1
|
Remon J, Bortolot M, Bironzo P, Cortiula F, Menis J, Brandao M, Naidoo J, van Geel R, Reguart N, Arrieta O, Mountzios G, Hendriks LEL, Besse B. De-Escalation Strategies With Immune Checkpoint Blockers in Non-Small Cell Lung Cancer: Do We Already Have Enough Evidence? J Clin Oncol 2025; 43:1148-1156. [PMID: 39836933 DOI: 10.1200/jco-24-02347] [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: 10/20/2024] [Revised: 11/05/2024] [Accepted: 11/27/2024] [Indexed: 01/23/2025] Open
Abstract
Immune checkpoint blockers (ICBs) have revolutionized the treatment of non-small cell lung cancer (NSCLC). Currently, one-dose-fits-all maximalist regimens have been considered the standard of care, with ICBs administered at flat doses regardless of patients' weight. Treatment duration with ICBs is often arbitrary across stages, ranging from a fixed time point to until disease progression or unacceptable toxicity. However, the pharmacokinetic and pharmacodynamic properties of ICBs differ significantly from those of traditional cytotoxic drugs and the approved and selected doses on the basis of the maximum tolerated dose are often overestimated as there is limited evidence supporting a direct relationship between therapeutic intensity and outcomes. This can lead to overtreatment of patients, resulting in an increased risk of toxicity without enhanced efficacy. In addition, the use of these drugs is associated with significant costs that burden the global health care system and exacerbate disparities in access to care. De-escalating treatment by reducing the dose, duration, and frequency of administration of ICBs could optimize treatment efficacy, reduce toxicities, improve patients' quality of life, and even decrease costs. Ultimately, de-escalation strategies may help to reduce treatment inequalities and to improve drug access worldwide. The aim of this review is to summarize and discuss the main issues and challenges regarding the de-escalation of ICBs in patients with NSCLC, focusing on dose-intensity reduction and treatment duration selection. Moreover, we assess the economic impact of implementing de-escalation approaches.
Collapse
Affiliation(s)
- Jordi Remon
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Martina Bortolot
- Department of Pulmonary Diseases, Maastricht University Medical Centre+, GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Paolo Bironzo
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
- Department of Oncology, University of Turin, Turin, Italy
| | - Francesco Cortiula
- Department of Radiation Oncology (Maastro), Maastricht University Medical Centre (+), GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
- University Hospital of Udine, Department of Oncology, Udine, Italy
| | - Jessica Menis
- Medical Oncology Department, University and Hospital Trust of Verona, Verona, Italy
| | - Mariana Brandao
- Institute Jules Bordet-Hôpital Universitaire de Bruxelles, Brussels, Belgium
| | - Jarushka Naidoo
- Beaumont Hospital and RCSI University of Health Sciences, Dublin, Ireland
| | - Robin van Geel
- Department of Clinical Pharmacy & Toxicology, Maastricht University Medical Centre+, Maastricht, the Netherlands
- CARIM School for Cardiovascular Disease, Maastricht University, Maastricht, the Netherlands
| | - Noemi Reguart
- Department of Oncology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Oscar Arrieta
- Thoracic Oncology Unit, Instituto Nacional de Cancerología (INCan), Mexico City, Mexico
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, Athens, Greece
| | - Lizza E L Hendriks
- Department of Pulmonary Diseases, Maastricht University Medical Centre+, GROW Research Institute for Oncology and Reproduction, Maastricht, the Netherlands
| | - Benjamin Besse
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
2
|
Parisi FM, Lentini M, Chiesa-Estomba CM, Mayo-Yanez M, Leichen JR, White M, Giurdanella G, Cocuzza S, Bianco MR, Fakhry N, Maniaci A. Liquid Biopsy in HPV-Associated Head and Neck Cancer: A Comprehensive Review. Cancers (Basel) 2025; 17:977. [PMID: 40149311 PMCID: PMC11940600 DOI: 10.3390/cancers17060977] [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: 01/24/2025] [Revised: 02/25/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
Objectives: Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer globally, with HPV-positive cases emerging as a distinct subtype with unique clinical and molecular characteristics. Current diagnostic methods, including tissue biopsy and imaging, face limitations in terms of invasiveness, static disease assessment, and difficulty in distinguishing recurrence from treatment-related changes. This review aimed to assess the potential of liquid biopsy as a minimally invasive tool for the diagnosis, treatment monitoring, and surveillance of HPV-associated HNSCC. Methods: This systematic review analyzed literature from PubMed/MEDLINE, Embase, and Web of Science, focusing on original research and reviews related to liquid biopsy applications in HPV-positive HNSCC. Included studies were evaluated based on the robustness of the study design, clinical relevance, and analytical performance of liquid biopsy technologies. Biomarker types, detection methods, and implementation strategies were assessed to identify advancements and challenges in this field. Results: Liquid biopsy technologies, including circulating HPV DNA, ctDNA, and extracellular vesicles, demonstrated high sensitivity (90-95%) and specificity (>98%) in detecting HPV-positive HNSCC. These methods enabled real-time monitoring of tumor dynamics, early detection of recurrence, and insights into treatment resistance. Longitudinal analysis revealed that biomarker clearance during treatment correlates strongly with patient outcomes. Conclusions: Liquid biopsy is a transformative diagnostic and monitoring tool for HPV-associated HNSCC, offering minimally invasive, real-time insights into tumor biology. While challenges remain in standardization and clinical implementation, ongoing research and technological innovations hold promise for integrating liquid biopsy into personalized cancer care, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Federica Maria Parisi
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, ENT Section, University of Catania, 95125 Catania, Italy; (F.M.P.); (S.C.)
| | - Mario Lentini
- Department of Otolaryngology, ASP 7, Ragusa Hospital, 97100 Ragusa, Italy
| | - Carlos M. Chiesa-Estomba
- Department of Otorhinolaryngology-Head and Neck Surgery, Hospital Universitario Donostia, 20001 San Sebastian, Spain
| | - Miguel Mayo-Yanez
- Otorhinolaryngology-Head and Neck Surgery Department, Complexo Hospitalario Universitario A Coruña (CHUAC), 15006 La Coruña, Spain;
- Otorhinolaryngology-Head and Neck Surgery Department, Hospital San Rafael (HSR) de A Coruña, 15006 La Coruña, Spain
- Otorhinolaryngology-Head and Neck Surgery Research Group, Institute of Biomedical Research of A Coruña, (INIBIC), Complexo Hospitalario Universitario de A Corñna (CHUAC), Universidade da Corñna (UDC), 15494 La Coruña, Spain
| | - Jerome R. Leichen
- Department of Human Anatomy and Experimental Oncology, UMONS Research Institute for Health Sciences and Technology, University of Mons (UMons), 7011 Mons, Belgium;
| | - Matthew White
- Division of Otorhinolaryngology, Head and Neck Surgery, University of Cape Town, Cape Town 8001, South Africa;
| | - Giovanni Giurdanella
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy;
| | - Salvatore Cocuzza
- Department of Medical and Surgical Sciences and Advanced Technologies “G.F. Ingrassia”, ENT Section, University of Catania, 95125 Catania, Italy; (F.M.P.); (S.C.)
| | - Maria Rita Bianco
- Otolaryngology-Department of Health Science, University of Catanzaro, 88100 Catanzaro, Italy;
| | - Nicolas Fakhry
- Department of Oto-Rhino-Laryngology Head and Neck Surgery, La Conception University Hospital, AP-HM, Aix Marseille Université, 13006 Marseille, France;
| | - Antonino Maniaci
- Department of Medicine and Surgery, University of Enna “Kore”, 94100 Enna, Italy;
| |
Collapse
|
3
|
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
|
4
|
Xu L, Si H, Zhuang F, Li C, Zhang L, Zhao Y, Chen T, Dong Y, Wang T, Hou L, Hu T, Sun T, She Y, Hu X, Xie D, Wu J, Wu C, Zhao D, Chen C. Predicting therapeutic response to neoadjuvant immunotherapy based on an integration model in resectable stage IIIA (N2) non-small cell lung cancer. J Thorac Cardiovasc Surg 2025; 169:242-253.e4. [PMID: 38763304 DOI: 10.1016/j.jtcvs.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/28/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024]
Abstract
OBJECTIVE Accurately predicting response during neoadjuvant chemoimmunotherapy for resectable non-small cell lung cancer remains clinically challenging. In this study, we investigated the effectiveness of blood-based tumor mutational burden (bTMB) and a deep learning (DL) model in predicting major pathologic response (MPR) and survival from a phase 2 trial. METHODS Blood samples were prospectively collected from 45 patients with stage IIIA (N2) non-small cell lung cancer undergoing neoadjuvant chemoimmunotherapy. An integrated model, combining the computed tomography-based DL score, bTMB, and clinical factors, was developed to predict tumor response to neoadjuvant chemoimmunotherapy. RESULTS At baseline, bTMB were detected in 77.8% (35 of 45) of patients. Baseline bTMB ≥11 mutations/megabase was associated with significantly greater MPR rates (77.8% vs 38.5%, P = .042), and longer disease-free survival (P = .043), but not overall survival (P = .131), compared with bTMB <11 mutations/megabase in 35 patients with bTMB available. The developed DL model achieved an area under the curve of 0.703 in all patients. Importantly, the predictive performance of the integrated model improved to an area under the curve of 0.820 when combining the DL score with bTMB and clinical factors. Baseline circulating tumor DNA (ctDNA) status was not associated with pathologic response and survival. Compared with ctDNA residual, ctDNA clearance before surgery was associated with significantly greater MPR rates (88.2% vs 11.1%, P < .001) and improved disease-free survival (P = .010). CONCLUSIONS The integrated model shows promise as a predictor of tumor response to neoadjuvant chemoimmunotherapy. Serial ctDNA dynamics provide a reliable tool for monitoring tumor response.
Collapse
Affiliation(s)
- Long Xu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Haojie Si
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fenghui Zhuang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chongwu Li
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Lei Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yue Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yichen Dong
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tingting Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Likun Hou
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Tao Hu
- Department of Medicine, Amoy Diagnostics Co, Ltd, Xiamen, China
| | - Tianlin Sun
- Department of Medicine, Amoy Diagnostics Co, Ltd, Xiamen, China
| | - Yunlang She
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xuefei Hu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Dong Xie
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
5
|
Acker F, Sebastian M. Can circulating tumor DNA guide treatment de-escalation in metastatic lung adenocarcinoma harboring actionable genomic alterations? Transl Lung Cancer Res 2024; 13:3831-3834. [PMID: 39830755 PMCID: PMC11736600 DOI: 10.21037/tlcr-24-861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/04/2024] [Indexed: 01/22/2025]
Affiliation(s)
- Fabian Acker
- Department of Medicine II, Hematology and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| | - Martin Sebastian
- Department of Medicine II, Hematology and Oncology, Goethe University Frankfurt, University Hospital, Frankfurt, Germany
| |
Collapse
|
6
|
Li S, Yuan T, Yuan J, Zhu B, Chen D. Opportunities and challenges of using circulating tumor DNA to predict lung cancer immunotherapy efficacy. J Cancer Res Clin Oncol 2024; 150:501. [PMID: 39545998 PMCID: PMC11568038 DOI: 10.1007/s00432-024-06030-8] [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: 02/27/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
Immune checkpoint inhibitors (ICIs), particularly anti-programmed death 1 (PD-1)/ programmed death ligand 1 (PD-L1) antibodies, have led to significant progress in lung cancer treatment. However, only a minority of patients have responses to these therapies. Detecting peripheral blood of circulating tumor DNA (ctDNA) allows minimally invasive diagnosis, characterization, and monitoring of lung cancer. ctDNA has potential to be a prognostic biomarker and a predictor of the response to ICI therapy since it can indicate the genomic status and tumor burden. Recent studies on lung cancer have shown that pretreatment ctDNA analysis can detect residual proliferative disease in the adjuvant immunotherapy setting and evaluate tumor burden in patients with metastatic disease. Early ctDNA dynamics can not only predict the clinical outcome of ICI therapy but also help distinguish between pseudoprogression and real progression. Furthermore, in addition to quantitative assessment, ctDNA can also detect genetic predictors of response to ICI therapy. However, barriers still exist in the application of ctDNA analysis in clinical lung cancer treatment. The predictive value of ctDNA in lung cancer immunotherapy requires further identification and resolution of these challenges. This review aims to summarize the existing data of ctDNA analysis in patients receiving immunotherapy for lung cancer, understand the limitations of clinical treatment, and discuss future research directions.
Collapse
Affiliation(s)
- Shanshan Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Ting Yuan
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China
| | - Jing Yuan
- Center for Joint Surgery, Department of Orthopedic Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| | - Degao Chen
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, 400037, China.
| |
Collapse
|
7
|
Jun S, Shukla NA, Durm G, Hui AB, Cao S, Ganti AK, Jabbour SK, Kunder C, Alizadeh AA, Hanna NH, Diehn M. Analysis of Circulating Tumor DNA Predicts Outcomes of Short-Course Consolidation Immunotherapy in Unresectable Stage III NSCLC. J Thorac Oncol 2024; 19:1427-1437. [PMID: 38971369 PMCID: PMC11666285 DOI: 10.1016/j.jtho.2024.06.024] [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: 02/29/2024] [Revised: 06/13/2024] [Accepted: 06/30/2024] [Indexed: 07/08/2024]
Abstract
INTRODUCTION The current standard of care for patients with inoperable stage III non-small cell lung cancer includes chemoradiotherapy (CRT) followed by 1 year of checkpoint inhibitor (CPI) therapy. Nevertheless, the optimal duration of consolidation CPI remains unknown. Here, we characterized the relationship between circulating tumor DNA (ctDNA) minimal residual disease (MRD) and clinical outcomes of patients with unresectable locally advanced non-small cell lung cancer treated on a phase 2 trial of short-course consolidation immunotherapy after CRT, with the goal of testing whether ctDNA may be able to identify patients who do not require a full year of treatment. METHODS Plasma samples for ctDNA analysis were collected from patients on the Big Ten Cancer Research Consortium LUN 16-081 trial after completion of CRT, before day 1 of cycle 2 (C2D1) of CPI (i.e., 1 mo after treatment start), and at the end of up to 6 months of treatment. Tumor-informed ctDNA MRD analysis was performed using cancer personalized profiling by deep sequencing. Levels of ctDNA at each time point were correlated with clinical outcomes. RESULTS Detection of ctDNA predicted significantly inferior progression-free survival after completion of CRT (24-mo 29% versus 65%, p = 0.0048), before C2D1 of CPI (24-mo 0% versus 72%, p < 0.0001) and at the end of CPI (24-mo 15% versus 67%, p = 0.0011). In addition, patients with decreasing or undetectable ctDNA levels after 1 cycle of CPI had improved outcomes compared with patients with increasing ctDNA levels (24-mo progression-free survival 72% versus 0%, p < 0.0001). Progression of disease occurred within less than 12 months of starting CPI in all patients with increasing ctDNA levels at C2D1. CONCLUSIONS Detection of ctDNA before, during, or after 6 months of consolidation CPI is strongly associated with inferior outcomes. Our findings suggest that analysis of ctDNA MRD may enable personalizing the duration of consolidation immunotherapy treatment.
