1
|
Gianmarco M, Carolina P, Gregorio M, Michela V, Monica P, Claire GG, Michele M, Giulia M, Roberta M, Cinzia A, Lorena B, Marcello T, Fabiana P, Roberta M. Circulating tumor DNA monitoring in advanced mutated melanoma (LIQUID-MEL). THE JOURNAL OF LIQUID BIOPSY 2025; 8:100295. [PMID: 40276578 PMCID: PMC12019447 DOI: 10.1016/j.jlb.2025.100295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 04/07/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025]
Abstract
Introduction Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of metastatic melanoma, but a percentage of patients did not show benefit. Circulating tumor DNA (ctDNA) has emerged as a potential non-invasive tool for monitoring disease evolution and treatment response. The present study aimed to evaluate the clinical utility of ctDNA dynamics in patients with metastatic melanoma receiving ICIs, while exploring its role in the oncological course. Materials and methods The LIQUID-MEL study is a prospective, single-centre pilot study including patients with BRAF/NRAS-mutant metastatic melanoma. ctDNA was quantified using digital droplet PCR (ddPCR) at four different time points. Uni- and multivariable Cox regression models were used to assess the correlation between shedding and progression-free survival (PFS), and overall survival (OS). Results Overall, 23 patients were included. At baseline, ctDNA was detectable in 5/23 (21.7 %) cases. Baseline ctDNA shedding was associated with shorter PFS (3.88 months vs. 0.69 months, p=0.012). A strong numerical trend was observed also in OS (12.66 months vs. 2.53 months, p=0.287). Shedding at baseline did not demonstrate independent prognostic or predictive value in the uni- and multivariable analysis. The longitudinal analysis revealed intriguing patterns of ctDNA shedding in individual patients. Conclusion ctDNA detectability and its dynamic changes during treatment may have potential clinical utility in patients with metastatic melanoma, offering a valuable non-invasive tool for monitoring disease and treatment response. The small sample size limited the statistical power of the analysis. Further studies with larger cohorts are needed to validate its role in routine clinical practice.
Collapse
Affiliation(s)
| | - Palazzi Carolina
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Monica Gregorio
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Verzè Michela
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Pluchino Monica
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | | | - Maffezzoli Michele
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Portsmouth Hospital University NHS Trust, Portsmouth, United Kingdom
| | - Mazzaschi Giulia
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Manuguerra Roberta
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Azzoni Cinzia
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Bottarelli Lorena
- Pathology Unit, Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Tiseo Marcello
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Perrone Fabiana
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| | - Minari Roberta
- Medical Oncology Unit, University Hospital of Parma, Parma, Italy
| |
Collapse
|
2
|
Han B, Shen C, Hu H, Zhang J, Xie X, Mo Q, Deng Y. Circulating Tumor DNA Predicts Conversion Therapy Response and Prognosis in Initially Unresectable Colorectal Liver-Limited Metastases: A Retrospective Study. Br J Hosp Med (Lond) 2025; 86:1-16. [PMID: 40135301 DOI: 10.12968/hmed.2024.0695] [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] [Indexed: 03/27/2025]
Abstract
Aims/Background Effective molecular biomarkers for predicting prognosis and guiding treatment in patients with initially unresectable colorectal liver metastases (CRLMs) undergoing conversion therapy are currently lacking. This study investigated the predictive value of circulating tumor DNA (ctDNA) conversion therapy outcomes in initially unresectable CRLMs. Methods A retrospective analysis was conducted on 81 patients with CRLMs treated at the Sixth Affiliated Hospital, Sun Yat-sen University from January 2017 to April 2021. The relationships between baseline and treatment ctDNA levels and clinical responses were evaluated using group comparisons based on data type. The impact of ctDNA on survival outcomes was analyzed through Cox regression survival analysis. Results Analysis of 81 patients with ctDNA-positive at baseline showed that patients in the ctDNA low-level group had a significantly longer median progression-free survival (mPFS) (p = 0.039). Among 45 patients who underwent ctDNA testing during systemic therapy, the proportion of patients in the ctDNA-negative group receiving local ablative treatment (LAT) was significantly higher (70.0% vs 26.7%, p = 0.006). Furthermore, 50% of patients in the ctDNA-negative group achieved no evidence of disease (NED) status, compared to 6.7% in the ctDNA-positive group (p = 0.004). Both mPFS and median overall survival (mOS) were significantly longer in ctDNA-negative patients compared to ctDNA-positive patients (p < 0.05). Of the 61 patients who underwent LAT, 37 received ctDNA testing at the same time as imaging assessment for NED. The proportion of patients with ctDNA clearance who achieved NED status was markedly higher than that of patients with ctDNA non-clearance (78.6% vs 33.3%, p = 0.036). Patients with ctDNA clearance demonstrated significantly improved mOS compared to those with ctDNA non-clearance (not reached vs 30.1 months, p = 0.036). Conclusion Dynamic changes in ctDNA levels can predict both long-term survival and the effectiveness of conversion therapy in patients with initially unresectable CRLMs.
Collapse
Affiliation(s)
- Bohan Han
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Cailu Shen
- Department of Medical Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu, China
| | - Huabin Hu
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Jianwei Zhang
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiaoyu Xie
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qinli Mo
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yanhong Deng
- Department of Medical Oncology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| |
Collapse
|
3
|
Alexander EM, Miller HA, Egger ME, Smith ML, Yaddanapudi K, Linder MW. The Correlation between Plasma Circulating Tumor DNA and Radiographic Tumor Burden. J Mol Diagn 2024; 26:952-961. [PMID: 39181324 PMCID: PMC11524323 DOI: 10.1016/j.jmoldx.2024.07.001] [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: 01/23/2024] [Revised: 06/05/2024] [Accepted: 07/15/2024] [Indexed: 08/27/2024] Open
Abstract
Conventional blood-based biomarkers and radiographic imaging are excellent for use in monitoring different aspects of malignant disease, but given their specific shortcomings, their integration with other, complementary markers such as plasma circulating tumor DNA (ctDNA) will be beneficial toward a precision medicine-driven future. Plasma ctDNA analysis utilizes the measurement of cancer-specific molecular alterations in a variety of bodily fluids released by dying tumor cells to monitor and profile response to therapy, and is being employed in several clinical scenarios. Plasma concentrations of ctDNA have been reported to correlate with tumor burden. However, the strength of this association is generally poor and highly variable, confounding the interpretation of longitudinal plasma ctDNA measurements in conjunction with routine radiographic assessments. Herein is discussed what is currently understood with respect to the fundamental characteristics of tumor growth that dictate plasma ctDNA concentrations, with a perspective on its interpretation in conjunction with radiographically determined tumor burden assessments.
Collapse
Affiliation(s)
- Evan M Alexander
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, Kentucky
| | - Hunter A Miller
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, Kentucky
| | - Michael E Egger
- Hiram C. Polk, Jr, MD, Department of Surgery, University of Louisville, Louisville, Kentucky; UofL Health-Brown Cancer Center, University of Louisville, Louisville, Kentucky
| | - Melissa L Smith
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, Kentucky; Department of Biochemistry and Molecular Genetics, University of Louisville, Louisville, Kentucky; Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky
| | - Kavitha Yaddanapudi
- UofL Health-Brown Cancer Center, University of Louisville, Louisville, Kentucky; Department of Microbiology and Immunology, University of Louisville, Louisville, Kentucky; Department of Medicine, University of Louisville, Louisville, Kentucky
| | - Mark W Linder
- Department of Pathology and Laboratory Medicine, University of Louisville, Louisville, Kentucky; UofL Health-Brown Cancer Center, University of Louisville, Louisville, Kentucky.
| |
Collapse
|
4
|
Slusher N, Jones N, Nonaka T. Liquid biopsy for diagnostic and prognostic evaluation of melanoma. Front Cell Dev Biol 2024; 12:1420360. [PMID: 39156972 PMCID: PMC11327088 DOI: 10.3389/fcell.2024.1420360] [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: 04/20/2024] [Accepted: 07/03/2024] [Indexed: 08/20/2024] Open
Abstract
Melanoma is the most aggressive form of skin cancer, and the majority of cases are associated with chronic or intermittent sun exposure. The incidence of melanoma has grown exponentially over the last 50 years, especially in populations of fairer skin, at lower altitudes and in geriatric populations. The gold standard for diagnosis of melanoma is performing an excisional biopsy with full resection or an incisional tissue biopsy. However, due to their invasiveness, conventional biopsy techniques are not suitable for continuous disease monitoring. Utilization of liquid biopsy techniques represent substantial promise in early detection of melanoma. Through this procedure, tumor-specific components shed into circulation can be analyzed for not only diagnosis but also treatment selection and risk assessment. Additionally, liquid biopsy is significantly less invasive than tissue biopsy and offers a novel way to monitor the treatment response and disease relapse, predicting metastasis.
Collapse
Affiliation(s)
- Nicholas Slusher
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Nicholas Jones
- School of Medicine, Louisiana State University Health Shreveport, Shreveport, LA, United States
| | - Taichiro Nonaka
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, United States
- Feist-Weiller Cancer Center, Louisiana State University Health Shreveport, Shreveport, LA, United States
| |
Collapse
|
5
|
Yao S, Han Y, Yang M, Jin K, Lan H. Integration of liquid biopsy and immunotherapy: opening a new era in colorectal cancer treatment. Front Immunol 2023; 14:1292861. [PMID: 38077354 PMCID: PMC10702507 DOI: 10.3389/fimmu.2023.1292861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 11/03/2023] [Indexed: 12/18/2023] Open
Abstract
Immunotherapy has revolutionized the conventional treatment approaches for colorectal cancer (CRC), offering new therapeutic prospects for patients. Liquid biopsy has shown significant potential in early screening, diagnosis, and postoperative monitoring by analyzing circulating tumor cells (CTC) and circulating tumor DNA (ctDNA). In the era of immunotherapy, liquid biopsy provides additional possibilities for guiding immune-based treatments. Emerging technologies such as mass spectrometry-based detection of neoantigens and flow cytometry-based T cell sorting offer new tools for liquid biopsy, aiming to optimize immune therapy strategies. The integration of liquid biopsy with immunotherapy holds promise for improving treatment outcomes in colorectal cancer patients, enabling breakthroughs in early diagnosis and treatment, and providing patients with more personalized, precise, and effective treatment strategies.
Collapse
Affiliation(s)
- Shiya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Yuejun Han
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Mengxiang Yang
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Ketao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Huanrong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Tan XL, Le A, Tang H, Brown M, Scherrer E, Han J, Jiang R, Diede SJ, Shui IM. Burden and Risk Factors of Brain Metastases in Melanoma: A Systematic Literature Review. Cancers (Basel) 2022; 14:6108. [PMID: 36551594 PMCID: PMC9777047 DOI: 10.3390/cancers14246108] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/07/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022] Open
Abstract
Melanoma can frequently metastasize to the brain with severe consequences. However, variation of melanoma brain metastases (MBM) development among populations is not well studied, and underlying mechanisms and risk factors for MBM development are not consistently documented. We conducted a systematic literature review (SLR) including a total of 39 articles to evaluate the proportion of melanoma patients who are diagnosed with, or develop, brain metastases, and summarize the risk factors of MBM. The average proportion of MBM was calculated and weighted by the sample size of each study. Meta-analyses were conducted for the selected risk factors using a random-effects model. The proportion of MBM at diagnosis was 33% (975 with MBM out of 2948 patients) among patients with cutaneous melanoma (excluding acral) and 23% (651/2875) among patients with cutaneous mixed with other types of melanoma. The proportion at diagnosis was lower among populations with mucosal (9/96, 9%) or uveal (4/184, 2%) melanoma and among populations outside the United States and Europe. Meta-analysis demonstrated that male vs. female gender and left-sided tumors vs. right-sided were significantly associated with increased risk of melanoma brain metastases. These data may help clinicians to assess an individual patient's risk of developing melanoma brain metastases.
Collapse
Affiliation(s)
| | - Amy Le
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| | - Huilin Tang
- Integrative Precision Health, LLC, Carmel, IN 46032, USA
| | - Madeline Brown
- Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN 46202, USA
| | - Emilie Scherrer
- Merck & Co., Inc., Rahway, NJ 07065, USA
- Seagen Inc., Bothell, WA 98021, USA
| | - Jiali Han
- Integrative Precision Health, LLC, Carmel, IN 46032, USA
| | | | | | | |
Collapse
|
7
|
Pitfalls and Rewards of Setting Up a Liquid Biopsy Approach for the Detection of Driver Mutations in Circulating Tumor DNAs: Our Institutional Experience. J Pers Med 2022; 12:jpm12111845. [PMID: 36579573 PMCID: PMC9692455 DOI: 10.3390/jpm12111845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 10/30/2022] [Accepted: 11/01/2022] [Indexed: 11/10/2022] Open
Abstract
We describe our institutional experience of developing a liquid biopsy approach using circulating tumor DNA (ctDNA) analysis for personalized medicine in cancer patients, focusing on the hurdles encountered during the multistep process in order to benefit other investigators wishing to set up this type of study in their institution. Blood samples were collected at the time of cancer surgery from 209 patients with one of nine different cancer types. Extracted tumor DNA and circulating cell-free DNA were sequenced using cancer-specific panels and the Illumina MiSeq machine. Almost half of the pairs investigated were uninformative, mostly because there was no trackable pathogenic mutation detected in the original tumor. The pairs with interpretable data corresponded to 107 patients. Analysis of 48 gene sequences common to both panels was performed and revealed that about 40% of these pairs contained at least one driver mutation detected in the DNA extracted from plasma. Here, we describe the choice of our overall approach, the selection of the cancer panels, and the difficulties encountered during the multistep process, including the use of several tumor types and in the data analysis. We also describe some case reports using longitudinal samples, illustrating the potential advantages and rewards in performing ctDNA sequencing to monitor tumor burden or guide treatment for cancer patients.
