1
|
Jiao W, Zhao L, Mei J, Zhong J, Yu Y, Bi N, Zhang L, Wang L, Fu X, Wang J, Lu S, Liu L, Gao S. Clinical practice guidelines for perioperative multimodality treatment of non-small cell lung cancer. Chin Med J (Engl) 2025:00029330-990000000-01521. [PMID: 40246578 DOI: 10.1097/cm9.0000000000003635] [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: 01/07/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Lung cancer is currently the most prevalent malignancy and the leading cause of cancer deaths worldwide. Although the early stage non-small cell lung cancer (NSCLC) presents a relatively good prognosis, a considerable number of lung cancer cases are still detected and diagnosed at locally advanced or late stages. Surgical treatment combined with perioperative multimodality treatment is the mainstay of treatment for locally advanced NSCLC and has been shown to improve patient survival. Following the standard methods of neoadjuvant therapy, perioperative management, postoperative adjuvant therapy, and other therapeutic strategies are important for improving patients' prognosis and quality of life. However, controversies remain over the perioperative management of NSCLC and presently consensus and standardized guidelines are lacking for addressing critical clinical issues in multimodality treatment. METHODS The working group consisted of 91 multidisciplinary experts from thoracic surgery, medical oncology, radiotherapy, epidemiology, and psychology. This guideline was developed using the Grading of Recommendations Assessment, Development, and Evaluation (GRADE) system. The clinical questions were collected and selected based on preliminary open-ended questionnaires and subsequent discussions during the Guideline Working Group meetings. PubMed, Web of Science, Cochrane Library, Scopus, and China National Knowledge Infrastructure (CNKI) were searched for available evidence. The GRADE system was used to evaluate the quality of evidence and grade the strengths of recommendations. Finally, the recommendations were developed through a structured consensus-building process. RESULTS The Guideline Development Group initially collected a total of 62 important clinical questions. After a series of consensus-building conferences, 24 clinical questions were identified and corresponding recommendation were ultimately developed, focusing on neoadjuvant therapy, perioperative management, adjuvant therapy, postoperative psychological rehabilitation, prognosis assement, and follow-up protocols for NSCLC. CONCLUSIONS This guideline puts forward reasonable recommendations focusing on neoadjuvant therapy, perioperative management, adjuvant therapy, postoperative psychological rehabilitation, prognosis assessment, and follow-up protocol of NSCLC. It standardizes perioperative multimodality treatment and provides guidance for clinical practice among thoracic surgeons, medical oncologists, and radiotherapists, aiming to reduce postoperative recurrence, improve patient survival, accelerate recovery, and minimize postoperative complications such as atelectasis.
Collapse
Affiliation(s)
- Wenjie Jiao
- Department of Thoracic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266000, China
| | - Liang Zhao
- 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 100021, China
| | - Jiandong Mei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jia Zhong
- CAMS Key Laboratory of Translational Research on Lung Cancer, 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 100021, China
| | - Yongfeng Yu
- Department of Medical Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Nan Bi
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100021, China
| | - Lan Zhang
- Psychological Health Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Lvhua Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong 518117, China
| | - Xiaolong Fu
- Department of Radiation Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jie Wang
- CAMS Key Laboratory of Translational Research on Lung Cancer, 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 100021, China
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Lunxu Liu
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Shugeng Gao
- 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 100021, China
| |
Collapse
|
2
|
Schuurbiers MMF, Smith CG, Hartemink KJ, Rintoul RC, Gale D, Monkhorst K, Mandos BLR, Paterson AL, van den Broek D, Rosenfeld N, van den Heuvel MM, On Behalf of the LEMA Study Group and the LUCID Study Group. Recurrence prediction using circulating tumor DNA in patients with early-stage non-small cell lung cancer after treatment with curative intent: A retrospective validation study. PLoS Med 2025; 22:e1004574. [PMID: 40233104 PMCID: PMC12021277 DOI: 10.1371/journal.pmed.1004574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 04/24/2025] [Accepted: 03/13/2025] [Indexed: 04/17/2025] Open
Abstract
BACKGROUND Despite treatment with curative intent, many patients with localized non-small cell lung cancer (NSCLC) develop recurrence. The current challenge is to identify high-risk patients to guide adjuvant treatment. Identification of residual disease by detection of circulating tumor DNA (ctDNA) may allow more accurate clinical decision-making, but its reliability in NSCLC is not established. We aimed to build on previous data to validate a tissue-informed personalized ctDNA assay, to predict recurrence in patients with early-stage disease. METHODS AND FINDINGS Tumor tissue and plasma was collected from patients with stage 0-III NSCLC enrolled to LEMA (Lung cancer Early Molecular Assessment trial, NCT02894853). Serial plasma was collected before and after definitive treatment, with the latter including key timeframes of interest (1-3 days post-treatment, between 14 and 122 days after treatment end, and ≥14 days after treatment end). Somatic mutations identified by tumor exome sequencing were used to design patient-specific assays, to analyze ctDNA. Results were compared and combined with an independent dataset (LUCID; LUng Cancer CIrculating Tumour Dna study, NCT04153526). In LEMA, 130 patients (57% male; median age 66 years (range 44-82); 69% adenocarcinoma, 22% squamous cell carcinoma (SCC); 3%/49%/19%/29% with stage 0/I/II/III) were treated with curative intent. Tumor tissue originated from surgical resection or diagnostic biopsy in 118 and 12 patients respectively. LUCID included 88 patients (51% male; median age 72 years (range 44-88); 63% adenocarcinoma, 31% SCC; 49%/28%/23% with stage I/II/III). Before treatment, ctDNA was detected in 48% LEMA and 51% LUCID patients. Sensitivity, specificity, positive and negative predictive value of ctDNA detection post-treatment (≥1 positive sample ≥14 days after treatment end) to predict recurrence were 61%, 97%, 92% and 84% for LEMA and 64%, 96%, 90% and 83% for LUCID. In the combined cohort, ctDNA detection after treatment was associated with shorter recurrence-free survival (hazard ratio (HR) 11.4 (95% confidence interval (CI) [7.0,18.7]; p < 0.001)) and overall survival (HR 8.1 (95% CI [4.6,14.2]; p < 0.001)), accounting for guarantee-time bias. Of note, a key limitation of this work was the irregular sample collection schedules, during routine follow-up visits, of both studies. CONCLUSIONS ctDNA detection predicted recurrence in independent retrospective cohorts with notable reproducibility, including near-identical detection rates and predictive values, confirming its ability to differentiate patients at high- versus low risk of recurrence. Our results support the potential of tissue-informed ctDNA analysis as a decision-support tool for adjuvant therapy in NSCLC.
Collapse
MESH Headings
- Humans
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/blood
- Carcinoma, Non-Small-Cell Lung/therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Circulating Tumor DNA/blood
- Circulating Tumor DNA/genetics
- Lung Neoplasms/genetics
- Lung Neoplasms/blood
- Lung Neoplasms/therapy
- Lung Neoplasms/pathology
- Male
- Female
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/blood
- Neoplasm Recurrence, Local/diagnosis
- Aged
- Middle Aged
- Retrospective Studies
- Neoplasm Staging
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Aged, 80 and over
- Adult
- Reproducibility of Results
- Predictive Value of Tests
Collapse
Affiliation(s)
| | | | - Koen J. Hartemink
- Department of Surgery, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Robert C. Rintoul
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
- Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
| | - Davina Gale
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Kim Monkhorst
- Department of Pathology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Bas L. R. Mandos
- Department of Pulmonary Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Anna L. Paterson
- Department of Histopathology, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Daan van den Broek
- Department of Laboratory Medicine, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Nitzan Rosenfeld
- Cancer Research UK Cambridge Centre, Cambridge, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
- Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | | | | |
Collapse
|
3
|
Hait S, Noronha V, Chowdhury A, Chaudhary A, Bawaskar B, Dahimbekar G, Ahmad S, Joshi A, Patil V, Menon N, Shah M, Kaushal R, Choughule A, Bharde A, Khandare J, Shafi G, Lakhwani D, Desai S, Chandrani P, Prabhash K, Dutt A. The impact of co-occurring tumor suppressor mutations with mEGFR as early indicators of relapse in lung cancer. ESMO Open 2025; 10:104479. [PMID: 40088801 PMCID: PMC11937282 DOI: 10.1016/j.esmoop.2025.104479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/28/2025] [Accepted: 02/03/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND Lung adenocarcinoma frequently presents with EGFR mutations, often progressing on EGFR tyrosine kinase inhibitors (TKIs) despite an initial response. Progression is frequently driven by additional genetic changes, including mutations in tumor suppressor genes (TSGs). Understanding the role of these concurrent TSG mutations can help elucidate resistance mechanisms and guide the development of more effective treatment approaches. MATERIALS AND METHODS We examined survival outcomes in 483 EGFR-mutant (mEGFR) patients from the GENIE BPC non-small-cell lung cancer (NSCLC) dataset. To understand the mutational landscape and clonal dynamics, whole exome sequencing (WES) was carried out on 48 tumor samples from 16 mEGFR patients at both baseline and post-relapse. A comprehensive gene panel was applied to 200 liquid biopsy samples obtained longitudinally from 25 patients to track clonal evolution. RESULTS mEGFR patients with co-occurring TSG mutations exhibited significantly worse outcomes. In the GENIE dataset, overall survival (OS) was shorter [51.11 versus 99.3 months; hazard ratio (HR) 1.8, confidence interval (CI) 1.22-2.75, P = 0.003] and progression-free survival (PFS) was reduced (9.83 versus 11.48 months; HR 1.4, CI 1.03-1.91, P = 0.026). WES analysis revealed 17 TSG mutations that were retained and showed clonal enrichment, particularly in early relapse (progression within 10 months of TKI initiation) or intermediate-stage relapse (relapse occurred between 10 and 20 months), indicated by increased variant allele frequency and their presence was strongly linked to early relapse. Longitudinal clonal studies further confirmed that TSG mutations co-occurring with mEGFR were often truncal, predominantly in early relapsers. Survival analysis using this subset of 17 TSGs showed significantly shorter OS (55.26 versus 99.3 months; HR 1.7, CI 1.12-2.65, P = 0.011) and PFS (9.67 versus 13.12 months; HR 1.5, CI 1.08-2.10, P = 0.013). CONCLUSIONS A set of 17 co-occurring TSG mutations has been identified as key biomarkers for early relapse in mEGFR lung adenocarcinoma. Longitudinal genomic monitoring, with a focus on clonal evolution, offers valuable insights that can inform personalized treatment strategies and potentially improve patient outcomes.