Collapse
Affiliation(s)
- Soyeong Jun
- Department of Radiation Oncology, Stanford University, Stanford, California; Stanford Cancer Institute, Stanford University, Stanford, California
| | - Nikhil A Shukla
- Community Hospital Oncology Physicians, Community Health Network MD Anderson Comprehensive Cancer Center, Indianapolis, Indiana
| | - Greg Durm
- Division of Hematology/Oncology, IU Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Angela B Hui
- Department of Radiation Oncology, Stanford University, Stanford, California; Stanford Cancer Institute, Stanford University, Stanford, California
| | - Sha Cao
- Division of Hematology/Oncology, IU Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Apar Kishor Ganti
- Division of Oncology-Hematology, Department of Internal Medicine, VA Nebraska Western Iowa Health Care System and University of Nebraska Medical Center, Omaha, Nebraska
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers University, New Brunswick, New Jersey
| | - Christian Kunder
- Department of Pathology, Stanford University, Stanford, California
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, California; Division of Oncology, Department of Medicine, Stanford University, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California
| | - Nasser H Hanna
- Division of Hematology/Oncology, IU Simon Comprehensive Cancer Center, Indianapolis, Indiana
| | - Maximilian Diehn
- Department of Radiation Oncology, Stanford University, Stanford, California; Stanford Cancer Institute, Stanford University, Stanford, California; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, California.
| |
Collapse
|
8
|
Xu J, Wan R, Cai Y, Cai S, Wu L, Li B, Duan J, Cheng Y, Li X, Wang X, Han L, Wu X, Fan Y, Yu Y, Lv D, Shi J, Huang J, Zhou S, Han B, Sun G, Guo Q, Ji Y, Zhu X, Hu S, Zhang W, Wang Q, Jia Y, Wang Z, Song Y, Wu J, Shi M, Li X, Han Z, Liu Y, Yu Z, Liu AW, Wang X, Zhou C, Zhong D, Miao L, Zhang Z, Zhao H, Yang J, Wang D, Wang Y, Li Q, Zhang X, Ji M, Yang Z, Cui J, Gao B, Wang B, Liu H, Nie L, He M, Jin S, Gu W, Shu Y, Zhou T, Feng J, Yang X, Huang C, Zhu B, Yao Y, Yu J, Yao S, Shen R, Wang Z, Wang J. Circulating tumor DNA-based stratification strategy for chemotherapy plus PD-1 inhibitor in advanced non-small-cell lung cancer. Cancer Cell 2024; 42:1598-1613.e4. [PMID: 39255777 DOI: 10.1016/j.ccell.2024.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 07/19/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Stratification strategies for chemotherapy plus PD-1 inhibitors in advanced non-small-cell lung cancer (NSCLC) are critically demanded. We performed high-throughput panel-based deep next-generation sequencing and low-pass whole genome sequencing on prospectively collected circulating tumor DNA (ctDNA) specimens from 460 patients in the phase 3 CHOICE-01 study at different time points. We identified predictive markers for chemotherapy plus PD-1 inhibitor, including ctDNA status and genomic features such as blood-based tumor mutational burden, intratumor heterogeneity, and chromosomal instability. Furthermore, we established an integrated ctDNA-based stratification strategy, blood-based genomic immune subtypes (bGIS) scheme, to distinguish patients who benefit from the addition of PD-1 inhibitor to first-line chemotherapy. Moreover, we demonstrated potential applications for the dynamic monitoring of ctDNA. Overall, we proposed a potential therapeutic algorithm based on the ctDNA-based stratification strategy, shedding light on the individualized management of immune-chemotherapies for patients with advanced NSCLC.
Collapse
Affiliation(s)
- Jiachen Xu
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, 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, China
| | - Rui Wan
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, 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, China
| | - Yiran Cai
- Burning Rock Biotech, Guangzhou, China
| | | | - Lin Wu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Baolan Li
- Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jianchun Duan
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, 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, China
| | | | - Xiaoling Li
- Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Xicheng Wang
- The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou, China
| | - Liang Han
- Xuzhou Central Hospital, Xuzhou, China
| | - Xiaohong Wu
- Jiangnan University Affiliated Hospital, Wuxi, China
| | - Yun Fan
- Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou, China
| | - Yan Yu
- Harbin Medical University Cancer Hospital, Harbin, China
| | - Dongqing Lv
- Taizhou Hospital of Zhejiang Province, Linhai, China
| | | | - Jianjin Huang
- The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Shaozhang Zhou
- Guangxi Medical University Affiliated Tumor Hospital, Nanning, China
| | - Baohui Han
- Shanghai Chest Hospital, Shanghai, China
| | - Guogui Sun
- Tangshan People's Hospital, Tangshan, China
| | - Qisen Guo
- Shangdong Cancer Hospital, Jinan, China
| | - Youxin Ji
- Qingdao Central Hospital, Qingdao, China
| | - Xiaoli Zhu
- Zhongda Hospital Southeast University, Nanjing, China
| | - Sheng Hu
- Hubei Cancer Hospital, Wuhan, China
| | - Wei Zhang
- The First Affiliated Hospital of Nanchang University, Nanchang, China
| | | | - Yuming Jia
- The Second People's Hospital of Yibin, Yibin, China
| | - Ziping Wang
- Peking University Cancer Hospital, Beijing, China
| | - Yong Song
- Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jingxun Wu
- The First Affiliated Hospital of Xiamen University, Xiamen, China
| | - Meiqi Shi
- Jiangsu Cancer Hospital, Nanjing, China
| | - Xingya Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhigang Han
- Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Yunpeng Liu
- The First Hospital of China Medical University, Shenyang, China
| | - Zhuang Yu
- The Affiliated Hospital of Qingdao University, Qingdao, China
| | - An-Wen Liu
- The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiuwen Wang
- Qilu Hospital of Shandong University, Jinan, China
| | - Caicun Zhou
- Shanghai Pulmonary Hospital, Shanghai, China
| | | | - Liyun Miao
- Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | | | - Hui Zhao
- The Second Hospital of Anhui Medical University, Hefei, China
| | - Jun Yang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dong Wang
- Army Medical Center of PLA, Daping Hospital, Daping, China
| | - Yingyi Wang
- Peking Union Medical College Hospital, Beijing, China
| | - Qiang Li
- Shanghai East Hospital of Tongji University, Shanghai, China
| | | | - Mei Ji
- The First People's Hospital of Changzhou, Changzhou, China
| | - Zhenzhou Yang
- The Second Affiliated Hospital of Chongqing University, Chongqing, China
| | - Jiuwei Cui
- The First Hospital of Jilin University, Jilin, China
| | - Beili Gao
- Ruijin Hospital Shanghai Jiaotong University, School of Medicine, Shanghai, China
| | - Buhai Wang
- Subei People's Hospital of Jiangsu Province, Yanghzou, China
| | - Hu Liu
- Anhui Provincial Cancer Hospital, Hefei, China
| | - Lei Nie
- Shanxi Provincial Tumor Hospital, Xian, China
| | - Mei He
- Shanxi Provincial People's Hospital, Taiyuan, China
| | - Shi Jin
- Cancer Hospital of Chinese Academy of Medical Sciences, Shenzhen Center, Shenzhen, China
| | - Wei Gu
- Nanjing First Hospital, Nanjing, China
| | - Yongqian Shu
- The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tong Zhou
- ChangZhou Cancer Hospital, Changzhou, China
| | - Jian Feng
- Affiliated Hospital of Nantong University, Nantong, China
| | | | | | - Bo Zhu
- Xinqiao Hospital of Army Medical University, Chongqing, China
| | - Yu Yao
- First Affiliated Hospital of Xi'an Jiaotong University, Xian, China
| | - Jianjun Yu
- Shanghai Junshi Biosciences, Shanghai, China
| | - Sheng Yao
- Shanghai Junshi Biosciences, Shanghai, China
| | | | - Zhijie Wang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, 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, China.
| | - Jie Wang
- State Key Laboratory of Molecular Oncology, CAMS Key Laboratory of Translational Research on Lung Cancer, 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, China.
| |
Collapse
|
9
|
Murciano-Goroff YR, Hui ABY, Araujo Filho JA, Hamilton EG, Chabon JJ, Moding EJ, Bonilla RF, Lebow ES, Gomez D, Rimner A, Ginsberg MS, Offin M, Kundra R, Allaj V, Norton L, Reis-Filho JS, Razavi P, Drilon A, Jones DR, Isbell JM, Lai WV, Rudin CM, Alizadeh AA, Li BT, Diehn M. Early Circulating Tumor DNA Shedding Kinetics for Prediction of Platinum Sensitivity in Patients With Small Cell Lung Cancer. JCO Precis Oncol 2024; 8:e2400216. [PMID: 39231375 PMCID: PMC11376985 DOI: 10.1200/po.24.00216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/06/2024] [Accepted: 07/18/2024] [Indexed: 09/06/2024] Open
Abstract
PURPOSE Small cell lung cancer (SCLC) is characterized by rapid progression after platinum resistance. Circulating tumor (ctDNA) dynamics early in treatment may help determine platinum sensitivity. MATERIALS AND METHODS Serial plasma samples were collected from patients receiving platinum-based chemotherapy for SCLC on the first 3 days of cycle one and on the first days of subsequent cycles with paired samples collected both before and again after infusions. Tumor-informed plasma analysis was carried out using CAncer Personalized Profiling by deep Sequencing (CAPP-Seq). The mean variant allele frequency (VAF) of all pretreatment mutations was tracked in subsequent blood draws and correlated with radiologic response. RESULTS ctDNA kinetics were assessed in 122 samples from 21 patients. Pretreatment VAF did not differ significantly between patients who did and did not respond to chemotherapy (mean 22.5% v 4.6%, P = .17). A slight increase in ctDNA on cycle 1, day 1 immediately post-treatment was seen in six of the seven patients with available draws (fold change from baseline: 1.01-1.44), half of whom achieved a response. All patients who responded had a >2-fold decrease in mean VAF on cycle 2 day 1 (C2D1). Progression-free survival (PFS) and overall survival (OS) were significantly longer in patients with a >2-fold decrease in mean VAF after one treatment cycle (6.8 v 2.6 months, log-rank P = .0004 and 21.7 v 6.4 months, log rank P = .04, respectively). CONCLUSION A >2-fold decrease in ctDNA concentration was observed by C2D1 in all patients who were sensitive to platinum-based therapy and was associated with longer PFS and OS.
Collapse
Affiliation(s)
- Yonina R Murciano-Goroff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Angela B-Y Hui
- Stanford Cancer Institute, Stanford University, Stanford, CA
| | - Jose A Araujo Filho
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Jacob J Chabon
- Stanford Cancer Institute, Stanford University, Stanford, CA
| | - Everett J Moding
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Rene F Bonilla
- Department of Radiation Oncology, Stanford University, Stanford, CA
| | - Emily S Lebow
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michelle S Ginsberg
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Michael Offin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Ritika Kundra
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Viola Allaj
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Larry Norton
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Pedram Razavi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Alexander Drilon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - David R Jones
- Department of Surgery, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - James M Isbell
- Department of Surgery, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - W Victoria Lai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Ash A Alizadeh
- Stanford Cancer Institute, Stanford University, Stanford, CA
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA
| | - Bob T Li
- Department of Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, New York, NY
| | - Maximilian Diehn
- Stanford Cancer Institute, Stanford University, Stanford, CA
- Department of Radiation Oncology, Stanford University, Stanford, CA
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA
| |
Collapse
|
10
|
Jiang H. Latest Research Progress of Liquid Biopsy in Tumor-A Narrative Review. Cancer Manag Res 2024; 16:1031-1042. [PMID: 39165347 PMCID: PMC11335005 DOI: 10.2147/cmar.s479338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Human life expectancy is significantly impacted by cancer, with liquid biopsy emerging as an advantageous method for cancer detection because of its noninvasive nature, high accuracy, ease of sampling, and cost-effectiveness compared with conventional tissue biopsy techniques. Liquid biopsy shows promise in early cancer detection, real-time monitoring, and personalized treatment for various cancers, including lung, cervical, and prostate cancers, and offers innovative approaches for cancer diagnosis and management. By utilizing circulating tumor DNA, circulating tumor cells, and exosomes as biomarkers, liquid biopsy enables the tracking of cancer progression. Various techniques commonly used in life sciences research, such as polymerase chain reaction (PCR), next-generation sequencing (NGS), and droplet digital PCR, are employed to assess cancer progression on the basis of different indicators. This review examines the latest advancements in liquid biopsy markers-circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes-for cancer diagnosis over the past three years, with a focus on their detection methodologies and clinical applications. It encapsulates the pivotal aims of liquid biopsy, including early detection, therapy response prediction, treatment monitoring, prognostication, and its relevance in minimal residual disease, while also addressing the challenges facing routine clinical adoption. By combining the latest research advancements and practical clinical experiences, this work focuses on discussing the clinical significance of DNA methylation biomarkers and their applications in tumor screening, auxiliary diagnosis, companion diagnosis, and recurrence monitoring. These discussions may help enhance the application of liquid biopsy throughout the entire process of tumor diagnosis and treatment, thereby providing patients with more precise and effective treatment plans.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Urology, The Fifth Affiliated Hospital of Zunyi Medical University (Zhuhai Sixth People’s Hospital), Zhuhai, People’s Republic of China
| |
Collapse
|
11
|
Fu R, Xiong Y, Cai M, Li F, Chen R, Wu Y, Zhong W. Evaluation of molecular residual disease in operable non-small cell lung cancer with gene fusions, MET exon skipping or de novo MET amplification. Front Med 2024; 18:735-743. [PMID: 38805102 DOI: 10.1007/s11684-024-1060-z] [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: 09/27/2023] [Accepted: 01/17/2024] [Indexed: 05/29/2024]
Abstract
Gene fusions and MET alterations are rare and difficult to detect in plasma samples. The clinical detection efficacy of molecular residual disease (MRD) based on circulating tumor DNA (ctDNA) in patients with non-small cell lung cancer (NSCLC) with these mutations remains unknown. This prospective, non-intervention study recruited 49 patients with operable NSCLC with actionable gene fusions (ALK, ROS1, RET, and FGFR1), MET exon 14 skipping or de novo MET amplification. We analyzed 43 tumor tissues and 111 serial perioperative plasma samples using 1021- and 338-gene panels, respectively. Detectable MRD correlated with a significantly higher recurrence rate (P < 0.001), yielding positive predictive values of 100% and 90.9%, and negative predictive values of 82.4% and 86.4% at landmark and longitudinal time points, respectively. Patients with detectable MRD showed reduced disease-free survival (DFS) compared to those with undetectable MRD (P < 0.001). Patients who harbored tissue-derived fusion/MET alterations in their MRD had reduced DFS compared to those who did not (P = 0.05). To our knowledge, this is the first comprehensive study on ctDNA-MRD clinical detection efficacy in operable NSCLC patients with gene fusions and MET alterations. Patients with detectable tissue-derived fusion/MET alterations in postoperative MRD had worse clinical outcomes.
Collapse
Affiliation(s)
- Rui Fu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | | | - Miao Cai
- Geneplus-Beijing, Beijing, 102206, China
| | - Fang Li
- Geneplus-Beijing, Beijing, 102206, China
| | | | - Yilong Wu
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wenzhao Zhong
- School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- Guangdong Lung Cancer Institute, Guangdong Provincial Key Laboratory of Translational Medicine in Lung Cancer, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China.
| |
Collapse
|
12
|
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
|
13
|
Yeghaian M, Tareco Bucho TM, de Bruin M, Schmitz A, Bodalal Z, Smit EF, Beets-Tan RGH, van den Broek D, Trebeschi S. Can blood-based markers predict RECIST progression in non-small cell lung cancer treated with immunotherapy? J Cancer Res Clin Oncol 2024; 150:329. [PMID: 38922374 PMCID: PMC11208229 DOI: 10.1007/s00432-024-05814-2] [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: 12/01/2023] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
PURPOSE In this study, we aimed to evaluate the potential of routine blood markers, serum tumour markers and their combination in predicting RECIST-defined progression in patients with stage IV non-small cell lung cancer (NSCLC) undergoing treatment with immune checkpoint inhibitors. METHODS We employed time-varying statistical models and machine learning classifiers in a Monte Carlo cross-validation approach to investigate the association between RECIST-defined progression and blood markers, serum tumour markers and their combination, in a retrospective cohort of 164 patients with NSCLC. RESULTS The performance of the routine blood markers in the prediction of progression free survival was moderate. Serum tumour markers and their combination with routine blood markers generally improved performance compared to routine blood markers alone. Elevated levels of C-reactive protein (CRP) and alkaline phosphatase (ALP) ranked as the top predictive routine blood markers, and CYFRA 21.1 was consistently among the most predictive serum tumour markers. Using these classifiers to predict overall survival yielded moderate to high performance, even when cases of death-defined progression were excluded. Performance varied across the treatment journey. CONCLUSION Routine blood tests, especially when combined with serum tumour markers, show moderate predictive value of RECIST-defined progression in NSCLC patients receiving immune checkpoint inhibitors. The relationship between overall survival and RECIST-defined progression may be influenced by confounding factors.
Collapse
Affiliation(s)
- Melda Yeghaian
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Radiology Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Teresa M Tareco Bucho
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Radiology Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Melissa de Bruin
- Radiology Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Alexander Schmitz
- Radiology Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Zuhir Bodalal
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Radiology Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Egbert F Smit
- Pulmonology Department, Leiden University Medical Center, Leiden, The Netherlands
| | - Regina G H Beets-Tan
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Radiology Department, Netherlands Cancer Institute, Amsterdam, The Netherlands
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Daan van den Broek
- Department of Laboratory Medicine, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Stefano Trebeschi
- GROW Research Institute for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands.