Collapse
|
8
|
Promising Blood-Based Biomarkers for Melanoma: Recent Progress of Liquid Biopsy and Its Future Perspectives. Curr Treat Options Oncol 2022; 23:562-577. [PMID: 35298769 DOI: 10.1007/s11864-022-00948-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/16/2021] [Indexed: 12/11/2022]
Abstract
OPINION STATEMENT Because the recent success of novel therapeutic approaches has dramatically changed the clinical management of melanoma, less invasive and repeatable monitoring tools that can predict the disease status, drug resistance, and the development of side effects are increasingly needed. As liquid biopsy has enabled us to diagnose and monitor disease status less invasively, substantial attention has been directed toward this technique, which is gaining importance as a diagnostic and/or prognostic tool. It is evident that microRNA, cell-free DNA, and circulating tumor cells obtained via liquid biopsy are promising diagnostic and prognostic tools for melanoma, and they also have utility for monitoring the disease status and predicting drug effects. Although current challenges exist for each biomarker, such as poor sensitivity and/or specificity and technical problems, recent technical advances have increasingly improved these aspects. For example, next-generation sequencing technology for detecting microRNAs or cell-free DNA enabled high-throughput analysis and provided significantly higher sensitivity. In particular, cancer personalized profiling by deep sequencing for quantifying cell-free DNA is a promising method for high-throughput analysis that provides real-time comprehensive data for patients at various disease stages. For wide clinical implementation, it is necessary to increase the sensitivity for the markers and standardize the assay procedures to make them reproducible, valid, and inexpensive; however, the broad clinical application of liquid biopsy could occur quickly. This review focuses on the significance of liquid biopsy, particularly related to the use of blood samples from patients with melanoma, and discusses its future perspectives.
Collapse
|
9
|
Berger F, Marce M, Delaloge S, Hardy-Bessard AC, Bachelot T, Bièche I, Pradines A, De La Motte Rouge T, Canon JL, André F, Arnould L, Clatot F, Lemonnier J, Marques S, Bidard FC. Randomised, open-label, multicentric phase III trial to evaluate the safety and efficacy of palbociclib in combination with endocrine therapy, guided by ESR1 mutation monitoring in oestrogen receptor-positive, HER2-negative metastatic breast cancer patients: study design of PADA-1. BMJ Open 2022; 12:e055821. [PMID: 35241469 PMCID: PMC8896060 DOI: 10.1136/bmjopen-2021-055821] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
INTRODUCTION The combination of a CDK4/6 inhibitor with an aromatase inhibitor (AI) has recently become the gold standard for AI-sensitive first line treatment of oestrogen receptor-positive (ER+) HER2-negative (HER2-) advanced breast cancer. However, most patients receiving this combination will ultimately progress and require further therapies.Several studies have demonstrated that the onset of a ESR1 gene mutation lead to AIs resistance in the advanced setting. ESR1 mutations can be detected in circulating tumour DNA (ctDNA) using a digital PCR assay. Our study aims to prove the clinical efficacy of periodic monitoring for emerging or rise of ESR1 mutations in ctDNA to trigger an early change from AI plus palbociclib to fulvestrant plus palbociclib treatment while assessing global safety. METHODS PADA-1 is a randomised, open-label, multicentric, phase III trial conducted in patients receiving AI and palbociclib as first line therapy for metastatic ER +HER2- breast cancer. 1000 patients will be included and treated with palbociclib in combination with an AI. Patients will be screened for circulating blood ESR1 mutation detection at regular intervals. Patients for whom a rising circulating ESR1 mutation is detected without tumour progression (up to N=200) will be randomised (1:1) between (1) Arm A: no modification of therapy; and (2) Arm B: palbociclib in combination with fulvestrant, a selective ER down-regulator. At tumour progression, an optional crossover will be offered to patients randomised in arm A. The coprimary endpoints are (1) Grade ≥3 haematological toxicities and their associations with baseline characteristics and (2) progression-free survival in randomised patients. ETHICS AND DISSEMINATION The study has been approved by the French medicines agency (ANSM) and by an ethics committee (ref 01/17_1 CPP Ouest-IV Nantes) in January 2017. The trial results will be published in academic conference presentations and international peer-reviewed journals. TRIAL REGISTRATION NUMBERS EudraCT: 2016-004360-18; NCT03079011.
Collapse
Affiliation(s)
- Frédérique Berger
- Biometry Unit, Institut Curie, PSL Research University, Paris and Saint-Cloud, France
| | - Margaux Marce
- Biometry Unit, Data Center, Institut Régional du Cancer de Montpellier, Montpellier, France
| | | | | | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Ivan Bièche
- Pharmacogenomic Unit, Genetics laboratory, Department of Diagnostic and Theranostic Medicine, Institut Curie and PSL University, Paris, France
| | - Anne Pradines
- INSERM U1037 CNRS ERL5294 UPS, Cancer Research Center of Toulouse, Toulouse, France
- Prospective Biology Unit, Medical Laboratory, Claudius Regaud Institute, Toulouse University Cancer Institute (IUCT-O), Toulouse, France
| | | | - Jean-Luc Canon
- Department of Medical Oncology, Grand Hôpital de Charleroi, Charleroi, Belgique
| | - Fabrice André
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Laurent Arnould
- Department of Pathology, Centre Georges François Leclerc, Dijon, France
| | - Florian Clatot
- Department of Medical Oncology, Centre Henri Becquerel, Rouen, France
| | | | | | - François-Clement Bidard
- Department of Medical Oncology, Institut Curie, UVSQ/Paris Saclay University, Saint Cloud, France
- Circulating Tumor Biomarkers laboratory, Inserm CIC-BT 1428, Institut Curie, Paris, France
| |
Collapse
|
10
|
Tivey A, Britton F, Scott JA, Rothwell D, Lorigan P, Lee R. Circulating Tumour DNA in Melanoma-Clinic Ready? Curr Oncol Rep 2022; 24:363-373. [PMID: 35133615 PMCID: PMC8885536 DOI: 10.1007/s11912-021-01151-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2021] [Indexed: 01/05/2023]
Abstract
PURPOSE OF REVIEW Liquid biopsies, including circulating tumour DNA (ctDNA), can inform a variety of clinical questions. This review examines the potential role of ctDNA as a clinical tool to inform clinical decision-making from early to late stage cutaneous melanoma. RECENT FINDINGS In pre-clinical studies, ctDNA has been shown to detect minimal residual disease and molecular relapse; predict and monitor response to therapy; and identify key resistance mechanisms. Here, we examine the potential utility of ctDNA and discuss its limitations for use in patients with melanoma. We present novel clinical trials, which are testing its value as a tool to augment clinical decision-making. Finally, we discuss the steps that are needed to ensure that ctDNA is used optimally in order to improve outcomes for patients with melanoma. Preclinical studies have shown that ctDNA has huge potential to provide real-time information about disease status in patients with melanoma. It is now time to test it rigorously within clinical trials to assess how it can be optimally used to benefit patients in the clinic.
Collapse
Affiliation(s)
- Ann Tivey
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- Division of Cancer Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Fiona Britton
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Julie-Ann Scott
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
| | - Dominic Rothwell
- Division of Cancer Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Nucleic Acids Biomarker Team, Cancer Research UK Manchester Institute, Cancer Biomarker Centre, The University of Manchester, Alderley Park, SK10 4TG, UK
| | - Paul Lorigan
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK
- Division of Cancer Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Rebecca Lee
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK.
- Division of Cancer Sciences, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester, M20 4BX, UK.
| |
Collapse
|
11
|
Kalita-de Croft P, Joshi V, Saunus JM, Lakhani SR. Emerging Biomarkers for Diagnosis, Prevention and Treatment of Brain Metastases-From Biology to Clinical Utility. Diseases 2022; 10:11. [PMID: 35225863 PMCID: PMC8884016 DOI: 10.3390/diseases10010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/18/2022] [Accepted: 01/27/2022] [Indexed: 11/17/2022] Open
Abstract
Primary malignancies of the lung, skin (melanoma), and breast have higher propensity for metastatic spread to the brain. Advances in molecular tumour profiling have aided the development of targeted therapies, stereotactic radiotherapy, and immunotherapy, which have led to some improvement in patient outcomes; however, the overall prognosis remains poor. Continued research to identify new prognostic and predictive biomarkers is necessary to further impact patient outcomes, as this will enable better risk stratification at the point of primary cancer diagnosis, earlier detection of metastatic deposits (for example, through surveillance), and more effective systemic treatments. Brain metastases exhibit considerable inter- and intratumoural heterogeneity-apart from distinct histology, treatment history and other clinical factors, the metastatic brain tumour microenvironment is incredibly variable both in terms of subclonal diversity and cellular composition. This review discusses emerging biomarkers; specifically, the biological context and potential clinical utility of tumour tissue biomarkers, circulating tumour cells, extracellular vesicles, and circulating tumour DNA.
Collapse
Affiliation(s)
- Priyakshi Kalita-de Croft
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Vaibhavi Joshi
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Jodi M. Saunus
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
| | - Sunil R. Lakhani
- UQ Centre for Clinical Research, The University of Queensland Faculty of Medicine, Herston, QLD 4029, Australia; (V.J.); (J.M.S.)
- Pathology Queensland, The Royal Brisbane and Women’s Hospital Herston, Herston, QLD 4029, Australia
| |
Collapse
|
12
|
Stadler JC, Belloum Y, Deitert B, Sementsov M, Heidrich I, Gebhardt C, Keller L, Pantel K. Current and Future Clinical Applications of ctDNA in Immuno-Oncology. Cancer Res 2022; 82:349-358. [PMID: 34815256 PMCID: PMC9397642 DOI: 10.1158/0008-5472.can-21-1718] [Citation(s) in RCA: 89] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 10/06/2021] [Accepted: 11/09/2021] [Indexed: 01/07/2023]
Abstract
Testing peripheral blood for circulating tumor DNA (ctDNA) offers a minimally invasive opportunity to diagnose, characterize, and monitor the disease in individual cancer patients. ctDNA can reflect the actual tumor burden and specific genomic state of disease and thus might serve as a prognostic and predictive biomarker for immune checkpoint inhibitor (ICI) therapy. Recent studies in various cancer entities (e.g., melanoma, non-small cell lung cancer, colon cancer, and urothelial cancer) have shown that sequential ctDNA analyses allow for the identification of responders to ICI therapy, with a significant lead time to imaging. ctDNA assessment may also help distinguish pseudoprogression under ICI therapy from real progression. Developing dynamic changes in ctDNA concentrations as a potential surrogate endpoint of clinical efficacy in patients undergoing adjuvant immunotherapy is ongoing. Besides overall ctDNA burden, further ctDNA characterization can help uncover tumor-specific determinants (e.g., tumor mutational burden and microsatellite instability) of responses or resistance to immunotherapy. In future studies, standardized ctDNA assessments need to be included in interventional clinical trials across cancer entities to demonstrate the clinical utility of ctDNA as a biomarker for personalized cancer immunotherapy.
Collapse
Affiliation(s)
- Julia-Christina Stadler
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yassine Belloum
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Deitert
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mark Sementsov
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabel Heidrich
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Keller
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Corresponding Authors: Klaus Pantel, Institute for Tumor Biologie, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, Hamburg, 20246, Germany. E-mail: ; and Laura Keller, E-mail:
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,Corresponding Authors: Klaus Pantel, Institute for Tumor Biologie, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, Hamburg, Hamburg, 20246, Germany. E-mail: ; and Laura Keller, E-mail:
| |
Collapse
|
13
|
Gracie L, Pan Y, Atenafu EG, Ward DG, Teng M, Pallan L, Stevens NM, Khoja L. Circulating tumour DNA (ctDNA) in metastatic melanoma, a systematic review and meta-analysis. Eur J Cancer 2021; 158:191-207. [PMID: 34757258 DOI: 10.1016/j.ejca.2021.09.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 08/31/2021] [Accepted: 09/16/2021] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Circulating tumour DNA (ctDNA) is an emerging biomarker in melanoma. We performed a systematic review and meta-analysis to explore its clinical utility as a prognostic, pharmacodynamic (PD) and predictive biomarker. METHODS A systematic search was conducted from Jan 2015 to April 2021, of the electronic databases PubMed, Cochrane Library and Ovid MEDLINE to identify studies. Studies were restricted to those published in English within the last 5 years, evaluating ctDNA in humans in ≥10 patients. Survival data were extracted for meta-analysis using pooled treatment effect (TE), i.e. log hazard ratios (HRs) and corresponding standard error of TE for progression-free survival or overall survival differences in patients who were ctDNA positive or negative. PRISMA statement guidelines were followed. RESULTS A meta-analysis of 19 studies grouped according to methodology of ctDNA detection, revealed a combined estimate for HR of progression-free survival (13 studies using droplet digital Polymerase Chain Reaction (ddPCR) methodology (N = 1002) of 2.10 (95% CI: 1.71-2.59) revealing a poorer prognosis when ctDNA was detected. This result was confirmed in the smaller analysis of (non-ddPCR, N = 347) five studies: HR = 2.45 (95% CI: 1.29-4.63). Similar findings were found in the overall survival analysis of nine studies (ddPCR methodology, N = 841) where the combined HR was 2.78 (95% CI: 2.21-3.49) and of the five studies (non-ddPCR methodology, N = 326) where the combined HR was 2.58 (95% CI: 1.74-3.84). Serial ctDNA levels on treatment showed a pharmacodynamic role reflecting response or resistance earlier than radiological assessment. CONCLUSIONS Circulating tumour DNA is a predictive, prognostic and PD biomarker in melanoma. Technical standardisation of assays is required before clinical adoption.