Collapse
Affiliation(s)
- S Hait
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - V Noronha
- Homi Bhabha National Institute, Training School Complex, Mumbai, India; Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - A Chowdhury
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - A Chaudhary
- Department of Genetics, University of Delhi South Campus, New Delhi, India
| | - B Bawaskar
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India
| | - G Dahimbekar
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India
| | - S Ahmad
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - A Joshi
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - V Patil
- Homi Bhabha National Institute, Training School Complex, Mumbai, India; Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - N Menon
- Homi Bhabha National Institute, Training School Complex, Mumbai, India; Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - M Shah
- Homi Bhabha National Institute, Training School Complex, Mumbai, India; Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - R Kaushal
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India; Department of Pathology, Tata Memorial Hospital, Mumbai, India
| | - A Choughule
- Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India
| | - A Bharde
- OneCell Diagnostics, Pune, India
| | | | - G Shafi
- OneCell Diagnostics, Pune, India
| | - D Lakhwani
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India
| | - S Desai
- Integrated Cancer Genomics Laboratory, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India; Homi Bhabha National Institute, Training School Complex, Mumbai, India
| | - P Chandrani
- Homi Bhabha National Institute, Training School Complex, Mumbai, India; Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India; Computational Biology, Bioinformatics and Crosstalk Lab, Advanced Centre for Treatment, Research, and Education in Cancer, Navi Mumbai, India
| | - K Prabhash
- Homi Bhabha National Institute, Training School Complex, Mumbai, India; Department of Medical Oncology, Tata Memorial Hospital, Mumbai, India.
| | - A Dutt
- Department of Genetics, University of Delhi South Campus, New Delhi, India.
| |
Collapse
|
4
|
Zhang J, Liu G, Wang D, Bu C, Lv G, Zhang Z, Guo G, Gao Y, Yan Z, Ma C, Liu G, Zhao R, Wang M, Bu X. Dynamic Tumor in Situ Fluid Circulating Tumor DNA Postsurgery Effectively Predicts Recurrence and Clinical Benefits for Glioblastomas. Neurosurgery 2025:00006123-990000000-01532. [PMID: 40042315 DOI: 10.1227/neu.0000000000003384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 12/02/2024] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Glioblastoma (GBM) recurrence after surgery remains a significant clinical challenge because of limited early detection methods and effective molecular markers. This study investigates the potential of dynamic tumor in situ fluid circulating tumor DNA (TISF-ctDNA) as a tool for monitoring molecular residual disease and evaluating treatment efficacy in postsurgical patients with GBM. METHODS In this prospective cohort study, 75 resectable glioma patients were enrolled between October 2019 to June 2023. Primary tumor tissues and postsurgical TISF samples were collected, along with cranial imaging. TISF-ctDNA was identified through targeted next-generation sequencing, with positivity defined by the presence of one or more variants in the TISF matching those of the primary tumors or an increase of ≥10 single-nucleotide variants across consecutive assessments. RESULTS TISF-ctDNA positivity was identified in 62.2% of patients in the early postsurgical period, indicating a high prevalence of molecular residual disease. Patients with positive TISF-ctDNA showed a significantly higher risk of recurrence (hazard ratio 2.512, 95% CI 1.264-4.993, P = .0054). Conversion to TISF-ctDNA negativity post-treatment was associated with improved overall survival, highlighting its role in evaluating treatment response. Multivariate analysis revealed that TISF-ctDNA positivity was an independent predictor of progression-free survival during adjuvant therapy, with high predictive accuracy (sensitivity 86.2%, specificity 100%). Importantly, TISF-ctDNA positivity preceded imaging signs of recurrence by a median of 71 days. In this study, no significant impact of specific tumor markers in each subcohort (EGFR, TP53, PTEN, NF1) on progression-free survival or overall survival was observed. CONCLUSION TISF-ctDNA is a promising biomarker for monitoring GBM recurrence and evaluating clinical benefit. It provides insight into tumor genomic evolution and identifies patients who may benefit from adjuvant chemotherapy. TISF-ctDNA positivity is a robust prognostic indicator for patient outcomes and a marker for molecular recurrence after frontline therapy.
Collapse
Affiliation(s)
- Jiubing Zhang
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
| | - Guanzheng Liu
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
| | - Dayang Wang
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurosurgery, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Chaojie Bu
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Psychological Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Guangming Lv
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
| | - Ziyue Zhang
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
| | - Guangzhong Guo
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yushuai Gao
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhaoyue Yan
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
| | - Chunxiao Ma
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurosurgery, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Gang Liu
- Department of Center for Clinical Single Cell Biomedicine, Department of Oncology, Clinical Research Center, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Ruijiao Zhao
- Department of Pathology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Meiyun Wang
- Department of Radiology, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Radiology, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xingyao Bu
- Department of Neurosurgery, Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Engineering Research Center for Precision Diagnosis and Treatment of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Glioma Clinical Diagnosis and Treatment Center of Henan Province, Henan Provincial People's Hospital, Zhengzhou, China
- Department of Neurosurgery, Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| |
Collapse
|
5
|
Normanno N, Morabito A, Rachiglio AM, Sforza V, Landi L, Bria E, Delmonte A, Cappuzzo F, De Luca A. Circulating tumour DNA in early stage and locally advanced NSCLC: ready for clinical implementation? Nat Rev Clin Oncol 2025; 22:215-231. [PMID: 39833354 DOI: 10.1038/s41571-024-00985-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2024] [Indexed: 01/22/2025]
Abstract
Circulating tumour DNA (ctDNA) can be released by cancer cells into biological fluids through apoptosis, necrosis or active release. In patients with non-small-cell lung cancer (NSCLC), ctDNA levels correlate with clinical and pathological factors, including histology, tumour size and proliferative status. Currently, ctDNA analysis is recommended for molecular profiling in patients with advanced-stage NSCLC. In this Review, we summarize the increasing evidence suggesting that ctDNA has potential clinical applications in the management of patients with early stage and locally advanced NSCLC. In those with early stage NSCLC, detection of ctDNA before and/or after surgery is associated with a greater risk of disease recurrence. Longitudinal monitoring after surgery can further increase the prognostic value of ctDNA testing and enables detection of disease recurrence earlier than the assessment of clinical or radiological progression. In patients with locally advanced NSCLC, the detection of ctDNA after chemoradiotherapy is also associated with a greater risk of disease progression. Owing to the limited number of patients enrolled and the different technologies used for ctDNA testing in most of the clinical studies performed thus far, their results are not sufficient to currently support the routine clinical use of ctDNA monitoring in patients with early stage or locally advanced NSCLC. Therefore, we discuss the need for interventional studies to provide evidence for implementing ctDNA testing in this setting.
Collapse
Affiliation(s)
- Nicola Normanno
- Scientific Directorate, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) "Dino Amadori", Meldola, Italy.
| | - Alessandro Morabito
- Thoracic Department, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Vincenzo Sforza
- Thoracic Department, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| | - Lorenza Landi
- Clinical Trials Center: Phase 1 and Precision Medicine, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Emilio Bria
- Medical Oncology Unit, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
- Medical Oncology, Ospedale Isola Tiberina Gemelli Isola, Roma, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS IRST "Dino Amadori", Meldola, Italy
| | - Federico Cappuzzo
- Division of Medical Oncology 2, IRCCS "Regina Elena" National Cancer Institute, Rome, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
6
|
Veccia A, Dipasquale M, Lorenzi M, Monteverdi S, Kinspergher S, Zambotti E, Caffo O. Osimertinib in the Treatment of Epidermal Growth Factor Receptor-Mutant Early and Locally Advanced Stages of Non-Small-Cell Lung Cancer: Current Evidence and Future Perspectives. Cancers (Basel) 2025; 17:668. [PMID: 40002263 PMCID: PMC11853037 DOI: 10.3390/cancers17040668] [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: 01/12/2025] [Revised: 02/07/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
The treatment of epidermal growth factor receptor (EGFR)-mutant non-small-cell lung cancer (NSCLC) patients was dramatically revolutionized by the introduction of EGFR tyrosine kinase inhibitors in clinical practice, both in advanced and locally advanced/early stages. The present work focuses on osimertinib use in locally advanced and early NSCLC stages. Phase 3 clinical trials have supported the use of osimertinib as the new standard of care, both in the adjuvant setting and in locally advanced disease. The ADAURA study reported an overall survival (OS) advantage for adjuvant osimertinib in completely resected stage II-IIIA EGFR-mutant tumors, while the LAURA study proved a statistically significant benefit in progression-free survival (PFS) and a delay of central nervous system metastasis development in EGFR-mutant patients treated with osimertinib maintenance after concurrent chemoradiotherapy for locally advanced disease. In the neoadjuvant setting, data on osimertinib's efficacy are conflicting; therefore, the Neo-ADAURA study is evaluating the efficacy and safety of neoadjuvant osimertinib alone or in combination with chemotherapy in patients with stage II-IIIB NSCLC and common EGFR mutations. We discuss several issues that need to be clarified, such as the efficacy of the drug on uncommon mutations, the long-term impact on survival, and the management of resistance mechanisms. Moreover, we report the studies that are trying to identify potential biomarkers of response, such as the circulating tumor DNA (ctDNA), with the aim of selecting patients who will benefit most from osimertinib.
Collapse
Affiliation(s)
- Antonello Veccia
- Medical Oncology, Santa Chiara Hospital, Largo Medaglie d’Oro 1, 38122 Trento, Italy; (M.D.); (M.L.); (S.M.); (S.K.); (E.Z.); (O.C.)
| | | | | | | | | | | | | |
Collapse
|
7
|
Saw SPL, Zhong WZ, Fu R, Li MSC, Goto Y, Fox SB, Yatabe Y, Ong BH, Ng CSH, Lee DDW, Cam Phuong P, Park IK, Yang JCH, Tsuboi M, Tho LM, John T, Hsu HH, Tan DSW, Mok TSK, Reungwetwattana T, Singh N. Asian Thoracic Oncology Research Group expert consensus statement on the peri-operative management of non-small cell lung cancer. Lung Cancer 2025; 200:108076. [PMID: 39799810 DOI: 10.1016/j.lungcan.2024.108076] [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: 09/27/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 01/15/2025]
Abstract
The peri-operative management of non-small cell lung cancer (NSCLC) in earlier stage disease has seen significant advances in recent years with the incorporation of immune checkpoint inhibitors and targeted therapy. However, many unanswered questions and challenges remain, including the application of clinical trial data to routine clinical practice. Recognising the unique demographic profile of Asian patients with NSCLC and heterogeneous healthcare systems, the Asian Thoracic Oncology Research Group (ATORG) convened a consensus meeting in Singapore on 26 April 2024 to discuss relevant issues spanning diagnostic testing to post-neoadjuvant treatment considerations and future directions. An interdisciplinary group of 19 experts comprising medical oncologists, thoracic surgeons, radiation oncologists, pulmonologists and pathologists from Singapore, Hong Kong, Mainland China, Korea, Japan, Taiwan, India, Malaysia, Thailand, Vietnam and Australia met to discuss emerging data, identify existing gaps in clinical care and develop a multidisciplinary, multinational expert consensus statement on the peri-operative management of NSCLC tailored to the Asia-Pacific region.
Collapse
Affiliation(s)
- Stephanie P L Saw
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore 168583, Singapore.
