1
|
Kurma K, Eslami-S Z, Alix-Panabières C, Cayrefourcq L. Liquid biopsy: paving a new avenue for cancer research. Cell Adh Migr 2024; 18:1-26. [PMID: 39219215 PMCID: PMC11370957 DOI: 10.1080/19336918.2024.2395807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 09/04/2024] Open
Abstract
The current constraints associated with cancer diagnosis and molecular profiling, which rely on invasive tissue biopsies or clinical imaging, have spurred the emergence of the liquid biopsy field. Liquid biopsy involves the extraction of circulating tumor cells (CTCs), circulating free or circulating tumor DNA (cfDNA or ctDNA), circulating cell-free RNA (cfRNA), extracellular vesicles (EVs), and tumor-educated platelets (TEPs) from bodily fluid samples. Subsequently, these components undergo molecular characterization to identify biomarkers that are critical for early cancer detection, prognosis, therapeutic assessment, and post-treatment monitoring. These innovative biosources exhibit characteristics analogous to those of the primary tumor from which they originate or interact. This review comprehensively explores the diverse technologies and methodologies employed for processing these biosources, along with their principal clinical applications.
Collapse
Affiliation(s)
- Keerthi Kurma
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Zahra Eslami-S
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Laure Cayrefourcq
- Laboratory of Rare Human Circulating Cells (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- CREEC/CANECEV, MIVEGEC (CREES),
University of Montpellier, CNRS, IRD, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| |
Collapse
|
2
|
Bravo AC, Morão B, Luz A, Dourado R, Oliveira B, Guedes A, Moreira-Barbosa C, Fidalgo C, Mascarenhas-Lemos L, Costa-Santos MP, Maio R, Paulino J, Viana Baptista P, Fernandes AR, Cravo M. Bringing Hope to Improve Treatment in Pancreatic Ductal Adenocarcinoma-A New Tool for Molecular Profiling of KRAS Mutations in Tumor and Plasma Samples. Cancers (Basel) 2024; 16:3544. [PMID: 39456638 PMCID: PMC11506488 DOI: 10.3390/cancers16203544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/10/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Pancreatic ductal adenocarcinoma (PDAC) incidence is rising, and prognosis remains poor due to late diagnosis and limited effective therapies. Currently, patients are treated based on TNM staging, without molecular tumor characterization. This study aimed to validate a technique that combines the amplification refractory mutation system (ARMS) with high-resolution melting analysis (HRMA) for detecting mutations in codon 12 of KRAS in tumor and plasma, and to assess its prognostic value. METHODS Prospective study including patients with newly diagnosed PDAC with tumor and plasma samples collected before treatment. Mutations in codon 12 of KRAS (G12D, G12V, G12C, and G12R) were detected using ARMS-HRMA and compared to Sanger sequencing (SS). Univariate and multivariate analyses were used to evaluate the prognostic significance of these mutations. RESULTS A total of 88 patients, 93% with ECOG-PS 0-1, 57% with resectable disease. ARMS-HRMA technique showed a higher sensitivity than SS, both in tumor and plasma (77% vs. 51%; 25 vs. 0%, respectively). The most frequent mutation was G12D (n = 32, 36%), followed by G12V (n = 22, 25%). On multivariate analysis, patients with G12D and/or G12C mutations, either in tumor or plasma, had lower PFS (HR 1.792, 95% CI 1.061-3.028, p = 0.029; HR 2.081, 95% CI 1.014-4.272, p = 0.046, respectively) and lower OS (HR 1.757, 95% CI 1.013-3.049, p = 0.045; HR 2.229, 95% CI 1.082-4.594, p = 0.030, respectively). CONCLUSIONS ARMS-HRMA is a rapid and cost-effective method for detecting KRAS mutations in PDAC patients, offering the potential for stratifying prognosis and guiding treatment decisions. The presence of G12D and G12C mutations in both tumor and plasma is associated with a poorer prognosis.
Collapse
Affiliation(s)
- Ana Catarina Bravo
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
| | - Bárbara Morão
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
| | - André Luz
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Rúben Dourado
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Beatriz Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Ana Guedes
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz Learning Health, Luz Saúde, 1500-650 Lisboa, Portugal
| | - Catarina Moreira-Barbosa
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz Learning Health, Luz Saúde, 1500-650 Lisboa, Portugal
| | - Catarina Fidalgo
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
| | - Luís Mascarenhas-Lemos
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
- Catolica Medical School, 1649-023 Lisboa, Portugal
| | | | - Rui Maio
- Hospital Beatriz Ângelo, 2674-514 Loures, Portugal; (A.C.B.); (B.M.); (A.G.); (C.M.-B.); (C.F.); (R.M.)
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
| | - Jorge Paulino
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- NOVA Medical School, 1169-056 Lisboa, Portugal
| | - Pedro Viana Baptista
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Alexandra R. Fernandes
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal; (A.L.); (R.D.); (B.O.); (P.V.B.); (A.R.F.)
- UCIBIO—Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, NOVA University Lisbon, 2829-516 Caparica, Portugal
| | - Marília Cravo
- Hospital da Luz, 1500-650 Lisboa, Portugal; (L.M.-L.); (J.P.)
- Lisbon School of Medicine, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
3
|
Wu J, Zhang S, Yu S, An G, Wang Y, Yu Y, Liang L, Wang Y, Xu X, Xiong Y, Shao D, Shi Z, Li N, Wang J, Jin D, Liu T, Cui Y. Nivolumab plus anlotinib hydrochloride in advanced gastric adenocarcinoma and esophageal squamous cell carcinoma: the phase II OASIS trial. Nat Commun 2024; 15:8876. [PMID: 39406730 PMCID: PMC11480398 DOI: 10.1038/s41467-024-53109-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024] Open
Abstract
Vascular endothelial growth factor inhibitors, including tyrosine kinase inhibitors (TKIs), possess immunomodulatory properties and have shown promising outcomes when combined with anti-PD-1 antibodies. The OASIS phase II trial (NCT04503967) is designed to determine the clinical activity and safety of nivolumab (anti-PD-1) and anlotinib hydrochloride (a multi-targets TKI) as second-line or above therapy in patients with advanced gastric adenocarcinoma (GAC) and esophageal squamous cell carcinoma (ESCC). From December 2020 to September 2022, 45 patients with GAC and 3 with ESCC were enrolled in this study. The pre-specified endpoints were reached, with the primary endpoint of overall response rate achieving 29.2%. For secondary objectives, disease control rate was 64.6%; median progression-free survival was 4.0 months; and median overall survival was 11.1 months with a manageable toxicity profile. The exploratory analyses unveiled that the balance of gut bacteria and the presence of a pre-existing immune signature characterized by a high percentage of CD68+PD-L1+ PD-1+ macrophages and low pretreatment variant allele frequencies (VAF), as well as low expression of certain cytokines were significantly associated with improved clinical outcomes in patients with GAC.
Collapse
Affiliation(s)
- Jing Wu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Medical Oncology, Shanghai geriatric medical center, Shanghai, China
| | - Shilong Zhang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Shan Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Guo An
- BGI Genomics, Shenzhen, China
- Clin Lab, BGI Genomics, Shanghai, China
| | - Yi Wang
- Department of Tumor Screening and Prevention, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yiyi Yu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Li Liang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaojing Xu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - YanShi Xiong
- BGI Genomics, Shenzhen, China
- Clin Lab, BGI Genomics, Shanghai, China
| | - Di Shao
- BGI Genomics, Shenzhen, China
| | | | - Nannan Li
- BGI Genomics, Shenzhen, China
- BGI Research, Shenzhen, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jingyuan Wang
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dawei Jin
- BGI Genomics, Shenzhen, China.
- Clin Lab, BGI Genomics, Shanghai, China.
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Yuehong Cui
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
4
|
Botta GP, Abdelrahim M, Drengler RL, Aushev VN, Esmail A, Laliotis G, Brewer CM, George GV, Abbate SM, Chandana SR, Tejani MA, Malla M, Bansal D, Rivero-Hinojosa S, Spickard E, McCormick N, Cecchini M, Lacy J, Fei N, Kasi PM, Kasi A, Dayyani F, Hanna DL, Sharma S, Malhotra M, Aleshin A, Liu MC, Jurdi A. Association of personalized and tumor-informed ctDNA with patient survival outcomes in pancreatic adenocarcinoma. Oncologist 2024; 29:859-869. [PMID: 39022993 PMCID: PMC11449101 DOI: 10.1093/oncolo/oyae155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 04/30/2024] [Indexed: 07/20/2024] Open
Abstract
INTRODUCTION Personalized and tumor-informed circulating tumor DNA (ctDNA) testing is feasible and allows for molecular residual disease (MRD) identification in patients with pancreatic ductal adenocarcinoma (PDAC). METHODS In this retrospective analysis of commercial cases from multiple US institutions, personalized, tumor-informed, whole-exome sequenced, and germline-controlled ctDNA levels were quantified and analyzed in patients with PDAC. Plasma samples (n = 1329) from 298 clinically validated patients were collected at diagnosis, perioperatively (MRD-window; within 2-12 weeks after surgery, before therapy), and during surveillance (>12 weeks post-surgery if no ACT or starting 4 weeks post-ACT) from November 2019 to March 2023. RESULTS Of the initially diagnosed patients with stages I-III PDAC who went for resection, the median follow-up time from surgery was 13 months (range 0.1-214). Positive ctDNA detection rates were 29% (29/100) and 29.6% (45/152) during the MRD and surveillance windows, respectively. Positive ctDNA detection was significantly associated with shorter DFS within the MRD window (median DFS of 6.37 months for ctDNA-positive vs 33.31 months for ctDNA-negative patients; HR: 5.45, P < .0001) as well as during the surveillance period (median DFS: 11.40 months for ctDNA-positive vs NR for ctDNA-negative; HR: 12.38, P < .0001). Additionally, DFS was significantly better with KRAS wildtype status followed by KRASG12R (HR: 0.99, P = .97), KRASG12D (HR: 1.42, P = .194), and worse with KRASG12V (HR: 2.19, P = .002) status. In multivariate analysis, ctDNA detection at surveillance was found to be the most significant prognostic factor for recurrence (HR: 24.28, P < .001). CONCLUSIONS Perioperative tumor-informed ctDNA detection in PDAC is feasible across all stages and is associated with patient survival outcomes.
