and significantly improved the outcomes even in advanced stage of disease. Identification of genomic alterations predicting response to specific targeted treatment is the key to success for this "personalized treatment" approach. Availability of multiple choices of therapeutic options for specific genomic alterations highlight the importance of optimum sequencing of drugs. Multiplex gene testing has become mandatory in view of constantly increasing number of therapeutic targets and effective treatment options. Influence of genomic characteristics on response to immunotherapy further makes comprehensive genomic profiling necessary before therapeutic decision making. A comprehensive elucidation of resistance mechanisms and directed treatments have made the continuum of care possible and transformed this deadly disease into a chronic condition. Liquid biopsy-based approach has made the dynamic monitoring of disease possible and enabled treatment optimizations accordingly. Current lung cancer management is the perfect example of "precision-medicine" in clinical oncology.
Collapse
Affiliation(s)
- Neha Pathak
- Department of Medical Oncology, Dr. B.R.A.I.R.C.H., All India Institute of Medical Sciences, New Delhi, India
| | - Sindhura Chitikela
- Department of Medical Oncology, Dr. B.R.A.I.R.C.H., All India Institute of Medical Sciences, New Delhi, India
| | - Prabhat Singh Malik
- Department of Medical Oncology, Dr. B.R.A.I.R.C.H., All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
24
|
Coleman N, Hong L, Zhang J, Heymach J, Hong D, Le X. Beyond epidermal growth factor receptor: MET amplification as a general resistance driver to targeted therapy in oncogene-driven non-small-cell lung cancer. ESMO Open 2021; 6:100319. [PMID: 34837746 PMCID: PMC8637467 DOI: 10.1016/j.esmoop.2021.100319] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/24/2021] [Accepted: 10/27/2021] [Indexed: 12/16/2022] Open
Abstract
The rapidly changing treatment paradigm for patients with metastatic oncogene-driven lung cancer continues to evolve, and consequently our understanding of the landscape of resistance must also advance. MET amplification is an established and frequent driver of resistance in EGFR-mutant non-small-cell lung cancer (NSCLC). Recently, the combination of MET proto-oncogene (MET) and epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) has shown promise in overcoming this molecularly defined resistance in clinical trials, and this combination strategy is being pursued in ongoing trials. Emerging data also demonstrate MET amplification as a resistance driver to TKI-treated ALK-, RET-, and ROS-1-fusion NSCLC, consistently at the range of 15%, while the resistance profiling data are maturing for other molecular targets. In this review, we discuss MET amplification as a driver of acquired resistance in well-defined molecular subsets of NSCLC, explore the biology behind this mechanism of resistance, and summarize the recently published clinical data, including the proposed combination strategies in the clinic achieving success in overcoming acquired MET amplification-dependent resistance. Understanding mechanisms of resistance in oncogene-driven lung cancer is crucial. MET amplification is a recurrent driver of resistance, across molecularly defined subsets of NSCLC. Overcoming this resistance in clinical trials, using combination strategies, is currently being pursued. We explore the biology behind this mechanism of resistance and summarize recent successes in the clinic.
Collapse
Affiliation(s)
- N Coleman
- Department of Investigational Cancer Therapeutics (Phase I Program), University of Texas MD Anderson Cancer Center, Houston, USA
| | - L Hong
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Zhang
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - J Heymach
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA
| | - D Hong
- Department of Investigational Cancer Therapeutics (Phase I Program), University of Texas MD Anderson Cancer Center, Houston, USA
| | - X Le
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
25
|
Rolfo C, Mack P, Scagliotti GV, Aggarwal C, Arcila ME, Barlesi F, Bivona T, Diehn M, Dive C, Dziadziuszko R, Leighl N, Malapelle U, Mok T, Peled N, Raez LE, Sequist L, Sholl L, Swanton C, Abbosh C, Tan D, Wakelee H, Wistuba I, Bunn R, Freeman-Daily J, Wynes M, Belani C, Mitsudomi T, Gandara D. Liquid Biopsy for Advanced NSCLC: A Consensus Statement From the International Association for the Study of Lung Cancer. J Thorac Oncol 2021; 16:1647-1662. [PMID: 34246791 DOI: 10.1016/j.jtho.2021.06.017] [Citation(s) in RCA: 355] [Impact Index Per Article: 88.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/03/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022]
Abstract