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Rui Fu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Molly S C Li
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong China
| | - Yasushi Goto
- National Cancer Center Hospital, Department of Thoracic Oncology, 5-1-1, Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Stephen B Fox
- Department of Pathology, Peter MacCallum Cancer Centre and University of Melbourne, Australia
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center, Tokyo, Japan
| | - Boon-Hean Ong
- Department of Cardiothoracic Surgery, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore
| | - Calvin S H Ng
- Department of Surgery, The Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - David D W Lee
- Department of Clinical Oncology, University of Malaya, Kuala Lumpur, Malaysia
| | - Pham Cam Phuong
- The Nuclear Medicine and Oncology Center, Bach Mai Hospital, 78 Giai Phong Street, Dong Da, Hanoi, Viet Nam
| | - In Kyu Park
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Hospital, Seoul, Korea
| | - James C H Yang
- Department of Oncology, National Taiwan University Hospital, Taiwan
| | - Masahiro Tsuboi
- Department of Thoracic Surgery and Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Lye Mun Tho
- Department of Oncology, Beacon Hospital, Petaling Jaya, Malaysia
| | - Thomas John
- Medical Oncologist, Peter MacCallum Cancer Centre and University of Melbourne, Australia
| | - Hsao-Hsun Hsu
- Department of Surgical Oncology and Surgery, National Taiwan University Cancer Center and College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Daniel S W Tan
- Division of Medical Oncology, National Cancer Centre Singapore, 30 Hospital Boulevard, Singapore 168583, Singapore
| | - Tony S K Mok
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong China
| | - Thanyanan Reungwetwattana
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Navneet Singh
- Lung Cancer Clinic, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
8
|
Miyoshi T, Tsuboi M. Postoperative adjuvant therapy with molecularly targeted agents for non-small cell lung cancer. Int J Clin Oncol 2025; 30:210-214. [PMID: 39692834 DOI: 10.1007/s10147-024-02671-4] [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: 08/28/2024] [Accepted: 11/25/2024] [Indexed: 12/19/2024]
Abstract
The efficacy of molecularly targeted agents has been established in advanced lung cancer, and their indications have recently expanded to include perioperative treatment of resectable lung cancer. For epidermal growth factor receptor (EGFR) mutation-positive patients, postoperative adjuvant therapy with EGFR-tyrosine kinase inhibitors (EGFR-TKIs) is available in Japan following the results of the ADAURA trial. In addition to EGFR-TKIs, postoperative adjuvant therapy with TKIs targeting anaplastic lymphoma kinase (ALK) and rearranged during transfection (RET) is expected to be established. On the other hand, because adjuvant chemotherapy is ineffective in patients who have been completely cured of cancer through surgery alone, the balance between efficacy and adverse effects must be considered, and further studies will be needed to determine the necessary and sufficient dosage and the appropriate duration of administration. In addition, the cost of adjuvant chemotherapy has recently become an issue that cannot be overlooked. Therefore, it will be imperative to develop biomarkers to effectively narrow down the patients who benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Tomohiro Miyoshi
- Division of Thoracic Surgery, Department of Thoracic Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| | - Masahiro Tsuboi
- Division of Thoracic Surgery, Department of Thoracic Oncology, National Cancer Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan
| |
Collapse
|
9
|
Passarella G, Canova S, Abbate MI, Caspani G, Sala L, Russo A, Muscolino P, Colonese F, Cortinovis DL. The hype around ctDNA guiding an informed perioperative therapeutic strategy in early-stage non-small cell lung cancer. Discov Oncol 2025; 16:100. [PMID: 39881042 PMCID: PMC11780067 DOI: 10.1007/s12672-025-01826-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) remains a dire disease being the first cause of cancer death among both genders. Early-stage NSCLC often has better treatment outcomes despite it being a highly heterogeneous disease. So far, the neo-adjuvant chemotherapy strategies have led to a small benefit with an improvement of 5% in overall survival as an absolute benefit. Recently, the introduction of immune checkpoint inhibitors combined with chemotherapy has shown robust efficacy in terms of event-free survival and overall survival. Thus, these combinations are today considered a new standard of care in early-stage NSCLC. The application of these strategies to all-comer population lead to confounding definitive results regarding the efficacy and predictive biomarkers are urgently needed balancing the promise of healing than toxicities. At present, the clinical staging TNM system guides the clinical choice, however it is not entirely sufficient. Circulant tumoral DNA (ctDNA) emerged as a promising prognostic and predictive biomarker that may guide the future perioperative strategy and pave the way to personalized medicine also in this exciting field. This narrative review aims to put in the context the employment of ctDNA, give some perspective and suggestions weighing the pros and cons of this technique for our tomorrow clinical practice.
Collapse
Affiliation(s)
- Gaia Passarella
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
- Medicine and Surgery Department, University of Brescia, Brescia, Italy.
| | - Stefania Canova
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy.
| | - Maria Ida Abbate
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Giulia Caspani
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Medicine and Surgery Department, University of Milano Bicocca, Milan, Italy
| | - Luca Sala
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Alessandro Russo
- Department of Medical Oncology, Humanitas Istituto Clinico Catanese, Misterbianco, Catania, Italy
| | - Paola Muscolino
- Department of Human Pathology "G. Barresi", Medical Oncology Specialization School, University of Messina, Messina, Italy
| | - Francesca Colonese
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Diego Luigi Cortinovis
- Medical Oncology Unit, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
- Medicine and Surgery Department, University of Milano Bicocca, Milan, Italy
| |
Collapse
|
10
|
Black JRM, Bartha G, Abbott CW, Boyle SM, Karasaki T, Li B, Chen R, Harris J, Veeriah S, Colopi M, Bakir MA, Liu WK, Lyle J, Navarro FCP, Northcott J, Pyke RM, Hill MS, Thol K, Huebner A, Bailey C, Colliver EC, Martínez-Ruiz C, Grigoriadis K, Pawlik P, Moore DA, Marinelli D, Shutkever OG, Murphy C, Sivakumar M, Shaw JA, Hackshaw A, McGranahan N, Jamal-Hanjani M, Frankell AM, Chen RO, Swanton C. Ultrasensitive ctDNA detection for preoperative disease stratification in early-stage lung adenocarcinoma. Nat Med 2025; 31:70-76. [PMID: 39806071 PMCID: PMC11750713 DOI: 10.1038/s41591-024-03216-y] [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: 09/21/2023] [Accepted: 07/29/2024] [Indexed: 01/16/2025]
Abstract
Circulating tumor DNA (ctDNA) detection can predict clinical risk in early-stage tumors. However, clinical applications are constrained by the sensitivity of clinically validated ctDNA detection approaches. NeXT Personal is a whole-genome-based, tumor-informed platform that has been analytically validated for ultrasensitive ctDNA detection at 1-3 ppm of ctDNA with 99.9% specificity. Through an analysis of 171 patients with early-stage lung cancer from the TRACERx study, we detected ctDNA pre-operatively within 81% of patients with lung adenocarcinoma (LUAD), including 53% of those with pathological TNM (pTNM) stage I disease. ctDNA predicted worse clinical outcome, and patients with LUAD with <80 ppm preoperative ctDNA levels (the 95% limit of detection of a ctDNA detection approach previously published in TRACERx) experienced reduced overall survival compared with ctDNA-negative patients with LUAD. Although prospective studies are needed to confirm the clinical utility of the assay, these data show that our approach has the potential to improve disease stratification in early-stage LUADs.
Collapse
Affiliation(s)
- James R M Black
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | | | | | | | - Takahiro Karasaki
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Thoracic Surgery, Respiratory Center, Toranomon Hospital, Tokyo, Japan
| | | | - Rui Chen
- Personalis Inc., Fremont, CA, USA
| | | | - Selvaraju Veeriah
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Martina Colopi
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Maise Al Bakir
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Wing Kin Liu
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | | | | | | | | | - Mark S Hill
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Kerstin Thol
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Ariana Huebner
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Chris Bailey
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Emma C Colliver
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Carlos Martínez-Ruiz
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Kristiana Grigoriadis
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Piotr Pawlik
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - David A Moore
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
- Department of Cellular Pathology, University College London Hospitals, London, UK
| | - Daniele Marinelli
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Department of Experimental Medicine, Sapienza University, Rome, Italy
| | - Oliver G Shutkever
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Cian Murphy
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | - Monica Sivakumar
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Jacqui A Shaw
- Leicester NIHR BRC & University of Leicester, Leicester, UK
| | - Allan Hackshaw
- Cancer Research UK & UCL Cancer Trials Centre, London, UK
| | - Nicholas McGranahan
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Genome Evolution Research Group, Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
| | - Mariam Jamal-Hanjani
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Metastasis Laboratory, University College London Cancer Institute, London, UK
- Department of Oncology, University College London Hospitals, London, UK
| | - Alexander M Frankell
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK
| | | | - Charles Swanton
- Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, London, UK.
- Cancer Evolution and Genome Instability Laboratory., The Francis Crick Institute, London, UK.
- Department of Oncology, University College London Hospitals, London, UK.
| |
Collapse
|
11
|
Kang YK, Shin DH, Park JY, Hwang CS, Lee HJ, Lee JH, Kim JY, Na J. Comparison of tissue-based and plasma-based testing for EGFR mutation in non-small cell lung cancer patients. J Pathol Transl Med 2025; 59:60-67. [PMID: 39815744 PMCID: PMC11736281 DOI: 10.4132/jptm.2024.10.01] [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: 06/26/2024] [Revised: 09/25/2024] [Accepted: 09/30/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Epidermal growth factor receptor (EGFR) gene mutation testing is crucial for the administration of tyrosine kinase inhibitors to treat non-small cell lung cancer. In addition to traditional tissue-based tests, liquid biopsies using plasma are increasingly utilized, particularly for detecting T790M mutations. This study compared tissue- and plasma-based EGFR testing methods. METHODS A total of 248 patients were tested for EGFR mutations using tissue and plasma samples from 2018 to 2023 at Pusan National University Yangsan Hospital. Tissue tests were performed using PANAmutyper, and plasma tests were performed using the Cobas EGFR Mutation Test v2. RESULTS All 248 patients underwent tissue-based EGFR testing, and 245 (98.8%) showed positive results. Of the 408 plasma tests, 237 (58.1%) were positive. For the T790M mutation, tissue biopsies were performed 87 times in 69 patients, and 30 positive cases (38.6%) were detected. Plasma testing for the T790M mutation was conducted 333 times in 207 patients, yielding 62 positive results (18.6%). Of these, 57 (27.5%) were confirmed to have the mutation via plasma testing. Combined tissue and plasma tests for the T790M mutation were positive in nine patients (13.4%), while 17 (25.4%) were positive in tissue only and 12 (17.9%) in plasma only. This mutation was not detected in 28 patients (43.3%). CONCLUSIONS Although the tissue- and plasma-based tests showed a sensitivity of 37.3% and 32.8%, respectively, combined testing increased the detection rate to 56.7%. Thus, neither test demonstrated superiority, rather, they were complementary.