Collapse
Affiliation(s)
- Gregory P Botta
- Division of Hematology and Oncology, Department of Medicine, University of
California San Diego Moores Cancer Center, La Jolla, CA
92037, United States
| | - Maen Abdelrahim
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist
Cancer Center, Houston, TX 77030, United States
| | - Ronald L Drengler
- Medical Oncology, The START Center for Cancer Care,
San Antonio, TX 78229, United
States
| | | | - Abdullah Esmail
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist
Cancer Center, Houston, TX 77030, United States
| | | | | | - Giby V George
- Oncology, Natera, Inc., Austin, TX
78753, United States
| | - Steven M Abbate
- Medical Oncology, The START Center for Cancer Care,
San Antonio, TX 78229, United
States
| | - Sreenivasa R Chandana
- Department of Gastrointestinal Medical Oncology, Cancer & Hematology
Centers of Western Michigan, Grand Rapids, MI
49546, United States
| | - Mohamedtaki A Tejani
- Hematology and Oncology, Advent Health Cancer Institute,
Orlando, FL 32804, United
States
| | - Midhun Malla
- Section of Hematology and Oncology, University of Alabama at
Birmingham, AL 35233,United
States
| | - Dhruv Bansal
- Department of Hematology-Oncology, Saint Luke’s Cancer
Institute, Kansas City, MO 64111, United States
| | | | - Erik Spickard
- Oncology, Natera, Inc., Austin, TX
78753, United States
| | | | - Michael Cecchini
- Department of Internal Medicine (Medical Oncology), Yale University
School of Medicine, New Haven, CT 06520, United States
| | - Jill Lacy
- Department of Internal Medicine (Medical Oncology), Yale University
School of Medicine, New Haven, CT 06520, United States
| | - Naomi Fei
- Division of Hematology, Oncology and Blood and Marrow Transplantation,
University of Iowa Hospitals and Clinics, Iowa City, IA
52242, United States
| | - Pashtoon Murtaza Kasi
- Division of Internal Medicine, Department of Oncology/Hematology, Weill
Cornell Medicine, New York, NY 10021, United States
| | - Anup Kasi
- Division of Medical Oncology, Department of Medicine, Kansas University
Cancer Center, Kansas City, KS 66160, United States
| | - Farshid Dayyani
- Chao Family Comprehensive Cancer Center, University of California
Irvine, Orange, CA 92868, United States
| | - Diana L Hanna
- Division of Medical Oncology, Norris Cancer Center, Keck Medicine of
USC, Los Angeles, CA 90033, United States
| | - Shruti Sharma
- Oncology, Natera, Inc., Austin, TX
78753, United States
| | | | | | - Minetta C Liu
- Oncology, Natera, Inc., Austin, TX
78753, United States
| | - Adham Jurdi
- Oncology, Natera, Inc., Austin, TX
78753, United States
| |
Collapse
|
5
|
Munnings R, Gibbs P, Lee B. Evolution of Liquid Biopsies for Detecting Pancreatic Cancer. Cancers (Basel) 2024; 16:3335. [PMID: 39409954 PMCID: PMC11475855 DOI: 10.3390/cancers16193335] [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: 09/06/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a lethal malignancy characterised by late diagnosis and poor prognosis. Despite advancements, current diagnostic and prognostic strategies remain limited. Liquid biopsy techniques, including circulating tumour DNA (ctDNA), circulating tumour cells (CTCs), circulating tumour exosomes, and proteomics, offer potential solutions to improve PDAC diagnosis, prognostication, and management. A systematic search of Ovid MEDLINE identified studies published between 2019 and 2024, focusing on liquid biopsy biomarkers for PDAC. A total of 49 articles were included. ctDNA research shows some promise in diagnosing and prognosticating PDAC, especially through detecting mutant KRAS in minimal residual disease assays. CTC analyses had low sensitivity for early-stage PDAC and inconsistent prognostic results across subpopulations. Exosomal studies revealed diverse biomarkers with some diagnostic and prognostic potential. Proteomics, although relatively novel, has demonstrated superior accuracy in PDAC diagnosis, including early detection, and notable prognostic capacity. Proteomics combined with CA19-9 analysis has shown the most promising results to date. An update on multi-cancer early detection testing, given its significance for population screening, is also briefly discussed. Liquid biopsy techniques offer promising avenues for improving PDAC diagnosis, prognostication, and management. In particular, proteomics shows considerable potential, yet further research is needed to validate existing findings and comprehensively explore the proteome using an unbiased approach.
Collapse
Affiliation(s)
- Ryan Munnings
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Department of Medical Education, Melbourne Medical School, Parkville, VIC 3052, Australia
| | - Peter Gibbs
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Western Health, Footscray, VIC 3011, Australia
| | - Belinda Lee
- Walter & Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
- Peter MacCallum Cancer Centre, Parkville, VIC 3052, Australia
- Northern Health, Epping, VIC 3076, Australia
| |
Collapse
|
6
|
Aliseda D, Martí-Cruchaga P, Zozaya G, Blanco N, Ponz M, Chopitea A, Rodríguez J, Castañón E, Pardo F, Rotellar F. Neoadjuvant therapy versus upfront surgery in resectable pancreatic cancer: reconstructed patient-level meta-analysis of randomized clinical trials. BJS Open 2024; 8:zrae087. [PMID: 39329454 PMCID: PMC11428068 DOI: 10.1093/bjsopen/zrae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 05/22/2024] [Accepted: 07/03/2024] [Indexed: 09/28/2024] Open
Abstract
BACKGROUND Neoadjuvant treatment has shown promising results in patients with borderline resectable pancreatic ductal adenocarcinoma. The potential benefits of neoadjuvant treatment on long-term overall survival in patients with resectable pancreatic ductal adenocarcinoma have not yet been established. The aim of this study was to compare long-term overall survival of patients with resectable pancreatic ductal adenocarcinoma based on whether they received neoadjuvant treatment or underwent upfront surgery. METHODS A systematic review including randomized clinical trials on the overall survival outcomes between neoadjuvant treatment and upfront surgery in patients with resectable pancreatic ductal adenocarcinoma was conducted up to 1 August 2023 from PubMed, MEDLINE and Web of Science databases. Patient-level survival data was extracted and reconstructed from available Kaplan-Meier curves. A frequentist one-stage meta-analysis was employed, using Cox-based models and a non-parametric method (restricted mean survival time), to assess the difference in overall survival between groups. A Bayesian meta-analysis was also conducted. RESULTS Five randomized clinical trials comprising 625 patients were included. Among patients with resectable pancreatic ductal adenocarcinoma, neoadjuvant treatment was not significantly associated with a reduction in the hazard of death compared with upfront surgery (shared frailty HR 0.88, 95% c.i. 0.72 to 1.08, P = 0.223); this result was consistent in the non-parametric restricted mean survival time model (+2.41 months, 95% c.i. -1.22 to 6.04, P < 0.194), in the sensitivity analysis that excluded randomized clinical trials with a high risk of bias (shared frailty HR 0.91 (95% c.i. 0.72 to 1.15; P = 0.424)) and in the Bayesian analysis with a posterior shared frailty HR of 0.86 (95% c.i. 0.70 to 1.05). CONCLUSION Neoadjuvant treatment does not demonstrate a survival advantage over upfront surgery for patients with resectable pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Daniel Aliseda
- HPB and Liver Transplant Unit, Department of General Surgery, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Pablo Martí-Cruchaga
- HPB and Liver Transplant Unit, Department of General Surgery, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
| | - Gabriel Zozaya
- HPB and Liver Transplant Unit, Department of General Surgery, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
| | - Nuria Blanco
- HPB and Liver Transplant Unit, Department of General Surgery, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
| | - Mariano Ponz
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
- Department of Oncology, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Ana Chopitea
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
- Department of Oncology, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Javier Rodríguez
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
- Department of Oncology, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Eduardo Castañón
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
- Department of Oncology, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
| | - Fernando Pardo
- HPB and Liver Transplant Unit, Department of General Surgery, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
| | - Fernando Rotellar
- HPB and Liver Transplant Unit, Department of General Surgery, Clinica Universidad de Navarra, University of Navarra, Pamplona, Spain
- Institute of Health Research of Navarra (IdisNA), Pamplona, Spain
| |
Collapse
|
7
|
Theparee T, Akroush M, Sabatini LM, Wang V, Mangold KA, Joseph N, Stocker SJ, Freedman A, Helseth DL, Talamonti MS, Kaul KL. Cell free DNA in patients with pancreatic adenocarcinoma: clinicopathologic correlations. Sci Rep 2024; 14:15744. [PMID: 38977725 PMCID: PMC11231234 DOI: 10.1038/s41598-024-65562-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 06/20/2024] [Indexed: 07/10/2024] Open
Abstract
Detection of circulating tumor DNA (ctDNA) from plasma cell free DNA (cfDNA) has shown promise for diagnosis, therapeutic targeting, and prognosis. This study explores ctDNA detection by next generation sequencing (NGS) and associated clinicopathologic factors in patients with pancreatic adenocarcinoma (PDAC). Patients undergoing surgical exploration or resection of pancreatic lesions were enrolled with informed consent. Plasma samples (4-6 ml) were collected prior to surgery and cfDNA was recovered from 95 plasma samples. Adequate cfDNA for NGS (20 ng) was obtained from 81 patients. NGS was performed using the Oncomine Lung cfDNA assay on the Ion Torrent S5 sequencing platform. Twenty-five patients (30.9%) had detectable mutations in KRAS and/or TP53 with allele frequencies ranging from 0.05 to 8.5%, while mutations in other genes were detected less frequently and always along with KRAS or TP53. Detectable ctDNA mutations were more frequent in patients with poorly differentiated tumors, and patients without detectable ctDNA mutations showed longer survival (medians of 10.5 months vs. 18 months, p = 0.019). The detection of circulating tumor DNA in pancreatic adenocarcinomas is correlated with worse survival outcomes.