Collapse
Affiliation(s)
- Lara Gracie
- College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Yi Pan
- Centre for Computational Biology and Institute of Immunology and Immunotherapy, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Eshetu G Atenafu
- Biostatistics Department, Princess Margaret Cancer Center, University Health Network, University Way, Toronto, Ontario, Canada
| | - Douglas G Ward
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Mabel Teng
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Department of Oncology, Birmingham B15 2TH, UK
| | - Lallit Pallan
- University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital, Department of Oncology, Birmingham B15 2TH, UK
| | - Neil M Stevens
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - Leila Khoja
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
14
|
Kamińska P, Buszka K, Zabel M, Nowicki M, Alix-Panabières C, Budna-Tukan J. Liquid Biopsy in Melanoma: Significance in Diagnostics, Prediction and Treatment Monitoring. Int J Mol Sci 2021; 22:9714. [PMID: 34575876 PMCID: PMC8468624 DOI: 10.3390/ijms22189714] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/25/2021] [Accepted: 09/06/2021] [Indexed: 02/06/2023] Open
Abstract
Liquid biopsy is a common term referring to circulating tumor cells and other biomarkers, such as circulating tumor DNA (ctDNA) or extracellular vesicles. Liquid biopsy presents a range of clinical advantages, such as the low invasiveness of the blood sample collection and continuous control of the tumor progression. In addition, this approach enables the mechanisms of drug resistance to be determined in various methods of cancer treatment, including immunotherapy. However, in the case of melanoma, the application of liquid biopsy in patient stratification and therapy needs further investigation. This review attempts to collect all of the relevant and recent information about circulating melanoma cells (CMCs) related to the context of malignant melanoma and immunotherapy. Furthermore, the biology of liquid biopsy analytes, including CMCs, ctDNA, mRNA and exosomes, as well as techniques for their detection and isolation, are also described. The available data support the notion that thoughtful selection of biomarkers and technologies for their detection can contribute to the development of precision medicine by increasing the efficacy of cancer diagnostics and treatment.
Collapse
Affiliation(s)
- Paula Kamińska
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.K.); (K.B.); (M.N.)
| | - Karolina Buszka
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.K.); (K.B.); (M.N.)
| | - Maciej Zabel
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Góra, 65-046 Zielona Góra, Poland;
| | - Michał Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.K.); (K.B.); (M.N.)
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, 34093 Montpellier, France;
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, IRD, 34000 Montpellier, France
| | - Joanna Budna-Tukan
- Department of Histology and Embryology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (P.K.); (K.B.); (M.N.)
| |
Collapse
|
15
|
Lin C, Liu X, Zheng B, Ke R, Tzeng CM. Liquid Biopsy, ctDNA Diagnosis through NGS. Life (Basel) 2021; 11:life11090890. [PMID: 34575039 PMCID: PMC8468354 DOI: 10.3390/life11090890] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/08/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022] Open
Abstract
Liquid biopsy with circulating tumor DNA (ctDNA) profiling by next-generation sequencing holds great promise to revolutionize clinical oncology. It relies on the basis that ctDNA represents the real-time status of the tumor genome which contains information of genetic alterations. Compared to tissue biopsy, liquid biopsy possesses great advantages such as a less demanding procedure, minimal invasion, ease of frequent sampling, and less sampling bias. Next-generation sequencing (NGS) methods have come to a point that both the cost and performance are suitable for clinical diagnosis. Thus, profiling ctDNA by NGS technologies is becoming more and more popular since it can be applied in the whole process of cancer diagnosis and management. Further developments of liquid biopsy ctDNA testing will be beneficial for cancer patients, paving the way for precision medicine. In conclusion, profiling ctDNA with NGS for cancer diagnosis is both biologically sound and technically convenient.
Collapse
Affiliation(s)
- Chen Lin
- School of Medicine, Huaqiao University, Quanzhou 362021, China; (C.L.); (X.L.)
| | - Xuzhu Liu
- School of Medicine, Huaqiao University, Quanzhou 362021, China; (C.L.); (X.L.)
| | - Bingyi Zheng
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China;
- Xiamen Key Laboratory of Cancer Cell Theranostics and Clinical Translation, Xiamen 361102, China
| | - Rongqin Ke
- School of Medicine, Huaqiao University, Quanzhou 362021, China; (C.L.); (X.L.)
- Correspondence: (R.K.); (C.-M.T.)
| | - Chi-Meng Tzeng
- Translational Medicine Research Center, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China;
- Xiamen Key Laboratory of Cancer Cell Theranostics and Clinical Translation, Xiamen 361102, China
- Correspondence: (R.K.); (C.-M.T.)
| |
Collapse
|
16
|
Tolmeijer SH, Koornstra RHT, de Groot JWB, Geerlings MJ, van Rens DH, Boers-Sonderen MJ, Schalken JA, Gerritsen WR, Ligtenberg MJL, Mehra N. Plasma BRAF Mutation Detection for the Diagnostic and Monitoring Trajectory of Patients with LDH-High Stage IV Melanoma. Cancers (Basel) 2021; 13:3913. [PMID: 34359813 PMCID: PMC8345527 DOI: 10.3390/cancers13153913] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 01/16/2023] Open
Abstract
For patients with newly diagnosed metastatic melanoma, rapid BRAF mutation (mBRAF) assessment is vital to promptly initiate systemic therapy. Additionally, blood-based biomarkers are desired to monitor and predict treatment response. Circulating tumor DNA (ctDNA) has shown great promise for minimally invasive mBRAF assessment and treatment monitoring, but validation studies are needed. This prospective study utilized longitudinal plasma samples at regular timepoints (0, 6, 12, 18 weeks) to address the clinical validity of ctDNA measurements in stage IV melanoma patients with elevated serum lactate dehydrogenase (LDH > 250U/L) starting first-line systemic treatment. Using droplet digital PCR, the plasma mBRAF abundance was assessed in 53 patients with a BRAFV600 tissue mutation. Plasma mBRAF was detected in 50/51 patients at baseline (98% sensitivity; median fraction abundance of 19.5%) and 0/17 controls (100% specificity). Patients in whom plasma mBRAF became undetectable during the first 12-18 weeks of treatment had a longer progression-free survival (30.2 vs. 4.0 months; p < 0.001) and cancer-specific survival (not reached vs. 10.2 months; p < 0.001) compared to patients with detectable mBRAF. The ctDNA dynamics outperformed LDH and S100 dynamics. These results confirm the clinical validity of ctDNA measurements as a minimally invasive biomarker for the diagnostic and monitoring trajectory of patients with LDH-high stage IV melanoma.
Collapse
Affiliation(s)
- Sofie H. Tolmeijer
- Department of Medical Oncology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Rutger H. T. Koornstra
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
- Department of Medical Oncology, Rijnstate Hospital, 6815 AD Arnhem, The Netherlands
| | | | - Maartje J. Geerlings
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (M.J.G.); (M.J.L.L.)
| | - Dirk H. van Rens
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
| | - Marye J. Boers-Sonderen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
| | - Jack A. Schalken
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands;
| | - Winald R. Gerritsen
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
| | - Marjolijn J. L. Ligtenberg
- Department of Human Genetics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (M.J.G.); (M.J.L.L.)
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (R.H.T.K.); (D.H.v.R.); (M.J.B.-S.); (W.R.G.)
| |
Collapse
|
17
|
Longitudinal Relationship between Idylla Plasma ctBRAF V600 Mutation Detection and Tumor Burden in Patients with Metastatic Melanoma. Mol Diagn Ther 2021; 25:361-371. [PMID: 33970440 DOI: 10.1007/s40291-021-00528-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Circulating tumor DNA (ctDNA) may complement radiography for interim assessment of patients with cancer. OBJECTIVE Our objective was to explore the relationship between changes in plasma ctDNA versus radiographic imaging among patients with metastatic melanoma. METHODS Using the Idylla system, we measured B-Raf proto-oncogene (BRAF) V600 ctDNA in plasma from 15 patients with BRAF V600E/K-positive primary tumors undergoing standard-of-care monitoring, including cross-sectional computed tomography (CT) imaging. BRAF V600 mutant allele frequency (%MAF) was calculated from the Idylla Cq values and directly measured using droplet digital polymerase chain reaction (ddPCR). RESULTS The Idylla ctDNA assay demonstrated 91% sensitivity, 96% specificity, 91% positive predictive value, and 96% negative predictive value for the presence of > 93 mm metastatic disease. Qualitative ctDNA results corresponded to changes in RECIST (Response Evaluation Criteria in Solid Tumors) 1.1 status determined by CT imaging in 11 of 15 subjects (73%). Calculated %MAF results correlated with ddPCR (R2 = 0.94) and provided evidence of progressive disease 55 and 97 days in advance of CT imaging for two subjects with persistently positive qualitative results. CONCLUSIONS Overall, interim ctDNA results provided evidence of partial response or progressive disease an average of 82 days before radiography. This pilot study supports the feasibility of using the Idylla plasma BRAF V600 ctDNA assay as a complement to CT scanning for routine monitoring of therapeutic response. Somatic mutation quantification based on Cq values shows promise for identifying disease progression and warrants further validation.
Collapse
|
18
|
Zhang Y, Han X, Shao Y. The ROC of Cox proportional hazards cure models with application in cancer studies. LIFETIME DATA ANALYSIS 2021; 27:195-215. [PMID: 33507457 DOI: 10.1007/s10985-021-09516-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 01/13/2021] [Indexed: 06/12/2023]
Abstract
With recent advancement in cancer screening and treatment, many patients with cancers are identified at early stage and clinically cured. Importantly, uncured patients should be treated timely before the cancer progresses to advanced stages for which therapeutic options are rather limited. It is also crucial to identify uncured subjects among patients with early-stage cancers for clinical trials to develop effective adjuvant therapies. Thus, it is of interest to develop statistical predictive models with as high accuracy as possible in predicting the latent cure status. The receiver operating characteristic curve (ROC) and the area under the ROC curve (AUC) are among the most widely used statistical metrics for assessing predictive accuracy or discriminatory power for a dichotomous outcome (cured/uncured). Yet the conventional AUC cannot be directly used due to incompletely observed cure status. In this article, we proposed new estimates of the ROC curve and its AUC for predicting latent cure status in Cox proportional hazards (PH) cure models and transformation cure models. We developed explicit formulas to estimate sensitivity, specificity, the ROC and its AUC without requiring to know the patient cure status. We also developed EM type estimates to approximate sensitivity, specificity, ROC and AUC conditional on observed data. Numerical studies were used to assess their finite-sample performance of the proposed methods. Both methods are consistent and have similar efficiency as shown in our numerical studies. A melanoma dataset was used to demonstrate the utility of the proposed estimates of the ROC curve for the latent cure status. We also have developed an [Formula: see text] package called [Formula: see text] to efficiently compute the proposed estimates.
Collapse
Affiliation(s)
- Yilong Zhang
- Department of Biostatistics and Research Decision Sciences, Merck & Co., Inc, Kenilworth, NJ, USA
| | - Xiaoxia Han
- Department of Public Health Sciences, Henry Ford Health System, Detroit, MI, USA
| | - Yongzhao Shao
- Departments of Population Health & Environmental Medicine, NYU Grossman School of Medicine, 180 Madison Ave, 4th Floor, Suite 455, New York, NY, 10016, USA.
| |
Collapse
|
19
|
Cheng ML, Pectasides E, Hanna GJ, Parsons HA, Choudhury AD, Oxnard GR. Circulating tumor DNA in advanced solid tumors: Clinical relevance and future directions. CA Cancer J Clin 2021; 71:176-190. [PMID: 33165928 DOI: 10.3322/caac.21650] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/21/2020] [Accepted: 09/14/2020] [Indexed: 02/06/2023] Open
Abstract
The application of genomic profiling assays using plasma circulating tumor DNA (ctDNA) is rapidly evolving in the management of patients with advanced solid tumors. Diverse plasma ctDNA technologies in both commercial and academic laboratories are in routine or emerging use. The increasing integration of such testing to inform treatment decision making by oncology clinicians has complexities and challenges but holds significant potential to substantially improve patient outcomes. In this review, the authors discuss the current role of plasma ctDNA assays in oncology care and provide an overview of ongoing research that may inform real-world clinical applications in the near future.
Collapse
Affiliation(s)
- Michael L Cheng
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Eirini Pectasides
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Glenn J Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Heather A Parsons
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Atish D Choudhury
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey R Oxnard
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
20
|
He X, Sun X, Shao Y. Network-based survival analysis to discover target genes for developing cancer immunotherapies and predicting patient survival. J Appl Stat 2021; 48:1352-1373. [PMID: 35444359 DOI: 10.1080/02664763.2020.1812543] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recently, cancer immunotherapies have been life-savers, however, only a fraction of treated patients have durable responses. Consequently, statistical methods that enable the discovery of target genes for developing new treatments and predicting patient survival are of importance. This paper introduced a network-based survival analysis method and applied it to identify candidate genes as possible targets for developing new treatments. RNA-seq data from a mouse study was used to select differentially expressed genes, which were then translated to those in humans. We constructed a gene network and identified gene clusters using a training set of 310 human gliomas. Then we conducted gene set enrichment analysis to select the gene clusters with significant biological function. A penalized Cox model was built to identify a small set of candidate genes to predict survival. An independent set of 690 human glioma samples was used to evaluate predictive accuracy of the survival model. The areas under time-dependent ROC curves in both the training and validation sets are more than 90%, indicating strong association between selected genes and patient survival. Consequently, potential biomedical interventions targeting these genes might be able to alter their expressions and prolong patient survival.
Collapse
|
21
|
Pilla L, Alberti A, Di Mauro P, Gemelli M, Cogliati V, Cazzaniga ME, Bidoli P, Maccalli C. Molecular and Immune Biomarkers for Cutaneous Melanoma: Current Status and Future Prospects. Cancers (Basel) 2020; 12:E3456. [PMID: 33233603 PMCID: PMC7699774 DOI: 10.3390/cancers12113456] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 12/18/2022] Open
Abstract
Advances in the genomic, molecular and immunological make-up of melanoma allowed the development of novel targeted therapy and of immunotherapy, leading to changes in the paradigm of therapeutic interventions and improvement of patients' overall survival. Nevertheless, the mechanisms regulating either the responsiveness or the resistance of melanoma patients to therapies are still mostly unknown. The development of either the combinations or of the sequential treatment of different agents has been investigated but without a strongly molecularly motivated rationale. The need for robust biomarkers to predict patients' responsiveness to defined therapies and for their stratification is still unmet. Progress in immunological assays and genomic techniques as long as improvement in designing and performing studies monitoring the expression of these markers along with the evolution of the disease allowed to identify candidate biomarkers. However, none of them achieved a definitive role in predicting patients' clinical outcomes. Along this line, the cross-talk of melanoma cells with tumor microenvironment plays an important role in the evolution of the disease and needs to be considered in light of the role of predictive biomarkers. The overview of the relationship between the molecular basis of melanoma and targeted therapies is provided in this review, highlighting the benefit for clinical responses and the limitations. Moreover, the role of different candidate biomarkers is described together with the technical approaches for their identification. The provided evidence shows that progress has been achieved in understanding the molecular basis of melanoma and in designing advanced therapeutic strategies. Nevertheless, the molecular determinants of melanoma and their role as biomarkers predicting patients' responsiveness to therapies warrant further investigation with the vision of developing more effective precision medicine.