Collapse
Affiliation(s)
- Yoon Kyung Kang
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Korea
| | - Dong Hoon Shin
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Korea
| | - Joon Young Park
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Korea
| | - Chung Su Hwang
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Korea
| | - Hyun Jung Lee
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Korea
| | - Jung Hee Lee
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Korea
| | - Jee Yeon Kim
- Department of Pathology, Pusan National University School of Medicine, Yangsan, Korea
| | - JooYoung Na
- Department of Forensic Medicine, Pusan National University School of Medicine, Yangsan, Korea
| |
Collapse
|
12
|
Gristina V, Russo G, Bazan Russo TD, Busuito G, Iannì G, Pisapia P, Scimone C, Palumbo L, Incorvaia L, Badalamenti G, Galvano A, Bazan V, Russo A, Troncone G, Malapelle U, Pepe F. Recent advances in the use of liquid biopsy for the diagnosis and treatment of lung cancer. Expert Rev Respir Med 2024; 18:991-1001. [PMID: 39491533 DOI: 10.1080/17476348.2024.2423824] [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: 09/03/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
INTRODUCTION In the era of precision medicine, liquid biopsy rapidly emerges as an integrative diagnostic tool to successfully stratify solid tumor patients in accordance with molecular fingerprinting. As the matter of fact, a plethora of analytes may be isolated from liquid biosources supporting the potential application of liquid biopsy in several clinical scenarios. Despite this promising role, liquid biopsy is drastically affected by low abundance of analytes in biological matrix requiring highly sensitive technologies, trained personnel, and optimized diagnostic procedures to successfully administrate this revolutionary diagnostic tool in clinical practice. AREAS COVERED This review aims to investigate the recent advancements in technical approaches available to manage liquid biopsy samples, particularly focusing on their application in LC diagnosis and treatment. EXPERT OPINION The rapidly evolving scenario of liquid biopsy-based approaches is revolutionizing clinical administration of lung cancer patients. Of note, the integration of genomic, epigenomic, and transcriptomic markers lays the basis for 'comprehensive' molecular fingerprinting of lung cancer patients. Here, the next-generation technologies are fundamental in molecular profiling in diagnostic routine biofluids.
Collapse
Affiliation(s)
- Valerio Gristina
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Gianluca Russo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Tancredi Didier Bazan Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giulia Busuito
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giuliana Iannì
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Pasquale Pisapia
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Claudia Scimone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lucia Palumbo
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Lorena Incorvaia
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Antonio Galvano
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Viviana Bazan
- Department of Biomedicine, Neuroscience and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, Italy
| | - Antonio Russo
- Department of Precision Medicine in Medical, Surgical and Critical Care (Me.Pre.C.C.), University of Palermo, Palermo, Italy
| | - Giancarlo Troncone
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University Federico II of Naples, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University Federico II of Naples, Naples, Italy
| |
Collapse
|
13
|
Chen C, Chen S, Fu Y, Wei Y, Xie L, Chen M. Electrochemical sensing technology based on a ligation-initiated LAMP-assisted CRISPR/Cas12a system for high-specificity detection of EGFR E746-A750 deletion mutation. Biosens Bioelectron 2024; 263:116635. [PMID: 39116629 DOI: 10.1016/j.bios.2024.116635] [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: 04/27/2024] [Revised: 07/24/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Epidermal growth factor receptor (EGFR) mutation status is pivotal in predicting the efficacy of tyrosine kinase inhibitor treatments against tumors. Among EGFR mutations, the E746-A750 deletion is particularly common and accurately quantifying it can guide targeted therapies. This study introduces a novel visual sensing technology using the clustered regularly interspaced short palindromic repeats (CRISPR)/Cas12a system guided by ligation-initiated loop-mediated isothermal amplification (LAMP) to detect the del E746-A750 mutation in EGFR. Conventional LAMP primers were simplified by designing a pair of target-specific stem-loop DNA probes, enabling selective amplification of the target DNA. The CRISPR/Cas12a system was employed to identify the target nucleic acid and activate Cas12a trans-cleavage activity, thereby enhancing the specificity of the assay. Furthermore, the biosensor utilized high-performance nanomaterials such as triangular gold nanoparticles and graphdiyne, known for their large specific surface area, to enhance sensitivity effectively as a sensing platform. The proposed biosensor demonstrated outstanding specificity, achieving a low detection limit of 17 fM (S/N = 3). Consequently, this innovative strategy not only expands the application scope of CRISPR/Cas12a technology but also introduces a promising approach for clinical diagnostics in modern medicine.
Collapse
Affiliation(s)
- Cizhi Chen
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Siyu Chen
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Yang Fu
- School of Bioscience and Technology, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Yuxin Wei
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Linzhi Xie
- School of Public Health, Chengdu Medical College, Chengdu, Sichuan, 610500, PR China
| | - Mei Chen
- Clinical Laboratory, Clinical Medical College and the First Affiliated Hospital of Chengdu Medical College, Chengdu, Sichuan, 610500, PR China.
| |
Collapse
|
14
|
Ge Q, Zhang ZY, Li SN, Ma JQ, Zhao Z. Liquid biopsy: Comprehensive overview of circulating tumor DNA (Review). Oncol Lett 2024; 28:548. [PMID: 39319213 PMCID: PMC11420644 DOI: 10.3892/ol.2024.14681] [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: 06/21/2024] [Accepted: 08/29/2024] [Indexed: 09/26/2024] Open
Abstract
Traditional tumor diagnosis methods rely on tissue biopsy, which can be invasive and unsuitable for long-term monitoring of tumor dynamics. The advent of liquid biopsy has notably improved the overall management of patients with cancer. Liquid biopsy techniques primarily involve detection of circulating tumor cells (CTCs) and circulating tumor DNA (ctDNA). The present review focuses on ctDNA because of its significance in tumor diagnosis, monitoring and treatment. The use of ctDNA-based liquid biopsy offers several advantages, including non-invasive or minimally invasive collection methods, the ability to conduct repeated assessment and comprehensive insights into tumor biology. It serves crucial roles in disease management by facilitating screening of high-risk patients, dynamically monitoring therapeutic responses and diagnosis. Furthermore, ctDNA can be used to demonstrate pseudo-progression, monitor postoperative tumor status and guide adaptive treatment plans. The present study provides a comprehensive review of ctDNA, exploring its origins, metabolism, detection methods, clinical role and the current challenges associated with its application.
Collapse
Affiliation(s)
- Qian Ge
- Graduate School, Xi'an Medical University, Xi'an, Shaanxi 710000, P.R. China
| | - Zhi-Yun Zhang
- Graduate School, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, P.R. China
| | - Suo-Ni Li
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Jie-Qun Ma
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| | - Zheng Zhao
- Department of Internal Medicine, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi 710000, P.R. China
| |
Collapse
|
15
|
Teng F, Ju X, Gao Z, Xu J, Li Y, Wang Y, Zou B, Yu J. Perioperative immunotherapy for patients with EGFR mutant non-small cell lung cancer: Unexpected potential benefits. Biochim Biophys Acta Rev Cancer 2024; 1879:189194. [PMID: 39413856 DOI: 10.1016/j.bbcan.2024.189194] [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: 05/30/2024] [Revised: 09/01/2024] [Accepted: 10/07/2024] [Indexed: 10/18/2024]
Abstract
Given that immunotherapy has resulted in a significant overall survival (OS) benefit in advanced-stage disease, it is of notable interest to determine the effectiveness of these agents in early-stage non-small cell lung cancer (NSCLC). The potential exists for the immunotherapeutic approach in early-stage NSCLC to mirror the paradigm seen in advanced NSCLC, wherein survival enhancements have notably benefited the majority of patients. However, their performance in early-stage epidermal growth factor receptor (EGFR) mutant NSCLC is controversial. In the limited studies that included patients with EGFR mutation status, we found unexpected, good survival benefits of perioperative immune checkpoint inhibitors (ICIs) in resectable EGFR-positive NSCLC, which is controversial with those in advanced EGFR-mutant NSCLC. It is possible because of the shift toward immunosuppression that the immune environment undergoes during tumor progression. In the early disease stages, the anti-tumor immune response can be activated with fewer hindrances. In the context of EGFR mutant tumors, intratumor genetic heterogeneity can generate treatment-sensitive and -resistant subclones. The subclonality of the resistant subclone is pivotal in therapy response, with tyrosine kinase inhibitors (TKIs) selectively controlling EGFR-mutant cell proliferation and "competitive release" potentially explaining lower pathological responses in adjuvant TKIs trials. This review delves into emerging data on perioperative treatment modalities for early-stage EGFR mutant NSCLC, exploring unique mechanisms and predictive biomarkers to guide perioperative management strategies.
Collapse
Affiliation(s)
- Feifei Teng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China.
| | - Xiao Ju
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhenhua Gao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Junhao Xu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yikun Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yungang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bingwen Zou
- Department of Radiation Oncology, West China Hospital of Sichuan University, Sichuan, China
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China; Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
16
|
Chang ZH, Zhu TF, Ou W, Jiang H, Wang SY. A real-world retrospective study to assess efficacy and safety of alectinib as adjuvant therapy in IB-IIIB NSCLC patients harboring ALK rearrangement. Front Oncol 2024; 14:1422035. [PMID: 39497711 PMCID: PMC11532029 DOI: 10.3389/fonc.2024.1422035] [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: 04/23/2024] [Accepted: 10/07/2024] [Indexed: 11/07/2024] Open
Abstract
Background Alectinib has demonstrated promising disease-free survival (DFS) benefit for early-stage non-small cell lung cancer (NSCLC) patients with ALK rearrangement positive in phase 3 ALINA trial. However, real-world evidence for the efficacy and safety of alectinib in early-stage ALK-positive NSCLC is limited. Materials and methods We retrospectively reviewed 68 patients with stage IB-IIIB ALK-positive NSCLC who underwent complete pulmonary resections from April 2010 to July 2023 at a single institution. 38 (55.9%) enrolled patients had N2 lymph node metastasis, and 17 (24.9%) patients had multi-station N2 metastasis. Patients were stratified into two groups according to the adjuvant treatment regimen, with 19 patients in the alectinib group and 49 patients in the chemotherapy group. There were no significant differences in clinicopathological characteristics between the two groups. After curative resection surgery, patients in alectinib group received oral alectinib at a dose of 600 mg twice daily and patients in chemotherapy group received platinum-based doublet chemotherapy regimen every 3 weeks for 4 cycles. The primary endpoint was 3-year DFS. The Kaplan-Meier method was used to estimate DFS and overall survival (OS). Safety analyses were conducted by comparing the incidence of adverse events between the two groups. Results At the last follow-up date (January 22th, 2024), A total of 1 (5.3%) and 28 (57.1%) DFS events were observed in alectinib group and chemotherapy group respectively. The 3-year DFS showed significant improvement in the alectinib group compared with chemotherapy group (91.7% vs 60.7%, P=0.051). In the IIIAN2 subgroup, the 3-year DFS rate in the alectinib group reached a satisfactory 87.5%. In both groups, the majority of AEs were graded as level 1 or 2, No grade 3-4 AEs were observed in alectinib group. Conclusion Alectinib, as adjuvant therapy, demonstrated favorable efficacy and manageable safety in patients with completely resected ALK-positive stage I B-IIIB non-small cell lung cancer. A limitation of this study is the small sample size, and a larger-scale real-world sample study is needed to further evaluate the efficacy and safety of alectinib as adjuvant therapy.