Collapse
Affiliation(s)
- Talent Theparee
- Department of Pathology and Laboratory Medicine, NorthShore University Health System, Evanston, IL, USA
- Department of Pathology, Chulalongkorn University Faculty of Medicine, Bangkok, Thailand
| | - Michael Akroush
- Department of Pathology and Laboratory Medicine, NorthShore University Health System, Evanston, IL, USA
| | - Linda M Sabatini
- Department of Pathology and Laboratory Medicine, NorthShore University Health System, Evanston, IL, USA
| | - Vivien Wang
- Department of Pathology and Laboratory Medicine, NorthShore University Health System, Evanston, IL, USA
| | - Kathy A Mangold
- Department of Pathology and Laboratory Medicine, NorthShore University Health System, Evanston, IL, USA
| | - Nora Joseph
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI, USA
| | - Susan Jane Stocker
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA
| | - Alexa Freedman
- Northwestern University School of Medicine, Chicago, IL, USA
| | - Donald L Helseth
- Department of Pathology and Laboratory Medicine, NorthShore University Health System, Evanston, IL, USA
| | - Mark S Talamonti
- Department of Surgery, NorthShore University Health System, Evanston, IL, USA
| | - Karen L Kaul
- Department of Pathology and Laboratory Medicine, NorthShore University Health System, Evanston, IL, USA.
| |
Collapse
|
8
|
Turabi K, Klute K, Radhakrishnan P. Decoding the Dynamics of Circulating Tumor DNA in Liquid Biopsies. Cancers (Basel) 2024; 16:2432. [PMID: 39001494 PMCID: PMC11240538 DOI: 10.3390/cancers16132432] [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: 05/25/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
Circulating tumor DNA (ctDNA), a fragment of tumor DNA found in the bloodstream, has emerged as a revolutionary tool in cancer management. This review delves into the biology of ctDNA, examining release mechanisms, including necrosis, apoptosis, and active secretion, all of which offer information about the state and nature of the tumor. Comprehensive DNA profiling has been enabled by methods such as whole genome sequencing and methylation analysis. The low abundance of the ctDNA fraction makes alternative techniques, such as digital PCR and targeted next-generation exome sequencing, more valuable and accurate for mutation profiling and detection. There are numerous clinical applications for ctDNA analysis, including non-invasive liquid biopsies for minimal residual disease monitoring to detect cancer recurrence, personalized medicine by mutation profiling for targeted therapy identification, early cancer detection, and real-time evaluation of therapeutic response. Integrating ctDNA analysis into routine clinical practice creates promising avenues for successful and personalized cancer care, from diagnosis to treatment and follow-up.
Collapse
Affiliation(s)
- Khadija Turabi
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Kelsey Klute
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
9
|
Lee MR, Woo SM, Kim MK, Han S, Park S, Lee WJ, Lee D, Choi SI, Choi W, Yoon K, Chun JW, Kim Y, Kong S. Application of plasma circulating KRAS mutations as a predictive biomarker for targeted treatment of pancreatic cancer. Cancer Sci 2024; 115:1283-1295. [PMID: 38348576 PMCID: PMC11007020 DOI: 10.1111/cas.16104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/11/2024] [Accepted: 01/27/2024] [Indexed: 04/12/2024] Open
Abstract
Kirsten rat sarcoma viral oncogene homolog (KRAS) mutations in circulating tumor deoxyribonucleic acid (ctDNA) have been reported as representative noninvasive prognostic markers for pancreatic ductal adenocarcinoma (PDAC). Here, we aimed to evaluate single KRAS mutations as prognostic and predictive biomarkers, with an emphasis on potential therapeutic approaches to PDAC. A total of 128 patients were analyzed for multiple or single KRAS mutations (G12A, G12C, G12D, G12R, G12S, G12V, and G13D) in their tumors and plasma using droplet digital polymerase chain reaction (ddPCR). Overall, KRAS mutations were detected by multiplex ddPCR in 119 (93%) of tumor DNA and 68 (53.1%) of ctDNA, with a concordance rate of 80% between plasma ctDNA and tumor DNA in the metastatic stage, which was higher than the 44% in the resectable stage. Moreover, the prognostic prediction of both overall survival (OS) and progression-free survival (PFS) was more relevant using plasma ctDNA than tumor DNA. Further, we evaluated the selective tumor-suppressive efficacy of the KRAS G12C inhibitor sotorasib in a patient-derived organoid (PDO) from a KRAS G12C-mutated patient using a patient-derived xenograft (PDX) model. Sotorasib showed selective inhibition in vitro and in vivo with altered tumor microenvironment, including fibroblasts and macrophages. Collectively, screening for KRAS single mutations in plasma ctDNA and the use of preclinical models of PDO and PDX with genetic mutations would impact precision medicine in the context of PDAC.
Collapse
Affiliation(s)
- Mi Rim Lee
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Molecular Imaging Branch, Division of Convergence TechnologyResearch Institute of National Cancer CenterGoyangKorea
| | - Sang Myung Woo
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
- Immuno‐Oncology Branch, Division of Rare and Refractory CenterResearch Institute of National Cancer CenterGoyangKorea
| | - Min Kyeong Kim
- Targeted Therapy Branch, Division of Rare and Refractory CenterResearch Institute of National Cancer CenterGoyangKorea
| | - Sung‐Sik Han
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
| | - Sang‐Jae Park
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
| | - Woo Jin Lee
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
- Interventional Medicine Branch, Division of Clinical ResearchResearch Institute of National Cancer CenterGoyangKorea
| | - Dong‐eun Lee
- Biostatistics Collaboration TeamResearch Core Center, National Cancer CenterGoyangKorea
| | - Sun Il Choi
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Molecular Imaging Branch, Division of Convergence TechnologyResearch Institute of National Cancer CenterGoyangKorea
- Henan Key Laboratory of Brain Targeted Bio‐Nanomedicine, School of Life Sciences & School of PharmacyHenan UniversityKaifengHenanChina
| | - Wonyoung Choi
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Center for Clinical Trials, Hospital, National Cancer CenterGoyangKorea
- Cancer Molecular Biology Branch, Division of Cancer BiologyResearch Institute of National Cancer CenterGoyangKorea
| | - Kyong‐Ah Yoon
- College of Veterinary MedicineKonkuk UniversitySeoulKorea
| | - Jung Won Chun
- Center for Liver and Pancreatobiliary Cancer, Hospital, National Cancer CenterGoyangKorea
- Interventional Medicine Branch, Division of Clinical ResearchResearch Institute of National Cancer CenterGoyangKorea
| | - Yun‐Hee Kim
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Molecular Imaging Branch, Division of Convergence TechnologyResearch Institute of National Cancer CenterGoyangKorea
| | - Sun‐Young Kong
- Department of Cancer Biomedical ScienceNational Cancer Center Graduate School of Cancer Science and PolicyGoyangKorea
- Targeted Therapy Branch, Division of Rare and Refractory CenterResearch Institute of National Cancer CenterGoyangKorea
- Department of Laboratory MedicineHospital, National Cancer CenterGoyangKorea
| |
Collapse
|
10
|
Adashek JJ, Kato S, Sicklick JK, Lippman SM, Kurzrock R. If it's a target, it's a pan-cancer target: Tissue is not the issue. Cancer Treat Rev 2024; 125:102721. [PMID: 38522181 PMCID: PMC11093268 DOI: 10.1016/j.ctrv.2024.102721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 03/26/2024]
Abstract
Cancer is traditionally diagnosed and treated on the basis of its organ of origin (e.g., lung or colon cancer). However, organ-of-origin diagnostics does not reveal the underlying oncogenic drivers. Fortunately, molecular diagnostics have advanced at a breathtaking pace, and it is increasingly apparent that cancer is a disease of the genome. Hence, we now have multiple genomic biomarker-based, tissue-agnostic Food and Drug Administration approvals for both gene- and immune-targeted therapies (larotrectinib/entrectinib, for NTRK fusions; selpercatinib, RET fusions; dabrafenib plus trametinib, BRAFV600E mutations; pembrolizumab/dostarlimab, microsatellite instability; and pembrolizumab for high tumor mutational burden; pemigatinib is also approved for FGFR1-rearranged myeloid/lymphoid neoplasms). There are emerging targets as well, including but not limited to ALK, BRCA and/or homologous repair deficiency, ERBB2 (HER2), IDH1/2, KIT, KRASG12C, NRG1, and VHL. Many tissue-agnostic approvals center on rare/ultra-rare biomarkers (often < 1 % of cancers), necessitating screening hundreds of tumors to find a single one harboring the cognate molecular alteration. Approval has generally been based on small single-arm studies (<30-100 patients) with high response rates (>30 % to > 75 %) of remarkable durability. Because of biomarker rarity, single-gene testing is not practical; next generation sequencing of hundreds of genes must be performed to obtain timely answers. Resistance to biomarker-driven therapeutics is often due to secondary mutations or co-driver gene defects; studies are now addressing the need for customized drug combinations matched to the complex molecular alteration portfolio in each tumor. Future investigation should expand tissue-agnostic therapeutics to encompass both hematologic and solid malignancies and include biomarkers beyond those that are DNA-based.
Collapse
Affiliation(s)
- Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA; Department of Surgery, Division of Surgical Oncology, University of California San Diego, UC San Diego Health, San Diego, CA, USA; Department of Pharmacology, University of California San Diego, UC San Diego Health, San Diego, CA, USA
| | - Scott M Lippman
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee Wisconsin, USA; WIN Consortium, Paris France; University of Nebraska, United States.
| |
Collapse
|
11
|
Huang L, Lv Y, Guan S, Yan H, Han L, Wang Z, Han Q, Dai G, Shi Y. High somatic mutations in circulating tumor DNA predict response of metastatic pancreatic ductal adenocarcinoma to first-line nab-paclitaxel plus S-1: prospective study. J Transl Med 2024; 22:184. [PMID: 38378604 PMCID: PMC10877900 DOI: 10.1186/s12967-024-04989-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 02/12/2024] [Indexed: 02/22/2024] Open
Abstract
AIMS We previously showed that the nab-paclitaxel plus S-1 (NPS) regimen had promising effects against metastatic pancreatic ducal adenocarcinoma (mPDAC), whose efficacy however could not be precisely predicted by routine biomarkers. This prospective study aimed to investigate the values of mutations in circulating tumor DNA (ctDNA) and their dynamic changes in predicting response of mPDAC to NPS chemotherapy. METHODS Paired tumor tissue and blood samples were prospectively collected from patients with mPDAC receiving first-line NPS chemotherapy, and underwent next-generation sequencing with genomic profiling of 425 genes for ctDNA. High mutation allelic frequency (MAF) was defined as ≥ 30% and ≥ 5% in tumor tissue and blood, respectively. Kappa statistics were used to assess agreement between mutant genes in tumor and ctDNA. Associations of mutations in ctDNA and their dynamic changes with tumor response, overall survival (OS), and progression-free survival (PFS) were assessed using the Kaplan-Meier method, multivariable-adjusted Cox proportional hazards regression, and longitudinal data analysis. RESULTS 147 blood samples and 43 paired tumor specimens from 43 patients with mPDAC were sequenced. The most common driver genes with high MAF were KRAS (tumor, 35%; ctDNA, 37%) and TP53 (tumor, 37%; ctDNA, 33%). Mutation rates of KRAS and TP53 in ctDNA were significantly higher in patients with liver metastasis, with baseline CA19-9 ≥ 2000 U/mL, and/or without an early CA19-9 response. κ values for the 5 most commonly mutated genes between tumor and ctDNA ranged from 0.48 to 0.76. MAFs of the genes mostly decreased sequentially during subsequent measurements, which significantly correlated with objective response, with an increase indicating cancer progression. High mutations of KRAS and ARID1A in both tumor and ctDNA, and of TP53, CDKN2A, and SMAD4 in ctDNA but not in tumor were significantly associated with shorter survival. When predicting 6-month OS, AUCs for the 5 most commonly mutated genes in ctDNA ranged from 0.59 to 0.84, larger than for genes in tumor (0.56 to 0.71) and for clinicopathologic characteristics (0.51 to 0.68). Repeated measurements of mutations in ctDNA significantly differentiated survival and tumor response. Among the 31 patients with ≥ 2 ctDNA tests, longitudinal analysis of changes in gene MAF showed that ctDNA progression was 60 and 58 days ahead of radiologic and CA19-9 progression for 48% and 42% of the patients, respectively. CONCLUSIONS High mutations of multiple driving genes in ctDNA and their dynamic changes could effectively predict response of mPDAC to NPS chemotherapy, with promising reliable predictive performance superior to routine clinicopathologic parameters. Inspiringly, longitudinal ctDNA tracking could predict disease progression about 2 months ahead of radiologic or CA19-9 evaluations, with the potential to precisely devise individualized therapeutic strategies for mPDAC.