Although precision medicine has had a mixed impact on the clinical management of patients with advanced-stage cancer overall, for NSCLC, and more specifically for lung adenocarcinoma, the advances have been dramatic, largely owing to the genomic complexity and growing number of druggable oncogene drivers. Furthermore, although tumor tissue is historically the "accepted standard" biospecimen for these molecular analyses, there are considerable innate limitations. Thus, liquid biopsy represents a practical alternative source for investigating tumor-derived somatic alterations. Although data are most robust in NSCLC, patients with other cancer types may also benefit from this minimally invasive approach to facilitate selection of targeted therapies. The liquid biopsy approach includes a variety of methodologies for circulating analytes. From a clinical point of view, plasma circulating tumor DNA is the most extensively studied and widely adopted alternative to tissue tumor genotyping in solid tumors, including NSCLC, first entering clinical practice for detection of EGFR mutations in NSCLC. Since the publication of the first International Association for the Study of Lung Cancer (IASLC) liquid biopsy statement in 2018, several additional advances have been made in this field, leading to changes in the therapeutic decision-making algorithm for advanced NSCLC and prompting this 2021 update. In view of the novel and impressive technological advances made in the past few years, the growing clinical application of plasma-based, next-generation sequencing, and the recent Food and Drug and Administration approval in the United States of two different assays for circulating tumor DNA analysis, IASLC revisited the role of liquid biopsy in therapeutic decision-making in a recent workshop in October 2020 and the question of "plasma first" versus "tissue first" approach toward molecular testing for advanced NSCLC. Moreover, evidence-based recommendations from IASLC provide an international perspective on when to order which test and how to interpret the results. Here, we present updates and additional considerations to the previous statement article as a consensus from a multidisciplinary and international team of experts selected by IASLC.
Collapse
Affiliation(s)
- Christian Rolfo
- Center for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Philip Mack
- Center for Thoracic Oncology, The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Giorgio V Scagliotti
- Department of Oncology, University of Turin, San Luigi Hospital, Orbassano, Italy
| | - Charu Aggarwal
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Maria E Arcila
- Department of Pathology, Molecular Diagnostics Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Fabrice Barlesi
- CRCM, CNRS, INSERM, Aix Marseille University, Marseille, France; Department of Medical Oncology, Gustave Roussy Cancer Campus, Villejuif, France
| | - Trever Bivona
- Department of Medicine, University of California San Francisco, San Francisco, California; Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California; Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, California
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, California
| | - Caroline Dive
- Cancer Research UK Manchester Institute Cancer Biomarker Centre, University of Manchester, Manchester, United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University of Manchester, Manchester, United Kingdom
| | - Rafal Dziadziuszko
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Gdańsk, Poland
| | - Natasha Leighl
- Princess Margaret Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Tony Mok
- State Key Laboratory of Translational Oncology, Chinese University of Hong Kong, Hong Kong
| | - Nir Peled
- The Legacy Heritage Oncology Center and Dr. Larry Norton Institute, Soroka University Medical Center, Beer-Sheva, Israel
| | - Luis E Raez
- Thoracic Oncology Program, Memorial Cancer Institute/Memorial Health Care System, Florida International University, Miami, Florida
| | - Lecia Sequist
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts; Department of Biostatistics, Massachusetts General Hospital, Boston, Massachusetts; Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, United Kingdom; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Chris Abbosh
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, United Kingdom
| | - Daniel Tan
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore; Duke-NUS Medical School, National University of Singapore, Singapore, Singapore
| | - Heather Wakelee
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Ignacio Wistuba
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rebecca Bunn
- International Association for the Study of Lung Cancer, Aurora, Colorado
| | | | - Murry Wynes
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Chandra Belani
- Department of Medicine Penn State College of Medicine, Penn State Cancer Institute, Hershey, Pennsylvania
| | - Tetsuya Mitsudomi
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of Medicine, Ohno-Higashi, Osaka-Sayama, Japan
| | - David Gandara
- Division of Hematology/Oncology, UC Davis Comprehensive Cancer Center, Sacramento, California.