- Radiology Department, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
14
|
Song Y, Loomans-Kropp H, Baugher RN, Somerville B, Baxter SS, Kerr TD, Plona TM, Mellott SD, Young TB, Lawhorn HE, Wei L, Hu Q, Liu S, Hutson A, Pinto L, Potter JD, Sei S, Gelincik O, Lipkin SM, Gebert J, Kloor M, Shoemaker RH. Frameshift mutations in peripheral blood as a biomarker for surveillance of Lynch syndrome. J Natl Cancer Inst 2024; 116:957-965. [PMID: 38466935 PMCID: PMC11160491 DOI: 10.1093/jnci/djae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/06/2024] [Accepted: 02/27/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Lynch syndrome is a hereditary cancer predisposition syndrome caused by germline mutations in DNA mismatch repair genes, which lead to high microsatellite instability and frameshift mutations at coding mononucleotide repeats in the genome. Recurrent frameshift mutations in these regions are thought to play a central role in the increased risk of various cancers, but no biomarkers are currently available for the surveillance of high microsatellite instability-associated cancers. METHODS A frameshift mutation-based biomarker panel was developed and validated by targeted next-generation sequencing of supernatant DNA from cultured high microsatellite instability colorectal cancer cells. This panel supported selection of 122 frameshift mutation targets as potential biomarkers. This biomarker panel was then tested using matched tumor, adjacent normal tissue, and buffy coat samples (53 samples) and blood-derived cell-free DNA (cfDNA) (38 samples) obtained from 45 high microsatellite instability and mismatch repair-deficient patients. We also sequenced cfDNA from 84 healthy participants to assess background noise. RESULTS Recurrent frameshift mutations at coding mononucleotide repeats were detectable not only in tumors but also in cfDNA from high microsatellite instability and mismatch repair-deficient patients, including a Lynch syndrome carrier, with a varying range of target detection (up to 85.2%), whereas they were virtually undetectable in healthy participants. Receiver operating characteristic curve analysis showed high sensitivity and specificity (area under the curve = 0.94) of the investigated panel. CONCLUSIONS We demonstrated that frameshift mutations can be detected in cfDNA from high microsatellite instability and mismatch repair-deficient patients and asymptomatic carriers. The 122-target frameshift mutation panel described here has promise as a tool for improved surveillance of high microsatellite instability and mismatch repair-deficient patients, with the potential to reduce the frequency of invasive screening methods for this high-cancer-risk cohort.
Collapse
Affiliation(s)
- Yurong Song
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Holli Loomans-Kropp
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
- Now at Department of Internal Medicine, The Ohio State University, Columbus, OH, USA
| | - Ryan N Baugher
- Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Brandon Somerville
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Shaneen S Baxter
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Travis D Kerr
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Teri M Plona
- Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Stephanie D Mellott
- Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Todd B Young
- Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Heidi E Lawhorn
- Molecular Diagnostics Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Lei Wei
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Qiang Hu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Song Liu
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Alan Hutson
- Department of Biostatistics and Bioinformatics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Ligia Pinto
- Vaccine, Immunity and Cancer Directorate, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Research Centre for Hauora and Health, Massey University, Wellington, New Zealand
- School of Public Health, University of Washington, Seattle, WA, USA
| | - Shizuko Sei
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Ozkan Gelincik
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Steven M Lipkin
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Johannes Gebert
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Kloor
- Department of Applied Tumor Biology, Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Robert H Shoemaker
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
15
|
Wang H, Zhang Y, Zhang H, Cao H, Mao J, Chen X, Wang L, Zhang N, Luo P, Xue J, Qi X, Dong X, Liu G, Cheng Q. Liquid biopsy for human cancer: cancer screening, monitoring, and treatment. MedComm (Beijing) 2024; 5:e564. [PMID: 38807975 PMCID: PMC11130638 DOI: 10.1002/mco2.564] [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: 04/23/2023] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/30/2024] Open
Abstract
Currently, tumor treatment modalities such as immunotherapy and targeted therapy have more stringent requirements for obtaining tumor growth information and require more accurate and easy-to-operate tumor information detection methods. Compared with traditional tissue biopsy, liquid biopsy is a novel, minimally invasive, real-time detection tool for detecting information directly or indirectly released by tumors in human body fluids, which is more suitable for the requirements of new tumor treatment modalities. Liquid biopsy has not been widely used in clinical practice, and there are fewer reviews of related clinical applications. This review summarizes the clinical applications of liquid biopsy components (e.g., circulating tumor cells, circulating tumor DNA, extracellular vesicles, etc.) in tumorigenesis and progression. This includes the development process and detection techniques of liquid biopsies, early screening of tumors, tumor growth detection, and guiding therapeutic strategies (liquid biopsy-based personalized medicine and prediction of treatment response). Finally, the current challenges and future directions for clinical applications of liquid biopsy are proposed. In sum, this review will inspire more researchers to use liquid biopsy technology to promote the realization of individualized therapy, improve the efficacy of tumor therapy, and provide better therapeutic options for tumor patients.
Collapse
Affiliation(s)
- Hao Wang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Yi Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Hao Zhang
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Hui Cao
- Department of PsychiatryThe School of Clinical Medicine, Hunan University of Chinese MedicineChangshaChina
- Department of PsychiatryBrain Hospital of Hunan Province (The Second People’s Hospital of Hunan Province)ChangshaChina
| | - Jinning Mao
- Health Management CenterThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Xinxin Chen
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Liangchi Wang
- Department of NeurosurgeryFengdu People's Hospital, ChongqingChongqingChina
| | - Nan Zhang
- College of Life Science and TechnologyHuazhong University of Science and TechnologyWuhanChina
| | - Peng Luo
- Department of OncologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Ji Xue
- Department of NeurosurgeryTraditional Chinese Medicine Hospital Dianjiang ChongqingChongqingChina
| | - Xiaoya Qi
- Health Management CenterThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Xiancheng Dong
- Department of Cerebrovascular DiseasesDazhou Central HospitalSichuanChina
| | - Guodong Liu
- Department of NeurosurgeryThe Second Affiliated Hospital, Chongqing Medical UniversityChongqingChina
| | - Quan Cheng
- Department of NeurosurgeryXiangya Hospital, Central South UniversityChangshaChina
- National Clinical Research Center for Geriatric DisordersXiangya Hospital, Central South UniversityChangshaChina
| |
Collapse
|
16
|
Gouda MA, Janku F, Wahida A, Buschhorn L, Schneeweiss A, Abdel Karim N, De Miguel Perez D, Del Re M, Russo A, Curigliano G, Rolfo C, Subbiah V. Liquid Biopsy Response Evaluation Criteria in Solid Tumors (LB-RECIST). Ann Oncol 2024; 35:267-275. [PMID: 38145866 DOI: 10.1016/j.annonc.2023.12.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/17/2023] [Accepted: 12/09/2023] [Indexed: 12/27/2023] Open
Abstract
Current evaluation of treatment response in solid tumors depends on dynamic changes in tumor diameters as measured by imaging. However, these changes can only be detected when there are enough macroscopic changes in tumor volume, which limits the usability of radiological response criteria in evaluating earlier stages of disease response and necessitates much time to lapse for gross changes to be notable. One promising approach is to incorporate dynamic changes in circulating tumor DNA (ctDNA), which occur early in the course of therapy and can predict tumor responses weeks before gross size changes manifest. However, several issues need to be addressed before recommending the implementation of ctDNA response criteria in daily clinical practice such as clinical, biological, and regulatory challenges and, most importantly, the need to standardize/harmonize detection methods and ways to define ctDNA response and/or progression for precision oncology. Herein, we review the use of liquid biopsy (LB) to evaluate response in solid tumors and propose a plan toward standardization of LB-RECIST.
Collapse
Affiliation(s)
- M A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston
| | - F Janku
- Monte Rosa Therapeutics, Boston, USA
| | - A Wahida
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - L Buschhorn
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - A Schneeweiss
- Division of Gynecological Oncology, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - N Abdel Karim
- Inova Schar Cancer Institute, Fairfax, (5)University of Virginia, Charlottesville
| | - D De Miguel Perez
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - M Del Re
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - A Russo
- Medical Oncology Unit, Papardo Civil Hospital and Department of Human Pathology, University of Messina, Messina
| | - G Curigliano
- Department of Oncology and Hemato-Oncology, University of Milano, Milano; Division of Early Drug Development for Innovative Therapies, European Institute of Oncology, IRCCS, Milano, Italy
| | - C Rolfo
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai, New York, USA
| | - V Subbiah
- Sarah Cannon Research Institute, Nashville, USA.
| |
Collapse
|
17
|
Murray JC, Sivapalan L, Hummelink K, Balan A, White JR, Niknafs N, Rhymee L, Pereira G, Rao N, Weksler B, Bahary N, Phallen J, Leal A, Bartlett DL, Marrone KA, Naidoo J, Goel A, Levy B, Rosner S, Hann CL, Scott SC, Feliciano J, Lam VK, Ettinger DS, Li QK, Illei PB, Monkhorst K, Scharpf RB, Brahmer JR, Velculescu VE, Zaidi AH, Forde PM, Anagnostou V. Elucidating the Heterogeneity of Immunotherapy Response and Immune-Related Toxicities by Longitudinal ctDNA and Immune Cell Compartment Tracking in Lung Cancer. Clin Cancer Res 2024; 30:389-403. [PMID: 37939140 PMCID: PMC10792359 DOI: 10.1158/1078-0432.ccr-23-1469] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 09/05/2023] [Accepted: 11/03/2023] [Indexed: 11/10/2023]
Abstract
PURPOSE Although immunotherapy is the mainstay of therapy for advanced non-small cell lung cancer (NSCLC), robust biomarkers of clinical response are lacking. The heterogeneity of clinical responses together with the limited value of radiographic response assessments to timely and accurately predict therapeutic effect-especially in the setting of stable disease-calls for the development of molecularly informed real-time minimally invasive approaches. In addition to capturing tumor regression, liquid biopsies may be informative in capturing immune-related adverse events (irAE). EXPERIMENTAL DESIGN We investigated longitudinal changes in circulating tumor DNA (ctDNA) in patients with metastatic NSCLC who received immunotherapy-based regimens. Using ctDNA targeted error-correction sequencing together with matched sequencing of white blood cells and tumor tissue, we tracked serial changes in cell-free tumor load (cfTL) and determined molecular response. Peripheral T-cell repertoire dynamics were serially assessed and evaluated together with plasma protein expression profiles. RESULTS Molecular response, defined as complete clearance of cfTL, was significantly associated with progression-free (log-rank P = 0.0003) and overall survival (log-rank P = 0.01) and was particularly informative in capturing differential survival outcomes among patients with radiographically stable disease. For patients who developed irAEs, on-treatment peripheral blood T-cell repertoire reshaping, assessed by significant T-cell receptor (TCR) clonotypic expansions and regressions, was identified on average 5 months prior to clinical diagnosis of an irAE. CONCLUSIONS Molecular responses assist with the interpretation of heterogeneous clinical responses, especially for patients with stable disease. Our complementary assessment of the peripheral tumor and immune compartments provides an approach for monitoring of clinical benefits and irAEs during immunotherapy.
Collapse
Affiliation(s)
- Joseph C. Murray
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Lung Cancer Precision Medicine Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lavanya Sivapalan
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Karlijn Hummelink
- Antoni van Leeuwenhoek Nederlands Kanker Instituut, Amsterdam, the Netherlands
| | - Archana Balan
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - James R. White
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Noushin Niknafs
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Lamia Rhymee
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gavin Pereira
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Nisha Rao
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Benny Weksler
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Nathan Bahary
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Jillian Phallen
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alessandro Leal
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David L. Bartlett
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Kristen A. Marrone
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Lung Cancer Precision Medicine Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jarushka Naidoo
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Beaumont RCSI Cancer Centre, Dublin, Ireland
| | - Akul Goel
- California Institute of Technology, 1200 E California Blvd, Pasadena, California
| | - Benjamin Levy
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Samuel Rosner
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christine L. Hann
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Susan C. Scott
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Josephine Feliciano
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Vincent K. Lam
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - David S. Ettinger
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qing Kay Li
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Peter B. Illei
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland
| | - Kim Monkhorst
- Antoni van Leeuwenhoek Nederlands Kanker Instituut, Amsterdam, the Netherlands
| | - Robert B. Scharpf
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Julie R. Brahmer
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Lung Cancer Precision Medicine Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Victor E. Velculescu
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ali H. Zaidi
- Allegheny Health Network Cancer Institute, Allegheny Health Network, Pittsburgh, Pennsylvania
| | - Patrick M. Forde
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, Maryland
- The Lung Cancer Precision Medicine Center of Excellence, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
18
|
Tsai CJ, Yang JT, Shaverdian N, Patel J, Shepherd AF, Eng J, Guttmann D, Yeh R, Gelblum DY, Namakydoust A, Preeshagul I, Modi S, Seidman A, Traina T, Drullinsky P, Flynn J, Zhang Z, Rimner A, Gillespie EF, Gomez DR, Lee NY, Berger M, Robson ME, Reis-Filho JS, Riaz N, Rudin CM, Powell SN. Standard-of-care systemic therapy with or without stereotactic body radiotherapy in patients with oligoprogressive breast cancer or non-small-cell lung cancer (Consolidative Use of Radiotherapy to Block [CURB] oligoprogression): an open-label, randomised, controlled, phase 2 study. Lancet 2024; 403:171-182. [PMID: 38104577 PMCID: PMC10880046 DOI: 10.1016/s0140-6736(23)01857-3] [Citation(s) in RCA: 102] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/27/2023] [Accepted: 08/31/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND Most patients with metastatic cancer eventually develop resistance to systemic therapy, with some having limited disease progression (ie, oligoprogression). We aimed to assess whether stereotactic body radiotherapy (SBRT) targeting oligoprogressive sites could improve patient outcomes. METHODS We did a phase 2, open-label, randomised controlled trial of SBRT in patients with oligoprogressive metastatic breast cancer or non-small-cell lung cancer (NSCLC) after having received at least first-line systemic therapy, with oligoprogression defined as five or less progressive lesions on PET-CT or CT. Patients aged 18 years or older were enrolled from a tertiary cancer centre in New York, NY, USA, and six affiliated regional centres in the states of New York and New Jersey, with a 1:1 randomisation between standard of care (standard-of-care group) and SBRT plus standard of care (SBRT group). Randomisation was done with a computer-based algorithm with stratification by number of progressive sites of metastasis, receptor or driver genetic alteration status, primary site, and type of systemic therapy previously received. Patients and investigators were not masked to treatment allocation. The primary endpoint was progression-free survival, measured up to 12 months. We did a prespecified subgroup analysis of the primary endpoint by disease site. All analyses were done in the intention-to-treat population. The study is registered with ClinicalTrials.gov, NCT03808662, and is complete. FINDINGS From Jan 1, 2019, to July 31, 2021, 106 patients were randomly assigned to standard of care (n=51; 23 patients with breast cancer and 28 patients with NSCLC) or SBRT plus standard of care (n=55; 24 patients with breast cancer and 31 patients with NSCLC). 16 (34%) of 47 patients with breast cancer had triple-negative disease, and 51 (86%) of 59 patients with NSCLC had no actionable driver mutation. The study was closed to accrual before reaching the targeted sample size, after the primary efficacy endpoint was met during a preplanned interim analysis. The median follow-up was 11·6 months for patients in the standard-of-care group and 12·1 months for patients in the SBRT group. The median progression-free survival was 3·2 months (95% CI 2·0-4·5) for patients in the standard-of-care group versus 7·2 months (4·5-10·0) for patients in the SBRT group (hazard ratio [HR] 0·53, 95% CI 0·35-0·81; p=0·0035). The median progression-free survival was higher for patients with NSCLC in the SBRT group than for those with NSCLC in the standard-of-care group (10·0 months [7·2-not reached] vs 2·2 months [95% CI 2·0-4·5]; HR 0·41, 95% CI 0·22-0·75; p=0·0039), but no difference was found for patients with breast cancer (4·4 months [2·5-8·7] vs 4·2 months [1·8-5·5]; 0·78, 0·43-1·43; p=0·43). Grade 2 or worse adverse events occurred in 21 (41%) patients in the standard-of-care group and 34 (62%) patients in the SBRT group. Nine (16%) patients in the SBRT group had grade 2 or worse toxicities related to SBRT, including gastrointestinal reflux disease, pain exacerbation, radiation pneumonitis, brachial plexopathy, and low blood counts. INTERPRETATION The trial showed that progression-free survival was increased in the SBRT plus standard-of-care group compared with standard of care only. Oligoprogression in patients with metastatic NSCLC could be effectively treated with SBRT plus standard of care, leading to more than a four-times increase in progression-free survival compared with standard of care only. By contrast, no benefit was observed in patients with oligoprogressive breast cancer. Further studies to validate these findings and understand the differential benefits are warranted. FUNDING National Cancer Institute.