Collapse
Affiliation(s)
- Lei Huang
- Medical Center on Aging of Ruijin Hospital, MCARJH, Shanghai Jiaotong University School of Medicine, 197 Ruijin Er Road, Shanghai, 200025, China.
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yao Lv
- Department of Medical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Shasha Guan
- Department of Medical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Huan Yan
- Department of Medical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Lu Han
- Department of Medical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China
| | - Zhikuan Wang
- Department of Medical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Quanli Han
- Department of Medical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Guanghai Dai
- Department of Medical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Yan Shi
- Department of General Surgery, Shanghai Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, 358 Datong Road, Gaoqiao Town, Shanghai, 200137, China.
| |
Collapse
|
12
|
Rompen IF, Habib JR, Wolfgang CL, Javed AA. Anatomical and Biological Considerations to Determine Resectability in Pancreatic Cancer. Cancers (Basel) 2024; 16:489. [PMID: 38339242 PMCID: PMC10854859 DOI: 10.3390/cancers16030489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/11/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains associated with poor outcomes with a 5-year survival of 12% across all stages of the disease. These poor outcomes are driven by a delay in diagnosis and an early propensity for systemic dissemination of the disease. Recently, aggressive surgical approaches involving complex vascular resections and reconstructions have become more common, thus allowing more locally advanced tumors to be resected. Unfortunately, however, even after the completion of surgery and systemic therapy, approximately 40% of patients experience early recurrence of disease. To determine resectability, many institutions utilize anatomical staging systems based on the presence and extent of vascular involvement of major abdominal vessels around the pancreas. However, these classification systems are based on anatomical considerations only and do not factor in the burden of systemic disease. By integrating the biological criteria, we possibly could avoid futile resections often associated with significant morbidity. Especially patients with anatomically resectable disease who have a heavy burden of radiologically undetected systemic disease most likely do not derive a survival benefit from resection. On the contrary, we could offer complex resections to those who have locally advanced or oligometastatic disease but have favorable systemic biology and are most likely to benefit from resection. This review summarizes the current literature on defining anatomical and biological resectability in patients with pancreatic cancer.
Collapse
Affiliation(s)
- Ingmar F. Rompen
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
- Department of General, Visceral, and Transplantation Surgery, Heidelberg University Hospital, 69120 Heidelberg, Germany
| | - Joseph R. Habib
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
| | - Christopher L. Wolfgang
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
| | - Ammar A. Javed
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA
| |
Collapse
|
13
|
Zhang XJ, Fei H, Sun CY, Li ZF, Li Z, Guo CG, Zhao DB. Novel prognostic score based on the preoperative total bilirubin-albumin ratio and fibrinogen-albumin ratio in ampullary adenocarcinoma. World J Gastrointest Surg 2023; 15:2247-2258. [PMID: 37969714 PMCID: PMC10642462 DOI: 10.4240/wjgs.v15.i10.2247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/22/2023] [Accepted: 09/04/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND The preoperative total bilirubin-albumin ratio (TBAR) and fibrinogen-albumin ratio (FAR) have been proven to be valuable prognostic factors in various cancers. AIM To detect the prognostic value of TBAR and FAR in ampullary adenocarcinoma (AC) patients who underwent curative pancreaticoduodenectomy. METHODS AC patients who underwent curative pancreaticoduodenectomy in the National Cancer Center of China between 1998 and 2020 were retrospectively reviewed. The prognostic cutoff values of TBAR and FAR were determined through the best survival separation model. Then, a novel prognostic score combining TBAR and FAR was calculated and validated through the logistic regression analysis and Cox regression analysis. RESULTS A total of 188 AC patients were enrolled in the current study. The best cutoff values of TBAR and FAR for predicting overall survival were 1.7943 and 0.1329, respectively. AC patients were divided into a TBAR-low group (score = 0) vs a TBAR-high group (score = 1) and a FAR-low group (score = 0) vs a FAR-high group (score = 1). The total score was calculated as a novel prognostic factor. Multivariable logistic regression analysis revealed that a high score was an independent protective factor for recurrence [score = 1 vs score = 0: Odds ratio (OR) = 0.517, P = 0.046; score = 2 vs score = 0 OR = 0.236, P = 0.038]. In addition, multivariable survival analysis also demonstrated that a high score was an independent protective factor in AC patients (score = 2 vs score = 0: Hazard ratio = 0.230, P = 0.046). CONCLUSION A novel prognostic score based on preoperative TBAR and FAR has been demonstrated to have good predictive power in AC patients who underwent curative pancreaticoduodenectomy. However, more studies with larger samples are needed to validate this conclusion.
Collapse
Affiliation(s)
- Xiao-Jie Zhang
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - He Fei
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chong-Yuan Sun
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ze-Feng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zheng Li
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Chun-Guang Guo
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Dong-Bing Zhao
- Department of Pancreatic and Gastric Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| |
Collapse
|
14
|
Ma Y, Gan J, Bai Y, Cao D, Jiao Y. Minimal residual disease in solid tumors: an overview. Front Med 2023; 17:649-674. [PMID: 37707677 DOI: 10.1007/s11684-023-1018-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/24/2023] [Indexed: 09/15/2023]
Abstract
Minimal residual disease (MRD) is termed as the small numbers of remnant tumor cells in a subset of patients with tumors. Liquid biopsy is increasingly used for the detection of MRD, illustrating the potential of MRD detection to provide more accurate management for cancer patients. As new techniques and algorithms have enhanced the performance of MRD detection, the approach is becoming more widely and routinely used to predict the prognosis and monitor the relapse of cancer patients. In fact, MRD detection has been shown to achieve better performance than imaging methods. On this basis, rigorous investigation of MRD detection as an integral method for guiding clinical treatment has made important advances. This review summarizes the development of MRD biomarkers, techniques, and strategies for the detection of cancer, and emphasizes the application of MRD detection in solid tumors, particularly for the guidance of clinical treatment.
Collapse
Affiliation(s)
- Yarui Ma
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jingbo Gan
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Yinlei Bai
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Dandan Cao
- Genetron Health (Beijing) Co. Ltd., Beijing, 102206, China
| | - Yuchen Jiao
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
15
|
Cassese G, Han HS, Yoon YS, Lee JS, Lee B, Cubisino A, Panaro F, Troisi RI. Role of neoadjuvant therapy for nonmetastatic pancreatic cancer: Current evidence and future perspectives. World J Gastrointest Oncol 2023; 15:911-924. [PMID: 37389109 PMCID: PMC10302990 DOI: 10.4251/wjgo.v15.i6.911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 02/17/2023] [Accepted: 04/24/2023] [Indexed: 06/14/2023] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is one of the most common and lethal human cancers worldwide. Surgery followed by adjuvant chemotherapy offers the best chance of a long-term survival for patients with PDAC, although only approximately 20% of the patients have resectable tumors when diagnosed. Neoadjuvant chemotherapy (NACT) is recommended for borderline resectable pancreatic cancer. Several studies have investigated the role of NACT in treating resectable tumors based on the recent advances in PDAC biology, as NACT provides the potential benefit of selecting patients with favorable tumor biology and controls potential micro-metastases in high-risk patients with resectable PDAC. In such challenging cases, new potential tools, such as ct-DNA and molecular targeted therapy, are emerging as novel therapeutic options that may improve old paradigms. This review aims to summarize the current evidence regarding the role of NACT in treating non-metastatic pancreatic cancer while focusing on future perspectives in light of recent evidence.
Collapse
Affiliation(s)
- Gianluca Cassese
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive HPB Surgery and Transplantation Service, Federico II University Hospital, Naples 80131, Italy
| | - Ho-Seong Han
- Department of Surgery, Seoul National University College of Medicine, Seongnam 13620, Gyeonggi-do, South Korea
| | - Yoo-Seok Yoon
- Department of Surgery, Seoul National University College of Medicine, Seongnam 13620, Gyeonggi-do, South Korea
| | - Jun Suh Lee
- Department of Surgery, Seoul National University College of Medicine, Seongnam 13620, Gyeonggi-do, South Korea
| | - Boram Lee
- Department of Surgery, Seoul National University College of Medicine, Seongnam 13620, Gyeonggi-do, South Korea
| | - Antonio Cubisino
- Department of HPB Surgery and Transplantation, Beaujon Hospital, Clichy 92110, France
| | - Fabrizio Panaro
- Department of Digestive Surgery and Liver Transplantation, CHU Montpellier, Montpellier 34100, France
| | - Roberto Ivan Troisi
- Department of Clinical Medicine and Surgery, Division of Minimally Invasive HPB Surgery and Transplantation Service, Federico II University Hospital, Naples 80131, Italy
| |
Collapse
|
16
|
Zhang Z, Zhang H, Liao X, Tsai HI. KRAS mutation: The booster of pancreatic ductal adenocarcinoma transformation and progression. Front Cell Dev Biol 2023; 11:1147676. [PMID: 37152291 PMCID: PMC10157181 DOI: 10.3389/fcell.2023.1147676] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/10/2023] [Indexed: 05/09/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most common type of pancreatic cancer. It has a poor response to conventional therapy and has an extremely poor 5-year survival rate. PDAC is driven by multiple oncogene mutations, with the highest mutation frequency being observed in KRAS. The KRAS protein, which binds to GTP, has phosphokinase activity, which further activates downstream effectors. KRAS mutation contributes to cancer cell proliferation, metabolic reprogramming, immune escape, and therapy resistance in PDAC, acting as a critical driver of the disease. Thus, KRAS mutation is positively associated with poorer prognosis in pancreatic cancer patients. This review focus on the KRAS mutation patterns in PDAC, and further emphases its role in signal transduction, metabolic reprogramming, therapy resistance and prognosis, hoping to provide KRAS target therapy strategies for PDAC.