Collapse
Affiliation(s)
- Lorenzo Pilla
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Andrea Alberti
- Medical Oncology Unit, Department of Medical and Surgical Specialties, Radiological Health Science and Public Health, University of Brescia, ASST Ospedali Civili, 25123 Brescia, Italy;
| | - Pierluigi Di Mauro
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Maria Gemelli
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Viola Cogliati
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Marina Elena Cazzaniga
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Paolo Bidoli
- Division of Medical Oncology, San Gerardo Hospital, University of Milano-Bicocca School of Medicine, 20900 Monza, Italy; (P.D.M.); (M.G.); (V.C.); (M.E.C.); (P.B.)
| | - Cristina Maccalli
- Laboratory of Immune and Biological Therapy, Research Department, Sidra Medicine, Doha 26999, Qatar;
| |
Collapse
|
22
|
Marczynski GT, Laus AC, Dos Reis MB, Reis RM, Vazquez VDL. Circulating tumor DNA (ctDNA) detection is associated with shorter progression-free survival in advanced melanoma patients. Sci Rep 2020; 10:18682. [PMID: 33122747 PMCID: PMC7596487 DOI: 10.1038/s41598-020-75792-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 10/20/2020] [Indexed: 02/07/2023] Open
Abstract
BRAF, NRAS and TERT mutations occur in more than 2/3 of melanomas. Its detection in patient's blood, as circulating tumor DNA (ctDNA), represents a possibility for identification and monitoring of metastatic disease. We proposed to standardize a liquid biopsy platform to identify hotspot mutations in BRAF, NRAS and TERT in plasma samples from advanced melanoma patients and investigate whether it was associated to clinical outcome. Firstly, we performed digital polymerase chain reaction using tumor cell lines for validation and determination of limit of detection (LOD) of each assay and screened plasma samples from healthy individuals to determine the limit of blank (LOB). Then, we selected 19 stage III and IV patients and determined the somatic mutations status in tumor tissue and track them in patients' plasma. We established a specific and sensitive methodology with a LOD ranging from 0.13 to 0.37%, and LOB ranging from of 0 to 5.201 copies/reaction. Somatic mutations occurred in 17/19 (89%) patients, of whom seven (41%) had ctDNA detectable their paired plasma. ctDNA detection was associated with shorter progression free survival (p = 0.01). In conclusion, our data support the use of ctDNA as prognosis biomarker, suggesting that patients with detectable levels have an unfavorable outcome.
Collapse
Affiliation(s)
- Gabriella Taques Marczynski
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela St, Barretos, SP, 14784-400, Brazil
| | - Ana Carolina Laus
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela St, Barretos, SP, 14784-400, Brazil
| | - Mariana Bisarro Dos Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela St, Barretos, SP, 14784-400, Brazil
| | - Rui Manuel Reis
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela St, Barretos, SP, 14784-400, Brazil
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Vinicius de Lima Vazquez
- Molecular Oncology Research Center, Barretos Cancer Hospital, 1331, Antenor Duarte Villela St, Barretos, SP, 14784-400, Brazil.
- Surgery Department of Melanoma, Sarcoma and Mesenchymal Tumors, Barretos Cancer Hospital, Barretos, Brazil.
- Barretos School of Health Sciences Dr. Paulo Prata - FACISB, Barretos, Brazil.
| |
Collapse
|
23
|
Sacco A, Forgione L, Carotenuto M, De Luca A, Ascierto PA, Botti G, Normanno N. Circulating Tumor DNA Testing Opens New Perspectives in Melanoma Management. Cancers (Basel) 2020; 12:E2914. [PMID: 33050536 PMCID: PMC7601606 DOI: 10.3390/cancers12102914] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/06/2020] [Accepted: 10/08/2020] [Indexed: 12/21/2022] Open
Abstract
Malignant melanoma accounts for about 1% of all skin cancers, but it causes most of the skin cancer-related deaths. Circulating tumor DNA (ctDNA) testing is emerging as a relevant tool for the diagnosis and monitoring of cancer. The availability of highly sensitive techniques, including next generation sequencing (NGS)-based panels, has increased the fields of application of ctDNA testing. While ctDNA-based tests for the early detection of melanoma are not available yet, perioperative ctDNA analysis in patients with surgically resectable melanoma offers relevant prognostic information: i) the detection of ctDNA before surgery correlates with the extent and the aggressiveness of the disease; ii) ctDNA testing after surgery/adjuvant therapy identifies minimal residual disease; iii) testing ctDNA during the follow-up can detect a tumor recurrence, anticipating clinical/radiological progression. In patients with advanced melanoma, several studies have demonstrated that the analysis of ctDNA can better depict tumor heterogeneity and provides relevant prognostic information. In addition, ctDNA testing during treatment allows assessing the response to systemic therapy and identifying resistance mechanisms. Although validation in prospective clinical trials is needed for most of these approaches, ctDNA testing opens up new scenarios in the management of melanoma patients that could lead to improvements in the diagnosis and therapy of this disease.
Collapse
Affiliation(s)
- Alessandra Sacco
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.S.); (L.F.); (M.C.); (A.D.L.)
| | - Laura Forgione
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.S.); (L.F.); (M.C.); (A.D.L.)
| | - Marianeve Carotenuto
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.S.); (L.F.); (M.C.); (A.D.L.)
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.S.); (L.F.); (M.C.); (A.D.L.)
| | - Paolo A. Ascierto
- Department of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Napoli, Italy;
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori IRCCS Fondazione Pascale, 80131 Napoli, Italy;
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (A.S.); (L.F.); (M.C.); (A.D.L.)
| |
Collapse
|
24
|
Kozak K, Kowalik A, Gos A, Wasag B, Lugowska I, Jurkowska M, Krawczynska N, Kosela-Paterczyk H, Switaj T, Teterycz P, Klimczak A, Siedlecki JA, Chlopek M, Kalisz J, Limon J, Rutkowski P. Cell-free DNA BRAF V600E measurements during BRAF inhibitor therapy of metastatic melanoma: long-term analysis. TUMORI JOURNAL 2020; 106:300891619900928. [PMID: 32026754 DOI: 10.1177/0300891619900928] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVE We assessed the status of the BRAF V600E mutation in cell-free circulating tumor DNA (cfDNA) isolated from the plasma of patients with metastatic melanoma treated with the BRAF inhibitor vemurafenib, collected at different time points during therapy to evaluate the sensitivity and specificity of quantitative polymerase chain reaction and droplet digital polymerase chain reaction (ddPCR) and the correlation between the level of plasma cfDNA p.V600E and the long-term clinical outcome. METHODS cfDNA in patients with BRAF-mutated melanoma (n = 62) was analyzed at baseline and at 4-8 weeks from the start of vemurafenib therapy. BRAF mutations were assessed using tumor tissue-derived DNA and circulating cfDNA from plasma samples. Quantification of BRAF V600E was performed in cfDNA using ddPCR. RESULTS cfDNA V600E was detected in the plasma of 48/62 (77%) patients at baseline and in 18/62 (29%) patients after 4-8 weeks of treatment. Patients positive for BRAF mutations in cfDNA at baseline had shorter progression-free survival (PFS) and overall survival (OS) compared with patients with undetectable cfDNA BRAF mutations. Undetectable cfDNA p.V600E at baseline and after 4-8 weeks of therapy was associated with the best prognosis. When treated as a continuous variable, the log-transformed concentration of baseline cfDNA p.V600E was significantly associated with both PFS and OS. This effect was retained in the multivariate OS Cox model adjusted for Eastern Cooperative Oncology Group performance status, the presence of brain metastases, patient age, and previous systemic treatment. CONCLUSIONS Monitoring of plasma BRAF p.V600E cfDNA concentrations in patients with metastatic melanoma on targeted therapy may have prognostic value. Undetectable cfDNA p.V600E before and during treatment was associated with a favorable prognosis.
Collapse
Affiliation(s)
- Katarzyna Kozak
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, Kielce, Poland
| | - Aleksandra Gos
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Bartosz Wasag
- Department of Molecular Biology, Medical University of Gdansk, Gdansk, Poland
| | - Iwona Lugowska
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | | | - Natalia Krawczynska
- Department of Molecular Biology, Medical University of Gdansk, Gdansk, Poland
| | - Hanna Kosela-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Tomasz Switaj
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Paweł Teterycz
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Anna Klimczak
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Janusz A Siedlecki
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| | - Małgorzata Chlopek
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, Kielce, Poland
| | - Joanna Kalisz
- Department of Molecular Diagnostics, Holy Cross Cancer Centre, Kielce, Poland
| | - Janusz Limon
- Department of Molecular Biology, Medical University of Gdansk, Gdansk, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Institute Oncology Center, Warsaw, Poland
| |
Collapse
|
25
|
Said R, Guibert N, Oxnard GR, Tsimberidou AM. Circulating tumor DNA analysis in the era of precision oncology. Oncotarget 2020; 11:188-211. [PMID: 32010431 PMCID: PMC6968778 DOI: 10.18632/oncotarget.27418] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 12/16/2019] [Indexed: 12/13/2022] Open
Abstract
The spatial and temporal genomic heterogeneity of various tumor types and advances in technology have stimulated the development of circulating tumor DNA (ctDNA) genotyping. ctDNA was developed as a non-invasive, cost-effective alternative to tumor biopsy when such biopsy is associated with significant risk, when tumor tissue is insufficient or inaccessible, and/or when repeated assessment of tumor molecular abnormalities is needed to optimize treatment. The role of ctDNA is now well established in the clinical decision in certain alterations and tumors, such as the epidermal growth factor receptor (EGFR) mutation in non-small cell lung cancer and the v-Ki-ras2 kirsten rat sarcoma viral oncogene homolog (KRAS) mutation in colorectal cancer. The role of ctDNA analysis in other tumor types remains to be validated. Evolving data indicate the association of ctDNA level with tumor burden, and the usefulness of ctDNA analysis in assessing minimal residual disease, in understanding mechanisms of resistance to treatment, and in dynamically guiding therapy. ctDNA analysis is increasingly used to select therapy. Carefully designed clinical trials that use ctDNA analysis will increase the rate of patients who receive targeted therapy, will elucidate our understanding of evolution of tumor biology and will accelerate drug development and implementation of precision medicine. In this article we provide a critical overview of clinical trials and evolving data of ctDNA analysis in specific tumors and across tumor types.
Collapse
Affiliation(s)
- Rabih Said
- Department of Investigational Cancer Therapeutics, Phase I Clinical Trials Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, St. George Hospital University Medical Center, University of Balamand, Beirut, Lebanon
- Co-authorship
| | - Nicolas Guibert
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Thoracic Oncology, Toulouse University Hospital, Toulouse, France
- Co-authorship
| | - Geoffrey R. Oxnard
- Lowe Center for Thoracic Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Apostolia M. Tsimberidou
- Department of Investigational Cancer Therapeutics, Phase I Clinical Trials Program, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
26
|
Herbreteau G, Charpentier S, Vallée A, Denis MG. Use of circulating tumoral DNA to guide treatment for metastatic melanoma. Pharmacogenomics 2019; 20:1259-1270. [DOI: 10.2217/pgs-2019-0097] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The management of metastatic cutaneous melanoma is conditioned by the identification of BRAF-activating mutations in tumor DNA. Tumor genotyping is usually performed on DNA extracted from tissue samples. However, these invasive samples are rarely repeated during follow-up, and their analysis requires a sample pre-treatment which may take several weeks. Circulating tumor DNA (ctDNA), released into blood by cancer cells, is a good alternative to tissue sampling. ctDNA is not subject to tumor heterogeneity, and can be analyzed rapidly, making possible the detection of mutations in emergency or in patients whose tumor cannot be sampled. ctDNA can also be analyzed repeatedly during follow-up, for postresection minimal residual disease assessment, for therapeutic response monitoring and for early relapse detection.
Collapse
Affiliation(s)
- Guillaume Herbreteau
- Laboratoire de Biochimie et Plateforme de Génétique Moléculaire des Cancers, CHU Nantes, Nantes, France
- Centre de Recherche en Cancérologie et Immunologie, CRCINA, INSERM U1232, Nantes, France
| | - Sandrine Charpentier
- Laboratoire de Biochimie et Plateforme de Génétique Moléculaire des Cancers, CHU Nantes, Nantes, France
- Centre de Recherche en Cancérologie et Immunologie, CRCINA, INSERM U1232, Nantes, France
| | - Audrey Vallée
- Laboratoire de Biochimie et Plateforme de Génétique Moléculaire des Cancers, CHU Nantes, Nantes, France
- Centre de Recherche en Cancérologie et Immunologie, CRCINA, INSERM U1232, Nantes, France
| | - Marc G Denis
- Laboratoire de Biochimie et Plateforme de Génétique Moléculaire des Cancers, CHU Nantes, Nantes, France
- Centre de Recherche en Cancérologie et Immunologie, CRCINA, INSERM U1232, Nantes, France
| |
Collapse
|
27
|
Forthun RB, Hovland R, Schuster C, Puntervoll H, Brodal HP, Namløs HM, Aasheim LB, Meza-Zepeda LA, Gjertsen BT, Knappskog S, Straume O. ctDNA detected by ddPCR reveals changes in tumour load in metastatic malignant melanoma treated with bevacizumab. Sci Rep 2019; 9:17471. [PMID: 31767937 PMCID: PMC6877652 DOI: 10.1038/s41598-019-53917-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 11/05/2019] [Indexed: 12/26/2022] Open
Abstract
Bevacizumab is included in an increasing number of clinical trials. To find biomarkers to predict and monitor treatment response, cancer and angiogenesis relevant mutations in tumour and circulating tumour DNA (ctDNA) were investigated in 26 metastatic melanoma patients treated with bevacizumab. Patients with >1% BRAF/NRAS ctDNA at treatment start had significantly decreased progression free survival (PFS) and overall survival (OS) (PFS: p = 0.019, median 54 vs 774 days, OS: p = 0.026, median 209 vs 1064 days). Patients with >1% BRAF/NRAS ctDNA during treatment showed similar results (PFS: p = 0.002, OS: p = 0.003). ≤1% BRAF/NRAS ctDNA and normal lactate dehydrogenase (LDH) levels both significantly predicted increased response to treatment, but BRAF/NRAS ctDNA was better at predicting response compared to LDH at treatment start (OR 16.94, p = 0.032 vs OR 4.57, p = 0.190), and at predicting PFS (HR 6.76, p = 0.002) and OS (HR 6.78, p = 0.002) during therapy. ctDNA BRAF p.V600D/E/K and NRAS p.G12V/p.Q61K/L/R were better biomarkers for response prediction than TERT promoter mutations (OR 1.50, p = 0.657). Next generation sequencing showed that all patients with ≥2 mutations in angiogenesis-relevant genes had progressive disease, but did not reveal other biomarkers identifying responders. To conclude, ctDNA and LDH are useful biomarkers for both monitoring and predicting response to bevacizumab.