Collapse
Affiliation(s)
| | | | | | | | - Si-Yu Wang
- Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
17
|
Marinello A, Tagliamento M, Pagliaro A, Conci N, Cella E, Vasseur D, Remon J, Levy A, Dall'Olio FG, Besse B. Circulating tumor DNA to guide diagnosis and treatment of localized and locally advanced non-small cell lung cancer. Cancer Treat Rev 2024; 129:102791. [PMID: 38963991 DOI: 10.1016/j.ctrv.2024.102791] [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: 04/23/2024] [Revised: 06/15/2024] [Accepted: 06/22/2024] [Indexed: 07/06/2024]
Abstract
Liquid biopsy is a minimally invasive method for biomarkers detection in body fluids, particularly in blood, which offers an elevated and growing number of clinical applications in oncology. As a result of the improvement in the techniques for DNA analysis, above all next-generation sequencing (NGS) assays, circulating tumor DNA (ctDNA) has become the most informing tumor-derived material for most types of cancer, including non-small cell lung cancer (NSCLC). Although ctDNA concentration is higher in patients with advanced tumors, it can be detected even in patients with early-stage disease. Therefore, numerous clinical applications of ctDNA in the management of early-stage lung cancer are emerging, such as lung cancer screening, the identification of minimal residual disease (MRD), and the prediction of relapse before radiologic progression. Moreover, a high number of clinical trials are ongoing to better define the impact of ctDNA evaluation in this setting. Aim of this review is to offer a comprehensive overview of the most relevant implementations in using ctDNA for the management of early-stage lung cancer, addressing available data, technical aspects, limitations, and future perspectives.
Collapse
Affiliation(s)
- Arianna Marinello
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; INSERM Unit 1030 - Molecular Radiotherapy and Therapeutic Innovation, Gustave Roussy, Villejuif, France
| | - Marco Tagliamento
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy.
| | - Arianna Pagliaro
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France; Department of Medical Oncology, IRCCS Istituto Clinico Humanitas, Rozzano, Italy
| | - Nicole Conci
- Department of Medical Oncology, IRCCS Sant'Orsola-Malpighi, Bologna, Italy
| | - Eugenia Cella
- Department of Internal Medicine and Medical Specialties, University of Genova, Genova, Italy
| | - Damien Vasseur
- Department of Medical Biology and Pathology, Gustave Roussy, Villejuif, France
| | - Jordi Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Antonin Levy
- Department of Radiotherapy, Gustave Roussy, Villejuif, France
| | | | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
18
|
Lavaud P, Bortolot M, Zullo L, O’Reilly D, Naidoo J, Mountzios G, Mercier O, Hendriks LEL, Remon J. Early-Stage Non-Small Cell Lung Cancer: New Challenges with Immune Checkpoint Blockers and Targeted Therapies. Cancers (Basel) 2024; 16:2779. [PMID: 39199552 PMCID: PMC11353229 DOI: 10.3390/cancers16162779] [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: 07/19/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
The recent advent of tyrosine kinase inhibitors (TKIs) and immune checkpoint blockers (ICBs) in early-stage non-small cell lung cancer (NSCLC) has dramatically modified treatment strategies by improving the prognosis in this setting. Osimertinib and alectinib, both TKIs, have shown significant improvements in outcomes for patients with resected EGFR- and ALK-positive NSCLC, respectively, changing the standard of care in these subgroups. More recently, the LAURA trial showed the efficacy of osimertinib after chemoradiotherapy in patients with unresectable stage III NSCLC harboring EGFR mutations. Numerous trials are still ongoing to investigate neoadjuvant/perioperative TKIs in several oncogene-driven NSCLC. In addition, several ICBs have been tested and approved as adjuvant (atezolizumab and pembrolizumab), neoadjuvant (nivolumab), and perioperative treatments (pembrolizumab) for patients with resectable early-stage NSCLC. Despite these advances, many challenges remain regarding the use of TKIs and ICBs in this setting, including the optimal duration of adjuvant TKI or induction ICB therapy, the role of minimal residual disease to identify patients at high-risk of disease relapse and to guide adjuvant treatment decisions, and the role of adjuvant chemotherapy in resected oncogene-driven NSCLC. Furthermore, potential predictive biomarkers for efficacy are needed to eventually intensify the entire perioperative strategies. This review aims to summarize and discuss the available evidence, the ongoing trials, and the challenges associated with TKI- and ICB-based approaches in early-stage NSCLC.
Collapse
Affiliation(s)
- Pernelle Lavaud
- Gustave Roussy, Department of Cancer Medicine, Paris-Saclay University, 114, rue Edouard Vaillant, 94805 Villejuif, France (L.Z.)
| | - Martina Bortolot
- Department of Respiratory Medicine, Maastricht University Medical Centre, GROW School for Oncology and Reproduction, 6229 ER Maastricht, The Netherlands
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy
| | - Lodovica Zullo
- Gustave Roussy, Department of Cancer Medicine, Paris-Saclay University, 114, rue Edouard Vaillant, 94805 Villejuif, France (L.Z.)
| | - David O’Reilly
- Medical Oncology, Beaumont Hospital, RCSI University of Health Sciences, D02 YN77 Dublin, Ireland
| | - Jarushka Naidoo
- Medical Oncology, Beaumont Hospital, RCSI University of Health Sciences, D02 YN77 Dublin, Ireland
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, 11526 Athens, Greece
| | - Olaf Mercier
- Department of Thoracic Surgery, Hôpital Marie Lannelongue, 92350 Le Plessis-Robinson, France;
| | - Lizza E. L. Hendriks
- Department of Respiratory Medicine, Maastricht University Medical Centre, GROW School for Oncology and Reproduction, 6229 ER Maastricht, The Netherlands
| | - Jordi Remon
- Gustave Roussy, Department of Cancer Medicine, Paris-Saclay University, 114, rue Edouard Vaillant, 94805 Villejuif, France (L.Z.)
| |
Collapse
|
19
|
Maggie Liu SY, Jin ZY, Deng JY, Zhong SM, Ahn MJ, Horinouchi H, Li Y, Wu YL. Drug development and evidence for lung cancer targeted therapy in Eastern Asia. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2024; 49:101090. [PMID: 39381018 PMCID: PMC11459064 DOI: 10.1016/j.lanwpc.2024.101090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/25/2024] [Accepted: 04/29/2024] [Indexed: 10/10/2024]
Abstract
The development of targeted drugs in the Eastern Asia region is going through a flourishing stage. With the continuous advancement of technology and medical research, biotechnology companies and research institutions in the region have made significant progress in cancer field. The Eastern Asian region not only actively participates in clinical trials, but is also committed to developing personalized medical plans to meet the diverse genotypes and phenotypes of patients. The governments and enterprises are increasingly valuing innovation, strengthening international cooperation, and promoting drug development. This paper summarizes the development of genetic testing technology, targeted drugs approval, ongoing promising clinical trials in the field of lung cancer and the important progress made by governments in the Eastern Asian region, and proposed key factors that will contribute to the promising future prospects in the region. The targeted drug market in the Eastern Asian region is expected to drive the medical field forward.
Collapse
Affiliation(s)
- Si-Yang Maggie Liu
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Zhen-Yi Jin
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China
| | - Jia-Yi Deng
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Si-Min Zhong
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Myung-Ju Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Hidehito Horinouchi
- Department of Thoracic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Yangqiu Li
- Department of Hematology, The First Affiliated Hospital, Jinan University, Guangzhou, China
- Institute of Hematology, School of Medicine, Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
20
|
Zhu S, Wu R, Liu X, Xie B, Xie C, Li S, Wu Z, Zhang Z, Tang Z, Gu L. Clinical application of ctDNA in early diagnosis, treatment and prognosis of patients with non-small cell lung cancer. Future Oncol 2024; 20:2213-2224. [PMID: 39073412 PMCID: PMC11514542 DOI: 10.1080/14796694.2024.2376513] [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: 11/21/2023] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Lung cancer is one of the most common malignancies worldwide, with non-small cell lung cancer (NSCLC) being the most common type. As understanding of precise treatment options for NSCLC deepens, circulating tumor DNA (ctDNA) has emerged as a potential biomarker that has become a research hotspot and may represent a new approach for the individualized diagnosis and treatment of NSCLC. This article reviews the applications of ctDNA for the early screening of patients with NSCLC, guiding targeted therapy and immunotherapy, evaluating chemotherapy and postoperative efficacy, assessing prognosis and monitoring recurrence. With the in-depth study of the pathogenesis of NSCLC, plasma ctDNA may become an indispensable part of the precise treatment of NSCLC, which has great clinical application prospects.
Collapse
Affiliation(s)
- Shenyu Zhu
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Rongqian Wu
- Department of Endocrinology and Metabolism, Gaoxin Hospital of The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiangjin Liu
- Department of Thoracic Surgery, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Bin Xie
- First Clinical Medical College, The Gannan Medical University, Ganzhou, China
| | - Chunfa Xie
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Shulin Li
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Zhicheng Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Zuxiong Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Zhixian Tang
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| | - Liang Gu
- Department of Thoracic Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
- Ganzhou Key Lab of Brain Injury & Brain Protection, Ganzhou, China
| |
Collapse
|
21
|
Linares Díaz J, Edwards J, Deleu AL, Giaj-Levra N, Prisciandaro E, Roch B, Paesmans M, Berghmans T, Brandão M. What Does N2 Lymph Node Involvement Mean for Patients with Non-Small Cell Lung Cancer (NSCLC)?-A Review of Implications for Diagnosis and Treatment. Cancers (Basel) 2024; 16:2673. [PMID: 39123401 PMCID: PMC11311523 DOI: 10.3390/cancers16152673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Patients with stage III NSCLC with N2 lymph node involvement carry a complex and diverse disease entity. Challenges persist in the areas of diagnosis, staging, multimodal management, and the determination of surgical indications and resectability criteria. Therefore, this review focuses on the latest updates in N2 disease staging and its prognostic and treatment implications. Emphasis is placed on the importance of accurate staging using imaging modalities such as [18F]FDG-PET/CT as well as minimally invasive mediastinal staging endoscopic techniques. The evolving role of surgery in the management of N2 disease is also explored. The benefits of neoadjuvant and adjuvant treatments have been demonstrated, along with the efficacy of a combined multimodal approach with chemo-immunotherapy in the perioperative setting, reigniting the debate of N2 disease subsets and optimal treatment options. Furthermore, this review addresses the controversies surrounding surgical approaches in upfront "borderline" resectable stage III NSCLC as well as the benefits of combined chemoradiotherapy with consolidation immunotherapy for patients with unresectable tumors. In conclusion, personalized diagnostic and treatment approaches tailored to individual patient characteristics, resource availability, and institutional expertise are essential for optimizing outcomes in patients with stage III-N2 NSCLC.