Collapse
Affiliation(s)
- Zining Zhang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Heng Zhang
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Xiang Liao
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Hsiang-i Tsai
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
- Department of Medical Imaging, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
17
|
Kondo T, Kanai M, Matsubara J, Yamaguchi D, Ura T, Kou T, Itani T, Nomura M, Funakoshi T, Yokoyama A, Doi K, Tamaoki M, Yoshimura M, Uza N, Yamada T, Masui T, Minamiguchi S, Matsumoto S, Ishikawa H, Muto M. Association between homologous recombination gene variants and efficacy of oxaliplatin-based chemotherapy in advanced pancreatic cancer: prospective multicenter observational study. Med Oncol 2023; 40:144. [PMID: 37039943 DOI: 10.1007/s12032-023-02011-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023]
Abstract
Next-generation sequencing (NGS)-based gene profiling can identify patients with pancreatic cancer with homologous recombinant repair gene pathogenic variants (HRRv). Several retrospective studies have reported a positive association between HRRv and the efficacy of platinum-based chemotherapy. However, this association remains to be validated in a prospective study. This multicenter, prospective, observational study included patients with histologically confirmed unresectable or recurrent pancreatic cancer who required systemic chemotherapy. Patients who were oxaliplatin-naïve patients were eligible. The HRRv status was measured using a College of American Pathologists-accredited NGS panel. One-year overall survival rate (1yr-OS%) was calculated after initiation of oxaliplatin-based chemotherapy and was set as the primary endpoint. Forty patients were enrolled between August 2018 and March 2020. The NGS success rate was 95% (38/40). HRRv was detected in 11 patients (27.5%). Oxaliplatin-based chemotherapy was administered to 9 of 11 patients with HRRv (81.8%) and 15 of 29 patients with non-HRRv (51.7%). The 1yr-OS% after initiation of oxaliplatin-based chemotherapy was 44.4% [95% confidence interval (CI) 13.7-71.9] and 57.1% (95% CI 28.4-78.0) in HRRv-positive and -negative cohorts, respectively. These data suggested that HRRv status alone could not be a potential predictive marker of oxaliplatin-based chemotherapy in patients with advanced pancreatic cancer. These results were in line with the results of a recent phase II study reporting the limited efficacy of poly(adenosine diphosphate-ribose) polymerase inhibitor in patients with pancreatic cancer who harbored HRRv other than BRCA. Future studies investigating patients with biallelic HRRv in the first-line setting are warranted.Trial registration UMIN000033655.
Collapse
Affiliation(s)
- Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Masashi Kanai
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan.
| | - Junichi Matsubara
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Daisuke Yamaguchi
- Department of Medical Oncology, Kyoto-Katsura Hospital, Kyoto, Japan
| | - Takashi Ura
- Department of Clinical Oncology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Tadayuki Kou
- Department of Gastroenterology and Hepatology, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Toshinao Itani
- Department of Gastroenterology, Kobe City Nishi-Kobe Medical Center, Hyogo, Japan
| | - Motoo Nomura
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Taro Funakoshi
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Akira Yokoyama
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Keitaro Doi
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Masashi Tamaoki
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Michio Yoshimura
- Department of Radiation Oncology and Image-Applied Therapy, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Takahiro Yamada
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
- Division of Clinical Genetics, Hokkaido University Hospital, Hokkaido, Japan
| | - Toshihiko Masui
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Shigemi Matsumoto
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Hideki Ishikawa
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Manabu Muto
- Department of Clinical Oncology, Kyoto University Hospital, 54 Kawahara-Cho, Shogoin, Sakyo-Ku, Kyoto, 606-8507, Japan
| |
Collapse
|
18
|
Kung H, Yu J. Targeted therapy for pancreatic ductal adenocarcinoma: Mechanisms and clinical study. MedComm (Beijing) 2023; 4:e216. [PMID: 36814688 PMCID: PMC9939368 DOI: 10.1002/mco2.216] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 02/21/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and lethal malignancy with a high rate of recurrence and a dismal 5-year survival rate. Contributing to the poor prognosis of PDAC is the lack of early detection, a complex network of signaling pathways and molecular mechanisms, a dense and desmoplastic stroma, and an immunosuppressive tumor microenvironment. A recent shift toward a neoadjuvant approach to treating PDAC has been sparked by the numerous benefits neoadjuvant therapy (NAT) has to offer compared with upfront surgery. However, certain aspects of NAT against PDAC, including the optimal regimen, the use of radiotherapy, and the selection of patients that would benefit from NAT, have yet to be fully elucidated. This review describes the major signaling pathways and molecular mechanisms involved in PDAC initiation and progression in addition to the immunosuppressive tumor microenvironment of PDAC. We then review current guidelines, ongoing research, and future research directions on the use of NAT based on randomized clinical trials and other studies. Finally, the current use of and research regarding targeted therapy for PDAC are examined. This review bridges the molecular understanding of PDAC with its clinical significance, development of novel therapies, and shifting directions in treatment paradigm.
Collapse
Affiliation(s)
- Heng‐Chung Kung
- Krieger School of Arts and SciencesJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Jun Yu
- Departments of Medicine and OncologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
19
|
Umemoto K, Sunakawa Y, Ueno M, Furukawa M, Mizuno N, Sudo K, Kawamoto Y, Kajiwara T, Ohtsubo K, Okano N, Matsuhashi N, Itoh S, Matsumoto T, Shimizu S, Otsuru T, Hasegawa H, Okuyama H, Ohama H, Moriwaki T, Ohta T, Odegaard JI, Nakamura Y, Bando H, Yoshino T, Ikeda M, Morizane C. Clinical significance of circulating-tumour DNA analysis by metastatic sites in pancreatic cancer. Br J Cancer 2023; 128:1603-1608. [PMID: 36782009 PMCID: PMC10070329 DOI: 10.1038/s41416-023-02189-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Liquid biopsy is an alternative to tissue specimens for tumour genotyping. However, the frequency of genomic alterations with low circulating-tumour DNA (ctDNA) shedding is shown in pancreatic ductal adenocarcinoma (PDAC). We, therefore, investigated the prevalence of KRAS mutations and ctDNA fraction by the metastatic site in patients with PDAC. METHODS This study enrolled previously treated PDAC patients from a plasma genomic profiling study; ctDNA analysis was performed using Guardant360 at disease progression before initiating subsequent treatment. RESULTS In 512 patients with PDAC, KRAS mutations were detected in 57%. The frequency of KRAS mutation in ctDNA differed depending on the metastatic organ; among patients with single-organ metastasis (n = 296), KRAS mutation detection rate was significantly higher in patients with metastasis to the liver (78%). In addition, the median maximum variant allele frequency (VAF) was higher with metastasis to the liver (1.9%) than with metastasis to the lungs, lymph nodes, peritoneum or with locally advanced disease (0.2%, 0.4%, 0.2% and 0.3%, respectively). CONCLUSION The prevalence of KRAS mutations and maximum VAF were higher in patients with metastasis to the liver than in those with metastasis to other sites. This study indicated the clinical utility of ctDNA analysis, especially in PDAC with liver metastases.
Collapse
Affiliation(s)
- Kumiko Umemoto
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan.
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Masayuki Furukawa
- Department of Hepato-Biliary-Pancreatology, National Hospital Organization Kyushu Cancer Center, Fukuoka, Japan
| | - Nobumasa Mizuno
- Department of Gastroenterology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Kentaro Sudo
- Department of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Yasuyuki Kawamoto
- Division of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Koushiro Ohtsubo
- Division of Medical Oncology, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Mitaka, Japan
| | | | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toshihiko Matsumoto
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Satoshi Shimizu
- Department of Gastroenterology, Saitama Cancer Center, Saitama, Japan
| | - Toru Otsuru
- Department of Gastroenterological Surgery, Osaka University Graduate School of Medicine, Suita, Japan
| | - Hiroko Hasegawa
- Department of Gastroenterology and Hepatology, National Hospital Organization, Osaka National Hospital, Osaka, Japan
| | - Hiroyuki Okuyama
- Department of Clinical Oncology, Kagawa University, Kagawa, Japan
| | - Hideko Ohama
- Department of Gastroenterology, Osaka Medical College, Osaka, Japan
| | - Toshikazu Moriwaki
- Department of Gastroenterology, University of Tsukuba Hospital, Tsukuba, Japan
| | - Takashi Ohta
- Department of Clinical Oncology, Kansai Rosai Hospital, Hyogo, Japan
| | | | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Section, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masafumi Ikeda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
20
|
David P, Mittelstädt A, Kouhestani D, Anthuber A, Kahlert C, Sohn K, Weber GF. Current Applications of Liquid Biopsy in Gastrointestinal Cancer Disease-From Early Cancer Detection to Individualized Cancer Treatment. Cancers (Basel) 2023; 15:cancers15071924. [PMID: 37046585 PMCID: PMC10093361 DOI: 10.3390/cancers15071924] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 04/14/2023] Open
Abstract
Worldwide, gastrointestinal (GI) cancers account for a significant amount of cancer-related mortality. Tests that allow an early diagnosis could lead to an improvement in patient survival. Liquid biopsies (LBs) due to their non-invasive nature as well as low risk are the current focus of cancer research and could be a promising tool for early cancer detection. LB involves the sampling of any biological fluid (e.g., blood, urine, saliva) to enrich and analyze the tumor's biological material. LBs can detect tumor-associated components such as circulating tumor DNA (ctDNA), extracellular vesicles (EVs), and circulating tumor cells (CTCs). These components can reflect the status of the disease and can facilitate clinical decisions. LBs offer a unique and new way to assess cancers at all stages of treatment, from cancer screenings to prognosis to management of multidisciplinary therapies. In this review, we will provide insights into the current status of the various types of LBs enabling early detection and monitoring of GI cancers and their use in in vitro diagnostics.