Collapse
Affiliation(s)
- Rakel Brendsdal Forthun
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Randi Hovland
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
- Department of Biosciences, University of Bergen, Bergen, Norway
| | - Cornelia Schuster
- Centre of Cancer Biomarkers, CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
| | - Hanne Puntervoll
- Centre of Cancer Biomarkers, CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Hans Petter Brodal
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Heidi Maria Namløs
- Department of Tumour Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Lars Birger Aasheim
- Norwegian Cancer Genomics Consortium, Institute for Cancer Research, The Norwegian Radium Hospital/Oslo University Hospital, Oslo, Norway
| | - Leonardo A Meza-Zepeda
- Department of Tumour Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Norwegian Cancer Genomics Consortium, Institute for Cancer Research, The Norwegian Radium Hospital/Oslo University Hospital, Oslo, Norway
- Genomics Core Facility, Department of Core Facilities, Oslo University Hospital, Oslo, Norway
| | - Bjørn Tore Gjertsen
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
- Centre of Cancer Biomarkers, CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Stian Knappskog
- Department of Oncology, Haukeland University Hospital, Bergen, Norway
- K.G. Jebsen Center for Genome Directed Cancer Therapy, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Oddbjørn Straume
- Centre of Cancer Biomarkers, CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Department of Oncology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
28
|
Huertas CS, Calvo-Lozano O, Mitchell A, Lechuga LM. Advanced Evanescent-Wave Optical Biosensors for the Detection of Nucleic Acids: An Analytic Perspective. Front Chem 2019; 7:724. [PMID: 31709240 PMCID: PMC6823211 DOI: 10.3389/fchem.2019.00724] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022] Open
Abstract
Evanescent-wave optical biosensors have become an attractive alternative for the screening of nucleic acids in the clinical context. They possess highly sensitive transducers able to perform detection of a wide range of nucleic acid-based biomarkers without the need of any label or marker. These optical biosensor platforms are very versatile, allowing the incorporation of an almost limitless range of biorecognition probes precisely and robustly adhered to the sensor surface by covalent surface chemistry approaches. In addition, their application can be further enhanced by their combination with different processes, thanks to their integration with complex and automated microfluidic systems, facilitating the development of multiplexed and user-friendly platforms. The objective of this work is to provide a comprehensive synopsis of cutting-edge analytical strategies based on these label-free optical biosensors able to deal with the drawbacks related to DNA and RNA detection, from single point mutations assays and epigenetic alterations, to bacterial infections. Several plasmonic and silicon photonic-based biosensors are described together with their most recent applications in this area. We also identify and analyse the main challenges faced when attempting to harness this technology and how several innovative approaches introduced in the last years manage those issues, including the use of new biorecognition probes, surface functionalization approaches, signal amplification and enhancement strategies, as well as, sophisticated microfluidic solutions.
Collapse
Affiliation(s)
- Cesar S. Huertas
- Integrated Photonics and Applications Centre, School of Engineering, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| | - Olalla Calvo-Lozano
- Nanobiosensors and Bioanalytical Applications Group, Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, CIBER-BBN, Barcelona, Spain
| | - Arnan Mitchell
- Integrated Photonics and Applications Centre, School of Engineering, Royal Melbourne Institute of Technology University, Melbourne, VIC, Australia
| | - Laura M. Lechuga
- Nanobiosensors and Bioanalytical Applications Group, Catalan Institute of Nanoscience and Nanotechnology (ICN2), CSIC and the Barcelona Institute of Science and Technology, CIBER-BBN, Barcelona, Spain
| |
Collapse
|
29
|
Calbet-Llopart N, Potrony M, Tell-Martí G, Carrera C, Barreiro A, Aguilera P, Podlipnik S, Puig S, Malvehy J, Puig-Butillé JA. Detection of cell-free circulating BRAF V 600E by droplet digital polymerase chain reaction in patients with and without melanoma under dermatological surveillance. Br J Dermatol 2019; 182:382-389. [PMID: 31102256 DOI: 10.1111/bjd.18147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND The p.V600E mutation in the BRAF protein is the most frequent mutation in cutaneous melanoma and is a recurrent alteration found in common benign naevi. Analysis of the cell-free BRAF c.1799T>A, p.V600E mutation (cfBRAFV 600E ) in plasma has emerged as a biomarker for monitoring prognosis and treatment response in patients with melanoma. OBJECTIVES To quantify cfBRAFV 600E levels in plasma from patients with melanoma and from patients without melanoma undergoing regular follow-up of their melanocytic lesions, in order to assess the clinical significance of the test. METHODS We quantified cfBRAFV 600E by droplet digital polymerase chain reaction in plasma from 146 patients without melanoma undergoing continuous dermatological screening, from 26 stage III and seven stage IV patients with BRAF-mutant melanoma, and from 32 patients with melanoma who were free of disease for 3 or more years. RESULTS Among disease-free patients and individuals without melanoma, 52% presented a high naevus count (> 50) and 49% had clinically atypical naevi. cfBRAFV 600E was detected in 71% of patients with stage IV melanoma and 15% with stage III, and in 1·4% of individuals without melanoma. No cfBRAFV 600E mutation was detected in disease-free patients with melanoma. Individuals without melanoma had lower cfBRAFV 600E levels than patients with melanoma. We established a variant allelic frequency of 0·26% or 5 copies mL-1 of cfBRAFV 600E as the optimal cutoff value for identifying patients with melanoma with > 99% specificity. CONCLUSIONS This study suggests that naevus-related factors do not influence the detection of cfBRAFV 600E in individuals without melanoma, and supports the clinical diagnostic value of plasma cfBRAFV 600E quantification in patients with melanoma. What's already known about this topic? The analysis of the BRAF c.1799T>A (p.V600E) mutation in cell-free (cf)DNA has emerged as a potential biomarker for monitoring prognosis and treatment response in patients with metastatic BRAFV600E melanoma. The BRAFV600E alteration is a common genetic alteration found in benign proliferations such as melanocytic naevi. No information exists about the impact of the number of common acquired naevi or the presence of clinically atypical naevi in cfBRAFV600E detection in an individual. What does this study add? The cfBRAFV600E mutation is detected in plasma from a reduced number of individuals without melanoma undergoing continuous dermatological follow-up. A high number of naevi or the presence of clinically atypical naevi are factors that do not influence cfBRAFV600E detection in an individual. Both total cfBRAF concentration and cfBRAFV600E frequency are effective biomarkers in patients with advanced melanoma but not in patients at early stages or with micrometastases. What is the translational message? Detection of cfBRAFV600E in an individual is not influenced by naevus-related factors. cfBRAFV600E is a robust and reliable biomarker that can be used in dermatological surveillance programmes.
Collapse
Affiliation(s)
- N Calbet-Llopart
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - M Potrony
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
| | - G Tell-Martí
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
| | - C Carrera
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
| | - A Barreiro
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
| | - P Aguilera
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
| | - S Podlipnik
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain
| | - S Puig
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
| | - J Malvehy
- Dermatology Department, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
| | - J A Puig-Butillé
- Molecular Biology CORE Laboratory, Biochemistry and Molecular Genetics Department; Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
30
|
Ducreux M, Chamseddine A, Laurent-Puig P, Smolenschi C, Hollebecque A, Dartigues P, Samallin E, Boige V, Malka D, Gelli M. Molecular targeted therapy of BRAF-mutant colorectal cancer. Ther Adv Med Oncol 2019; 11:1758835919856494. [PMID: 31244912 PMCID: PMC6582307 DOI: 10.1177/1758835919856494] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 05/16/2019] [Indexed: 12/11/2022] Open
Abstract
Over the past two decades, the molecular characterization of metastatic colorectal cancer (mCRC) has been revolutionized by the routine implementation of RAS and BRAF tests. As a result, it is now known that patients with mCRC harboring BRAF mutations experience a poor prognosis. Although it accounts for only 10% of mCRC, this group is heterogeneous; only the BRAF-V600E mutation, also observed in melanoma, is associated with a very poor prognosis. In terms of treatment, these patients do not benefit from therapeutics targeting the epidermal growth factor receptor (EGFR). In first-line chemotherapy, there are two main options; the first one is to use a triple chemotherapy combination of 5-fluorouracil, irinotecan, and oxaliplatin, with the addition of bevacizumab, because post hoc analysis of randomized trials have reported interesting results. The other option is to use double chemotherapy plus bevacizumab, since anti-EGFR seems to have modest activity in these patients. Only a small percentage of patients who experience failure of this first-line treatment receive second-line treatment. Monotherapy with BRAF inhibitors has failed in this setting, and different combinations have also been tested. Using the rationale that BRAF inhibitor monotherapy fails due to feedback activation of the EGFR pathway, BRAF inhibitors have been combined with anti-EGFR agents plus or minus MEK inhibitors; however, the results did not live up to the hopes raised by the concept. To date, the best results in second-line treatment have been obtained with a combination of vemurafenib, cetuximab, and irinotecan. Despite these advances, further improvements are needed.
Collapse
Affiliation(s)
- Michel Ducreux
- Département d’Oncologie Médicale, Université Paris-Saclay, Gustave Roussy Cancer Campus Grand Paris, 114 rue Edouard Vaillant, Villejuif Cedex, 94805, France
| | - Ali Chamseddine
- Département d’Oncologie Médicale, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Pierre Laurent-Puig
- Département de Biologie, Assistance Publique Hôpitaux de Paris, Hôpital Européen Georges Pompidou, Paris, France
- Université Paris-Descartes, Paris, France; INSERM UMRS-1147, Paris, France
| | - Cristina Smolenschi
- Département d’Oncologie Médicale, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Antoine Hollebecque
- Département d’Oncologie Médicale, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
- Département d’Innovation Thérapeutique et des Essais Précoces (DITEP), Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Peggy Dartigues
- Département de Biopathologie, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Emmanuelle Samallin
- Département d’Oncologie Digestive, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - Valérie Boige
- Département d’Oncologie Médicale, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - David Malka
- Département d’Oncologie Médicale, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| | - Maximiliano Gelli
- Département de Chirurgie Viscérale, Gustave Roussy Cancer Campus Grand Paris, Villejuif, France
| |
Collapse
|
31
|
Zhang Y, Shao Y. Concordance measure and discriminatory accuracy in transformation cure models. Biostatistics 2019; 19:14-26. [PMID: 28481968 DOI: 10.1093/biostatistics/kxx016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Accepted: 02/22/2017] [Indexed: 01/03/2023] Open
Abstract
Many populations of early-stage cancer patients have non-negligible latent cure fractions that can be modeled using transformation cure models. However, there is a lack of statistical metrics to evaluate prognostic utility of biomarkers in this context due to the challenges associated with unknown cure status and heavy censorship. In this article, we develop general concordance measures as evaluation metrics for the discriminatory accuracy of transformation cure models including the so-called promotion time cure models and mixture cure models. We introduce explicit formulas for the consistent estimates of the concordance measures, and show that their asymptotically normal distributions do not depend on the unknown censoring distribution. The estimates work for both parametric and semiparametric transformation models as well as transformation cure models. Numerical feasibility of the estimates and their robustness to the censoring distributions are illustrated via simulation studies and demonstrated using a melanoma data set.
Collapse
Affiliation(s)
- Yilong Zhang
- Merck Research Laboratories, 126 E. Lincoln Ave., Rahway, NJ 07065, USA
| | - Yongzhao Shao
- Department of Population Health, New York University School of Medicine, 650 first ave 5th FL, New York, NY 10016, USA
| |
Collapse
|
32
|
Zhang L, Liang Y, Li S, Zeng F, Meng Y, Chen Z, Liu S, Tao Y, Yu F. The interplay of circulating tumor DNA and chromatin modification, therapeutic resistance, and metastasis. Mol Cancer 2019; 18:36. [PMID: 30849971 PMCID: PMC6408771 DOI: 10.1186/s12943-019-0989-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 02/26/2019] [Indexed: 02/07/2023] Open
Abstract
Peripheral circulating free DNA (cfDNA) is DNA that is detected in plasma or serum fluid with a cell-free status. For cancer patients, cfDNA not only originates from apoptotic cells but also from necrotic tumor cells and disseminated tumor cells that have escaped into the blood during epithelial-mesenchymal transition. Additionally, cfDNA derived from tumors, also known as circulating tumor DNA (ctDNA), carries tumor-associated genetic and epigenetic changes in cancer patients, which makes ctDNA a potential biomarker for the early diagnosis of tumors, monitory and therapeutic evaluations, and prognostic assessments, among others, for various kinds of cancer. Moreover, analyses of cfDNA chromatin modifications can reflect the heterogeneity of tumors and have potential for predicting tumor drug resistance.
Collapse
Affiliation(s)
- Lei Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yiyi Liang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Shifu Li
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Fanyuan Zeng
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yongan Meng
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Ziwei Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, 110 Xiangya Road, Changsha, 410078, Hunan, China.
- Department of Oncology, Institute of Medical Sciences, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China.