| |
Collapse
|
26
|
Phase 2 Study of Dabrafenib Plus Trametinib in Patients With BRAF V600E-Mutant Metastatic NSCLC: Updated 5-Year Survival Rates and Genomic Analysis. J Thorac Oncol 2021; 17:103-115. [PMID: 34455067 DOI: 10.1016/j.jtho.2021.08.011] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Dabrafenib plus trametinib was found to have robust antitumor activity in patients with BRAF V600E-mutant metastatic NSCLC (mNSCLC). We report updated survival analysis of a phase 2 study (NCT01336634) with a minimum of 5-year follow-up and updated genomic data. METHODS Pretreated (cohort B) and treatment-naive (cohort C) patients with BRAF V600E-mutant mNSCLC received dabrafenib 150 mg twice daily and trametinib 2 mg once daily. The primary end point was investigator-assessed overall response rate per Response Evaluation Criteria in Solid Tumors version 1.1. Secondary end points were duration of response, progression-free survival, overall survival, and safety. RESULTS At data cutoff, for cohorts B (57 patients) and C (36 patients), the median follow-up was 16.6 (range: 0.5-78.5) and 16.3 (range: 0.4-80) months, overall response rate (95% confidence interval [CI]) was 68.4% (54.8-80.1) and 63.9% (46.2-79.2), median progression-free survival (95% CI) was 10.2 (6.9-16.7) and 10.8 (7.0-14.5) months, and median overall survival (95% CI) was 18.2 (14.3-28.6) and 17.3 (12.3-40.2) months, respectively. The 4- and 5-year survival rates were 26% and 19% in pretreated patients and 34% and 22% in treatment-naive patients, respectively. A total of 17 patients (18%) were still alive. The most frequent adverse event was pyrexia (56%). Exploratory genomic analysis indicated that the presence of coexisting genomic alterations might influence clinical outcomes in these patients; however, these results require further investigation. CONCLUSIONS Dabrafenib plus trametinib therapy was found to have substantial and durable clinical benefit, with a manageable safety profile, in patients with BRAF V600E-mutant mNSCLC, regardless of previous treatment.
Collapse
|
27
|
Niu X, Sun Y, Planchard D, Chiu L, Bai J, Ai X, Lu S. Durable Response to the Combination of Atezolizumab With Platinum-Based Chemotherapy in an Untreated Non-Smoking Lung Adenocarcinoma Patient With BRAF V600E Mutation: A Case Report. Front Oncol 2021; 11:634920. [PMID: 34178624 PMCID: PMC8222507 DOI: 10.3389/fonc.2021.634920] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 05/19/2021] [Indexed: 12/26/2022] Open
Abstract
Background Immune checkpoint inhibitor (ICPi) has become a major treatment in advanced non-small cell lung cancer (NSCLC) and demonstrated a clinical benefit for NSCLC patients with high programmed death ligand-1 (PD-L1) expression without EGFR/ALK/ROS1 drivers; however, the benefit in BRAF V600E NSCLC is so far unknown. Here, we report a case of prolonged tumor response to the combination of immunotherapy with chemotherapy in a non-smoking BRAF V600E NSCLC patient. Materials and Methods We verify a co-expression of BRAF V600E mutation and PD-L1 high expression more than 50% on formalin-fixed paraffin-embedded tumor sample of a newly diagnosed lung adenocarcinoma patient by immunohistochemistry and BRAF V600E/EGFR/ALK/ROS1 Mutations Detection Kit. The tissue and liquid biopsies were further subjected to next-generation sequencing (NGS) for identification of mutations with progression on immunotherapy and BRAF inhibitor (BRAFi). The patient had provided written informed consent and authorized the publication of clinical case. Results We demonstrate the case of 62-year-old female non-smoker with high PD-L1 expression and BRAF V600E mutated NSCLC. The progression-free survival (PFS) of first-line combination of atezolizumab with platinum-based chemotherapy and sequential second-line treatment with BRAFi Vemurafenib are 20 and 5.5 months, respectively. Conclusion This case shows a durable response to ICPi in BRAF V600E non-smoking lung adenocarcinoma with PFS of 20 months under first-line atezolizumab plus chemotherapy treatment. The case supports the idea that the combination immunotherapy may be an attractive option for BRAF V600E mutated non-smoking NSCLC with high PD-L1 expression.