Collapse
Affiliation(s)
- Julio Linares Díaz
- Medical Oncology Department, Hospital La Fe, Avinguda de Fernando Abril Martorell 106, 46006 Valencia, Spain;
- Thoracic Oncology Functional Unit, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Rue Meylemeersch 90, 1070 Brussels, Belgium;
| | - John Edwards
- Department of Cardiothoracic Surgery, Sheffield Teaching Hospitals NHS Foundation Trust, Herries Road, Sheffield S5 5AU, UK;
| | - Anne-Leen Deleu
- Nuclear Medicine Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Rue Meylemeersch 90, 1070 Brussels, Belgium;
| | - Niccolo Giaj-Levra
- Advanced Radiation Oncology Department, IRCCS Sacro Cuore Don Calabria Hospital, 37024 Verona, Italy;
| | - Elena Prisciandaro
- Department of Thoracic Surgery, Hôpital Erasme, Hôpital Universitaire de Bruxelles, Route de Lennik 808, 1070 Brussels, Belgium;
| | - Benoit Roch
- Thoracic Oncology Unit, Department of Respiratory Diseases, University of Montpellier, CHU Montpellier, F-34295 Montpellier, France;
- Montpellier Cancer Research Institute, University of Montpellier, ICM, INSERM, F-34298 Montpellier, France
| | - Marianne Paesmans
- Statistics Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Rue Meylemeersch 90, 1070 Brussels, Belgium;
| | - Thierry Berghmans
- Thoracic Oncology Functional Unit, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Rue Meylemeersch 90, 1070 Brussels, Belgium;
| | - Mariana Brandão
- Thoracic Oncology Functional Unit, Institut Jules Bordet, Hôpital Universitaire de Bruxelles, Rue Meylemeersch 90, 1070 Brussels, Belgium;
| |
Collapse
|
22
|
Naso JR, Yip S, Hughesman C, Melosky B, Dowhy T, McConechy MK, English JC, Brasher PMA, Choi J, Grant K, Yee J, Lam S, McGuire A. Confirmation of Recurrent Lung Cancer Following Resection Using Liquid Biopsy, a Proof-of-Concept Real-World Study. Curr Oncol 2024; 31:4052-4062. [PMID: 39057174 PMCID: PMC11276127 DOI: 10.3390/curroncol31070302] [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: 06/16/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Appropriate management requires timely and accurate confirmation of non-small cell lung cancer (NSCLC) recurrence in patients who have had curative-intent surgical resection. We assessed the association between circulating tumor DNA (ctDNA) identified using amplicon sequencing and evidence of recurrence on CT surveillance. A prospective cohort study of NSCLC patients with early-stage disease undergoing curative-intent resection was conducted. Surveillance was performed post-operatively at pre-defined intervals with both liquid biopsy and chest CT imaging. Amplicon panel next-generation sequencing was performed on DNA and RNA from tumor tissue and on plasma cell-free DNA for tumor-informed ctDNA detection. Resected tumors from 78 NSCLC patients were analyzed. Alterations were detected on the DNA assay for 65 tumors and only on the RNA assay for 4 tumors. Of the 65 patients with alterations detected on the tumor DNA assay, 29 completed post-operative liquid biopsy testing. Four of those 29 patients had evidence of recurrence on imaging, of whom two had biopsy confirmation of recurrence and detectable ctDNA at the 12-month follow-up. Molecular confirmation of NSCLC recurrence can be provided through amplicon sequencing of plasma cell-free DNA in cases with imaging evidence of recurrence. Invasive tissue diagnosis may be avoidable in patients with ctDNA confirmation of recurrence that is suspected based on imaging. Further study of ctDNA assessment technologies in the setting of suspected recurrence is necessary to inform post-operative lung cancer surveillance guidelines.
Collapse
Affiliation(s)
- Julia R. Naso
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V5Z 4E6, Canada; (J.R.N.)
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Stephen Yip
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V5Z 4E6, Canada; (J.R.N.)
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
- Cancer Genetics and Genomics Laboratory, BC Cancer, Vancouver, BC V5Z 4E6, Canada
- Department of Pathology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Curtis Hughesman
- Department of Pathology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Barb Melosky
- Department of Medical Oncology, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Tanner Dowhy
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, BC V5Z 4S6, Canada
| | | | - John C. English
- Department of Pathology and Laboratory Medicine, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Penelope M. A. Brasher
- Centre for Clinical Epidemiology and Evaluation, Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| | - James Choi
- Division of Thoracic Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Kyle Grant
- Division of Thoracic Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - John Yee
- Division of Thoracic Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
| | - Stephen Lam
- Division of Respirology, Department of Medicine, BC Cancer, Vancouver, BC V5Z 4E6, Canada
| | - Anna McGuire
- Division of Thoracic Surgery, Department of Surgery, Vancouver General Hospital, Vancouver, BC V5Z 1M9, Canada
- Vancouver Coastal Health Research Institute, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
23
|
Zhang C, Sun YX, Yi DC, Jiang BY, Yan LX, Liu ZD, Peng LS, Zhang WJ, Sun H, Chen ZY, Wang DH, Peng D, Chen SA, Li SQ, Zhang Z, Tan XY, Yang J, Zhao ZY, Zhang WT, Su J, Li YS, Liao RQ, Dong S, Xu CR, Zhou Q, Yang XN, Wu YL, Zhang ZM, Zhong WZ. Neoadjuvant sintilimab plus chemotherapy in EGFR-mutant NSCLC: Phase 2 trial interim results (NEOTIDE/CTONG2104). Cell Rep Med 2024; 5:101615. [PMID: 38897205 PMCID: PMC11293361 DOI: 10.1016/j.xcrm.2024.101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/31/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024]
Abstract
The clinical efficacy of neoadjuvant immunotherapy plus chemotherapy remains elusive in localized epidermal growth factor receptor (EGFR)-mutant non-small cell lung cancer (NSCLC). Here, we report interim results of a Simon's two-stage design, phase 2 trial using neoadjuvant sintilimab with carboplatin and nab-paclitaxel in resectable EGFR-mutant NSCLC. All 18 patients undergo radical surgery, with one patient experiencing surgery delay. Fourteen patients exhibit confirmed radiological response, with 44% achieving major pathological response (MPR) and no pathological complete response (pCR). Similar genomic alterations are observed before and after treatment without influencing the efficacy of subsequent EGFR-tyrosine kinase inhibitors (TKIs) in vitro. Infiltration and T cell receptor (TCR) clonal expansion of CCR8+ regulatory T (Treg)hi/CXCL13+ exhausted T (Tex)lo cells define a subtype of EGFR-mutant NSCLC highly resistant to immunotherapy, with the phenotype potentially serving as a promising signature to predict immunotherapy efficacy. Informed circulating tumor DNA (ctDNA) detection in EGFR-mutant NSCLC could help identify patients nonresponsive to neoadjuvant immunochemotherapy. These findings provide supportive data for the utilization of neoadjuvant immunochemotherapy and insight into immune resistance in EGFR-mutant NSCLC.
Collapse
Affiliation(s)
- Chao Zhang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; School of Medicine, South China University of Technology, Guangzhou, China
| | - Yu-Xuan Sun
- School of Life Sciences, Peking University, Beijing, China
| | - Ding-Cheng Yi
- School of Life Sciences, Peking University, Beijing, China
| | - Ben-Yuan Jiang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Li-Xu Yan
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ze-Dao Liu
- School of Life Sciences, Peking University, Beijing, China
| | - Li-Shan Peng
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wen-Jie Zhang
- School of Life Sciences, Peking University, Beijing, China
| | - Hao Sun
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhi-Yong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Department of Radiation Therapy, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | | | - Di Peng
- Burning Rock Biotech, Guangzhou, China
| | | | - Si-Qi Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, China
| | - Ze Zhang
- Institute of Biomedical Research, Yunnan University, Kunming, China
| | - Xiao-Yue Tan
- PET Center, Department of Nuclear Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jie Yang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhang-Yi Zhao
- School of Life Sciences, Peking University, Beijing, China
| | - Wan-Ting Zhang
- School of Life Sciences, Peking University, Beijing, China
| | - Jian Su
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yang-Si Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ri-Qiang Liao
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Song Dong
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chong-Rui Xu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xue-Ning Yang
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Ze-Min Zhang
- School of Life Sciences, Peking University, Beijing, China; BIOPIC, Beijing Advanced Innovation Center for Genomics, Peking University, Beijing, China
| | - Wen-Zhao Zhong
- Department of Pulmonary Surgery, Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China; Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
24
|
Dong S, Wang Z, Zhang JT, Yan B, Zhang C, Gao X, Sun H, Li YS, Yan HH, Tu HY, Liu SYM, Gong Y, Gao W, Huang J, Liao RQ, Lin JT, Ke EE, Xu Z, Zhang X, Xia X, Li AN, Liu SY, Pan Y, Yang JJ, Zhong WZ, Yi X, Zhou Q, Yang XN, Wu YL. Circulating Tumor DNA-Guided De-Escalation Targeted Therapy for Advanced Non-Small Cell Lung Cancer: A Nonrandomized Controlled Trial. JAMA Oncol 2024; 10:932-940. [PMID: 38869865 DOI: 10.1001/jamaoncol.2024.1779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Importance Uninterrupted targeted therapy until disease progression or intolerable toxic effects is currently the routine therapy for advanced non-small cell lung cancer (NSCLC) involving driver gene variations. However, drug resistance is inevitable. Objective To assess the clinical feasibility of adaptive de-escalation tyrosine kinase inhibitor (TKI) treatment guided by circulating tumor DNA (ctDNA) for achieving complete remission after local consolidative therapy (LCT) in patients with advanced NSCLC. Design, Setting, and Participants This prospective nonrandomized controlled trial was conducted at a single center from June 3, 2020, to July 19, 2022, and included 60 patients with advanced NSCLC with driver variations without radiologically detectable disease after TKI and LCT. The median (range) follow-up time was 19.2 (3.8-29.7) months. Data analysis was conducted from December 15, 2022, to May 10, 2023. Intervention Cessation of TKI treatment and follow-up every 3 months. Treatment was restarted in patients with progressive disease (defined by the Response Evaluation Criteria in Solid Tumors 1.1 criteria), detectable ctDNA, or elevated carcinoembryonic antigen (CEA) levels, whichever manifested first, and treatment ceased if all indicators were negative during follow-up surveillance. Main Outcomes and Measures Progression-free survival (PFS). Secondary end points were objective response rate, time to next treatment, and overall survival. Results Among the total study sample of 60 participants (median [range] age, 55 [21-75] years; 33 [55%] were female), the median PFS was 18.4 (95% CI, 12.6-24.2) months and the median (range) total treatment break duration was 9.1 (1.5-28.1) months. Fourteen patients (group A) remained in TKI cessation with a median (range) treatment break duration of 20.3 (6.8-28.1) months; 31 patients (group B) received retreatment owing to detectable ctDNA and/or CEA and had a median PFS of 20.2 (95% CI, 12.9-27.4) months with a median (range) total treatment break duration of 8.8 (1.5-20.6) months; and 15 patients (group C) who underwent retreatment with TKIs due to progressive disease had a median PFS of 5.5 (95% CI, 1.5-7.2) months. For all participants, the TKI retreatment response rate was 96%, the median time to next treatment was 29.3 (95% CI, 25.3-35.2) months, and the data for overall survival were immature. Conclusions and Relevance The findings of this nonrandomized controlled trial suggest that this adaptive de-escalation TKI strategy for patients with NSCLC is feasible in those with no lesions after LCT and a negative ctDNA test result. This might provide a de-escalation treatment strategy guided by ctDNA for the subset of patients with advanced NSCLC. Trial Registration ClinicalTrials.gov Identifier: NCT03046316.