Collapse
Affiliation(s)
- Chiaojung Jillian Tsai
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| | - Jonathan T Yang
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Narek Shaverdian
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juber Patel
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Annemarie F Shepherd
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juliana Eng
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David Guttmann
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Randy Yeh
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daphna Y Gelblum
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Azadeh Namakydoust
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Isabel Preeshagul
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Shanu Modi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andrew Seidman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Tiffany Traina
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pamela Drullinsky
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhigang Zhang
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Andreas Rimner
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Erin F Gillespie
- Department of Radiation Oncology, University of Washington, Seattle, WA, USA
| | - Daniel R Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nancy Y Lee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mark E Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jorge S Reis-Filho
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Simon N Powell
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
19
|
Liu Y, Altreuter J, Bodapati S, Cristea S, Wong CJ, Wu CJ, Michor F. Predicting patient outcomes after treatment with immune checkpoint blockade: A review of biomarkers derived from diverse data modalities. CELL GENOMICS 2024; 4:100444. [PMID: 38190106 PMCID: PMC10794784 DOI: 10.1016/j.xgen.2023.100444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/12/2023] [Accepted: 10/24/2023] [Indexed: 01/09/2024]
Abstract
Immune checkpoint blockade (ICB) therapy targeting cytotoxic T-lymphocyte-associated protein 4, programmed death 1, and programmed death ligand 1 has shown durable remission and clinical success across different cancer types. However, patient outcomes vary among disease indications. Studies have identified prognostic biomarkers associated with immunotherapy response and patient outcomes derived from diverse data types, including next-generation bulk and single-cell DNA, RNA, T cell and B cell receptor sequencing data, liquid biopsies, and clinical imaging. Owing to inter- and intra-tumor heterogeneity and the immune system's complexity, these biomarkers have diverse efficacy in clinical trials of ICB. Here, we review the genetic and genomic signatures and image features of ICB studies for pan-cancer applications and specific indications. We discuss the advantages and disadvantages of computational approaches for predicting immunotherapy effectiveness and patient outcomes. We also elucidate the challenges of immunotherapy prognostication and the discovery of novel immunotherapy targets.
Collapse
Affiliation(s)
- Yang Liu
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Jennifer Altreuter
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Sudheshna Bodapati
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Simona Cristea
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Cheryl J Wong
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA 20115, USA
| | - Catherine J Wu
- Harvard Medical School, Boston, MA 02115, USA; The Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Franziska Michor
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA 20115, USA; The Eli and Edythe Broad Institute of MIT and Harvard, Cambridge, MA 02139, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA; Center for Cancer Evolution, Dana-Farber Cancer Institute, Boston, MA 02138, USA; The Ludwig Center at Harvard, Boston, MA 02115, USA.
| |
Collapse
|
20
|
Remon J, Lopez A, Planchard D, Besse B. Are we ready to escalate or de-escalate immune treatment strategies in NSCLC based on liquid biopsy data? Eur J Cancer 2023; 195:113369. [PMID: 37913681 DOI: 10.1016/j.ejca.2023.113369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023]
Affiliation(s)
- Jordi Remon
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif, France.
| | - Alvaro Lopez
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - David Planchard
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Benjamin Besse
- Paris-Saclay University, Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
21
|
Tang L, Li R, Wen H, Zhou Q, Xu C. Circulating tumor DNA (ctDNA)-based minimal residual disease in non-small cell lung cancer. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:207-214. [PMID: 39171282 PMCID: PMC11332816 DOI: 10.1016/j.pccm.2023.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Indexed: 08/23/2024]
Abstract
Lung cancer is the second most common cancer worldwide and the leading cause of cancer-related fatalities, with non-small cell lung cancer (NSCLC) accounting for 85% of all lung cancers. Over the past forty years, patients with NSCLC have had a 5-year survival rate of only 16%, despite improvements in chemotherapy, targeted therapy, and immunotherapy. Circulating tumor DNA (ctDNA) in blood can be used to identify minimal residual disease (MRD), and ctDNA-based MRD has been shown to be of significance in prognostic assessment, recurrence monitoring, risk of recurrence assessment, efficacy monitoring, and therapeutic intervention decisions in NSCLC. The level of MRD can be obtained by monitoring ctDNA to provide guidance for more precise and personalized treatment, the scientific feasibility of which could dramatically modify lung cancer treatment paradigm. In this review, we present a comprehensive review of MRD studies in NSCLC and focus on the application of ctDNA-based MRD in different stages of NSCLC in current clinical practice.
Collapse
Affiliation(s)
- Libo Tang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510080, China
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Ruiyang Li
- Youyang Hospital, First Affiliated Hospital of Chongqing Medical University, Chongqing 409800, China
| | - Huahai Wen
- Youyang Hospital, First Affiliated Hospital of Chongqing Medical University, Chongqing 409800, China
| | - Qing Zhou
- Cancer Hospital of Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Chongrui Xu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
22
|
Huang D, Lin G, Chu Q, Hu Y, Wang J, Wang Z, Yang F, Zhong W, Zhou C, Zhu B, Ai X, Cao B, Cao Y, Chen M, Chen X, Chu T, Duan J, Fan Y, Fang Y, Feng S, Feng W, Guo H, Han C, He Y, Hong S, Hu J, Huang M, Huang Y, Jiang D, Jiang K, Jiang R, Jin B, Jin S, Li J, Li M, Li Z, Li C, Lin J, Liu A, Liu SM, Liu Y, Liu Z, Liu Z, Liu Z, Liu Z, Liu Z, Lu Y, Lv T, Ma Z, Miao Q, Peng M, Pu X, Ren XB, Shan J, Shan J, Shen P, Shen B, Shi M, Song Y, Song Z, Su C, Sun J, Tian P, Wang J, Wang F, Wang H, Wang J, Wang Q, Wang W, Wang Y, Wu L, Wu F, Xia Y, Xie C, Xie C, Xin T, Xiong J, Xu H, Xu S, Xu Y, Xu B, Xu C, Yan X, Yang Z, Yao W, Yu Y, Feng Y, Yu Z, Yu Y, Yue D, Zhang H, Zhang H, Zhang L, Zhang L, Zhang Q, Zhang T, Zhang B, Zhao J, Zhao M, Zheng X, et alHuang D, Lin G, Chu Q, Hu Y, Wang J, Wang Z, Yang F, Zhong W, Zhou C, Zhu B, Ai X, Cao B, Cao Y, Chen M, Chen X, Chu T, Duan J, Fan Y, Fang Y, Feng S, Feng W, Guo H, Han C, He Y, Hong S, Hu J, Huang M, Huang Y, Jiang D, Jiang K, Jiang R, Jin B, Jin S, Li J, Li M, Li Z, Li C, Lin J, Liu A, Liu SM, Liu Y, Liu Z, Liu Z, Liu Z, Liu Z, Liu Z, Lu Y, Lv T, Ma Z, Miao Q, Peng M, Pu X, Ren XB, Shan J, Shan J, Shen P, Shen B, Shi M, Song Y, Song Z, Su C, Sun J, Tian P, Wang J, Wang F, Wang H, Wang J, Wang Q, Wang W, Wang Y, Wu L, Wu F, Xia Y, Xie C, Xie C, Xin T, Xiong J, Xu H, Xu S, Xu Y, Xu B, Xu C, Yan X, Yang Z, Yao W, Yu Y, Feng Y, Yu Z, Yu Y, Yue D, Zhang H, Zhang H, Zhang L, Zhang L, Zhang Q, Zhang T, Zhang B, Zhao J, Zhao M, Zheng X, Zhong F, Zhou J, Zhou P, Zhu Z, Zou J, Zou Z. Clinical definition of secondary resistance to immunotherapy in non-small cell lung cancer. Thorac Cancer 2023; 14:3421-3429. [PMID: 37963454 PMCID: PMC10693946 DOI: 10.1111/1759-7714.15157] [Show More Authors] [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: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/16/2023] Open
Abstract
Immune checkpoint inhibitors (PD-1/PD-L1 and CTLA-4 blockade) have revolutionized the treatment landscape in non-small cell lung cancer (NSCLC). Secondary resistance to immunotherapy (IO), which poses a substantial challenge in clinical settings, occurs in several initial responders. Currently, new treatment approaches have been extensively evaluated in investigational studies for these patients to tackle this difficult problem; however, the lack of consistency in clinical definition, uniform criteria for enrollment in clinical trials, and interpretation of results remain significant hurdles to progress. Thus, our expert panel comprehensively synthesized data from current studies to propose a practical clinical definition of secondary resistance to immunotherapy in NSCLC in metastatic and neoadjuvant settings. In addition to patients who received IO alone (including IO-IO combinations), we also generated a definition for patients treated with chemotherapy plus IO. This consensus aimed to provide guidance for clinical trial design and facilitate future discussions with investigators. It should be noted that additional updates in this consensus are required when new data is available.
Collapse
Affiliation(s)
- Dingzhi Huang
- Department of Thoracic OncologyTianjin Medical University Cancer Institute and HospitalTianjinPeople's Republic of China
| | - Gen Lin
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Qian Chu
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Yi Hu
- Senior Department of OncologyChinese PLA General HospitalBeijingPeople's Republic of China
| | - Jun Wang
- Department of OncologyThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJi'nanPeople's Republic of China
| | - Zhijie Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Fan Yang
- Department of Thoracic SurgeryPeking University People HospitalBeijingPeople's Republic of China
| | - Wenzhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouPeople's Republic of China
| | - Chengzhi Zhou
- Pulmonary and Critical Care Medicine, Guangzhou Institute of Respiratory Health, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, State Key Laboratory of Respiratory DiseasesThe First Affiliated Hospital of Guangzhou Medical UniversityGuangzhouPeople's Republic of China
| | - Bo Zhu
- Institute of Cancer, Xinqiao HospitalArmy Medical UniversityChongqingPeople's Republic of China
| | - Xinghao Ai
- Shanghai Lung Cancer Center, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Baoshan Cao
- Cancer centerPeking University Third Hospital/ Department of medical oncology and radiation sickness, Peking University Third HospitalBeijingPeople's Republic of China
| | - Yabing Cao
- Department of oncologyKiang Wu HospitalMacauPeople's Republic of China
| | - Mingqiu Chen
- Department of Thoracic Radiation Oncology, Clinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouPeople's Republic of China
| | - Xiaohui Chen
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouPeople's Republic of China
| | - Tianqing Chu
- Respiratory Department, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Jianchun Duan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Yun Fan
- Department of Medical OncologyZhejiang Cancer HospitalHangzhouPeople's Republic of China
| | - Yong Fang
- Department of Medical Oncology, Sir Run Run Shaw HospitalZhenjiang University School of MedicineHangzhouPeople's Republic of China
| | - Shuitu Feng
- Department of Medical OncologyFudan University Shanghai Cancer Center Xiamen HospitalXiamenPeople's Republic of China
| | - Weineng Feng
- Department of Pulmonary OncologyThe First People's Hospital of FoshanFoshanPeople's Republic of China
| | - Hui Guo
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anPeople's Republic of China
| | - Chengbo Han
- Department of OncologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Yong He
- Department of Respiratory Medicine, Xinqiao HospitalArmy Medical UniversityChongqingPeople's Republic of China
| | - Shaodong Hong
- State Key Laboratory of Oncology in Southern ChinaSun Yat‐sen University Cancer CenterGuangzhouPeople's Republic of China
| | - Jie Hu
- Zhongshan Hospital, Fudan UniversityShanghai Geriatric CenterShanghaiPeople's Republic of China
| | - Meijuan Huang
- Division of Thoracic Tumor Multimodality Treatment and Department of Medical Oncology, Cancer Center, West China HospitalSichuan UniversityChengduPeople's Republic of China
| | - Yan Huang
- State Key Laboratory of Oncology in Southern ChinaSun Yat‐sen University Cancer CenterGuangzhouPeople's Republic of China
| | - Da Jiang
- Department of OncologyThe Fourth Affiliated Hospital of Hebei Medical UniversityShijiazhuangPeople's Republic of China
| | - Kan Jiang
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Richeng Jiang
- Department of Thoracic OncologyTianjin Medical University Cancer Institute and HospitalTianjinPeople's Republic of China
| | - Bo Jin
- Department of Medical OncologyThe First affiliated hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Shi Jin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital &Shenzhen HospitalChinese Academy of Medical Sciences and Perking Union Medical CollegeShenzhenPeople's Republic of China
| | - Jisheng Li
- Department of Medical OncologyQilu Hospital of Shandong UniversityJi'nanPeople's Republic of China
| | - Min Li
- Department of Respiratory Medicine, Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Ziming Li
- Shanghai Lung Cancer Center, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Chao Li
- Department of PathologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Jie Lin
- Department of Medical OncologyThe Second Affiliated Hospital of Kunming Medical UniversityKunmingPeople's Republic of China
| | - Anwen Liu
- Department of Medical OncologyThe Second Affiliated Hospital of Nanchang UniversityNanchangPeople's Republic of China
| | - Si‐Yang Maggie Liu
- Department of Hematology, First Affiliated HospitalJi'nan UniversityGuangzhouPeople's Republic of China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Zhefeng Liu
- Senior Department of OncologyChinese PLA General HospitalBeijingPeople's Republic of China
| | - Zhe Liu
- Department of Medical Oncology, Beijing Chest HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Zhenhua Liu
- Department of Oncology, Shengli Clinical Medical College of Fujian Medical UniversityFujian Provincial HospitalFuzhouPeople's Republic of China
| | - Zhentian Liu
- Department of Thoracic Oncology,Jiangxi Cancer HospitalNanchangPeople's Republic of China
| | - Zhigang Liu
- Cancer CenterThe 10th Affiliated Hospital of Southern Medical UniversityDongguanPeople's Republic of China
| | - Yuping Lu
- Department of Abdominal OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingPeople's Republic of China
| | - Zhiyong Ma
- Department of Respiratory MedicineHenan Cancer Hospital /Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouPeople's Republic of China
| | - Qian Miao
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Min Peng
- Cancer centerRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
| | - Xingxiang Pu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaPeople's Republic of China
| | - Xiu Bao Ren
- Department of BiotherapyTianjin Medical University Cancer Institute and HospitalTianjinPeople's Republic of China
| | - Jianzhen Shan
- Department of Medical OncologyThe First Affiliated Hospital of Zhejiang UniversityZhejiangPeople's Republic of China
| | - Jinlu Shan
- Department of Medical Oncology, Daping HospitalArmy Medical UniversityChongqingPeople's Republic of China
| | - Peng Shen
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouPeople's Republic of China
| | - Bo Shen
- Department of Medical OncologyJiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingPeople's Republic of China
| | - Meiqi Shi
- Department of Medical OncologyJiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Affiliated Cancer Hospital of Nanjing Medical UniversityNanjingPeople's Republic of China
| | - Yong Song
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingPeople's Republic of China
| | - Zhengbo Song
- Department of Clinical TrialZhejiang Cancer HospitalHangzhouPeople's Republic of China
| | - ChunXia Su
- Department of Oncology, Shanghai Pulmonary Hospital & Thoracic Cancer InstituteTongji University School of MedicineShanghaiPeople's Republic of China
| | - Jianguo Sun
- Institute of Cancer, Xinqiao HospitalArmy Medical UniversityChongqingPeople's Republic of China
| | - Panwen Tian
- Department of Pulmonary and Critical Care Medicine, Lung Cancer Center, West China HospitalSichuan University, Precision Medicine Key Laboratory of Sichuan ProvinceChengduPeople's Republic of China
| | - Jinliang Wang
- Senior Department of OncologyChinese PLA General HospitalBeijingPeople's Republic of China
| | - Feng Wang
- Department of Thoracic Surgery, Clinical Oncology School of Fujian Medical UniversityFujian Cancer HospitalFuzhouPeople's Republic of China
| | - Huijuan Wang
- Department of Respiratory MedicineHenan Cancer Hospital /Affiliated Cancer Hospital of Zhengzhou UniversityZhengzhouPeople's Republic of China
| | - Jialei Wang
- Department of Thoracic Medical OncologyFudan University Shanghai Cancer CenterShanghaiPeople's Republic of China
| | - Qian Wang
- Department of Respiratory Medicine, Affiliated Hospital of Nanjing University of Chinese MedicineJiangsu Province Hospital of Chinese MedicineNanjingPeople's Republic of China
| | - Wenxian Wang
- Department of Medical OncologyZhejiang Cancer HospitalHangzhouPeople's Republic of China
| | - Yan Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingPeople's Republic of China
| | - Lin Wu
- Department of Thoracic Medical Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaPeople's Republic of China
| | - Fang Wu
- Department of Oncology, The Second Xiangya HospitalCentral South UniversityChangshaPeople's Republic of China
| | - Yang Xia
- Department of Respiratory and Critical Care MedicineSecond Affiliated Hospital of Zhejiang University School of MedicineHangzhouPeople's Republic of China
| | - Congying Xie