Collapse
Affiliation(s)
- Xiaomin Niu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yingjia Sun
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - David Planchard
- Gustave Roussy, Department of Medical Oncology, Thoracic Unit, Villejuif, France
| | | | - Jian Bai
- Berry Oncology Corporation, Beijing, China
| | - Xinghao Ai
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
28
|
Li M, Zhang X. BRAF Mutations and Resistance of Non-Small Cell Lung Cancer to BRAF-Targeted Therapies Using Liquid Biopsy. Asia Pac J Oncol Nurs 2021; 8:110-111. [PMID: 33688558 PMCID: PMC7934597 DOI: 10.4103/2347-5625.308303] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/18/2020] [Indexed: 01/26/2023] Open
Affiliation(s)
- Ming Li
- Department of Nursing, Zhangqiu People's Hospital of Jinan City, Jinan, China
| | - Xiaoying Zhang
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
29
|
Nokin MJ, Ambrogio C, Nadal E, Santamaria D. Targeting Infrequent Driver Alterations in Non-Small Cell Lung Cancer. Trends Cancer 2020; 7:410-429. [PMID: 33309239 DOI: 10.1016/j.trecan.2020.11.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/12/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023]
Abstract
The discovery of oncogenic driver mutations led to the development of targeted therapies with non-small cell lung cancer (NSCLC) being a paradigm for precision medicine in this setting. Nowadays, the number of clinical trials focusing on targeted therapies for uncommon drivers is growing exponentially, emphasizing the medical need for these patients. Unfortunately, similar to what is observed with most targeted therapies directed against a driver oncogene, the clinical response is almost always temporary and acquired resistance to these drugs invariably emerges. Here, we review the biology of infrequent genomic actionable alterations in NSCLC as well as the current and emerging therapeutic options for these patients. Mechanisms leading to acquired drug resistance and future challenges in the field are also discussed.
Collapse
Affiliation(s)
- Marie-Julie Nokin
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France
| | - Chiara Ambrogio
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ernest Nadal
- Department of Medical Oncology, Catalan Institute of Oncology, Clinical Research in Solid Tumors (CReST) Group, Oncobell Program, IDIBELL, L'Hospitalet, Barcelona, Spain.
| | - David Santamaria
- University of Bordeaux, INSERM U1218, ACTION Laboratory, IECB, 33600 Pessac, France.
| |
Collapse
|
30
|
Malapelle U, Rossi G, Pisapia P, Barberis M, Buttitta F, Castiglione F, Cecere FL, Grimaldi AM, Iaccarino A, Marchetti A, Massi D, Medicina D, Mele F, Minari R, Orlando E, Pagni F, Palmieri G, Righi L, Russo A, Tommasi S, Vermi W, Troncone G. BRAF as a positive predictive biomarker: Focus on lung cancer and melanoma patients. Crit Rev Oncol Hematol 2020; 156:103118. [PMID: 33038627 DOI: 10.1016/j.critrevonc.2020.103118] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 02/06/2023] Open
Abstract
In the era of personalized medicine, BRAF mutational assessment is mandatory in advanced-stage melanoma and non-small cell lung cancer (NSCLC) patients. The identification of actionable mutations is crucial for the adequate management of these patients. To date various drugs have been implemented in clinical practice. Similarly, various methods may be adopted for the identification of BRAF mutations. Here, we briefly review the current literature on BRAF in melanoma and NSCLC, focusing attention in particular on the different methods and drugs adopted in these patients. In addition, an overview of the real-world practice in different Italian laboratories with high expertise in molecular predictive pathology testing is provided.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Giulio Rossi
- Pathology Unit, Azienda USL Romagna, St. Maria delle Croci Hospital, Ravenna, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Massimo Barberis
- Unit of Histopathology and Molecular Diagnostics, European Institute of Oncology IRCCS, Milano, Italy
| | - Fiamma Buttitta
- Center for Advanced Studies and Technology (CAST) - Department of Medical, Oral and Biotechnological Sciences, University of Chieti, Italy
| | - Francesca Castiglione
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy
| | | | - Antonio Maria Grimaldi
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | - Antonino Iaccarino
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Antonio Marchetti
- Center for Advanced Studies and Technology (CAST) - Department of Medical, Oral and Biotechnological Sciences, University of Chieti, Italy
| | - Daniela Massi
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Daniela Medicina
- Section of Pathology, Asst Spedali Civili di Brescia, Brescia, Italy
| | - Fabio Mele
- Pathology Department, IRCCS-Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - Roberta Minari
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Elisabetta Orlando
- Department of Health Promotion, Mother and Child care, Internal Medicine and Medical Specialties (ProMISE), Unit of Anatomic Pathology, University of Palermo, Palermo, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, University Milan Bicocca, Milan, Italy
| | - Giuseppe Palmieri
- Unit of Cancer Genetics, Institute of Genetic and Biomedical Research (IRGB), National Research Council (CNR), Sassari, Italy
| | - Luisella Righi
- Department of Oncology, San Luigi Hospital, University of Turin, Turin, Italy
| | | | - Stefania Tommasi
- Pathology Department, IRCCS-Istituto Tumori 'Giovanni Paolo II', Bari, Italy
| | - William Vermi
- Section of Pathology, Asst Spedali Civili di Brescia, Brescia, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy.
| |
Collapse
|