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| | - Fenglei Yu
- Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
33
|
Váraljai R, Wistuba-Hamprecht K, Seremet T, Diaz JMS, Nsengimana J, Sucker A, Griewank K, Placke JM, Horn PA, von Neuhoff N, Shannan B, Chauvistré H, Vogel FCE, Horn S, Becker JC, Newton-Bishop J, Stang A, Neyns B, Weide B, Schadendorf D, Roesch A. Application of Circulating Cell-Free Tumor DNA Profiles for Therapeutic Monitoring and Outcome Prediction in Genetically Heterogeneous Metastatic Melanoma. JCO Precis Oncol 2019; 3:PO.18.00229. [PMID: 32914028 PMCID: PMC7446476 DOI: 10.1200/po.18.00229] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2018] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Circulating cell-free tumor DNA (ctDNA) reflects the heterogeneous spectrum of tumor-specific mutations, especially in systemic disease. We validated plasma-based assays that allow the dynamic quantitative detection of ctDNA as a prognostic biomarker for tumor load and prediction of therapy response in melanoma. MATERIALS AND METHODS We analyzed plasma-derived ctDNA from a large training cohort (n = 96) of patients with advanced-stage melanoma, with assays for the BRAF V600E and NRAS Q61 driver mutations as well as TERT C250T and TERT C228T promoter mutations. An independent patient cohort (n = 35) was used to validate the utility of ctDNA monitoring under mitogen-activated protein kinase-targeted or immune checkpoint therapies. RESULTS Elevated plasma ctDNA level at baseline was an independent prognostic factor of disease progression when compared with serum S100 and lactate dehydrogenase levels in multivariable analyses (hazard ratio [HR], 7.43; 95% CI, 1.01 to 55.19; P = .05). The change in ctDNA levels during therapy correlated with treatment response, where increasing ctDNA was predictive for shorter progression-free survival (eg, for BRAF V600E ctDNA, HR, 3.70; 95% CI, 1.86 to 7.34; P < .001). Increasing ctDNA levels predicted disease progression significantly earlier than did routine radiologic scans (P < .05), with a mean lead time of 3.5 months. NRAS-mutant ctDNA was detected in a significant proportion of patients with BRAF-mutant tumors under therapy, but unexpectedly also at baseline. In vitro sensitivity studies suggested that this represents higher-than-expected intratumoral heterogeneity. The detection of NRAS Q61 ctDNA in baseline samples of patients with BRAF V600E mutation who were treated with mitogen-activated protein kinase inhibitors significantly correlated with shorter progression-free survival (HR, 3.18; 95% CI, 1.31 to 7.68; P = .03) and shorter overall survival (HR, 4.08; 95% CI, 1.57 to 10.58; P = .01). CONCLUSION Our results show the potential role of ctDNA measurement as a sensitive monitoring and prediction tool for the early assessment of disease progression and therapeutic response in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Renáta Váraljai
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | | | - Teofila Seremet
- Universitair Ziekenhuis Brussel and Vrije Universiteit Brussel, Brussels, Belgium
| | - Joey Mark S. Diaz
- Leeds Institute of Cancer and Pathology,University of Leeds, Leeds, UK
| | | | - Antje Sucker
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Klaus Griewank
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Jan-Malte Placke
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Peter A. Horn
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Nils von Neuhoff
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Batool Shannan
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Heike Chauvistré
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Felix C. E. Vogel
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Susanne Horn
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Jürgen C. Becker
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | | | - Andreas Stang
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Bart Neyns
- Universitair Ziekenhuis Brussel and Vrije Universiteit Brussel, Brussels, Belgium
| | | | - Dirk Schadendorf
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| | - Alexander Roesch
- University Hospital of Essen, University Duisburg-Essen and German Cancer Consortium partner site Essen/Düsseldorf, Essen, Germany
| |
Collapse
|
34
|
Abstract
Genetic material derived from tumours is constantly shed into the circulation of cancer patients both in the form of circulating free nucleic acids and within circulating cells or extracellular vesicles. Monitoring cancer-specific genomic alterations, particularly mutant allele frequencies, in circulating nucleic acids allows for a non-invasive liquid biopsy for detecting residual disease and response to therapy. The advent of molecular targeted treatments and immunotherapies with increasing effectiveness requires corresponding effective molecular biology methods for the detection of biomarkers such as circulating nucleic acid to monitor and ultimately personalise therapy. The use of polymerase chain reaction (PCR)-based methods, such as droplet digital PCR, allows for a very sensitive analysis of circulating tumour DNA, but typically only a limited number of gene mutations can be detected in parallel. In contrast, next-generation sequencing allows for parallel analysis of multiple mutations in many genes. The development of targeted next-generation sequencing cancer gene panels optimised for the detection of circulating free DNA now provides both the flexibility of multiple mutation analysis coupled with a sensitivity that approaches or even matches droplet digital PCR. In this review, we discuss the advantages and disadvantages of these current molecular technologies in conjunction with how this field is evolving in the context of melanoma diagnosis, prognosis, and monitoring of response to therapy.
Collapse
|
35
|
Lu W, Burton L, Larkin J, Chapman PB, Ascierto PA, Ribas A, Robert C, Sosman JA, McArthur GA, Chang I, Caro I, Penuel E, Yan Y, Wongchenko MJ. Elevated Levels of BRAFV600 Mutant Circulating Tumor DNA and Circulating Hepatocyte Growth Factor Are Associated With Poor Prognosis in Patients With Metastatic Melanoma. JCO Precis Oncol 2018; 2:1-17. [DOI: 10.1200/po.17.00168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Purpose We performed a retrospective exploratory analysis to evaluate the prognostic and predictive effect of two circulating biomarkers, BRAFV600 mutant circulating tumor DNA (ctDNA) and circulating hepatocyte growth factor (cHGF), in metastatic melanoma. Materials and Methods This study evaluated patients from BRIM-3, a phase III trial comparing vemurafenib and dacarbazine in 675 patients with BRAFV600 mutated advanced melanoma. ctDNA was measured using droplet digital polymerase chain reaction, and cHGF was measured by enzyme-linked immunosorbent assay. Overall survival (OS) was estimated using the Kaplan-Meier method, and hazard ratios (HRs) were estimated using Cox proportional hazards modeling. Partitioning analysis was used to group patients into risk categories. Results Patients with elevated levels of baseline BRAFV600 ctDNA had significantly shorter median OS than those with undetectable levels of ctDNA (vemurafenib arm, 9.9 v 21.4 months, respectively, and dacarbazine arm: 6.1 v 21.0 months, respectively). Median OS was also shorter in patients with high levels of cHGF compared with those with low cHGF (vemurafenib arm, 11.9 v 17.3 months, respectively, and dacarbazine arm, 6.1 v 14.4 months, respectively). In a multivariable proportional hazards model with adjustment for lactate dehydrogenase, Eastern Cooperative Oncology Group status, disease stage, and treatment, ctDNA and cHGF were both independent prognostic factors for OS, (HR, 1.75; 95% CI, 1.35 to 2.28 for high v undetectable ctDNA; HR, 1.24; 95% CI, 1.00 to 1.53 for high v low cHGF). Using partitioning analysis, we found that patients with elevated ctDNA combined with elevated cHGF constituted the highest risk group with significantly shorter OS. Conclusion Here, we report that BRIM-3 patients with high levels of ctDNA and cHGF have worse OS regardless of treatment and that these factors are independent prognostic markers for metastatic melanoma.
Collapse
Affiliation(s)
- William Lu
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Luciana Burton
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - James Larkin
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Paul B. Chapman
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Paolo A. Ascierto
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Antoni Ribas
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Caroline Robert
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Jeffrey A. Sosman
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Grant A. McArthur
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Ilsung Chang
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Ivor Caro
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Elicia Penuel
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Yibing Yan
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| | - Matthew J. Wongchenko
- William Lu, Luciana Burton, Ilsung Chang, Ivor Caro, Elicia Penuel, Yibing Yan, and Matthew J. Wongchenko, Genentech, South San Francisco; Antoni Ribas, The Jonsson Comprehensive Cancer Center at University of California, Los Angeles, CA; James Larkin, The Royal Marsden NHS Foundation Trust, London, United Kingdom; Paul B. Chapman, Memorial Sloan Kettering Cancer Center, New York, NY; Paolo A. Ascierto, Istituto Nazionale Tumori Fondazione G. Pascale, Naples, Italy; Caroline Robert, Institut Gustave
| |
Collapse
|
36
|
Corless BC, Chang GA, Cooper S, Syeda MM, Shao Y, Osman I, Karlin-Neumann G, Polsky D. Development of Novel Mutation-Specific Droplet Digital PCR Assays Detecting TERT Promoter Mutations in Tumor and Plasma Samples. J Mol Diagn 2018; 21:274-285. [PMID: 30827467 DOI: 10.1016/j.jmoldx.2018.09.003] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/29/2018] [Accepted: 09/28/2018] [Indexed: 10/28/2022] Open
Abstract
Detecting mutations in the plasma of patients with solid tumors is becoming a valuable method of diagnosing and monitoring cancer. The TERT promoter is mutated at high frequencies in multiple cancer types, most commonly at positions -124 and -146 (designated C228T and C250T, respectively). Detection of these mutations has been challenging because of the high GC content of this region (approximately 80%). We describe development of novel probe-based droplet digital PCR assays that specifically detect and quantify these two mutations, along with the less common 242-243 CC>TT mutation, and demonstrate their application using human tumor and plasma samples from melanoma patients. Assay designs and running conditions were optimized using cancer cell line genomic DNAs with the C228T or C250T mutations. The limits of detection were 0.062% and 0.051% mutant allele fraction for the C228T and C250T assays, respectively. Concordance of 100% was observed between droplet digital PCR and sequencing-based orthogonal methods in the detection of TERT mutant DNA in 32 formalin-fixed, paraffin-embedded melanoma tumors. TERTmutant DNA was also identified in 21 of 27 plasma samples (78%) from patients with TERTmutant tumors, with plasma mutant allele fractions ranging from 0.06% to 15.3%. There were no false positives in plasma. These data demonstrate the potential of these assays to specifically detect and quantify TERTmutant DNA in tumors and plasma of cancer patients.
Collapse
Affiliation(s)
- Broderick C Corless
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Gregory A Chang
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Samantha Cooper
- Digital Biology Center, Bio-Rad Laboratories, Pleasanton, California
| | - Mahrukh M Syeda
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Yongzhao Shao
- Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | - Iman Osman
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York
| | | | - David Polsky
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York; Laura and Isaac Perlmutter Cancer Center, NYU Langone Health, New York, New York.
| |
Collapse
|
37
|
Ossandon MR, Agrawal L, Bernhard EJ, Conley BA, Dey SM, Divi RL, Guan P, Lively TG, McKee TC, Sorg BS, Tricoli JV. Circulating Tumor DNA Assays in Clinical Cancer Research. J Natl Cancer Inst 2018; 110:929-934. [PMID: 29931312 PMCID: PMC6136923 DOI: 10.1093/jnci/djy105] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 04/13/2018] [Accepted: 05/11/2018] [Indexed: 01/01/2023] Open
Abstract
The importance of circulating free DNA (cfDNA) in cancer clinical research was recognized in 1994 when a mutated RAS gene fragment was detected in a patient's blood sample. Up to 1% of the total circulating DNA in patients with cancer is circulating tumor DNA (ctDNA) that originates from tumor cells. As ctDNA is rapidly cleared from the blood stream and can be obtained by minimally invasive methods, it can be used as a dynamic cancer biomarker for cancer early detection, diagnosis, and treatment monitoring. Despite the potential for clinical use, few ctDNA assays have been cleared or approved by the US Food and Drug Administration. As tools for clinical and translational research, current ctDNA assays face some challenges, and more research is needed to advance use of these assays. On September 29-30, 2016, the Division of Cancer Treatment and Diagnosis at the National Cancer Institute convened a workshop entitled "Circulating Tumor DNA Assays in Clinical Cancer Research" to garner input from industry experts, academia, and government research and regulatory agencies to understand and promote the translation of ctDNA assays to clinical research, with potential to advance to use in clinical practice. This Commentary presents the topics of the workshop covered in the presentations and points made in the discussions that followed: 1) background on ctDNA, 2) potential clinical utility of ctDNA assays, 3) assay technology, 4) assay clinical and analytical validation, and 5) industry perspectives. Additional relevant information that has come to light since the workshop has been included.
Collapse
Affiliation(s)
- Miguel R Ossandon
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lokesh Agrawal
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Eric J Bernhard
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Barbara A Conley
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Sumana M Dey
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Rao L Divi
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Ping Guan
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tracy G Lively
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Tawnya C McKee
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Brian S Sorg
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - James V Tricoli
- National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
38
|
Illustrative cases for monitoring by quantitative analysis of BRAF/NRAS ctDNA mutations in liquid biopsies of metastatic melanoma patients who gained clinical benefits from anti-PD1 antibody therapy. Melanoma Res 2018; 28:65-70. [PMID: 29227333 DOI: 10.1097/cmr.0000000000000415] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Anti-programmed death 1 (PD-1) monoclonal antibodies improve the survival of metastatic melanoma patients. Predictive or monitoring biomarkers for response to this therapy could improve the clinical management of these patients. To date, no established biomarkers are available for monitoring the response to immunotherapy. Tumor- specific mutations in circulating tumor DNA (ctDNA) such as BRAF and NRAS mutations for melanoma patients have been proposed for monitoring of immunotherapy response. We present seven illustrative cases for the use of ctDNA BRAF and NRAS mutations' monitoring in plasma. The cases described exemplify four distinct clinical benefit patterns: rapid and durable complete response (CR), early progression, followed by CR, CR followed by early progression after interrupting treatment and long-term disease stabilization. These representative cases suggest that comprehensive BRAF/NRAS ctDNA monitoring during anti-PD1 therapy is informative and can be of added value for the monitoring of melanoma patients gaining clinical benefit on anti-PD1 treatment. An important advantage of our approach is that using the cartridge system on the Idylla platform for mutation analysis, the results become available the same day 2 h after plasma collection. Therefore, in the future, the ctDNA level can be an element in the clinical management of the patients.