- Department of Radiation and Medical OncologySecond Affiliated Hospital of Wenzhou Medical UniversityWenzhouPeople's Republic of China
| | - Conghua Xie
- Department of Pulmonary OncologyZhongnan Hospital of Wuhan UniversityWuhanPeople's Republic of China
| | - Tao Xin
- Department of OncologyThe Second Affiliated Hospital of Harbin Medical UniversityHarbinPeople's Republic of China
| | - Jianping Xiong
- Department of OncologyThe First Affiliated Hospital of Nanchang UniversityNanchangPeople's Republic of China
| | - Haipeng Xu
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Song Xu
- Department of Lung Cancer SurgeryTianjin Medical University General HospitalTianjinPeople's Republic of China
| | - Yiquan Xu
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Bin Xu
- Cancer centerRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
| | - Chunwei Xu
- Department of Respiratory Medicine, Affiliated Jinling HospitalMedical School of Nanjing UniversityNanjingPeople's Republic of China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu HospitalAir Force Medical UniversityXi'anPeople's Republic of China
| | - Zhenzhou Yang
- Department of Cancer CenterThe Second Affiliated Hospital of Chongqing Medical UniversityChongqingPeople's Republic of China
| | - Wenxiu Yao
- Department of Medical Oncology, Sichuan Cancer HospitalUniversity of Electronic Science and Technology of ChinaChengduPeople's Republic of China
| | - Yao Yu
- Department of Medical OncologyThe First Affiliated Hospital of Xi'an Jiaotong UniversityXi'anPeople's Republic of China
| | - Ye Feng
- Department of Medical Oncology, Xiamen Key Laboratory of Antitumor Drug Transformation ResearchThe First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen UniversityXiamenPeople's Republic of China
| | - Zongyang Yu
- Department of Respiratory Medicine, The 900th Hospital of the Joint Logistic Support ForcePeople's Liberation Army of ChinaFuzhouPeople's Republic of China
| | - Yongfeng Yu
- Shanghai Lung Cancer Center, Shanghai Chest HospitalShanghai Jiao Tong University School of MedicineShanghaiPeople's Republic of China
| | - Dongsheng Yue
- Department of Lung CancerTianjin Medical University Cancer Institute and HospitalTianjinPeople's Republic of China
| | - Haibo Zhang
- Department of OncologyGuangdong Provincial Hospital of Chinese MedicineGuangzhouPeople's Republic of China
| | - HongMei Zhang
- Department of Clinical Oncology, Xijing HospitalAir Force Medical UniversityXi'anPeople's Republic of China
| | - Li Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Longfeng Zhang
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Qiuyu Zhang
- Institute of ImmunotherapyFujian Medical UniversityFuzhouPeople's Republic of China
| | - Tongmei Zhang
- Department of Medical Oncology, Beijing Chest HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Bicheng Zhang
- Cancer centerRenmin Hospital of Wuhan UniversityWuhanPeople's Republic of China
| | - Jun Zhao
- Key Laboratory of Carcinogenesis and Translational Research(Ministry of Education/Beijing), Department I of Thoracic OncologyPeking University Cancer Hospital and InstituteBeijingPeople's Republic of China
| | - Mingfang Zhao
- Department of Medical OncologyThe First affiliated hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Xiaobin Zheng
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Fengqiao Zhong
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| | - Jin Zhou
- Department of Medical Oncology, Sichuan Cancer HospitalUniversity of Electronic Science and Technology of ChinaChengduPeople's Republic of China
| | - Penghui Zhou
- State Key Laboratory of Oncology in Southern ChinaSun Yat‐sen University Cancer CenterGuangzhouPeople's Republic of China
| | - Zhengfei Zhu
- Department of Radiation OncologyFudan University Shanghai Cancer CenterShanghaiPeople's Republic of China
| | - Juntao Zou
- Department of Respiratory MedicineThe First Affiliated Hospital of Nanchang UniversityNanchangPeople's Republic of China
| | - Zihua Zou
- Department of Thoracic OncologyClinical Oncology School of Fujian Medical University, Fujian Cancer HospitalFuzhouPeople's Republic of China
| |
Collapse
|
23
|
Azad TD, Ran KR, Liu J, Vattipally VN, Khela H, Leite E, Materi JD, Davidar AD, Bettegowda C, Theodore N. A future blood test for acute traumatic spinal cord injury. Biomarkers 2023; 28:703-713. [PMID: 38126897 DOI: 10.1080/1354750x.2023.2298650] [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: 10/21/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Acute spinal cord injury (SCI) requires prompt diagnosis and intervention to minimize the risk of permanent neurologic deficit. Presently, SCI diagnosis and interventional planning rely on magnetic resonance imaging (MRI), which is not always available or feasible for severely injured patients. Detection of disease-specific biomarkers in biofluids via liquid biopsy may provide a more accessible and objective means of evaluating patients with suspected SCI. Cell-free DNA, which has been used for diagnosing and monitoring oncologic disease, may detect damage to spinal cord neurons via tissue-specific methylation patterns. Other types of biomarkers, including proteins and RNA species, have also been found to reflect neuronal injury and may be included as part of a multi-analyte assay to improve liquid biopsy performance. The feasibility of implementing liquid biopsy into current practices of SCI management is supported by the relative ease of blood sample collection as well as recent advancements in droplet digital polymerase chain reaction technology. In this review, we detail the current landscape of biofluid biomarkers for acute SCI and propose a framework for the incorporation of a putative blood test into the clinical management of SCI.
Collapse
Affiliation(s)
- Tej D Azad
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Kathleen R Ran
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Jiaqi Liu
- Georgetown University School of Medicine, Washington, DC, USA
| | | | - Harmon Khela
- Department of Neurosurgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Enzo Leite
- Faculdade Pernambucana de Saúde (FPS), Recife, PE, Brazil
| | - Joshua D Materi
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - A Daniel Davidar
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Nicholas Theodore
- Department of Neurosurgery, Johns Hopkins Hospital, Baltimore, MD, USA
| |
Collapse
|
24
|
Zhao J, Reuther J, Scozzaro K, Hawley M, Metzger E, Emery M, Chen I, Barbosa M, Johnson L, O'Connor A, Washburn M, Hartje L, Reckase E, Johnson V, Zhang Y, Westheimer E, O'Callaghan W, Malani N, Chesh A, Moreau M, Daber R. Personalized Cancer Monitoring Assay for the Detection of ctDNA in Patients with Solid Tumors. Mol Diagn Ther 2023; 27:753-768. [PMID: 37632661 PMCID: PMC10590345 DOI: 10.1007/s40291-023-00670-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2023] [Indexed: 08/28/2023]
Abstract
BACKGROUND Highly sensitive molecular assays have been developed to detect plasma-based circulating tumor DNA (ctDNA), and emerging evidence suggests their clinical utility for monitoring minimal residual disease and recurrent disease, providing prognostic information, and monitoring therapy responses in patients with solid tumors. The Invitae Personalized Cancer Monitoring™ assay uses a patient-specific, tumor-informed variant signature identified through whole exome sequencing to detect ctDNA in peripheral blood of patients with solid tumors. METHODS The assay's tumor whole exome sequencing and ctDNA detection components were analytically validated using 250 unique human specimens and nine commercial reference samples that generated 1349 whole exome sequencing and cell-free DNA (cfDNA)-derived libraries. A comparison of tumor and germline whole exome sequencing was used to identify patient-specific tumor variant signatures and generate patient-specific panels, followed by targeted next-generation sequencing of plasma-derived cfDNA using the patient-specific panels with anchored multiplex polymerase chain reaction chemistry leveraging unique molecular identifiers. RESULTS Whole exome sequencing resulted in overall sensitivity of 99.8% and specificity of > 99.9%. Patient-specific panels were successfully designed for all 63 samples (100%) with ≥ 20% tumor content and 24 (80%) of 30 samples with ≥ 10% tumor content. Limit of blank studies using 30 histologically normal, formalin-fixed paraffin-embedded specimens resulted in 100% expected panel design failure. The ctDNA detection component demonstrated specificity of > 99.9% and sensitivity of 96.3% for a combination of 10 ng of cfDNA input, 0.008% allele frequency, 50 variants on the patient-specific panels, and a baseline threshold. Limit of detection ranged from 0.008% allele frequency when utilizing 60 ng of cfDNA input with 18-50 variants in the patient-specific panels (> 99.9% sensitivity) with a baseline threshold, to 0.05% allele frequency when using 10 ng of cfDNA input with an 18-variant panel with a monitoring threshold (> 99.9% sensitivity). CONCLUSIONS The Invitae Personalized Cancer Monitoring assay, featuring a flexible patient-specific panel design with 18-50 variants, demonstrated high sensitivity and specificity for detecting ctDNA at variant allele frequencies as low as 0.008%. This assay may support patient prognostic stratification, provide real-time data on therapy responses, and enable early detection of residual/recurrent disease.
Collapse
Affiliation(s)
- Jianhua Zhao
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA.
| | | | - Kaylee Scozzaro
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Megan Hawley
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Emily Metzger
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Matthew Emery
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Ingrid Chen
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | | | - Laura Johnson
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Alijah O'Connor
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Mike Washburn
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Luke Hartje
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Erik Reckase
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Verity Johnson
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
- Affiliated with Invitae Corp. at the time of the study, currently employees at Integrated DNA Technologies, 1710 Commercial Park, Coralville, IA, 52241, USA
| | - Yuhua Zhang
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | | | | | - Nirav Malani
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Adrian Chesh
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Michael Moreau
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| | - Robert Daber
- Invitae Corp., 1400 16th Street, San Francisco, CA, 94103, USA
| |
Collapse
|
25
|
Anagnostou V, Ho C, Nicholas G, Juergens RA, Sacher A, Fung AS, Wheatley-Price P, Laurie SA, Levy B, Brahmer JR, Balan A, Niknafs N, Avrutin E, Zhu L, Sausen M, Bradbury PA, O'Donnell-Tormey J, Gaudreau PO, Ding K, Dancey J. ctDNA response after pembrolizumab in non-small cell lung cancer: phase 2 adaptive trial results. Nat Med 2023; 29:2559-2569. [PMID: 37814061 PMCID: PMC10579094 DOI: 10.1038/s41591-023-02598-9] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 09/19/2023] [Indexed: 10/11/2023]
Abstract
Circulating tumor DNA (ctDNA) has shown promise in capturing primary resistance to immunotherapy. BR.36 is a multi-center, randomized, ctDNA-directed, phase 2 trial of molecular response-adaptive immuno-chemotherapy for patients with lung cancer. In the first of two independent stages, 50 patients with advanced non-small cell lung cancer received pembrolizumab as standard of care. The primary objectives of stage 1 were to ascertain ctDNA response and determine optimal timing and concordance with radiologic Response Evaluation Criteria in Solid Tumors (RECIST) response. Secondary endpoints included the evaluation of time to ctDNA response and correlation with progression-free and overall survival. Maximal mutant allele fraction clearance at the third cycle of pembrolizumab signified molecular response (mR). The trial met its primary endpoint, with a sensitivity of ctDNA response for RECIST response of 82% (90% confidence interval (CI): 52-97%) and a specificity of 75% (90% CI: 56.5-88.5%). Median time to ctDNA response was 2.1 months (90% CI: 1.5-2.6), and patients with mR attained longer progression-free survival (5.03 months versus 2.6 months) and overall survival (not reached versus 7.23 months). These findings are incorporated into the ctDNA-driven interventional molecular response-adaptive second stage of the BR.36 trial in which patients at risk of progression are randomized to treatment intensification or continuation of therapy. ClinicalTrials.gov ID: NCT04093167 .
Collapse
Affiliation(s)
- Valsamo Anagnostou
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Cheryl Ho
- BCCA-Vancouver Cancer Centre, Vancouver, BC, Canada
| | | | | | - Adrian Sacher
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Andrea S Fung
- Kingston Health Sciences Centre, Kingston, ON, Canada
| | | | | | - Benjamin Levy
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Julie R Brahmer
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Archana Balan
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Noushin Niknafs
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Egor Avrutin
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Liting Zhu
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Mark Sausen
- Personal Genome Diagnostics (LabCorp), Baltimore, MD, USA
| | - Penelope A Bradbury
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | | | | | - Keyue Ding
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Janet Dancey
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada.
| |
Collapse
|
26
|
Lin X, Zong C, Zhang Z, Fang W, Xu P. Progresses in biomarkers for cancer immunotherapy. MedComm (Beijing) 2023; 4:e387. [PMID: 37799808 PMCID: PMC10547938 DOI: 10.1002/mco2.387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 09/02/2023] [Accepted: 09/08/2023] [Indexed: 10/07/2023] Open
Abstract
Currently, checkpoint inhibitor-based immunotherapy has emerged as prevailing treatment modality for diverse cancers. However, immunotherapy as a first-line therapy has not consistently yielded durable responses. Moreover, the risk of immune-related adverse events increases with combination regimens. Thus, the development of predictive biomarkers is needed to optimize individuals benefit, minimize risk of toxicities, and guide combination approaches. The greatest focus has been on tumor programmed cell death-ligand 1 (PD-L1), microsatellite instability (MSI), and tumor mutational burden (TMB). However, there remains a subject of debate due to thresholds variability and significant heterogeneity. Major unmet challenges in immunotherapy are the discovery and validation of predictive biomarkers. Here, we show the status of tumor PD-L1, MSI, TMB, and emerging data on novel biomarker strategies with oncogenic signaling and epigenetic regulation. Considering the exploration of peripheral and intestinal immunity has served as noninvasive alternative in predicting immunotherapy, this review also summarizes current data in systemic immunity, encompassing solute PD-L1 and TMB, circulating tumor DNA and infiltrating lymphocytes, routine emerging inflammatory markers and cytokines, as well as gut microbiota. This review provides up-to-date information on the evolving field of currently available biomarkers in predicting immunotherapy. Future exploration of novel biomarkers is warranted.
Collapse
Affiliation(s)
- Xuwen Lin
- Department of Pulmonary and Critical Care MedicinePeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
- Department of Internal MedicineShantou University Medical CollegeShantouGuangdong ProvinceChina
| | - Chenyu Zong
- Department of Pulmonary and Critical Care MedicinePeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
- Department of Internal MedicineZunyi Medical UniversityZunyiGuizhou ProvinceChina
| | - Zhihan Zhang
- Department of Pulmonary and Critical Care MedicinePeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
| | - Weiyi Fang
- Cancer Research InstituteSchool of Basic Medical ScienceSouthern Medical UniversityGuangzhouGuangdong ProvinceChina
- Cancer CenterIntegrated Hospital of Traditional Chinese MedicineSouthern Medical UniversityGuangzhouGuangdong ProvinceChina
| | - Ping Xu
- Department of Pulmonary and Critical Care MedicinePeking University Shenzhen HospitalShenzhenGuangdong ProvinceChina
- Department of Internal MedicineZunyi Medical UniversityZunyiGuizhou ProvinceChina
| |
Collapse
|
27
|
Earland N, Zhang W, Usmani A, Nene A, Bacchiocchi A, Chen DY, Sznol M, Halaban R, Chaudhuri AA, Newman AM. CD4 T cells and toxicity from immune checkpoint blockade. Immunol Rev 2023; 318:96-109. [PMID: 37491734 PMCID: PMC10838135 DOI: 10.1111/imr.13248] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/27/2023] [Indexed: 07/27/2023]
Abstract
Immune-related toxicities, otherwise known as immune-related adverse events (irAEs), occur in a substantial fraction of cancer patients treated with immune checkpoint inhibitors (ICIs). Ranging from asymptomatic to life-threatening, ICI-induced irAEs can result in hospital admission, high-dose corticosteroid treatment, ICI discontinuation, and in some cases, death. A deeper understanding of the factors underpinning severe irAE development will be essential for improved irAE prediction and prevention, toward maximizing the benefits and safety profiles of ICIs. In recent work, we applied mass cytometry, single-cell RNA sequencing, single-cell V(D)J sequencing, bulk RNA sequencing, and bulk T-cell receptor (TCR) sequencing to identify pretreatment determinants of severe irAE development in patients with advanced melanoma. Across 71 patients separated into three cohorts, we found that two baseline features in circulation-elevated activated CD4 effector memory T-cell abundance and TCR diversity-are associated with severe irAE development, independent of the affected organ system within 3 months of ICI treatment initiation. Here, we provide an extended perspective on this work, synthesize and discuss related literature, and summarize practical considerations for clinical translation. Collectively, these findings lay a foundation for data-driven and mechanistic insights into irAE development, with the potential to reduce ICI morbidity and mortality in the future.