Collapse
Affiliation(s)
- Paul David
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anke Mittelstädt
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Dina Kouhestani
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Anna Anthuber
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Christoph Kahlert
- Department of Surgery, Carl Gustav Carus University Hospital, 01307 Dresden, Germany
| | - Kai Sohn
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, 70569 Stuttgart, Germany
| | - Georg F Weber
- Department of Surgery, University Hospital of Erlangen, Friedrich-Alexander University (FAU) Erlangen-Nürnberg, 91054 Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
21
|
Sellahewa R, Moghaddam SM, Lundy J, Jenkins BJ, Croagh D. Circulating Tumor DNA Is an Accurate Diagnostic Tool and Strong Prognostic Marker in Pancreatic Cancer. Pancreas 2023; 52:e188-e195. [PMID: 37751379 DOI: 10.1097/mpa.0000000000002239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
OBJECTIVE The objectives of the study are to investigate the sensitivity and specificity of circulating tumor DNA (ctDNA) for the diagnosis of pancreatic cancer and to assess the utility of ctDNA as a prognostic marker in this disease. METHODS Cell-free DNA was extracted from plasma of patients who underwent endoscopic ultrasound fine-needle aspiration or surgical resections for pancreatic cancer. The cell-free DNA was then analyzed using droplet digital polymerase chain reaction for KRAS G12/13 mutations. Eighty-one patients with pancreatic cancer and 30 patients with benign pancreatic disease were analyzed. RESULTS ctDNA KRAS G12/13 mutations were detected in 63% of all patients with pancreatic cancer and in 76% of those patients who also had KRAS G12/13 mutations detected in the pancreatic primary. Specificity and tissue concordance were both 100%. Circulating tumor DNA corresponded with tumor size and stage, and high ctDNA was associated with significantly worse prognosis on both univariate and multivariate testing. CONCLUSION Our study shows that ctDNA is an accurate diagnostic tool and strong prognostic marker in patients with pancreatic cancer. The continued investigation of ctDNA will enable its implementation in clinical practice to optimize the care and survival outcomes of patients with pancreatic cancer.
Collapse
|
22
|
Labiano I, Huerta AE, Arrazubi V, Hernandez-Garcia I, Mata E, Gomez D, Arasanz H, Vera R, Alsina M. State of the Art: ctDNA in Upper Gastrointestinal Malignancies. Cancers (Basel) 2023; 15:1379. [PMID: 36900172 PMCID: PMC10000247 DOI: 10.3390/cancers15051379] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/14/2023] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Circulating tumor DNA (ctDNA) has emerged as a promising non-invasive source to characterize genetic alterations related to the tumor. Upper gastrointestinal cancers, including gastroesophageal adenocarcinoma (GEC), biliary tract cancer (BTC) and pancreatic ductal adenocarcinoma (PADC) are poor prognostic malignancies, usually diagnosed at advanced stages when no longer amenable to surgical resection and show a poor prognosis even for resected patients. In this sense, ctDNA has emerged as a promising non-invasive tool with different applications, from early diagnosis to molecular characterization and follow-up of tumor genomic evolution. In this manuscript, novel advances in the field of ctDNA analysis in upper gastrointestinal tumors are presented and discussed. Overall, ctDNA analyses can help in early diagnosis, outperforming current diagnostic approaches. Detection of ctDNA prior to surgery or active treatment is also a prognostic marker that associates with worse survival, while ctDNA detection after surgery is indicative of minimal residual disease, anticipating in some cases the imaging-based detection of progression. In the advanced setting, ctDNA analyses characterize the genetic landscape of the tumor and identify patients for targeted-therapy approaches, and studies show variable concordance levels with tissue-based genetic testing. In this line, several studies also show that ctDNA serves to follow responses to active therapy, especially in targeted approaches, where it can detect multiple resistance mechanisms. Unfortunately, current studies are still limited and observational. Future prospective multi-center and interventional studies, carefully designed to assess the value of ctDNA to help clinical decision-making, will shed light on the real applicability of ctDNA in upper gastrointestinal tumor management. This manuscript presents a review of the evidence available in this field up to date.
Collapse
Affiliation(s)
- Ibone Labiano
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Ana Elsa Huerta
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
| | - Virginia Arrazubi
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Irene Hernandez-Garcia
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Elena Mata
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - David Gomez
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Hugo Arasanz
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Ruth Vera
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| | - Maria Alsina
- Oncobiona Group, Navarrabiomed-Instituto de Investigación Sanitaria de Navarra (IdiSNA), Irunlarrea 3, 31008 Pamplona, Spain
- Medical Oncology Department, Hospital Universitario de Navarra (HUN), Irunlarrea 3, 31008 Pamplona, Spain
| |
Collapse
|
23
|
Raufi AG, May MS, Hadfield MJ, Seyhan AA, El-Deiry WS. Advances in Liquid Biopsy Technology and Implications for Pancreatic Cancer. Int J Mol Sci 2023; 24:4238. [PMID: 36835649 PMCID: PMC9958987 DOI: 10.3390/ijms24044238] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 12/23/2022] [Accepted: 12/29/2022] [Indexed: 02/23/2023] Open
Abstract
Pancreatic cancer is a highly aggressive malignancy with a climbing incidence. The majority of cases are detected late, with incurable locally advanced or metastatic disease. Even in individuals who undergo resection, recurrence is unfortunately very common. There is no universally accepted screening modality for the general population and diagnosis, evaluation of treatment response, and detection of recurrence relies primarily on the use of imaging. Identification of minimally invasive techniques to help diagnose, prognosticate, predict response or resistance to therapy, and detect recurrence are desperately needed. Liquid biopsies represent an emerging group of technologies which allow for non-invasive serial sampling of tumor material. Although not yet approved for routine use in pancreatic cancer, the increasing sensitivity and specificity of contemporary liquid biopsy platforms will likely change clinical practice in the near future. In this review, we discuss the recent technological advances in liquid biopsy, focusing on circulating tumor DNA, exosomes, microRNAs, and circulating tumor cells.
Collapse
Affiliation(s)
- Alexander G. Raufi
- Division of Hematology/Oncology, Department of Medicine, Lifespan Health System, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University, Providence, RI 02903, USA
| | - Michael S. May
- Division of Hematology/Oncology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Matthew J. Hadfield
- Division of Hematology/Oncology, Department of Medicine, Lifespan Health System, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
| | - Attila A. Seyhan
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University, Providence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Wafik S. El-Deiry
- Division of Hematology/Oncology, Department of Medicine, Lifespan Health System, Providence, RI 02903, USA
- Legorreta Cancer Center, Brown University, Providence, RI 02903, USA
- Joint Program in Cancer Biology, Brown University, Providence, RI 02903, USA
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| |
Collapse
|
24
|
Shaya J, Kato S, Adashek JJ, Patel H, Fanta PT, Botta GP, Sicklick JK, Kurzrock R. Personalized matched targeted therapy in advanced pancreatic cancer: a pilot cohort analysis. NPJ Genom Med 2023; 8:1. [PMID: 36670111 PMCID: PMC9860045 DOI: 10.1038/s41525-022-00346-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/07/2022] [Indexed: 01/21/2023] Open
Abstract
Despite progress, 2-year pancreatic cancer survival remains dismal. We evaluated a biomarker-driven, combination/N-of-one strategy in 18 patients (advanced/metastatic pancreatic cancer) (from Molecular Tumor Board). Targeted agents administered/patient = 2.5 (median) (range, 1-4); first-line therapy (N = 5); second line, (N = 13). Comparing patients (high versus low degrees of matching) (matching score ≥50% versus <50%; reflecting number of alterations matched to targeted agents divided by number of pathogenic alterations), survival was significantly longer (hazard ratio [HR] 0.24 (95% confidence interval [CI], 0.078-0.76, P = 0.016); clinical benefit rates (CBR) (stable disease ≥6 months/partial/complete response) trended higher (45.5 vs 0.0%, P = 0.10); progression-free survival, HR, 95% CI, 0.36 (0.12-1.10) (p = 0.075). First versus ≥2nd-line therapy had higher CBRs (80.0 vs 7.7%, P = 0.008). No grade 3-4 toxicities occurred. The longest responder achieved partial remission (17.5 months) by co-targeting MEK and CDK4/6 alterations (chemotherapy-free). Therefore, genomically matched targeted agent combinations were active in these advanced pancreatic cancers. Larger prospective trials are warranted.
Collapse
Affiliation(s)
- Justin Shaya
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Shumei Kato
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA.
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, La Jolla, CA, USA.
| | - Jacob J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at The Johns Hopkins Hospital, Baltimore, MD, USA.
| | - Hitendra Patel
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Paul T Fanta
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Gregory P Botta
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Jason K Sicklick
- Center for Personalized Cancer Therapy, University of California San Diego Moores Cancer Center, La Jolla, CA, USA
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at The Johns Hopkins Hospital, Baltimore, MD, USA
- Department of Surgery, Division of Surgical Oncology, University of California San Diego, UC San Diego Health, San Diego, CA, USA
- Department of Pharmacology, University of California San Diego, UC San Diego Health, San Diego, CA, USA
| | - Razelle Kurzrock
- Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, WI, USA
- WIN Consortium, Paris, France
- University of Nebraska, Lincoln, NE, USA
| |
Collapse
|
25
|
Christenson ES, Lim SJ, Durham J, De Jesus-Acosta A, Bever K, Laheru D, Ryan A, Agarwal P, Scharpf RB, Le DT, Wang H. Cell-free DNA Predicts Prolonged Response to Multi-agent Chemotherapy in Pancreatic Ductal Adenocarcinoma. CANCER RESEARCH COMMUNICATIONS 2022; 2:1418-1425. [PMID: 36970054 PMCID: PMC10035498 DOI: 10.1158/2767-9764.crc-22-0343] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/18/2022] [Accepted: 10/18/2022] [Indexed: 11/07/2022]
Abstract
The treatment of metastatic pancreatic ductal adenocarcinoma (PDAC) is frequently characterized by significant toxicity and rapid development of resistance to current approved therapies. More reliable biomarkers of response are needed to guide clinical decision making. We evaluated cell-free DNA (cfDNA) using a tumor-agnostic platform and traditional biomarkers (CEA and CA19-9) levels in 12 patients treated at Johns Hopkins University on NCT02324543 "Study of Gemcitabine/Nab-Paclitaxel/Xeloda (GAX) in Combination With Cisplatin and Irinotecan in Subjects With Metastatic Pancreatic Cancer." The pretreatment values, levels after 2 months of treatment, and change in biomarker levels with treatment were compared with clinical outcomes to determine their predictive value. The variant allele frequency (VAF) of KRAS and TP53 mutations in cfDNA after 2 months of treatment was predictive of progression-free survival (PFS) and overall survival (OS). In particular, patients with a lower-than-average KRAS VAF after 2 months of treatment had a substantially longer PFS than patients with higher posttreatment KRAS VAF (20.96 vs. 4.39 months). Changes in CEA and CA19-9 after 2 months of treatment were also good predictors of PFS. Comparison via concordance index demonstrated KRAS or TP53 VAF after 2 months of treatment to be better predictors of PFS and OS than CA19-9 or CEA. This pilot study requires validation but suggests cfDNA measurement is a useful adjunct to traditional protein biomarkers and imaging evaluation and could distinguish between patients who are likely to achieve prolonged responses versus those that will have early progression and may benefit from a change in treatment approach. Significance We report on the association of cfDNA with response durability for patients undergoing treatment with a novel metronomic chemotherapy regimen (gemcitabine, nab-paclitaxel, capecitabine, cisplatin, irinotecan; GAX-CI) for metastatic PDAC. This investigation offers encouraging evidence that cfDNA may prove to be a valuable diagnostic tool to guide clinical management.