Collapse
Affiliation(s)
- Song Dong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhen Wang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jia-Tao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Bingfa Yan
- Geneplus-Beijing Institute, Beijing, China
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuan Gao
- Geneplus-Beijing Institute, Beijing, China
| | - Hao Sun
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yang-Si Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hong-Hong Yan
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Si-Yang Maggie Liu
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
- Chinese Thoracic Oncology Group, Guangzhou, Guangdong, China
| | - Yuhua Gong
- Geneplus-Beijing Institute, Beijing, China
| | - Wei Gao
- Geneplus-Beijing Institute, Beijing, China
| | - Jie Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Ri-Qiang Liao
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun-Tao Lin
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - E-E Ke
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zelong Xu
- Geneplus-Beijing Institute, Beijing, China
| | - Xue Zhang
- Geneplus-Beijing Institute, Beijing, China
| | | | - An-Na Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Si-Yang Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Pan
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Yi
- Geneplus-Beijing Institute, Beijing, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Chinese Thoracic Oncology Group, Guangzhou, Guangdong, China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Chinese Thoracic Oncology Group, Guangzhou, Guangdong, China
| |
Collapse
|
25
|
Liu Y, Lu XN, Guo HM, Bao C, Zhang J, Jin YN. Development and validation of a circulating tumor DNA-based optimization-prediction model for short-term postoperative recurrence of endometrial cancer. World J Clin Cases 2024; 12:3385-3394. [PMID: 38983398 PMCID: PMC11229938 DOI: 10.12998/wjcc.v12.i18.3385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Endometrial cancer (EC) is a common gynecological malignancy that typically requires prompt surgical intervention; however, the advantage of surgical management is limited by the high postoperative recurrence rates and adverse outcomes. Previous studies have highlighted the prognostic potential of circulating tumor DNA (ctDNA) monitoring for minimal residual disease in patients with EC. AIM To develop and validate an optimized ctDNA-based model for predicting short-term postoperative EC recurrence. METHODS We retrospectively analyzed 294 EC patients treated surgically from 2015-2019 to devise a short-term recurrence prediction model, which was validated on 143 EC patients operated between 2020 and 2021. Prognostic factors were identified using univariate Cox, Lasso, and multivariate Cox regressions. A nomogram was created to predict the 1, 1.5, and 2-year recurrence-free survival (RFS). Model performance was assessed via receiver operating characteristic (ROC), calibration, and decision curve analyses (DCA), leading to a recurrence risk stratification system. RESULTS Based on the regression analysis and the nomogram created, patients with postoperative ctDNA-negativity, postoperative carcinoembryonic antigen 125 (CA125) levels of < 19 U/mL, and grade G1 tumors had improved RFS after surgery. The nomogram's efficacy for recurrence prediction was confirmed through ROC analysis, calibration curves, and DCA methods, highlighting its high accuracy and clinical utility. Furthermore, using the nomogram, the patients were successfully classified into three risk subgroups. CONCLUSION The nomogram accurately predicted RFS after EC surgery at 1, 1.5, and 2 years. This model will help clinicians personalize treatments, stratify risks, and enhance clinical outcomes for patients with EC.
Collapse
Affiliation(s)
- Yuan Liu
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Xiao-Ning Lu
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Hui-Ming Guo
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Chan Bao
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Juan Zhang
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| | - Yu-Ni Jin
- Department of Gynaecology, The First Affiliated Hospital of Kunming Medical University, Kunming 650032, Yunnan Province, China
| |
Collapse
|
26
|
Chaft JE, Dziadziuszko R, Haddock Lobo Goulart B. Moving Immunotherapy Into the Treatment of Resectable Non-Small Cell Lung Cancer. Am Soc Clin Oncol Educ Book 2024; 44:e432500. [PMID: 38788177 DOI: 10.1200/edbk_432500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/26/2024]
Abstract
Clinical investigation of immune checkpoint inhibitors (ICIs) has expanded from indications in metastatic non-small cell lung cancer (NSCLC) to add to the treatment of early-stage or resectable NSCLC. Although completed randomized trials supported the approvals of some ICIs as perioperative therapies (ie, adjuvant, neoadjuvant, or neoadjuvant followed by adjuvant), ongoing trials are evaluating other anti-PD-(L)1 antibodies for similar indications, or in combination with stereotactic body radiotherapy (SBRT). The incorporation of immunotherapy brings potential to improve outcomes of patients with resectable NSCLC, but these advances have also increased the complexity of the treatment landscape and created important knowledge gaps. This article reviews the current standards for local therapies in NSCLC, describes the clinical trials exploring the combination of ICIs to SBRT, and explains the recent approvals of ICIs as perioperative therapies. A discussion follows to highlight three important areas of uncertainty: (1) the contribution of ICIs given in each treatment phase (neoadjuvant and adjuvant) to the overall effect of neoadjuvant chemoimmunotherapy followed by adjuvant ICIs; (2) the selection of regimens to serve as comparators in future randomized trials of perioperative therapies; and (3) the role of pathologic complete response as an intermediate end point and aid for selection of patients for adjuvant therapy. Moving forward, stakeholders will need to engage in concerted research efforts to address the relevant clinical questions regarding the optimal management of resectable NSCLC.
Collapse
Affiliation(s)
- Jamie E Chaft
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Rafal Dziadziuszko
- Dept. of Oncology and Radiotherapy and Early Phase Clinical Trials Center, Medical University of Gdansk, Gdańsk, Poland
| | | |
Collapse
|
27
|
Aguilar H, López-Roldán B, Vilalta-Lacarra A, Alkorta-Aranburu G, Claramunt R, López-Guerrero JA, Sandiego S, Gil-Bazo I. Liquid biopsy for monitoring minimal residual disease in localized and locally-advanced non-small cell lung cancer after radical-intent treatment. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100145. [PMID: 40027142 PMCID: PMC11863883 DOI: 10.1016/j.jlb.2024.100145] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/04/2024] [Accepted: 02/05/2024] [Indexed: 03/05/2025]
Abstract
Blood-based biomarkers investigation does not require invasive tissue biopsies and may explore diverse tumoral components such as proteins, microRNAs, circulating tumor cells, ctDNA, and exosomes and may better reflect tumor molecular heterogeneity, either temporal or spatial. ctDNA is related to tumor burden and represents a more objective measure of the total body disease burden than imaging findings. ctDNA profiling can be therefore useful to determine minimal residual disease (MRD), which is defined as the remaining tumor cells or tumor-derived material after definitive treatment in patients with no clinical evidence of disease. The detection of MRD is highly predictive of future disease recurrence. Although detectable MRD is associated with a poor prognosis, it is not clear whether MRD detection can guide therapy escalation to improve patient outcomes. In this review, we present four cases of epidermal growth factor receptor (EGFR) mutant NSCLC patients who received standard of care curative treatment and periodic radiological assessment and liquid biopsy analyses were carried out as follow-up. A tumor-informed 52 genes Oncomine Pan-Cancer Cell-Free assay (Thermo Fisher Scientific, Waltham, MA, USA), was used to identify single-nucleotide variants (SNVs), indels, copy number variations (CNVs), and RNA fusions in blood based liquid biopsy ctDNA in three of four patients. In one patient the approach used was through Commercial Kit Cobas EGFR Mutation Test v2 CE-IVD (Roche Diagnostics SL) that identifies 42 mutations in the EGFR gene. In one patient, an actionable oncogene driver alteration was identified in the ctDNA analysis, four months after radical intent concurrent chemoradiotherapy and six weeks before radiological distant relapse was clearly confirmed. There is no evidence of ctDNA or radiological disease relapse in the other three patients. Finally, a review of the literature addressing the potential value of MRD detection in this clinical setting is presented and discussed as well.
Collapse
Affiliation(s)
- Héctor Aguilar
- Department of Medical Oncology, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | | | | | | | - R. Claramunt
- Molecular Biology Laboratory, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - José Antonio López-Guerrero
- Molecular Biology Laboratory, Fundación Instituto Valenciano de Oncología, Valencia, Spain
- School of Medicine. Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
| | - S. Sandiego
- Department of Medical Oncology, Fundación Instituto Valenciano de Oncología, Valencia, Spain
| | - I. Gil-Bazo
- Department of Medical Oncology, Fundación Instituto Valenciano de Oncología, Valencia, Spain
- School of Medicine. Universidad Católica de Valencia San Vicente Mártir, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
28
|
Verma S, Young S, Kennedy TAC, Carvalhana I, Black M, Baer K, Churchman E, Warner A, Allan AL, Izaguirre-Carbonell J, Dhani H, Louie AV, Palma DA, Breadner DA. Detection of Circulating Tumor DNA After Stereotactic Ablative Radiotherapy in Patients With Unbiopsied Lung Tumors (SABR-DETECT). Clin Lung Cancer 2024; 25:e87-e91. [PMID: 38101984 DOI: 10.1016/j.cllc.2023.11.013] [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: 08/08/2023] [Revised: 11/22/2023] [Accepted: 11/27/2023] [Indexed: 12/17/2023]
Abstract
For patients with stage I/IIA non-small-cell lung cancer (NSCLC), surgical resection is the standard treatment. However, some of these patients are not candidates for surgery or refuse a surgical option. Definitive stereotactic ablative radiotherapy (SABR) is a standard approach in these patients. Approximately 15% of patients undergoing SABR for localized NSCLC will experience a recurrence within 2 years. Furthermore, many of these patients are deemed appropriate for SABR without a tissue diagnosis, based on the likelihood of malignancy which can be calculated by validated models. A liquid biopsy, detecting ctDNA, would be useful in early detection of recurrences, and documenting a cancer diagnosis in patients without a biopsy. This is a multi-institutional study enrolling patients with suspected stage I/IIA NSCLC and a pretreatment likelihood of malignancy of ≥60% using the validated models for patients without a tissue diagnosis, in cohort 1 (n = 45). The second cohort will consist of biopsied patients (n = 30-60). SABR will be delivered as per risk-adapted protocol. Plasma will be collected for ctDNA analysis prior to the first fraction of SABR, 24 to 72 hours after first fraction, and at 3, 6, 9, 12, 18, and 24-months. The patients will be followed up with imaging at 3, 6, 9, 12, 18, and 24-months. The primary objective is to assess whether a cancer detection liquid biopsy platform can predict recurrence of NSCLC. The secondary objectives are to assess the impact of SABR on detection rates of ctDNA in patients undergoing SABR and to correlate ctDNA positivity and pretreatment probability of malignancy (NCT05921474).