Collapse
|
39
|
Swe T, Kim KB. Update on systemic therapy for advanced cutaneous melanoma and recent development of novel drugs. Clin Exp Metastasis 2018; 35:503-520. [PMID: 30019239 DOI: 10.1007/s10585-018-9913-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 06/11/2018] [Indexed: 12/19/2022]
Abstract
Malignant melanoma is generally chemo- and radio-resistant, and patients with advanced melanoma have a poor prognosis. However, with our increased understanding of the checkpoint immune molecules and genetic alterations of melanoma cells, more effective immunotherapy, such as anti CTLA4 antibody and anti PD-1 antibodies, and targeted drug therapy, such as BRAF inhibitors and MEK inhibitors, have been developed, resulting in improved overall survival and quality of life of patients with advanced melanoma. In addition, emerging technologies to develop prognostic and predictive biomarkers for response to systemic therapy could help clinicians make more accurate assessments of the disease and formulate more effective treatment plans. In this review, current standard systemic therapy options and recently developed novel drugs for advanced melanoma are discussed.
Collapse
Affiliation(s)
- Thein Swe
- California Pacific Medical Center Research Institute, 2333 Buchanan St., San Francisco, CA, 94115, USA
| | - Kevin B Kim
- California Pacific Medical Center Research Institute, 2333 Buchanan St., San Francisco, CA, 94115, USA.
| |
Collapse
|
40
|
Lin SY, Huang SK, Huynh KT, Salomon MP, Chang SC, Marzese DM, Lanman RB, Talasaz A, Hoon DS. Multiplex Gene Profiling of Cell-Free DNA in Patients With Metastatic Melanoma for Monitoring Disease. JCO Precis Oncol 2018; 2:PO.17.00225. [PMID: 32913981 PMCID: PMC7446321 DOI: 10.1200/po.17.00225] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
PURPOSE Hotspot blood cell-free DNA (cfDNA) biomarker assays have limited utility in profiling tumor heterogeneity and burden and in capturing regional metastasis with low disease burden in patients with melanoma. We investigated the utility of a sensitive 54-cancer gene digital next-generation sequencing approach targeting blood cfDNA single nucleotide variants (SNVs) and copy number amplification for monitoring disease in patients with melanoma with regional or distant organ metastasis (DOM). PATIENTS AND METHODS A total of 142 blood samples were evaluated by digital next-generation sequencing across two patient cohorts. Cohort 1 contained 44 patients with stage II, III, or IV disease with matched tumor DNA at the time of surgery or DOM. Cohort 2 consisted of 12 overlapping patients who were longitudinally monitored after complete lymph node dissection to DOM. RESULTS In cohort 1, cfDNA SNVs were detected in 75% of patients. Tumor-cfDNA somatic SNV concordance was 85% at a variant allele fraction of ≥ 0.5%. An SNV load (number of unique SNVs detected) of greater than two SNVs and an SNV burden (total cumulative SNV VAF) of > 0.5% were significantly associated with worse overall survival (P < .05) in stage IV patients. In cohort 2, 98 longitudinal blood samples along with matched regional and distant metastases from 12 stage III patients were analyzed before complete lymph node dissection and throughout disease progression. cfDNA SNV levels correlated with tumor burden (P = .019), enabled earlier detection of recurrence compared with radiologic imaging (P < .01), captured tumor heterogeneity, and identified increasing SNVs levels before recurrence. CONCLUSION This study demonstrates significant utility for cfDNA profiling in patients with melanoma with regional and/or distant metastasis for earlier detection of recurrence and progression and in capturing tumor evolution and heterogeneity, thus impacting how patients with melanoma are monitored.
Collapse
Affiliation(s)
- Selena Y. Lin
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Sharon K. Huang
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Kelly T. Huynh
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Matthew P. Salomon
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Shu-Ching Chang
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Diego M. Marzese
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Richard B. Lanman
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - AmirAli Talasaz
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| | - Dave S.B. Hoon
- Selena Y. Lin, Sharon K. Huang, Kelly T. Huynh, Matthew P. Salomon, Diego M. Marzese, and Dave S.B. Hoon, John Wayne Cancer Institute at Providence Saint John’s Health Center, Santa Monica; Richard B. Lanman and AmirAli Talasaz, Guardant Health, Redwood City, CA; and Shu-Ching Chang, Medical Data Research Center at Providence Saint Joseph’s Health, Portland, OR
| |
Collapse
|
41
|
Lubitz CC, Zhan T, Gunda V, Amin S, Gigliotti BJ, Fingeret AL, Holm TM, Wachtel H, Sadow PM, Wirth LJ, Sullivan RJ, Panka DJ, Parangi S. Circulating BRAF V600E Levels Correlate with Treatment in Patients with Thyroid Carcinoma. Thyroid 2018; 28:328-339. [PMID: 29378474 PMCID: PMC5865613 DOI: 10.1089/thy.2017.0322] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND BRAFV600E is the most common mutation in papillary thyroid carcinoma (PTC) and can be associated with aggressive disease. Previously, a highly sensitive blood RNA-based BRAFV600E assay was reported. The objective of this study was to assess the correlation of BRAFV600E circulating tumor RNA levels with surgical and medical treatment. METHODS Circulating BRAFV600E levels were assessed in (i) a murine model of undifferentiated (anaplastic) thyroid carcinoma with known BRAFV600E mutation undergoing BRAFV600E-inhibitor (BRAFi) treatment, and (ii) in 111 patients enrolled prior to thyroidectomy (n = 86) or treatment of advanced recurrent or metastatic PTC (n = 25). Blood samples were drawn for BRAFV600E analysis before and after treatment. Testing characteristics were assessed and positivity criteria optimized. Changes in blood BRAFV600E values were assessed and compared to clinical characteristics and response to therapy. RESULTS In a murine model of anaplastic thyroid carcinoma with BRAFV600E mutation, blood BRAFV600E RNA correlated with tumor volume in animals treated with BRAFi. In tissue BRAFV600E-positive (n = 36) patients undergoing initial surgery for PTC, blood BRAFV600E levels declined postoperatively (median 370.0-178.5 fg/ng; p = 0.002). In four patients with metastatic or poorly differentiated thyroid carcinoma receiving targeted therapies, blood BRAFV600E declined following therapy and corresponded with radiographic evidence of partial response or stable disease. CONCLUSIONS This study shows the correlation of blood BRAFV600E levels in response to treatment in both an established animal model of thyroid cancer and in patients with BRAFV600E-positive tumors with all stages of disease. This assay represents an alternative biomarker in patients with positive thyroglobulin antibodies, and tumors, which do not express thyroglobulin.
Collapse
Affiliation(s)
- Carrie C. Lubitz
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
| | - Tiannan Zhan
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts
| | - Viswanath Gunda
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Salma Amin
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Abbey L. Fingeret
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Tammy M. Holm
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Heather Wachtel
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| | - Peter M. Sadow
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | - Lori J. Wirth
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Ryan J. Sullivan
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - David J. Panka
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Sareh Parangi
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts
| |
Collapse
|
42
|
Haselmann V, Gebhardt C, Brechtel I, Duda A, Czerwinski C, Sucker A, Holland-Letz T, Utikal J, Schadendorf D, Neumaier M. Liquid Profiling of Circulating Tumor DNA in Plasma of Melanoma Patients for Companion Diagnostics and Monitoring of BRAF Inhibitor Therapy. Clin Chem 2018; 64:830-842. [PMID: 29483107 DOI: 10.1373/clinchem.2017.281543] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 02/12/2018] [Indexed: 11/06/2022]
Abstract
BACKGROUND The current standard for determining eligibility of patients with metastatic melanoma for BRAF-targeted therapy is tissue-based testing of BRAF mutations. As patients are rarely rebiopsied, detection in blood might be advantageous by enabling a comprehensive assessment of tumor mutational status in real time and thereby representing a noninvasive biomarker for monitoring BRAF therapy. METHODS In all, 634 stage I to IV melanoma patients were enrolled at 2 centers, and 1406 plasma samples were prospectively collected. Patients were assigned to 3 separate study cohorts: study 1 for assessment of circulating tumor DNA (ctDNA) as part of companion diagnostics, study 2 for assessment of ctDNA for patients with low tumor burden and for follow-up, and study 3 for monitoring of resistance to BRAF inhibitor (BRAFi) or mitogen-activated protein kinase inhibitor therapy. RESULTS Overall, a high degree of concordance between plasma and tissue testing results was observed at 90.9% (study 1) and 90.1% (study 2), respectively. Interestingly, discrepant results were in some cases associated with nonresponse to BRAFi (n = 3) or a secondary BRAF-mutant malignancy (n = 5). Importantly, ctDNA results correlated with the clinical course of disease in 95.7% and with response to treatment. Significantly, the detection of BRAF mutant ctDNA preceded relapse assessed by Response Evaluation Criteria in Solid Tumors, and was more specific than serum S100 and lactate dehydrogenase. CONCLUSIONS Blood-based testing compares favorably with standard-of-care tissue-based BRAF mutation testing. Importantly, blood-based BRAF testing correlates with the clinical course, even for early-stage patients, and may be used to predict response to treatment, recurrence, and resistance before radioimaging under BRAFi therapy, thereby enabling considerable improvements in patient treatment.
Collapse
Affiliation(s)
- Verena Haselmann
- Department of Clinical Chemistry, University Medical Centre, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Christoffer Gebhardt
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,Department of Dermatology and Venereology, University Hospital Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Ingrid Brechtel
- Department of Clinical Chemistry, University Medical Centre, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Angelika Duda
- Department of Clinical Chemistry, University Medical Centre, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Claudia Czerwinski
- Department of Clinical Chemistry, University Medical Centre, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Antje Sucker
- Department of Dermatology, University of Essen Hospital, Essen, Germany and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Tim Holland-Letz
- German Cancer Research Center (DKFZ), Department of Biostatistics, Heidelberg, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center, Ruprecht-Karls University of Heidelberg, Mannheim, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University of Essen Hospital, Essen, Germany and German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Michael Neumaier
- Department of Clinical Chemistry, University Medical Centre, Ruprecht-Karls University of Heidelberg, Mannheim, Germany;
| |
Collapse
|
43
|
Almodovar K, Iams WT, Meador CB, Zhao Z, York S, Horn L, Yan Y, Hernandez J, Chen H, Shyr Y, Lim LP, Raymond CK, Lovly CM. Longitudinal Cell-Free DNA Analysis in Patients with Small Cell Lung Cancer Reveals Dynamic Insights into Treatment Efficacy and Disease Relapse. J Thorac Oncol 2018; 13:112-123. [PMID: 28951314 PMCID: PMC5827950 DOI: 10.1016/j.jtho.2017.09.1951] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 08/16/2017] [Accepted: 09/08/2017] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Patients with SCLC have a poor prognosis and limited treatment options. Because access to longitudinal tumor samples is very limited in patients with this disease, we chose to focus our studies on the characterization of plasma cell-free DNA (cfDNA) for rapid, noninvasive monitoring of disease burden. METHODS We developed a liquid biopsy assay that quantifies somatic variants in cfDNA. The assay detects single nucleotide variants, copy number alterations, and insertions or deletions in 14 genes that are frequently mutated in SCLC, including tumor protein p53 gene (TP53), retinoblastoma 1 gene (RB1), BRAF, KIT proto-oncogene receptor tyrosine kinase gene (KIT), notch 1 gene (NOTCH1), notch 2 gene (NOTCH2), notch 3 gene (NOTCH3), notch 4 gene (NOTCH4), phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha gene (PIK3CA), phosphatase and tensin homolog gene (PTEN), fibroblast growth factor receptor 1 gene (FGFR1), v-myc avian myelocytomatosis viral oncogene homolog gene (MYC), v-myc avian myelocytomatosis viral oncogene lung carcinoma derived homolog gene (MYCL1), and v-myc avian myelocytomatosis viral oncogene neuroblastoma derived homolog gene (MYCN). RESULTS Over the course of 26 months of peripheral blood collection, we examined 140 plasma samples from 27 patients. We detected disease-associated mutations in 85% of patient samples with mutant allele frequencies ranging from 0.1% to 87%. In our cohort, 59% of the patients had extensive-stage disease, and the most common mutations occurred in TP53 (70%) and RB1 (52%). In addition to mutations in TP53 and RB1, we detected alterations in 10 additional genes in our patient population (PTEN, NOTCH1, NOTCH2, NOTCH3, NOTCH4, MYC, MYCL1, PIK3CA, KIT, and BRAF). The observed allele frequencies and copy number alterations tracked closely with treatment responses. Notably, in several cases analysis of cfDNA provided evidence of disease relapse before conventional imaging. CONCLUSIONS These results suggest that liquid biopsies are readily applicable in patients with SCLC and can potentially provide improved monitoring of disease burden, depth of response to treatment, and timely warning of disease relapse in patients with this disease.
Collapse
Affiliation(s)
- Karinna Almodovar
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Wade T. Iams
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Catherine B. Meador
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Zhiguo Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Sally York
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Leora Horn
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Yingjun Yan
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | | | - Heidi Chen
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | - Yu Shyr
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN
| | | | | | - Christine M. Lovly
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN,Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN,Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN,Corresponding author: Christine M. Lovly, MD, PhD, Vanderbilt-Ingram Cancer Center, 2220 Pierce Avenue, 777 Preston Research Building, Nashville, TN 37232-6307, Phone 615-936-3457,
| |
Collapse
|
44
|
Tang H, Kong Y, Si L, Cui C, Sheng X, Chi Z, Dai J, Yu S, Ma M, Wu X, Yu J, Xu T, Yu H, Yan J, Guo J. Clinical significance of BRAF V600E mutation in circulating tumor DNA in Chinese patients with melanoma. Oncol Lett 2017; 15:1839-1844. [PMID: 29434880 DOI: 10.3892/ol.2017.7529] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 10/25/2017] [Indexed: 12/17/2022] Open
Abstract
The present study aimed to assess the B rapidly accelerated fibrosarcoma (BRAFV600E) status in plasma from Chinese patients with melanoma, and evaluated its prognostic value following treatment with BRAF inhibitors. Mutation-specific 3D digital polymerase chain reaction (dPCR) was used to quantify BRAFV600E in circulating tumor DNA (ctDNA) in 58 patients with melanoma, prior to treatment with BRAF inhibitors. Correlations between baseline ctDNA levels and clinicopathological characteristics and clinical benefits were then statistically analyzed. The concordance and sensitivity of BRAFV600E between ctDNA and tumor tissue were 70.2% and 76%, respectively, in 58 patients with melanoma. BRAFV600E mutation in ctDNA correlated with lactate dehydrogenase concentration (P=0.04) and Eastern Cooperative Oncology Group score (P=0.04). There was no correlation between BRAFV600E of ctDNA with response, progression-free survival (PFS), or overall survival (OS) following targeted therapy. The objective response rate, PFS and OS stratified by BRAFV600E of ctDNA were 30.0% vs. 56.7%, (P=0.3), 8.1 months vs. 6.7 months, (P=0.38) and 65.6 months vs. 42.3 months (P=0.52), respectively, for undetectable and mutant types. In conclusion, 3D dPCR is appropriate for ctDNA detection and BRAFV600E in ctDNA is a non-invasive biomarker in patients with melanoma.