Collapse
Affiliation(s)
- Noah Earland
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Wubing Zhang
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Abul Usmani
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Antonella Bacchiocchi
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - David Y. Chen
- Division of Dermatology, Washington University School of Medicine, St. Louis, MO, USA
- Siteman Cancer Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Mario Sznol
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Medicine, Division of Medical Oncology, Yale University School of Medicine, New Haven, CT, USA
| | - Ruth Halaban
- Yale Cancer Center, Yale University School of Medicine, New Haven, Connecticut, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Aadel A. Chaudhuri
- Division of Cancer Biology, Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Division of Biology and Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Computer Science and Engineering, Washington University in St. Louis, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Aaron M. Newman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| |
Collapse
|
28
|
Xia J, Zhang J, Xiong Y, Zhao J, Zhou Y, Jiang T, Zhu J. Circulating tumor DNA minimal residual disease in clinical practice of non-small cell lung cancer. Expert Rev Mol Diagn 2023; 23:913-924. [PMID: 37702546 DOI: 10.1080/14737159.2023.2252334] [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: 05/03/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023]
Abstract
INTRODUCTION The advance of diagnostics and treatments has greatly improved the prognosis of non-small cell lung cancer (NSCLC) patients. However, relapse and metastasis are still common problems encountered by NSCLC patients who have achieved complete remission. Therefore, overcoming the challenge of relapse and metastasis is particularly important for improving the prognosis of NSCLC patients. Research has shown that minimal residual disease (MRD) was a potential source of tumor relapse and metastasis, and circulating tumor DNA (ctDNA) MRD has obvious advantages in predicting the relapse and metastasis of NSCLC and evaluating treatment effectiveness. Therefore, dynamic monitoring of MRD is of great significance for NSCLC patient management strategies. AREAS COVERED We have reviewed articles related to NSCLC MRD included in PubMed and describes the biological significance and historical context of MRD research, reasons for using ctDNA to evaluate MRD, and potential value and challenges of ctDNA MRD in assessing relapse and metastasis of NSCLC, ultimately guiding clinical therapeutic strategies and management. EXPERT OPINION The standardized scope of ctDNA MRD detection for NSCLC requires more clinical research evidence to minimize study differences, making it possible to include in the clinical staging as a reliable indicator.
Collapse
Affiliation(s)
- Jinghua Xia
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jiao Zhang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yanlu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jinbo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yinxi Zhou
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jianfei Zhu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
- Department of Thoracic Surgery, Shaanxi Provincial People's Hospital, Xi'an, China
| |
Collapse
|
29
|
Bertoli E, De Carlo E, Basile D, Zara D, Stanzione B, Schiappacassi M, Del Conte A, Spina M, Bearz A. Liquid Biopsy in NSCLC: An Investigation with Multiple Clinical Implications. Int J Mol Sci 2023; 24:10803. [PMID: 37445976 DOI: 10.3390/ijms241310803] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/25/2023] [Accepted: 06/26/2023] [Indexed: 07/15/2023] Open
Abstract
Tissue biopsy is essential for NSCLC diagnosis and treatment management. Over the past decades, liquid biopsy has proven to be a powerful tool in clinical oncology, isolating tumor-derived entities from the blood. Liquid biopsy permits several advantages over tissue biopsy: it is non-invasive, and it should provide a better view of tumor heterogeneity, gene alterations, and clonal evolution. Consequentially, liquid biopsy has gained attention as a cancer biomarker tool, with growing clinical applications in NSCLC. In the era of precision medicine based on molecular typing, non-invasive genotyping methods became increasingly important due to the great number of oncogene drivers and the small tissue specimen often available. In our work, we comprehensively reviewed established and emerging applications of liquid biopsy in NSCLC. We made an excursus on laboratory analysis methods and the applications of liquid biopsy either in early or metastatic NSCLC disease settings. We deeply reviewed current data and future perspectives regarding screening, minimal residual disease, micrometastasis detection, and their implication in adjuvant and neoadjuvant therapy management. Moreover, we reviewed liquid biopsy diagnostic utility in the absence of tissue biopsy and its role in monitoring treatment response and emerging resistance in metastatic NSCLC treated with target therapy and immuno-therapy.
Collapse
Affiliation(s)
- Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Debora Basile
- Department of Medical Oncology, San Giovanni Di Dio Hospital, 88900 Crotone, Italy
| | - Diego Zara
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Monica Schiappacassi
- Molecular Oncology Unit, (OMMPPT) Department of Translational Research, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
30
|
Ancel J, Dormoy V, Raby BN, Dalstein V, Durlach A, Dewolf M, Gilles C, Polette M, Deslée G. Soluble biomarkers to predict clinical outcomes in non-small cell lung cancer treated by immune checkpoints inhibitors. Front Immunol 2023; 14:1171649. [PMID: 37283751 PMCID: PMC10239865 DOI: 10.3389/fimmu.2023.1171649] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 05/11/2023] [Indexed: 06/08/2023] Open
Abstract
Lung cancer remains the first cause of cancer-related death despite many therapeutic innovations, including immune checkpoint inhibitors (ICI). ICI are now well used in daily practice at late metastatic stages and locally advanced stages after a chemo-radiation. ICI are also emerging in the peri-operative context. However, all patients do not benefit from ICI and even suffer from additional immune side effects. A current challenge remains to identify patients eligible for ICI and benefiting from these drugs. Currently, the prediction of ICI response is only supported by Programmed death-ligand 1 (PD-L1) tumor expression with perfectible results and limitations inherent to tumor-biopsy specimen analysis. Here, we reviewed alternative markers based on liquid biopsy and focused on the most promising biomarkers to modify clinical practice, including non-tumoral blood cell count such as absolute neutrophil counts, platelet to lymphocyte ratio, neutrophil to lymphocyte ratio, and derived neutrophil to lymphocyte ratio. We also discussed soluble-derived immune checkpoint-related products such as sPD-L1, circulating tumor cells (detection, count, and marker expression), and circulating tumor DNA-related products. Finally, we explored perspectives for liquid biopsies in the immune landscape and discussed how they could be implemented into lung cancer management with a potential biological-driven decision.
Collapse
Affiliation(s)
- Julien Ancel
- Inserm UMR-S1250, P3Cell, University of Reims Champagne-Ardenne, SFR CAP-SANTE, Reims, France
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, Reims, France
| | - Valérian Dormoy
- Inserm UMR-S1250, P3Cell, University of Reims Champagne-Ardenne, SFR CAP-SANTE, Reims, France
| | - Béatrice Nawrocki Raby
- Inserm UMR-S1250, P3Cell, University of Reims Champagne-Ardenne, SFR CAP-SANTE, Reims, France
| | - Véronique Dalstein
- Inserm UMR-S1250, P3Cell, University of Reims Champagne-Ardenne, SFR CAP-SANTE, Reims, France
- Department of Biopathology, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, Reims, France
| | - Anne Durlach
- Inserm UMR-S1250, P3Cell, University of Reims Champagne-Ardenne, SFR CAP-SANTE, Reims, France
- Department of Biopathology, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, Reims, France
| | - Maxime Dewolf
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, Reims, France
| | - Christine Gilles
- Laboratory of Tumor and Development Biology, GIGA-Cancer, University of Liège, Liège, Belgium
| | - Myriam Polette
- Inserm UMR-S1250, P3Cell, University of Reims Champagne-Ardenne, SFR CAP-SANTE, Reims, France
- Department of Biopathology, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, Reims, France
| | - Gaëtan Deslée
- Inserm UMR-S1250, P3Cell, University of Reims Champagne-Ardenne, SFR CAP-SANTE, Reims, France
- Department of Respiratory Diseases, Centre Hospitalier Universitaire de Reims, Hôpital Maison Blanche, Reims, France
| |
Collapse
|
31
|
Carbonell C, Frigola J, Pardo N, Callejo A, Iranzo P, Valdivia A, Priano I, Cedrés S, Martinez-Marti A, Navarro A, Lenza L, Soleda M, Gonzalo-Ruiz J, Vivancos A, Sansó M, Carcereny E, Morán T, Amat R, Felip E. Dynamic changes in circulating tumor DNA assessed by shallow whole-genome sequencing associate with clinical efficacy of checkpoint inhibitors in NSCLC. Mol Oncol 2023; 17:779-791. [PMID: 36852704 PMCID: PMC10158763 DOI: 10.1002/1878-0261.13409] [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: 12/16/2022] [Revised: 01/17/2023] [Accepted: 02/27/2023] [Indexed: 03/01/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 axis are the main therapeutic option for patients with advanced non-small cell lung cancer (NSCLC) without a druggable oncogenic alteration. Nevertheless, only a portion of patients benefit from this type of treatment. Here, we assessed the value of shallow whole-genome sequencing (sWGS) on plasma samples to monitor ICI benefit. We applied sWGS on cell-free DNA (cfDNA) extracted from plasma samples of 45 patients with metastatic NSCLC treated with ICIs. Over 150 samples were obtained before ICI treatment initiation and at several time points throughout treatment. From sWGS data, we computed the tumor fraction (TFx) and somatic copy number alteration (SCNA) burden and associated them with ICI benefit and clinical features. TFx at baseline correlated with metastatic lesions at the bone and the liver, and high TFx (≥ 10%) associated with ICI benefit. Moreover, its assessment in on-treatment samples was able to better predict clinical efficacy, regardless of the TFx levels at baseline. Finally, for a subset of patients for whom SCNA burden could be computed, increased burden correlated with diminished benefit following ICI treatment. Thus, our data indicate that the analysis of cfDNA by sWGS enables the monitoring of two potential biomarkers-TFx and SCNA burden-of ICI benefit in a cost-effective manner, facilitating multiple serial-sample analyses. Larger cohorts will be needed to establish its clinical potential.
Collapse
Affiliation(s)
- Caterina Carbonell
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
| | - Joan Frigola
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
| | - Nuria Pardo
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| | - Ana Callejo
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| | - Patricia Iranzo
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| | - Augusto Valdivia
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| | - Ilaria Priano
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| | - Susana Cedrés
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| | - Alex Martinez-Marti
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| | - Alejandro Navarro
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| | - Laura Lenza
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
| | - Mireia Soleda
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
| | - Javier Gonzalo-Ruiz
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Laboratory, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
| | - Miriam Sansó
- Balearic Islands Health Research Institute (IdISBa), Palma de Mallorca, Spain
| | - Enric Carcereny
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Badalona Applied Research Group in Oncology, Institut Germans Trias i Pujol, Barcelona, Spain
| | - Teresa Morán
- Medical Oncology Department, Catalan Institute of Oncology Badalona, Hospital Universitari Germans Trias i Pujol, Badalona Applied Research Group in Oncology, Institut Germans Trias i Pujol, Barcelona, Spain
- Department of Medicine, Universitat Autònoma de Barcelona, Spain
| | - Ramon Amat
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
| | - Enriqueta Felip
- Thoracic Cancers Translational Genomics Unit, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Clinical Research Department, Vall d'Hebron Institut d'Oncologia (VHIO), Barcelona, Spain
- Oncology Department, Vall d'Hebron Barcelona Hospital Campus, Spain
| |
Collapse
|
32
|
Al-Showbaki L, Wilson B, Tamimi F, Molto C, Mittal A, Cescon DW, Amir E. Changes in circulating tumor DNA and outcomes in solid tumors treated with immune checkpoint inhibitors: a systematic review. J Immunother Cancer 2023; 11:jitc-2022-005854. [PMID: 36792122 PMCID: PMC9933752 DOI: 10.1136/jitc-2022-005854] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2023] [Indexed: 02/17/2023] Open
Abstract
BACKGROUND Quantification of circulating tumor DNA (ctDNA) levels is a reliable prognostic tool in several malignancies. Dynamic changes in ctDNA levels in response to treatment may also provide prognostic information. Here, we explore the value of changes in ctDNA levels in response to immune checkpoint inhibitors (ICIs). METHODS We searched MEDLINE (host: PubMed) for trials of ICIs in advanced solid tumors in which outcomes were reported based on change in ctDNA levels. ctDNA reduction was defined as reported in individual trials. Typically, this was either >50% reduction or a reduction to undetectable levels. We extracted HRs and related 95% CIs and/or p values comparing ctDNA reduction versus no reduction for progression-free survival (PFS) and/or overall survival (OS). Data were then pooled in a meta-analysis. Variation in effect size was examined using subgroup analyses. RESULTS Eighteen trials were included in the meta-analysis. ctDNA levels were detectable in all participants in all studies prior to initiation of ICIs. A reduction in ctDNA measured 6-16 weeks after starting treatment was associated with significantly better PFS (HR 0.20; 95% CI, 0.14 to 0.28; p<0.001). Similarly, OS was superior in patients with reduced ctDNA levels (HR 0.18; 95% CI, 0.12 to 0.26; p<0.001). The results were consistent across all disease sites, lines of treatment, magnitude of change (to undetectable vs >50% reduction) and whether treatment exposure comprised single or combination ICIs. CONCLUSIONS In advanced solid tumors, a reduction in ctDNA levels in response to ICIs is associated with substantial improvements in outcome. ctDNA change is an early response biomarker which may allow for de-escalation of cross-sectional imaging in patients receiving ICIs or support treatment de-escalation strategies.
Collapse
Affiliation(s)
- Laith Al-Showbaki
- Division of Hematology and Medical Oncology, Department of Medicine, The University of Jordan, Amman, Jordan
| | - Brooke Wilson
- Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Faris Tamimi
- Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Consolacion Molto
- Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Abhenil Mittal
- Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - David W Cescon
- Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Princess Margaret Hospital Cancer Centre, Toronto, Ontario, Canada
| |
Collapse
|
33
|
Li YZ, Kong SN, Liu YP, Yang Y, Zhang HM. Can Liquid Biopsy Based on ctDNA/cfDNA Replace Tissue Biopsy for the Precision Treatment of EGFR-Mutated NSCLC? J Clin Med 2023; 12:jcm12041438. [PMID: 36835972 PMCID: PMC9966257 DOI: 10.3390/jcm12041438] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/16/2023] [Accepted: 02/06/2023] [Indexed: 02/15/2023] Open
Abstract
More and more clinical trials have explored the role of liquid biopsy in the diagnosis and treatment of EGFR-mutated NSCLC. In certain circumstances, liquid biopsy has unique advantages and offers a new way to detect therapeutic targets, analyze drug resistance mechanisms in advanced patients, and monitor MRD in patients with operable NSCLC. Although its potential cannot be ignored, more evidence is needed to support the transition from the research stage to clinical application. We reviewed the latest progress in research on the efficacy and resistance mechanisms of targeted therapy for advanced NSCLC patients with plasma ctDNA EGFR mutation and the evaluation of MRD based on ctDNA detection in perioperative and follow-up monitoring.
Collapse
|
34
|
Sinoquet L, Jacot W, Quantin X, Alix-Panabières C. Liquid Biopsy and Immuno-Oncology for Advanced Nonsmall Cell Lung Cancer. Clin Chem 2023; 69:23-40. [PMID: 36322450 DOI: 10.1093/clinchem/hvac166] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/23/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND In the last decade, immune checkpoint inhibitors have revolutionized the treatment of metastatic nonsmall cell lung cancer without oncogenic addiction. Currently, programmed death ligand 1 (PD-L1) status, assessed in tissue biopsy samples, is the only test for guiding the prescription of these therapies in clinical practice. However, obtaining tumor tissue from patients with lung cancer is not always feasible and PD-L1 positivity is not a guarantee of immunotherapy efficacy. In this context, liquid biopsy, represented by several circulating biomarkers that reflect the tumor characteristics, is emerging as an interesting alternative approach. CONTENT We describe the main blood biomarkers evaluated in patients with metastatic nonsmall cell lung cancer before/during immune checkpoint inhibitor treatment, with a focus on circulating cell-free DNA, circulating tumor DNA (ctDNA), blood tumor mutational burden, and circulating tumor cells (CTCs). SUMMARY Monitoring of ctDNA and CTCs during immunotherapy may be a promising tool to help clinicians in therapeutic decision-making.