Collapse
Affiliation(s)
- Eric S Christenson
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Su Jin Lim
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jennifer Durham
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Ana De Jesus-Acosta
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Katherine Bever
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Daniel Laheru
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Amy Ryan
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Parul Agarwal
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Robert B Scharpf
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Dung T Le
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Hao Wang
- Bloomberg-Kimmel Institute, Sidney Kimmel Comprehensive Cancer Center, and The Skip Viragh Center for Pancreatic Cancer Research and Clinical Care, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
26
|
Systematic review and meta-analysis: Diagnostic performance of DNA alterations in pancreatic juice for the detection of pancreatic cancer. Pancreatology 2022; 22:973-986. [PMID: 35864067 DOI: 10.1016/j.pan.2022.06.260] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/20/2022] [Accepted: 06/22/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS Pancreatic cancer has a dismal prognosis. So far, imaging has been proven incapable of establishing an early enough diagnosis. Thus, biomarkers are urgently needed for early detection and improved survival. Our aim was to evaluate the pooled diagnostic performance of DNA alterations in pancreatic juice. METHODS A systematic literature search was performed in EMBASE, MEDLINE Ovid, Cochrane CENTRAL and Web of Science for studies concerning the diagnostic performance of DNA alterations in pancreatic juice to differentiate patients with high-grade dysplasia or pancreatic cancer from controls. Study quality was assessed using QUADAS-2. The pooled prevalence, sensitivity, specificity and diagnostic odds ratio were calculated. RESULTS Studies mostly concerned cell-free DNA mutations (32 studies: 939 cases, 1678 controls) and methylation patterns (14 studies: 579 cases, 467 controls). KRAS, TP53, CDKN2A, GNAS and SMAD4 mutations were evaluated most. Of these, TP53 had the highest diagnostic performance with a pooled sensitivity of 42% (95% CI: 31-54%), specificity of 98% (95%-CI: 92%-100%) and diagnostic odds ratio of 36 (95% CI: 9-133). Of DNA methylation patterns, hypermethylation of CDKN2A, NPTX2 and ppENK were studied most. Hypermethylation of NPTX2 performed best with a sensitivity of 39-70% and specificity of 94-100% for distinguishing pancreatic cancer from controls. CONCLUSIONS This meta-analysis shows that, in pancreatic juice, the presence of distinct DNA mutations (TP53, SMAD4 or CDKN2A) and NPTX2 hypermethylation have a high specificity (close to 100%) for the presence of high-grade dysplasia or pancreatic cancer. However, the sensitivity of these DNA alterations is poor to moderate, yet may increase if they are combined in a panel.
Collapse
|
27
|
Chawla A, Qadan M, Castillo CFD, Wo JY, Allen JN, Clark JW, Murphy JE, Catalano OA, Ryan DP, Ting DT, Deshpande V, Weekes CD, Parikh A, Lillemoe KD, Hong TS, Ferrone CR. Prospective Phase II Trials Validate the Effect of Neoadjuvant Chemotherapy on Pattern of Recurrence in Pancreatic Adenocarcinoma. Ann Surg 2022; 276:e502-e509. [PMID: 33086310 DOI: 10.1097/sla.0000000000004585] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The objective of this study was to characterize the patterns of first recurrence after curative-intent resection for pancreatic adenocarcinoma (PDAC). SUMMARY OF BACKGROUND DATA We evaluated the first site of recurrence after neoadjuvant treatment as locoregional (LR) or distant metastasis (DM). To validate our findings, we evaluated the pattern from 2 phase II clinical trials evaluating neoadjuvant chemotherapy (NAC) in PDAC. METHODS We identified site of first recurrence from a retrospective cohort of patients from 2011 to 2017 treated with NAC followed by chemoradiation and then an operation or an operation first followed by adjuvant therapy, and 2 separate prospective cohorts of patients derived from 2 phase II clinical trials evaluating patients treated with NAC in borderline-resectable and locally advanced PDAC. RESULTS In the retrospective cohorts, 160 out of 285 patients (56.1%) recurred after a median disease-free survival (mDFS) of 17.2 months. The pattern of recurrence was DM in 81.9% of patients, versus LR in 11.1%. This pattern was consistent in patients treated with upfront resection and adjuvant chemotherapy (DM 83.0%, LR 16.9%) regardless of margin-involvement (DM 80.1%, LR 19.4%). The use of NAC did not alter pattern of recurrence; 81.7% had DM and 18.3% had LR. This pattern also remained consistent regardless of margin-involvement (DM 94.1%, LR 5.9%). In the Phase II borderline-resectable trial (NCI# 01591733) cohort of 32 patients, the mDFS was 34.2 months. Pattern of recurrence remained predominantly DM (88.9%) versus LR (11.1%). In the Phase II locally-advanced trial (NCI# 01821729) cohort of 34 patients, the mDFS was 30.7 months. Although there was a higher rate of local recurrence in this cohort, pattern of first recurrence remained predominantly DM (66.6%) versus LR (33.3%) and remained consistent independent of margin-status. CONCLUSIONS The pattern of recurrence in PDAC is predominantly DM rather than LR, and is consistent regardless of the use of NAC and margin involvement.
Collapse
Affiliation(s)
- Akhil Chawla
- Division of Surgical Oncology, Department of Surgery, Northwestern Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Motaz Qadan
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Jennifer Y Wo
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jill N Allen
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Jeffrey W Clark
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Janet E Murphy
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Onofrio A Catalano
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David P Ryan
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - David T Ting
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Vikram Deshpande
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Colin D Weekes
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aparna Parikh
- Department of Medical Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Keith D Lillemoe
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Theodore S Hong
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Cristina R Ferrone
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
28
|
Yu SCY, Chan KCA. What is the Importance of Analyzing Tumor Tissues and Blood Cells in the Study of Circulating Tumor-Derived DNA? Clin Chem 2022; 68:1481-1483. [PMID: 36245241 DOI: 10.1093/clinchem/hvac168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/16/2022] [Indexed: 11/12/2022]
Affiliation(s)
- Stephanie C Y Yu
- Centre for Novostics, Hong Kong Science Park, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| | - K C Allen Chan
- Centre for Novostics, Hong Kong Science Park, Hong Kong SAR, China.,Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.,Department of Chemical Pathology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China.,State Key Laboratory of Translational Oncology, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong SAR, China
| |
Collapse
|
29
|
Nikanjam M, Kato S, Kurzrock R. Liquid biopsy: current technology and clinical applications. J Hematol Oncol 2022; 15:131. [PMID: 36096847 PMCID: PMC9465933 DOI: 10.1186/s13045-022-01351-y] [Citation(s) in RCA: 240] [Impact Index Per Article: 120.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 09/06/2022] [Indexed: 11/10/2022] Open
Abstract
Liquid biopsies are increasingly used for cancer molecular profiling that enables a precision oncology approach. Circulating extracellular nucleic acids (cell-free DNA; cfDNA), circulating tumor DNA (ctDNA), and circulating tumor cells (CTCs) can be isolated from the blood and other body fluids. This review will focus on current technologies and clinical applications for liquid biopsies. ctDNA/cfDNA has been isolated and analyzed using many techniques, e.g., droplet digital polymerase chain reaction, beads, emulsion, amplification, and magnetics (BEAMing), tagged-amplicon deep sequencing (TAm-Seq), cancer personalized profiling by deep sequencing (CAPP-Seq), whole genome bisulfite sequencing (WGBS-Seq), whole exome sequencing (WES), and whole genome sequencing (WGS). CTCs have been isolated using biomarker-based cell capture, and positive or negative enrichment based on biophysical and other properties. ctDNA/cfDNA and CTCs are being exploited in a variety of clinical applications: differentiating unique immune checkpoint blockade response patterns using serial samples; predicting immune checkpoint blockade response based on baseline liquid biopsy characteristics; predicting response and resistance to targeted therapy and chemotherapy as well as immunotherapy, including CAR-T cells, based on serial sampling; assessing shed DNA from multiple metastatic sites; assessing potentially actionable alterations; analyzing prognosis and tumor burden, including after surgery; interrogating difficult-to biopsy tumors; and detecting cancer at early stages. The latter can be limited by the small amounts of tumor-derived components shed into the circulation; furthermore, cfDNA assessment in all cancers can be confounded by clonal hematopoeisis of indeterminate potential, especially in the elderly. CTCs can be technically more difficult to isolate that cfDNA, but permit functional assays, as well as evaluation of CTC-derived DNA, RNA and proteins, including single-cell analysis. Blood biopsies are less invasive than tissue biopsies and hence amenable to serial collection, which can provide critical molecular information in real time. In conclusion, liquid biopsy is a powerful tool, and remarkable advances in this technology have impacted multiple aspects of precision oncology, from early diagnosis to management of refractory metastatic disease. Future research may focus on fluids beyond blood, such as ascites, effusions, urine, and cerebrospinal fluid, as well as methylation patterns and elements such as exosomes.