Collapse
Affiliation(s)
- Huan Tang
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Yan Kong
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Lu Si
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Chuanliang Cui
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xinan Sheng
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Zhihong Chi
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Jie Dai
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Sifan Yu
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Meng Ma
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xiaowen Wu
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Jiayi Yu
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Tianxiao Xu
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Huan Yu
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Junya Yan
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Jun Guo
- Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| |
Collapse
|
45
|
Momtaz P, Harding JJ, Ariyan C, Coit DG, Merghoub T, Gasmi B, You D, Viale A, Panageas KS, Samoila A, Postow MA, Wolchok JD, Chapman PB. Four-month course of adjuvant dabrafenib in patients with surgically resected stage IIIC melanoma characterized by a BRAFV600E/K mutation. Oncotarget 2017; 8:105000-105010. [PMID: 29285228 PMCID: PMC5739615 DOI: 10.18632/oncotarget.21072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 07/25/2017] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND We tested the hypothesis that a 4-month course of adjuvant dabrafenib in stage IIIC BRAF-mutated melanoma would improve 2 year RFS from 24% to 51%, and that tumor-derived cell free DNA (cfDNA) in plasma would correlate with and predict recurrence. METHODS Patients with stage IIIC BRAF V600E/K mutated melanoma who were free of disease after surgical resection received 4 months of adjuvant dabrafenib. Patients were evaluated with imaging at baseline, at the end of cycles 2, 4, 6, then every 3 months until disease relapse or 2 years, whichever came first. Serial blood samples were collected for evaluation of cfDNA at the same time. RESULTS 21/23 patients enrolled were evaluable; 2 patients withdrew consent during the first week of treatment. The 2 year RFS was 28.6% (95% CI 12-48%). The estimated overall survival at 2 years was 78% (95% CI 51-91%). cfDNA detection had a 53% sensitivity in relapsing patients but cfDNA detection did not provide lead-time advantage over CT scanning. CONCLUSION A 4-month course of adjuvant dabrafenib did not result in a detectable improvement in 2-year RFS. cfDNA was less sensitive than standard CT imaging and did not provide a lead-time advantage in detecting relapse.
Collapse
Affiliation(s)
- Parisa Momtaz
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - James J. Harding
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Charlotte Ariyan
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Daniel G. Coit
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Taha Merghoub
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Billel Gasmi
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Daoqi You
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Agnes Viale
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | | | - Michael A. Postow
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Jedd D. Wolchok
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Paul B. Chapman
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
46
|
Wong SQ, Raleigh JM, Callahan J, Vergara IA, Ftouni S, Hatzimihalis A, Colebatch AJ, Li J, Semple T, Doig K, Mintoff C, Sinha D, Yeh P, Silva MJ, Alsop K, Thorne H, Bowtell DD, Gyorki DE, Arnau GM, Cullinane C, Kee D, Brady B, Kelleher F, Dawson MA, Papenfuss AT, Shackleton M, Hicks RJ, McArthur GA, Sandhu S, Dawson SJ. Circulating Tumor DNA Analysis and Functional Imaging Provide Complementary Approaches for Comprehensive Disease Monitoring in Metastatic Melanoma. JCO Precis Oncol 2017; 1:1-14. [DOI: 10.1200/po.16.00009] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose Circulating tumor DNA (ctDNA) allows noninvasive disease monitoring across a range of malignancies. In metastatic melanoma, the extent to which ctDNA reflects changes in metabolic disease burden assessed by 18F-labeled fluorodeoxyglucose positron emission tomography (FDG-PET) is unknown. We assessed the role of ctDNA analysis in combination with FDG-PET to monitor tumor burden and genomic heterogeneity throughout treatment. Patients and Methods We performed a comprehensive analysis of serial ctDNA and FDG-PET in 52 patients who received systemic therapy for metastatic melanoma. Next-generation sequencing and digital polymerase chain reaction were used to analyze plasma samples from the cohort. Results ctDNA levels were monitored across patients with mutant BRAF, NRAS, and BRAF/NRAS wild type disease. Mutant BRAF and NRAS ctDNA levels correlated closely with changes in metabolic disease burden throughout treatment. TERT promoter mutant ctDNA levels also paralleled changes in tumor burden, which provide an alternative marker for disease monitoring. Of note, subcutaneous and cerebral disease sites were not well represented in plasma. Early changes in ctDNA and metabolic disease burden were important indicators of treatment response. Patients with an early decrease in ctDNA post-treatment had improved progression-free survival compared with patients in whom ctDNA levels remained unchanged or increased over time (hazard ratio, 2.6; P = .05). ctDNA analysis contributed key molecular information through the identification of putative resistance mechanisms to targeted therapy. A detailed comparison of the genomic architecture of plasma and multiregional tumor biopsy specimens at autopsy revealed the ability of ctDNA to comprehensively capture genomic heterogeneity across multiple disease sites. Conclusion The findings highlight the powerful role of ctDNA in metastatic melanoma as a complementary modality to functional imaging that allows real-time monitoring of both tumor burden and genomic changes throughout therapy.
Collapse
Affiliation(s)
- Stephen Q. Wong
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Jeanette M. Raleigh
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Jason Callahan
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Ismael A. Vergara
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Sarah Ftouni
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Athena Hatzimihalis
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Andrew J. Colebatch
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Jason Li
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Timothy Semple
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Kenneth Doig
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Christopher Mintoff
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Devbarna Sinha
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Paul Yeh
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Maria Joao Silva
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Kathryn Alsop
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Heather Thorne
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - David D. Bowtell
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - David E. Gyorki
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Gisela Mir Arnau
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Carleen Cullinane
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Damien Kee
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Benjamin Brady
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Fergal Kelleher
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Mark A. Dawson
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Anthony T. Papenfuss
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Mark Shackleton
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Rodney J. Hicks
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Grant A. McArthur
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Shahneen Sandhu
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - Sarah-Jane Dawson
- All authors: Peter MacCallum Cancer Centre; Kenneth Doig, Heather Thorne, David D. Bowtell, Carleen Cullinane, Mark A. Dawson, Anthony T. Papenfuss, Mark Shackleton, Rodney J. Hicks, Grant A. McArthur, Shahneen Sandhu, and Sarah-Jane Dawson, University of Melbourne; and Anthony T. Papenfuss, Walter Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| |
Collapse
|
47
|
Chang Y, Tolani B, Nie X, Zhi X, Hu M, He B. Review of the clinical applications and technological advances of circulating tumor DNA in cancer monitoring. Ther Clin Risk Manag 2017; 13:1363-1374. [PMID: 29066904 PMCID: PMC5644666 DOI: 10.2147/tcrm.s141991] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Circulating cell-free DNA (cfDNA) released by tumor cells, termed ctDNA, closely reflects the heterogeneity of primary cancers and their metastases. As a noninvasive, real-time monitoring biomarker, ctDNA is a promising tool for detecting driver gene mutations, assessing tumor burden and acquired resistance, and early diagnosis. However, isolation and enrichment of cfDNA is a big challenge due to the high degree of DNA fragmentation and its relatively low abundance in the bloodstream. This review aims to provide insights into the recent technological advances in acquisition of optimal quality cfDNA, the use of preservatives, isolation methods, processing timelines, and detection techniques. It also describes clinical applications of ctDNA in cancer patient management.
Collapse
Affiliation(s)
- Yi Chang
- Department of Respiratory Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Bhairavi Tolani
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Xiuhong Nie
- Department of Respiratory Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xiuyi Zhi
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Mu Hu
- Department of Thoracic Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Biao He
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
48
|
McEvoy AC, Calapre L, Pereira MR, Giardina T, Robinson C, Khattak MA, Meniawy TM, Pritchard AL, Hayward NK, Amanuel B, Millward M, Ziman M, Gray ES. Sensitive droplet digital PCR method for detection of TERT promoter mutations in cell free DNA from patients with metastatic melanoma. Oncotarget 2017; 8:78890-78900. [PMID: 29108273 PMCID: PMC5668006 DOI: 10.18632/oncotarget.20354] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Accepted: 07/25/2017] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Currently mainly BRAF mutant circulating tumor DNA (ctDNA) is utilized to monitor patients with melanoma. TERT promoter mutations are common in various cancers and found in up to 70% of melanomas, including half of BRAF wild-type cases. Therefore, a sensitive method for detection of TERT promoter mutations would increase the number of patients that could be monitored through ctDNA analysis. METHODS A droplet digital PCR (ddPCR) assay was designed for the concurrent detection of chr5:1,295,228 C>T and chr5:1,295,250 C>T TERT promoter mutations. The assay was validated using 39 melanoma cell lines and 22 matched plasma and tumor samples. In addition, plasma samples from 56 metastatic melanoma patients and 56 healthy controls were tested for TERT promoter mutations. RESULTS The established ddPCR assay detected TERT promoter mutations with a lower limit of detection (LOD) of 0.17%. Total concordance was demonstrated between ddPCR and Sanger sequencing in all cell lines except one, which carried a second mutation within the probe binding-site. Concordance between matched plasma and tumor tissue was 68% (15/22), with a sensitivity of 53% (95% CI, 27%-79%) and a specificity of 100% (95% CI, 59%-100%). A significantly longer PFS (p=0.028) was evident in ctDNA negative patients. Importantly, our TERT promoter mutations ddPCR assay allowed detection of ctDNA in 11 BRAF wild-type cases. CONCLUSIONS The TERT promoter mutation ddPCR assay offers a sensitive test for molecular analysis of melanoma tumors and ctDNA, with the potential to be applied to other cancers.
Collapse
Affiliation(s)
- Ashleigh C. McEvoy
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Leslie Calapre
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Michelle R. Pereira
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Tindaro Giardina
- Anatomical Pathology, PathWest, QEII Medical Centre, Nedlands, Western Australia, Australia
| | - Cleo Robinson
- Anatomical Pathology, PathWest, QEII Medical Centre, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Muhammad A. Khattak
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
- Department of Medical Oncology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Tarek M. Meniawy
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | | | - Nicholas K. Hayward
- QIMR Berghofer Medical Research Institute, Herston, Brisbane, QLD, Australia
| | - Benhur Amanuel
- Anatomical Pathology, PathWest, QEII Medical Centre, Nedlands, Western Australia, Australia
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Michael Millward
- School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Melanie Ziman
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, Western Australia, Australia
| | - Elin S. Gray
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| |
Collapse
|
49
|
Single-Color Digital PCR Provides High-Performance Detection of Cancer Mutations from Circulating DNA. J Mol Diagn 2017; 19:697-710. [PMID: 28818432 PMCID: PMC6593258 DOI: 10.1016/j.jmoldx.2017.05.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/05/2017] [Accepted: 05/09/2017] [Indexed: 02/02/2023] Open
Abstract
We describe a single-color digital PCR assay that detects and quantifies cancer mutations directly from circulating DNA collected from the plasma of cancer patients. This approach relies on a double-stranded DNA intercalator dye and paired allele-specific DNA primer sets to determine an absolute count of both the mutation and wild-type–bearing DNA molecules present in the sample. The cell-free DNA assay uses an input of 1 ng of nonamplified DNA, approximately 300 genome equivalents, and has a molecular limit of detection of three mutation DNA genome-equivalent molecules per assay reaction. When using more genome equivalents as input, we demonstrated a sensitivity of 0.10% for detecting the BRAF V600E and KRAS G12D mutations. We developed several mutation assays specific to the cancer driver mutations of patients' tumors and detected these same mutations directly from the nonamplified, circulating cell-free DNA. This rapid and high-performance digital PCR assay can be configured to detect specific cancer mutations unique to an individual cancer, making it a potentially valuable method for patient-specific longitudinal monitoring.
Collapse
|
50
|
Calapre L, Warburton L, Millward M, Ziman M, Gray ES. Circulating tumour DNA (ctDNA) as a liquid biopsy for melanoma. Cancer Lett 2017; 404:62-69. [PMID: 28687355 DOI: 10.1016/j.canlet.2017.06.030] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 06/16/2017] [Accepted: 06/16/2017] [Indexed: 01/10/2023]
Abstract
Circulating tumour DNA (ctDNA) has emerged as a promising blood-based biomarker for monitoring disease status of patients with advanced cancers. In melanoma, ctDNA has been shown to have clinical value as an alternative tumour source for the detection clinically targetable mutations for the assessment of response to therapy. This review provides a critical summary of the evidence that gives credence to the utility of ctDNA as a biomarker for monitoring of disease status in advanced melanoma and the steps required for its implementation into clinical settings.
Collapse
Affiliation(s)
- Leslie Calapre
- School of Medical Science, Edith Cowan University, Joondalup, WA, Australia.
| | - Lydia Warburton
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| | - Michael Millward
- Department of Medical Oncology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia; School of Medicine and Pharmacology, The University of Western Australia, Crawley, Western Australia, Australia
| | - Mel Ziman
- School of Medical Science, Edith Cowan University, Joondalup, WA, Australia; School of Pathology and Laboratory Medicine, University of Western Australia, Crawley, WA, Australia
| | - Elin S Gray
- School of Medical Science, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|