Collapse
Affiliation(s)
- Léa Sinoquet
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - William Jacot
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| | - Xavier Quantin
- Department of Medical Oncology, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Montpellier University, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France.,CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRD, Montpellier, France
| |
Collapse
|
35
|
Sivapalan L, Murray JC, Canzoniero JV, Landon B, Jackson J, Scott S, Lam V, Levy BP, Sausen M, Anagnostou V. Liquid biopsy approaches to capture tumor evolution and clinical outcomes during cancer immunotherapy. J Immunother Cancer 2023; 11:e005924. [PMID: 36657818 PMCID: PMC9853269 DOI: 10.1136/jitc-2022-005924] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 01/20/2023] Open
Abstract
Circulating cell-free tumor DNA (ctDNA) can serve as a real-time biomarker of tumor burden and provide unique insights into the evolving molecular landscape of cancers under the selective pressure of immunotherapy. Tracking the landscape of genomic alterations detected in ctDNA may reveal the clonal architecture of the metastatic cascade and thus improve our understanding of the molecular wiring of therapeutic responses. While liquid biopsies may provide a rapid and accurate evaluation of tumor burden dynamics during immunotherapy, the complexity of antitumor immune responses is not fully captured through single-feature ctDNA analyses. This underscores a need for integrative studies modeling the tumor and the immune compartment to understand the kinetics of tumor clearance in association with the quality of antitumor immune responses. Clinical applications of ctDNA testing in patients treated with immune checkpoint inhibitors have shown both predictive and prognostic value through the detection of genomic biomarkers, such as tumor mutational burden and microsatellite instability, as well as allowing for real-time monitoring of circulating tumor burden and the assessment of early on-therapy responses. These efforts highlight the emerging role of liquid biopsies in selecting patients for cancer immunotherapy, monitoring therapeutic efficacy, determining the optimal duration of treatment and ultimately guiding treatment selection and sequencing. The clinical translation of liquid biopsies is propelled by the increasing number of ctDNA-directed interventional clinical trials in the immuno-oncology space, signifying a critical step towards implementation of liquid biopsies in precision immuno-oncology.
Collapse
Affiliation(s)
- Lavanya Sivapalan
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Joseph C Murray
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jenna VanLiere Canzoniero
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Blair Landon
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | | | - Susan Scott
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Vincent Lam
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Benjamin P Levy
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mark Sausen
- Personal Genome Diagnostics, Baltimore, Maryland, USA
| | - Valsamo Anagnostou
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
36
|
Kansara M, Bhardwaj N, Thavaneswaran S, Xu C, Lee JK, Chang L, Madison RW, Lin F, Hsu E, Patel VK, Aleshin A, Oxnard GR, Simes J, Nimeiri H, Thomas DM. Early circulating tumor DNA dynamics as a pan-tumor biomarker for long-term clinical outcome in patients treated with durvalumab and tremelimumab. Mol Oncol 2022; 17:298-311. [PMID: 36426653 PMCID: PMC9892824 DOI: 10.1002/1878-0261.13349] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/13/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022] Open
Abstract
There is an urgent need to identify biomarkers of early response that can accurately predict the benefit of immune checkpoint inhibitors (ICI). Patients receiving durvalumab/tremelimumab had tumor samples sequenced before treatment (baseline) to identify variants for the design of a personalized circulating tumor (ctDNA) assay. ctDNA was assessed at baseline and at 4 and/or 8 weeks into treatment. Correlations between ctDNA changes to radiographic response and overall survival (OS) were made to assess potential clinical benefit. 35/40 patients (87.5%) had personalized ctDNA assays designed, and 29/35 (82.9%) had plasma available for baseline analysis, representing 16 unique solid tumor histologies. As early as 4 weeks after treatment, decline in ctDNA from baseline predicted improved OS (P = 0.0144; HR = 9.98) and ctDNA changes on treatment-supported and refined radiographic response calls. ctDNA clearance at any time through week 8 identified complete responders by a median lead time of 11.5 months ahead of radiographic imaging. ctDNA response monitoring is emerging as a dynamic, personalized biomarker method that may predict survival outcomes in patients with diverse solid tumor histologies, complementing and sometimes preceding standard-of-care imaging assessments.
Collapse
Affiliation(s)
- Maya Kansara
- The Kinghorn Cancer CentreGarvan Institute of Medical ResearchDarlinghurstNSWAustralia,Faculty of Medicine, St. Vincent's Clinical SchoolUNSW SydneyKensingtonNSWAustralia
| | | | - Subotheni Thavaneswaran
- The Kinghorn Cancer CentreGarvan Institute of Medical ResearchDarlinghurstNSWAustralia,Faculty of Medicine, St. Vincent's Clinical SchoolUNSW SydneyKensingtonNSWAustralia,National Health and Medical Research Council Clinical Trials CentreUniversity of SydneyNSWAustralia
| | - Chang Xu
- Foundation Medicine, Inc.CambridgeMAUSA
| | | | | | | | - Frank Lin
- The Kinghorn Cancer CentreGarvan Institute of Medical ResearchDarlinghurstNSWAustralia,Faculty of Medicine, St. Vincent's Clinical SchoolUNSW SydneyKensingtonNSWAustralia,National Health and Medical Research Council Clinical Trials CentreUniversity of SydneyNSWAustralia
| | - Eugene Hsu
- Radiology DepartmentSt Vincent's HospitalSydneyNSWAustralia
| | | | | | | | - John Simes
- National Health and Medical Research Council Clinical Trials CentreUniversity of SydneyNSWAustralia
| | | | - David M. Thomas
- The Kinghorn Cancer CentreGarvan Institute of Medical ResearchDarlinghurstNSWAustralia,Faculty of Medicine, St. Vincent's Clinical SchoolUNSW SydneyKensingtonNSWAustralia
| |
Collapse
|
37
|
Miller AM, Karajannis MA. Current Role and Future Potential of CSF ctDNA for the Diagnosis and Clinical Management of Pediatric Central Nervous System Tumors. J Natl Compr Canc Netw 2022; 20:1363-1369. [PMID: 36509077 PMCID: PMC10050207 DOI: 10.6004/jnccn.2022.7093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022]
Abstract
Most pediatric central nervous system (CNS) tumors are located in eloquent anatomic areas, making surgical resection and, in some cases, even biopsy risky or impossible. This diagnostic predicament coupled with the move toward molecular classification for diagnosis has exposed an urgent need to develop a minimally invasive means to obtain diagnostic information. In non-CNS solid tumors, the detection of circulating tumor DNA (ctDNA) in plasma and other bodily fluids has been incorporated into routine practice and clinical trial design for selection of molecular targeted therapy and longitudinal monitoring. For primary CNS tumors, however, detection of ctDNA in plasma has been challenging. This is likely related at least in part to anatomic factors such as the blood-brain barrier. Due to the proximity of primary CNS tumors to the cerebrospinal fluid (CSF) space, our group and others have turned to CSF as a rich alternative source of ctDNA. Although multiple studies at this time have demonstrated the feasibility of CSF ctDNA detection across multiple types of pediatric CNS tumors, the optimal role and utility of CSF ctDNA in the clinical setting has not been established. This review discusses the work-to-date on CSF ctDNA liquid biopsy in pediatric CNS tumors and the associated technical challenges, and reviews the promising opportunities that lie ahead for integration of CSF ctDNA liquid biopsy into clinical care and clinical trial design.
Collapse
Affiliation(s)
- Alexandra M. Miller
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | | |
Collapse
|
38
|
Fang X, Yu S, Jiang Y, Xiang Y, Lu K. Circulating tumor DNA detection in MRD assessment and diagnosis and treatment of non-small cell lung cancer. Front Oncol 2022; 12:1027664. [PMID: 36387176 PMCID: PMC9646858 DOI: 10.3389/fonc.2022.1027664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/11/2022] [Indexed: 11/24/2022] Open
Abstract
Circulating tumor DNA (ctDNA) has contributed immensely to the management of hematologic malignancy and is now considered a valuable detection tool for solid tumors. ctDNA can reflect the real-time tumor burden and be utilized for analyzing specific cancer mutations via liquid biopsy which is a non-invasive procedure that can be used with a relatively high frequency. Thus, many clinicians use ctDNA to assess minimal residual disease (MRD) and it serves as a prognostic and predictive biomarker for cancer therapy, especially for non-small cell lung cancer (NSCLC). Advanced methods have been developed to detect ctDNA, and recent clinical trials have shown the rationality and feasibility of ctDNA for identifying mutations and guiding treatments in NSCLC. Here, we have reviewed recently developed ctDNA detection methods and the importance of sequence analyses of ctDNA in NSCLC.
Collapse
Affiliation(s)
| | | | | | | | - Kaihua Lu
- Department of Oncology, The First Affiliated Hosptial of Nanjing Medicial University, Nanjing, China
| |
Collapse
|
39
|
Malapelle U, Pisapia P, Pepe F, Russo G, Buono M, Russo A, Gomez J, Khorshid O, Mack PC, Rolfo C, Troncone G. The evolving role of liquid biopsy in lung cancer. Lung Cancer 2022; 172:53-64. [PMID: 35998482 DOI: 10.1016/j.lungcan.2022.08.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 07/22/2022] [Accepted: 08/05/2022] [Indexed: 12/20/2022]
Abstract
Liquid biopsy has revolutionized the management of cancer patients. In particular, liquid biopsy-based testing has proven to be highly beneficial for identifying actionable cancer markers, especially when solid tissue biopsies are insufficient or unattainable. Beyond the predictive role, liquid biopsy may be a useful tool for comprehensive tumor genotyping, identification of emergent resistance mechanisms, monitoring of minimal residual disease, early detection, and cancer interception. The application of next generation sequencing to liquid biopsy has led to the "quantum leap" of predictive molecular pathology. Here, we review the evolving role of liquid biopsy in lung cancer.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy.
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Mauro Buono
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Jorge Gomez
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Ola Khorshid
- National Cancer Institute, Cairo University, Cairo, Egypt
| | - Philip C Mack
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Medical System & Icahn School of Medicine, New York, NY, USA
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, Naples, Italy
| |
Collapse
|
40
|
[Research Progress of Circulating Tumor DNA in Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:665-670. [PMID: 36172731 PMCID: PMC9549427 DOI: 10.3779/j.issn.1009-3419.2022.102.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
With the concept of "Precision Medicine" in malignant tumors popularized, many substances carrying valuable clinical information have emerged in the process of exploring the occurrence and development of tumors from a microscopic perspective. Circulating tumor DNA (ctDNA) is one of them. In various clinical stages of cancer, ctDNA exhibits rich diagnostic values including demonstrating the efficacy of treatment, predicting prognosis, and monitoring disease recurrence. This article mainly describes the application and research progress of ctDNA in different stages of clinical diagnosis and treatment of non-small cell lung cancer .
.
Collapse
|
41
|
García-Pardo M, Makarem M, Li JJN, Kelly D, Leighl NB. Integrating circulating-free DNA (cfDNA) analysis into clinical practice: opportunities and challenges. Br J Cancer 2022; 127:592-602. [PMID: 35347327 PMCID: PMC9381753 DOI: 10.1038/s41416-022-01776-9] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
In the current era of precision medicine, the identification of genomic alterations has revolutionised the management of patients with solid tumours. Recent advances in the detection and characterisation of circulating tumour DNA (ctDNA) have enabled the integration of liquid biopsy into clinical practice for molecular profiling. ctDNA has also emerged as a promising biomarker for prognostication, monitoring disease response, detection of minimal residual disease and early diagnosis. In this Review, we discuss current and future clinical applications of ctDNA primarily in non-small cell lung cancer in addition to other solid tumours.
Collapse
Affiliation(s)
- Miguel García-Pardo
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Maisam Makarem
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Janice J N Li
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Deirdre Kelly
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Natasha B Leighl
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada.
| |
Collapse
|
42
|
Mack PC, Miao J, Redman MW, Moon J, Goldberg SB, Herbst RS, Melnick MA, Walther Z, Hirsch FR, Politi K, Kelly K, Gandara DR. Circulating Tumor DNA Kinetics Predict Progression-Free and Overall Survival in EGFR TKI-Treated Patients with EGFR-Mutant NSCLC (SWOG S1403). Clin Cancer Res 2022; 28:3752-3760. [PMID: 35713632 PMCID: PMC9444942 DOI: 10.1158/1078-0432.ccr-22-0741] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/04/2022] [Accepted: 06/15/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE Dynamic changes in circulating tumor DNA (ctDNA) are under investigation as an early indicator of treatment outcome. EXPERIMENTAL DESIGN Serial plasma ctDNA (baseline, 8 weeks, and at progression) was prospectively incorporated into the SWOG S1403 clinical trial of afatinib ± cetuximab in tyrosine kinase inhibitor-naïve, EGFR mutation tissue-positive non-small cell lung cancer. RESULTS EGFR mutations were detected in baseline ctDNA in 77% (82/106) of patients, associated with the presence of brain and/or liver metastases and M1B stage. Complete clearance of EGFR mutations in ctDNA by 8 weeks was associated with a significantly decreased risk of progression, compared with those with persistent ctDNA at Cycle 3 Day 1 [HR, 0.23; 95% confidence interval (CI), 0.12-0.45; P < 0.0001], with a median progression-free survival (PFS) of 15.1 (95% CI, 10.6-17.5) months in the group with clearance of ctDNA versus 4.6 (1.7-7.5) months in the group with persistent ctDNA. Clearance was also associated with a decreased risk of death (HR, 0.44; 95% CI, 0.21-0.90), P = 0.02; median overall survival (OS): 32.6 (23.5-not estimable) versus 15.6 (4.9-28.3) months. CONCLUSIONS Plasma clearance of mutant EGFR ctDNA at 8 weeks was highly and significantly predictive of PFS and OS, outperforming RECIST response for predicting long-term benefit.
Collapse
Affiliation(s)
- Philip C. Mack
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Health System, New York City, NY
| | - Jieling Miao
- SWOG Statistical Center and Clinical Biostatistics, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Mary W. Redman
- SWOG Statistical Center and Clinical Biostatistics, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - James Moon
- SWOG Statistical Center and Clinical Biostatistics, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Sarah B. Goldberg
- Department of Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT
| | - Roy S. Herbst
- Department of Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT
| | - Mary Ann Melnick
- Department of Internal Medicine (Section of Medical Oncology), Yale School of Medicine, New Haven, CT
| | - Zenta Walther
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Fred R. Hirsch
- Center for Thoracic Oncology, Tisch Cancer Institute, Mount Sinai Health System, New York City, NY
| | - Katerina Politi
- Department of Pathology, Yale School of Medicine, New Haven, CT
| | - Karen Kelly
- Division of Hematology/Oncology, University of California at Davis, Sacramento, CA
| | - David R. Gandara
- Division of Hematology/Oncology, University of California at Davis, Sacramento, CA
| |
Collapse
|
43
|
Remon J, Levy A, Singh P, Hendriks LEL, Aldea M, Arrieta O. Current challenges of unresectable stage III NSCLC: are we ready to break the glass ceiling of the PACIFIC trial? Ther Adv Med Oncol 2022; 14:17588359221113268. [PMID: 35923929 PMCID: PMC9340398 DOI: 10.1177/17588359221113268] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/27/2022] [Indexed: 01/09/2023] Open
Abstract
Consolidation anti-programmed death-ligand 1 has become a new standard of care in unresectable stage III non-small cell lung cancer (NSCLC) following chemo-radiotherapy (CTRT), based on the results of two phase III trials. Advances remain however needed, in particular to reduce the risk of distant relapse and for treatment personalization. Newer strategies are currently being tested, including consolidation with dual immune checkpoint inhibitors (ICIs), concurrent chemo-radioimmunotherapy and (chemo)-immunotherapy induction before CTRT. One randomized phase II reported better outcomes with a double ICI consolidation as compared with durvalumab alone. Three nonrandomized phase II trials also suggested that concurrent ICI-CTRT was feasible. Within this review, we summarize the current evidence, highlight ongoing trials and discuss challenges that will ideally lead to a cure for more patients with unresectable stage III NSCLC.