Collapse
Affiliation(s)
- Saurav Verma
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Sympascho Young
- Division of Radiation Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Thomas A C Kennedy
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Ilda Carvalhana
- Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Morgan Black
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Kathie Baer
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Emma Churchman
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Andrew Warner
- Division of Radiation Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Alison L Allan
- London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada; Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | | | | | - Alexander V Louie
- Division of Radiation Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; Department of Radiation Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - David A Palma
- Division of Radiation Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada
| | - Daniel A Breadner
- Division of Medical Oncology, Department of Oncology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada; London Regional Cancer Program, London Health Sciences Centre, London, Ontario, Canada.
| |
Collapse
|
29
|
Desai A, Vázquez TA, Arce KM, Corassa M, Mack PC, Gray JE, Pellini B. ctDNA for the Evaluation and Management of EGFR-Mutant Non-Small Cell Lung Cancer. Cancers (Basel) 2024; 16:940. [PMID: 38473302 DOI: 10.3390/cancers16050940] [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: 12/30/2023] [Revised: 02/05/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Circulating tumor DNA (ctDNA) offers a new paradigm in optimizing treatment strategies for epidermal growth factor receptor (EGFR) mutant non-small cell lung cancer (NSCLC). Its potential spans early-stage disease, influencing adjuvant therapy, to advanced disease, where it aids in identifying genomic markers and resistance mechanisms. This review explores the evolving landscape of utilizing liquid biopsies, specifically circulating tumor DNA (ctDNA), in the management of NSCLC with EGFR mutations. While tissue-based genomic testing remains the cornerstone for clinical decision-making, liquid biopsies offer a well-validated, guideline-recommended alternative approach. Ongoing trials integrating ctDNA for EGFR-mutant NSCLC management are also discussed, shedding light on the potential of ctDNA in early-stage disease, including its applications in prognostication, risk stratification, and minimal residual disease detection post-curative intent treatment. For advanced disease, the role of ctDNA in identifying resistance mechanisms to EGFR tyrosine kinase inhibitors (TKIs) is explored, providing insights into disease progression and guiding treatment decisions. This review also addresses the challenges, including the limitations in sensitivity of current assays for disease recurrence detection, and calls for future studies to refine treatment approaches, standardize reporting, and explore alternative biofluids for enhanced sensitivity. A systematic approach is crucial to address barriers to ctDNA deployment, ensuring equitable access, and facilitating its integration into routine clinical practice.
Collapse
Affiliation(s)
- Aakash Desai
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Tadana A Vázquez
- School of Medicine, Ponce Health Sciences University, Ponce, PR 00716, USA
| | - Keishla M Arce
- School of Medicine, Ponce Health Sciences University, Ponce, PR 00716, USA
| | - Marcelo Corassa
- Thoracic Oncology Unit, BP-A Beneficência Portuguesa de São Paulo, São Paulo 01323-001, Brazil
| | - Philip C Mack
- Center for Thoracic Oncology, The Tisch Cancer Institute, Mount Sinai Health System, New York, NY 10029, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| | - Bruna Pellini
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
- Department of Oncologic Sciences, Morsani College of Medicine, University of South Florida, Tampa, FL 33602, USA
| |
Collapse
|
30
|
Hofman P, Berezowska S, Kazdal D, Mograbi B, Ilié M, Stenzinger A, Hofman V. Current challenges and practical aspects of molecular pathology for non-small cell lung cancers. Virchows Arch 2024; 484:233-246. [PMID: 37801103 PMCID: PMC10948551 DOI: 10.1007/s00428-023-03651-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 10/07/2023]
Abstract
The continuing evolution of treatment options in thoracic oncology requires the pathologist to regularly update diagnostic algorithms for management of tumor samples. It is essential to decide on the best way to use tissue biopsies, cytological samples, as well as liquid biopsies to identify the different mandatory predictive biomarkers of lung cancers in a short turnaround time. However, biological resources and laboratory member workforce are limited and may be not sufficient for the increased complexity of molecular pathological analyses and for complementary translational research development. In this context, the surgical pathologist is the only one who makes the decisions whether or not to send specimens to immunohistochemical and molecular pathology platforms. Moreover, the pathologist can rapidly contact the oncologist to obtain a new tissue biopsy and/or a liquid biopsy if he/she considers that the biological material is not sufficient in quantity or quality for assessment of predictive biomarkers. Inadequate control of algorithms and sampling workflow may lead to false negative, inconclusive, and incomplete findings, resulting in inappropriate choice of therapeutic strategy and potentially poor outcome for patients. International guidelines for lung cancer treatment are based on the results of the expression of different proteins and on genomic alterations. These guidelines have been established taking into consideration the best practices to be set up in clinical and molecular pathology laboratories. This review addresses the current predictive biomarkers and algorithms for use in thoracic oncology molecular pathology as well as the central role of the pathologist, notably in the molecular tumor board and her/his participation in the treatment decision-making. The perspectives in this setting will be discussed.
Collapse
Affiliation(s)
- Paul Hofman
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France.
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France.
| | - Sabina Berezowska
- Department of Laboratory Medicine and Pathology, Institute of Pathology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniel Kazdal
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Baharia Mograbi
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| | - Marius Ilié
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| | - Albrecht Stenzinger
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Centers for Personalized Medicine (ZPM), Heidelberg, Germany
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Véronique Hofman
- Côte d'Azur University, FHU OncoAge, IHU RespirERA, Laboratory of Clinical and Experimental Pathology, BB-0033-00025, Louis Pasteur Hospital, 30 avenue de la voie romaine, BP69, 06001, Nice cedex 01, France
- Côte d'Azur University, IRCAN, Inserm, CNRS 7284, U1081, Nice, France
| |
Collapse
|
31
|
Remon J, Saw SPL, Cortiula F, Singh PK, Menis J, Mountzios G, Hendriks LEL. Perioperative Treatment Strategies in EGFR-Mutant Early-Stage NSCLC: Current Evidence and Future Challenges. J Thorac Oncol 2024; 19:199-215. [PMID: 37783386 DOI: 10.1016/j.jtho.2023.09.1451] [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: 06/12/2023] [Revised: 08/21/2023] [Accepted: 09/27/2023] [Indexed: 10/04/2023]
Abstract
Treatment with 3 years of adjuvant osimertinib is considered a new standard in patients with completely resected stage I to IIIA NSCLC harboring a common sensitizing EGFR mutation. This therapeutic approach significantly prolonged the disease-free survival and the overall survival versus placebo and revealed a significant role in preventing the occurrence of brain metastases. However, many unanswered questions remain, including the optimal duration of this therapy, whether all patients benefit from adjuvant osimertinib, and the role of adjuvant chemotherapy in this population. Indeed, there is a renewed interest in neoadjuvant strategies with targeted therapies in resectable NSCLC harboring oncogenic drivers. In light of these considerations, we discuss the past and current treatment options, and the clinical challenges that should be addressed to optimize the treatment outcomes in this patient population.
Collapse
Affiliation(s)
- Jordi Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France.
| | - Stephanie P L Saw
- Department of Medical Oncology, National Cancer Centre Singapore, Duke-National University of Singapore Oncology Academic Clinical Programme, Singapore
| | | | - Pawan Kumar Singh
- Pandit Bhagwat Dayal Sharma Postgraduate Institute of Medical Science, Rothak, India
| | - Jessica Menis
- Medical Oncology Department, University and Hospital Trust of Verona, Verona, Italy
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | - Lizza E L Hendriks
- Department of Respiratory Medicine, Maastricht University Medical Centre, GROW School for Oncology and Reproduction, Maastricht, The Netherlands
| |
Collapse
|
32
|
Pezeshkian F, McAllister M, Singh A, Theeuwen H, Abdallat M, Figueroa PU, Gill RR, Kim AW, Jaklitsch MT. What's new in thoracic oncology. J Surg Oncol 2024; 129:128-137. [PMID: 38031889 DOI: 10.1002/jso.27535] [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: 11/03/2023] [Revised: 11/08/2023] [Accepted: 11/08/2023] [Indexed: 12/01/2023]
Abstract
Many changes have occurred in the field of thoracic surgery over the last several years. In this review, we will discuss new diagnostic techniques for lung cancer, innovations in surgery, and major updates on latest treatment options including immunotherapy. All these have significantly started to change our approach toward the management of lung cancer and have great potential to improve the lives of our patients afflicted with this disease.
Collapse
Affiliation(s)
- Fatemehsadat Pezeshkian
- Division of Thoracic Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Miles McAllister
- Division of Thoracic Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Anupama Singh
- Division of Thoracic Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Hailey Theeuwen
- Division of Thoracic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Mohammad Abdallat
- Division of Thoracic Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Paula Ugalde Figueroa
- Division of Thoracic Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ritu R Gill
- Department of Radiology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Anthony W Kim
- Division of Thoracic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Michael T Jaklitsch
- Division of Thoracic Surgery, Harvard Medical School, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Zavarykina TM, Lomskova PK, Pronina IV, Khokhlova SV, Stenina MB, Sukhikh GT. Circulating Tumor DNA Is a Variant of Liquid Biopsy with Predictive and Prognostic Clinical Value in Breast Cancer Patients. Int J Mol Sci 2023; 24:17073. [PMID: 38069396 PMCID: PMC10706922 DOI: 10.3390/ijms242317073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
This paper introduces the reader to the field of liquid biopsies and cell-free nucleic acids, focusing on circulating tumor DNA (ctDNA) in breast cancer (BC). BC is the most common type of cancer in women, and progress with regard to treatment has been made in recent years. Despite this, there remain a number of unresolved issues in the treatment of BC; in particular, early detection and diagnosis, reliable markers of response to treatment and for the prediction of recurrence and metastasis, especially for unfavorable subtypes, are needed. It is also important to identify biomarkers for the assessment of drug resistance and for disease monitoring. Our work is devoted to ctDNA, which may be such a marker. Here, we describe its main characteristics and potential applications in clinical oncology. This review considers the results of studies devoted to the analysis of the prognostic and predictive roles of various methods for the determination of ctDNA in BC patients. Currently known epigenetic changes in ctDNA with clinical significance are reviewed. The possibility of using ctDNA as a predictive and prognostic marker for monitoring BC and predicting the recurrence and metastasis of cancer is also discussed, which may become an important part of a precision approach to the treatment of BC.
Collapse
Affiliation(s)
- Tatiana M. Zavarykina
- N.M. Emanuel Institute of Biochemical Physics of the Russian Academy of Sciences, Moscow 119334, Russia;
- “B.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology of Ministry of Health of the Russian Federation, Moscow 117997, Russia; (S.V.K.); (G.T.S.)
| | - Polina K. Lomskova
- N.M. Emanuel Institute of Biochemical Physics of the Russian Academy of Sciences, Moscow 119334, Russia;
| | - Irina V. Pronina
- Institute of General Pathology and Pathophysiology, Moscow 125315, Russia;
| | - Svetlana V. Khokhlova
- “B.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology of Ministry of Health of the Russian Federation, Moscow 117997, Russia; (S.V.K.); (G.T.S.)
| | - Marina B. Stenina
- “N.N. Blokhin National Medical Research Center of Oncology of Ministry of Health of the Russian Federation, Moscow 115522, Russia;
| | - Gennady T. Sukhikh
- “B.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology of Ministry of Health of the Russian Federation, Moscow 117997, Russia; (S.V.K.); (G.T.S.)
| |
Collapse
|
34
|
Facchinetti F, Jänne PA, Tiseo M. Chasing EGFR Mutations in the Plasma of Patients With Resected NSCLC: Lessons in the ADAURA Era. J Thorac Oncol 2023; 18:1118-1120. [PMID: 37599043 DOI: 10.1016/j.jtho.2023.06.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 06/28/2023] [Indexed: 08/22/2023]
Affiliation(s)
- Francesco Facchinetti
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| | - Pasi A Jänne
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts; Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Marcello Tiseo
- Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy; Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|