Collapse
Affiliation(s)
- Jordi Remon
- Department of Medical Oncology, Centro Integral
Oncológico Clara Campal (HM-CIOCC), Hospital HM Nou Delfos, HM Hospitales,
Avinguda de Vallcarca, 151, Barcelona 08023, Spain
| | - Antonin Levy
- Department of Radiation Oncology, International
Center for Thoracic Cancers (CICT), Université Paris-Saclay, Gustave Roussy,
Gustave Roussy, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine,
Le Kremlin-Bicêtre, France
| | - Pawan Singh
- Department of Pulmonary and Critical Care
Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical
Sciences, Rohtak, Haryana, India
| | - Lizza E. L. Hendriks
- Department of Pulmonary Diseases, GROW–School
for Oncology and Reproduction, Maastricht University Medical Center,
Maastricht, The Netherlands
| | - Mihaela Aldea
- Department of Medical Oncology, International
Center for Thoracic Cancers (CICT), Université Paris-Saclay, Gustave Roussy,
Gustave Roussy, Villejuif, France
| | - Oscar Arrieta
- Thoracic Oncology Unit and Laboratory of
Personalized Medicine, Instituto Nacional de Cancerología, Mexico City,
Mexico
| |
Collapse
|
44
|
Gouda MA, Huang HJ, Piha-Paul SA, Call SG, Karp DD, Fu S, Naing A, Subbiah V, Pant S, Dustin DJ, Tsimberidou AM, Hong DS, Rodon J, Meric-Bernstam F, Janku F. Longitudinal Monitoring of Circulating Tumor DNA to Predict Treatment Outcomes in Advanced Cancers. JCO Precis Oncol 2022; 6:e2100512. [PMID: 35834760 PMCID: PMC9307306 DOI: 10.1200/po.21.00512] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/24/2022] [Accepted: 05/31/2022] [Indexed: 12/22/2022] Open
Abstract
PURPOSE The response to cancer therapies is typically assessed with radiologic imaging 6-10 weeks after treatment initiation. Circulating tumor DNA (ctDNA), however, has a short half-life, and dynamic changes in ctDNA quantity may allow for earlier assessment of the therapeutic response. METHODS Patients with advanced solid tumors referred to the Department of Investigational Cancer Therapeutics at The University of Texas MD Anderson Cancer Center were invited to participate in a liquid biopsy protocol for which serial blood samples were collected before, during, and after systemic therapy. We isolated ctDNA from serially collected plasma samples at baseline, mid-treatment, and first restaging. Genomically informed droplet digital polymerase chain reaction (ddPCR) was performed, and ctDNA quantities were reported as aggregate variant allele frequencies for all detected molecular aberrations. RESULTS We included 204 patients receiving 260 systemic therapies. The ctDNA detection rate was higher in progressors (patients with progressive disease) compared with nonprogressors (patients with stable disease, partial responses, or complete responses) at all time points (P < .009). Moreover, ctDNA detection was associated with a shorter median time-to-treatment failure (P ≤ .001). Positive delta and slope values for changes in ctDNA quantity were more frequent in progressors (P ≤ .03 and P < .001, respectively) and were associated with a shorter median time-to-treatment failure (P ≤ .014 and P < .001, respectively). Increasing ctDNA quantity was predictive of clinical and/or radiologic progressive disease in 73% of patients (median lead time, 23 days). CONCLUSION Detection of ctDNA and early dynamic changes in its quantity can predict the clinical outcomes of systemic therapies in patients with advanced solid tumors.
Collapse
Affiliation(s)
- Mohamed A. Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
- Department of Clinical Oncology, Faculty of Medicine, Menoufia University. Shebin Al-Kom, Egypt
| | - Helen J. Huang
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sarina A. Piha-Paul
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - S. Greg Call
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Daniel D. Karp
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Siqing Fu
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Aung Naing
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vivek Subbiah
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shubham Pant
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Derek J. Dustin
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Apostolia M. Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - David S. Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jordi Rodon
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Filip Janku
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
45
|
Sankar K, Zeinali M, Nagrath S, Ramnath N. Molecular biomarkers and liquid biopsies in lung cancer. Semin Oncol 2022; 49:275-284. [PMID: 35820969 DOI: 10.1053/j.seminoncol.2022.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 06/13/2022] [Accepted: 06/13/2022] [Indexed: 12/27/2022]
Abstract
Liquid biopsy refers to the identification of tumor-derived materials in body fluids including in blood circulation. In the age of immunotherapy and targeted therapies used for the treatment of advanced malignancies, molecular analysis of the tumor is considered a crucial step to guide management. In lung cancer, the concept of liquid biopsies is particularly relevant given the invasiveness of tumor biopsies in certain locations, and the potential risks of biopsy in a patient population with significant co-morbidities. Liquid biopsies have many advantages including non-invasiveness, lower cost, potential for genomic testing, ability to monitor tumor evolution through treatment, and the ability to overcome spatial and temporal intertumoral heterogeneity. The potential clinical applications of liquid biopsy are vast and include screening, detection of minimal residual disease and/or early relapse after curative intent treatment, monitoring response to immunotherapy, and identifying mutations that might be targetable or can confer resistance. Herein, we review the potential role of circulating tumor DNA and circulating tumor cells as forms of liquid biopsies and blood biomarkers in non-small cell lung cancer. We discuss the methodologies/platforms available for each, clinical applications, and limitations/challenges in incorporation into clinical practice. We additionally review emerging forms of liquid biopsies including tumor educated platelets, circular RNA, and exosomes.
Collapse
Affiliation(s)
- Kamya Sankar
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI
| | - Mina Zeinali
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI; Biointerfaces Institute, University of Michigan, Ann Arbor, MI; Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Sunitha Nagrath
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI; Biointerfaces Institute, University of Michigan, Ann Arbor, MI; Rogel Cancer Center, University of Michigan, Ann Arbor, MI
| | - Nithya Ramnath
- Division of Medical Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI.
| |
Collapse
|
46
|
Hegi-Johnson F, Rudd S, Hicks RJ, De Ruysscher D, Trapani JA, John T, Donnelly P, Blyth B, Hanna G, Everitt S, Roselt P, MacManus MP. Imaging immunity in patients with cancer using positron emission tomography. NPJ Precis Oncol 2022; 6:24. [PMID: 35393508 PMCID: PMC8989882 DOI: 10.1038/s41698-022-00263-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 02/24/2022] [Indexed: 12/26/2022] Open
Abstract
Immune checkpoint inhibitors and related molecules can achieve tumour regression, and even prolonged survival, for a subset of cancer patients with an otherwise dire prognosis. However, it remains unclear why some patients respond to immunotherapy and others do not. PET imaging has the potential to characterise the spatial and temporal heterogeneity of both immunotherapy target molecules and the tumor immune microenvironment, suggesting a tantalising vision of personally-adapted immunomodulatory treatment regimens. Personalised combinations of immunotherapy with local therapies and other systemic therapies, would be informed by immune imaging and subsequently modified in accordance with therapeutically induced immune environmental changes. An ideal PET imaging biomarker would facilitate the choice of initial therapy and would permit sequential imaging in time-frames that could provide actionable information to guide subsequent therapy. Such imaging should provide either prognostic or predictive measures of responsiveness relevant to key immunotherapy types but, most importantly, guide key decisions on initiation, continuation, change or cessation of treatment to reduce the cost and morbidity of treatment while enhancing survival outcomes. We survey the current literature, focusing on clinically relevant immune checkpoint immunotherapies, for which novel PET tracers are being developed, and discuss what steps are needed to make this vision a reality.
Collapse
Affiliation(s)
- Fiona Hegi-Johnson
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Stacey Rudd
- Department of Chemistry, University of Melbourne, Melbourne, VIC, Australia
| | - Rodney J Hicks
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Dirk De Ruysscher
- Department of Radiation Oncology (Maastro), GROW School for Oncology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Joseph A Trapani
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Thomas John
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Paul Donnelly
- Department of Chemistry, University of Melbourne, Melbourne, VIC, Australia
| | - Benjamin Blyth
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Gerard Hanna
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Sarah Everitt
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Peter Roselt
- Department of Cancer Imaging, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Michael P MacManus
- Department of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia.
- The Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
47
|
Lu C, Zhang YC, Chen ZH, Zhou Q, Wu YL. Applications of Circulating Tumor DNA in Immune Checkpoint Inhibition: Emerging Roles and Future Perspectives. Front Oncol 2022; 12:836891. [PMID: 35359372 PMCID: PMC8963952 DOI: 10.3389/fonc.2022.836891] [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: 12/16/2021] [Accepted: 02/11/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs), especially anti-programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) antibodies, have made dramatic progress in the treatment of lung cancer, especially for patients with cancers not driven by oncogenes. However, responses are limited to a subset of patients, and which subset of patients will optimally benefit from ICI remains unknown. With the advantage of being minimally invasive and dynamic, noninvasive biomarkers are promising candidates to predict response, monitor resistance, and track the evolution of lung cancer during ICI treatment. In this review, we focus on the application of circulating tumor DNA (ctDNA) in plasma in immunotherapy. We examine the potential of pre- and on-treatment features of ctDNA as biomarkers, and following multiparameter analysis, we determine the potential clinical value of integrating predictive liquid biomarkers of ICIs to optimize patient management. We further discuss the role of ctDNA in monitoring treatment resistance, as well as challenges in clinical translation.
Collapse
Affiliation(s)
- Chang Lu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yi-Chen Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhi-Hong Chen
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
48
|
Girardi DM, Niglio SA, Mortazavi A, Nadal R, Lara P, Pal SK, Saraiya B, Cordes L, Ley L, Ortiz OS, Cadena J, Diaz C, Bagheri H, Redd B, Steinberg SM, Costello R, Chan KS, Lee MJ, Lee S, Yu Y, Gurram S, Chalfin HJ, Valera V, Figg WD, Merino M, Toubaji A, Streicher H, Wright JJ, Sharon E, Parnes HL, Ning YM, Bottaro DP, Cao L, Trepel JB, Apolo AB. Cabozantinib plus Nivolumab Phase I Expansion Study in Patients with Metastatic Urothelial Carcinoma Refractory to Immune Checkpoint Inhibitor Therapy. Clin Cancer Res 2022; 28:1353-1362. [PMID: 35031545 PMCID: PMC9365339 DOI: 10.1158/1078-0432.ccr-21-3726] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/17/2021] [Accepted: 01/12/2022] [Indexed: 01/26/2023]
Abstract
PURPOSE This study investigated the efficacy and tolerability of cabozantinib plus nivolumab (CaboNivo) in patients with metastatic urothelial carcinoma (mUC) that progressed on checkpoint inhibition (CPI). PATIENTS AND METHODS A phase I expansion cohort of patients with mUC who received prior CPI was treated with cabozantinib 40 mg/day and nivolumab 3 mg/kg every 2 weeks until disease progression/unacceptable toxicity. The primary goal was objective response rate (ORR) per RECIST v.1.1. Secondary objectives included progression-free survival (PFS), duration of response (DoR), overall survival (OS), safety, and tolerability. RESULTS Twenty-nine out of 30 patients enrolled were evaluable for efficacy. Median follow-up was 22.2 months. Most patients (86.7%) received prior chemotherapy and all patients received prior CPI (median seven cycles). ORR was 16.0%, with one complete response and three partial responses (PR). Among 4 responders, 2 were primary refractory, 1 had a PR, and 1 had stable disease on prior CPI. Median DoR was 33.5 months [95% confidence interval (CI), 3.7-33.5], median PFS was 3.6 months (95% CI, 2.1-5.5), and median OS was 10.4 months (95% CI, 5.8-19.5). CaboNivo decreased immunosuppressive subsets such as regulatory T cells (Tregs) and increased potential antitumor immune subsets such as nonclassical monocytes and effector T cells. A lower percentage of monocytic myeloid-derived suppressor cells (M-MDSC) and polymorphonuclear MDSCs, lower CTLA-4 and TIM-3 expression on Tregs, and higher effector CD4+ T cells at baseline were associated with better PFS and/or OS. CONCLUSIONS CaboNivo was clinically active, well tolerated, and favorably modulated peripheral blood immune subsets in patients with mUC refractory to CPI.
Collapse
Affiliation(s)
- Daniel M. Girardi
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Scot A. Niglio
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, The Ohio State University, and the Comprehensive Cancer Center, Columbus, Ohio
| | - Rosa Nadal
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Primo Lara
- University of California Davis Comprehensive Cancer Center, Sacramento, California
| | - Sumanta K. Pal
- City of Hope Comprehensive Cancer Center, Duarte, California
| | - Biren Saraiya
- Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| | - Lisa Cordes
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Lisa Ley
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Olena Sierra Ortiz
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Jacqueline Cadena
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Carlos Diaz
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Hadi Bagheri
- Clinical Image Processing Service, Department of Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, Maryland
| | - Bernadette Redd
- Clinical Image Processing Service, Department of Radiology and Imaging Sciences, Clinical Center, NIH, Bethesda, Maryland
| | - Seth M. Steinberg
- Biostatistics and Data Management Section, Office of the Clinical Director, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Rene Costello
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Keith S. Chan
- Samuel Oschin Cancer Center, Cedars Sinai Medical Center, Los Angeles, California
| | - Min-Jung Lee
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Sunmin Lee
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Yunkai Yu
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Sandeep Gurram
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Heather J. Chalfin
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Vladimir Valera
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - William D. Figg
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Maria Merino
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Antoun Toubaji
- Laboratory of Pathology, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Howard Streicher
- Investigational Drug Branch, Cancer Therapy Evaluation Program, NCI, NIH, Rockville, Maryland
| | - John J. Wright
- Investigational Drug Branch, Cancer Therapy Evaluation Program, NCI, NIH, Rockville, Maryland
| | - Elad Sharon
- Investigational Drug Branch, Cancer Therapy Evaluation Program, NCI, NIH, Rockville, Maryland
| | - Howard L. Parnes
- Division of Cancer Prevention, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Yang-Min Ning
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Donald P. Bottaro
- Urologic Oncology Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Liang Cao
- Genetics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Jane B. Trepel
- Developmental Therapeutics Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Andrea B. Apolo
- Genitourinary Malignancies Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| |
Collapse
|
49
|
Song S, Bai M, Li X, Gong S, Yang W, Lei C, Tian H, Si M, Hao X, Guo T. Early Predictive Value of Circulating Biomarkers for Sorafenib in Advanced Hepatocellular Carcinoma. Expert Rev Mol Diagn 2022; 22:361-378. [PMID: 35234564 DOI: 10.1080/14737159.2022.2049248] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Sorafenib is currently the first-line therapeutic regimen for patients with advanced hepatocellular carcinoma (HCC). However, many patients did not experience any benefit and suffered extreme adverse events and heavy economic burden. Thus, the early identification of patients who are most likely to benefit from sorafenib is needed. AREAS COVERED This review focused on the clinical application of circulating biomarkers (including conventional biomarkers, immune biomarkers, genetic biomarkers, and some novel biomarkers) in advanced HCC patients treated with sorafenib. An online search on PubMed, Web of Science, Embase, and Cochrane Library was conducted from the inception to Aug 15, 2021. Studies investigating the predictive or prognostic value of these biomarkers were included. EXPERT OPINION The distinction of patients who may benefit from sorafenib treatment is of utmost importance. The predictive roles of circulating biomarkers could solve this problem. Many biomarkers can be obtained by liquid biopsy, which is a less or non-invasive approach. The short half-life of sorafenib could reflect the dynamic changes of tumor progression and monitor the treatment response. Circulating biomarkers obtained from liquid biopsy resulted as a promising assessment method in HCC, allowing for better treatment decisions in the near future.
Collapse
Affiliation(s)
- Shaoming Song
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.,Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
| | - Mingzhen Bai
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Xiaofei Li
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China
| | - Shiyi Gong
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China.,School of Basic Medical Sciences, Evidence-Based Medicine Center, Lanzhou University, Lanzhou, China
| | - Wenwen Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.,School of Basic Medical Sciences, Evidence-Based Medicine Center, Lanzhou University, Lanzhou, China
| | - Caining Lei
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China.,School of Basic Medical Sciences, Evidence-Based Medicine Center, Lanzhou University, Lanzhou, China
| | - Hongwei Tian
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.,Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China.,Key Laboratory of Molecular Diagnostics, and Precision Medicine of Surgical Oncology in Gansu Province, Lanzhou, China
| | - Moubo Si
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China.,Key Laboratory of Molecular Diagnostics, and Precision Medicine of Surgical Oncology in Gansu Province, Lanzhou, China
| | - Xiangyong Hao
- Department of General Surgery, Gansu Provincial Hospital, Lanzhou, China.,Key Laboratory of Molecular Diagnostics, and Precision Medicine of Surgical Oncology in Gansu Province, Lanzhou, China
| | - Tiankang Guo
- The First Clinical Medical College of Lanzhou University, Lanzhou, China.,Key Laboratory of Molecular Diagnostics, and Precision Medicine of Surgical Oncology in Gansu Province, Lanzhou, China
| |
Collapse
|
50
|
[Research Progress, Benefit Groups, Treatment Cycle and Efficacy Prediction
of Neoadjuvant Immunotherapy for Non-small Cell Lung Cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:92-101. [PMID: 35224962 PMCID: PMC8913292 DOI: 10.3779/j.issn.1009-3419.2022.101.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has dramatically changed the therapeutic outlook for patients with non-small cell lung cancer (NSCLC). Preoperative neoadjuvant immunotherapy has been paid more and more attention as an effective and safe treatment. Neoadjuvant immune therapy, however, the relevant research started late, relatively few research results and mainly focused on the small sample size of phase I and II studies, treatment itself exists many places it is not clear, also in benefit population screening, the respect such as the choice of treatment and curative effect prediction has not yet reached broad consensus. This paper reviews the important studies and recent achievements related to neoadjuvant immunotherapy, aiming to comprehensively discuss the procedures and existing problems of this kind of therapy from three aspects of beneficiary groups, treatment cycle and efficacy prediction.
.
Collapse
|