Collapse
Affiliation(s)
- Mina Nikanjam
- Division of Hematology-Oncology, University of California San Diego, La Jolla, 1200 Garden View Road, Encinitas, CA, 92024, USA.
| | - Shumei Kato
- Division of Hematology-Oncology, University of California San Diego, La Jolla, 1200 Garden View Road, Encinitas, CA, 92024, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center, Milwaukee, WI, USA.,WIN Consortium, Paris, France
| |
Collapse
|
30
|
Guan S, Deng G, Sun J, Han Q, Lv Y, Xue T, Ding L, Yang T, Qian N, Dai G. Evaluation of circulating tumor DNA as a prognostic biomarker for metastatic pancreatic adenocarcinoma. Front Oncol 2022; 12:926260. [PMID: 36081557 PMCID: PMC9446234 DOI: 10.3389/fonc.2022.926260] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
PurposePancreatic cancer is an aggressive solid tumor with a severe prognosis. Although tumor biomarkers are often used to identify advanced pancreatic cancer, this is not accurate, and the currently used biomarkers are not indicative of prognosis. The present study evaluated circulating tumor DNA (ctDNA) as a biomarker for prognosis prediction and disease monitoring in metastatic pancreatic adenocarcinoma (PAC).MethodsFrom 2017 to 2018, 40 patients with metastatic PAC were enrolled, and tumor tissue and blood samples were collected from 40 and 35 patients, respectively. CtDNA was sequenced by next-generation sequencing (NGS) with a 425-gene capture panel. The association of clinical characteristics, laboratory indicators, and dynamic ctDNA with patient outcomes was analyzed.ResultsMutations in KRAS (87.5%, N = 35) and TP53 (77.5%, N = 31) were most common in 40 tumor tissue. Patients’ ECOG score, CA19-9, CEA, neutrophil-lymphocyte ratio (NLR), platelet- lymphocyte ratio (PLR) levels and mutations in ≥ 3 driver genes were strongly correlated with patients’ overall survival (OS). Patients’ gender, ECOG score, CA19-9, and CEA levels were associated with progression-free survival (PFS) (P<0.05). In 35 blood samples, univariate analysis showed a significant association between ECOG score, CA19-9, KRAS or CDKN2A mutation in ctDNA and OS and between CA19-9, CDKN2A or SMAD4 mutation in ctDNA and PFS. Cox hazard proportion model showed that patients’ CDKN2A mutation in ctDNA (HR=16.1, 95% CI=4.4-59.1, P<0.001), ECOG score (HR=6.2, 95% CI=2.4-15.7, P<0.001) and tumor location (HR=0.4, 95% CI=0.1-0.9, P=0.027) were significantly associated with OS. Patients’ CDKN2A mutation in ctDNA (HR=6.8, 95% CI=2.3-19.9, P=0.001), SMAD4 mutation in ctDNA (HR=3.0, 95% CI=1.1-7.9, P=0.031) and metastatic organ (HR=0.4, 95% CI=0.2-1.0, P=0.046) were significantly associated with PFS. Longitudinal changes in gene mutation allelic frequency (MAF) value were evaluated in 24 patients. Detection of progression disease (PD) by ctDNA was 0.9 months earlier than by radiological imaging (mean PFS: 4.6m vs 5.5m, P=0.004, paired t-test).ConclusionsThe ctDNA has the potential as a specific survival predictive marker for metastatic PAC patients. Longitudinal ctDNA tracking could potentially help identify disease progression and be a valuable complement for routine clinical markers and imaging.
Collapse
Affiliation(s)
- Shasha Guan
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Guochao Deng
- Senior Department of Oncology, The Fifth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Jingjie Sun
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Quanli Han
- Senior Department of Oncology, The Fifth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Yao Lv
- Senior Department of Oncology, The Fifth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
| | - Tianhui Xue
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Lijuan Ding
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Tongxin Yang
- Department of Oncology, Hainan Hospital of Chinese People’s Liberation Army (PLA) General Hospital, Sanya, China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’ Liberation Army (PLA) General Hospital, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Niansong Qian, ; Guanghai Dai,
| | - Guanghai Dai
- Senior Department of Oncology, The Fifth Medical Center of Chinese People’s Liberation Army (PLA) General Hospital, Beijing, China
- *Correspondence: Niansong Qian, ; Guanghai Dai,
| |
Collapse
|
31
|
Li W, Zhang X, Li Y, Yue Q, Cui M, Liu J. Prognostic Value of KRAS Mutations in the Peripheral Blood of Patients with Pancreatic Cancer: a Systematic Review and Meta-analysis. Indian J Surg 2022. [DOI: 10.1007/s12262-021-03142-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
|
32
|
Ulanja MB, Moody AE, Beutler BD, Antwi-Amoabeng D, Rahman GA, Alese OB. Early-onset pancreatic cancer: a review of molecular mechanisms, management, and survival. Oncotarget 2022; 13:828-841. [PMID: 35720978 PMCID: PMC9200435 DOI: 10.18632/oncotarget.28242] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVES Early-onset pancreatic cancer (EOPC) - defined as pancreatic cancer diagnosed before the age of 50 years - is associated with a poor prognosis as compared to later-onset pancreatic cancer (LOPC). Emerging evidence suggests that EOPC may exhibit a genetic signature and tumor biology that is distinct from that of LOPC. We review genetic mutations that are more prevalent in EOPC relative to LOPC and discuss the potential impact of these mutations on treatment and survival. MATERIALS AND METHODS Using PubMed and Medline, the following terms were searched and relevant citations assessed: "early onset pancreatic cancer," "late onset pancreatic cancer," "pancreatic cancer," "pancreatic cancer genes," and "pancreatic cancer targeted therapy." RESULTS Mutations in CDKN2, FOXC2, and SMAD4 are significantly more common in EOPC as compared to LOPC. In addition, limited data suggest that PI3KCA mutations are more frequently observed in EOPC as compared to LOPC. KRAS mutations are relatively rare in EOPC. CONCLUSIONS Genetic mutations associated with EOPC are distinct from those of LOPC. The preponderance of the evidence suggest that poor outcomes in EOPC are related both to advanced stage of presentation and unique tumor biology. The molecular and genetic features of EOPC warrant further investigation in order to optimize management.
Collapse
Affiliation(s)
- Mark B. Ulanja
- Christus Ochsner Saint Patrick Hospital, Lake Charles, LA 70601, USA
| | - Alastair E. Moody
- Department of Anesthesiology, University of Utah, Salt Lake City, UT 84112, USA
| | - Bryce D. Beutler
- Department of Radiology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, USA
| | | | - Ganiyu A. Rahman
- Department of Surgery, University of Cape Coast, School of Medical Sciences, Cape Coast, Ghana
| | - Olatunji B. Alese
- Department of Hematology and Oncology, Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
33
|
Uson Junior PLS, Majeed U, Yin J, Botrus G, Sonbol MB, Ahn DH, Starr JS, Jones JC, Babiker H, Inabinett SR, Wylie N, Boyle AW, Bekaii-Saab TS, Gores GJ, Smoot R, Barrett M, Nagalo B, Meurice N, Elliott N, Petit J, Zhou Y, Arora M, Dumbauld C, Barro O, Baker A, Bogenberger J, Buetow K, Mansfield A, Mody K, Borad MJ. Cell-Free Tumor DNA Dominant Clone Allele Frequency Is Associated With Poor Outcomes in Advanced Biliary Cancers Treated With Platinum-Based Chemotherapy. JCO Precis Oncol 2022; 6:e2100274. [PMID: 35666960 PMCID: PMC9200394 DOI: 10.1200/po.21.00274] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 10/06/2021] [Accepted: 04/12/2022] [Indexed: 12/14/2022] Open
Abstract
PURPOSE This investigation sought to evaluate the prognostic value of pretreatment of circulating tumor DNA (ctDNA) in metastatic biliary tract cancers (BTCs) treated with platinum-based first-line chemotherapy treatment. MATERIALS AND METHODS We performed a retrospective analysis of 67 patients who underwent ctDNA testing before platinum-based chemotherapy for first-line treatment for metastatic BTC. For analysis, we considered the detected gene with highest variant allele frequency as the dominant clone allele frequency (DCAF). Results of ctDNA analysis were correlated with patients' demographics, progression-free survival (PFS), and overall survival (OS). RESULTS The median age of patients was 67 (27-90) years. Fifty-four (80.6%) of 67 patients evaluated had intrahepatic cholangiocarcinoma; seven had extrahepatic cholangiocarcinoma, and six gallbladder cancers. Forty-six (68.6%) of the patients were treated with cisplatin plus gemcitabine, and 16.4% of patients received gemcitabine and other platinum (carboplatin or oxaliplatin) combinations, whereas 15% of patients were treated on a clinical trial with gemcitabine and cisplatin plus additional agents (CX4945, PEGPH20, or nab-paclitaxel). TP53, KRAS, FGFR2, ARID1A, STK11, and IDH1 were the genes with highest frequency as DCAF. The median DCAF was 3% (0%-97%). DCAF > 3% was associated with worse OS (median OS: 10.8 v 18.8 months, P = .032). Stratifying DCAF in quartiles, DCAF > 10% was significantly related to worse PFS (median PFS: 3 months, P = .014) and worse OS (median OS: 7.0 months, P = .001). Each 1% increase in ctDNA was associated with a hazard ratio of 13.1 in OS when adjusting for subtypes, metastatic sites, size of largest tumor, age, sex, and CA19-9. CONCLUSION DCAF at diagnosis of advanced BTC can stratify patients who have worse outcomes when treated with upfront platinum-based chemotherapy. Each increase in %ctDNA decreases survival probabilities.
Collapse
Affiliation(s)
- Pedro Luiz Serrano Uson Junior
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Umair Majeed
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Jun Yin
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, MN
| | - Gehan Botrus
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Mohamad Bassam Sonbol
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Daniel H. Ahn
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Jason S. Starr
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Jeremy C. Jones
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Hani Babiker
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Samantha R. Inabinett
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Natasha Wylie
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Ashton W.R. Boyle
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Tanios S. Bekaii-Saab
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Gregory J. Gores
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Rochester, MN
| | - Rory Smoot
- Division of Medical Oncology, Mayo Clinic, Rochester, MN
| | - Michael Barrett
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Bolni Nagalo
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Nathalie Meurice
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Natalie Elliott
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Joachim Petit
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Yumei Zhou
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Mansi Arora
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Chelsae Dumbauld
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Oumar Barro
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - Alexander Baker
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | - James Bogenberger
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
| | | | | | - Kabir Mody
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL
| | - Mitesh J. Borad
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Molecular Medicine, Rochester, MN
- Mayo Clinic Cancer Center, Phoenix, AZ
| |
Collapse
|
34
|
Pietrasz D, Sereni E, Lancelotti F, Pea A, Luchini C, Innamorati G, Salvia R, Bassi C. Circulating tumour DNA: a challenging innovation to develop "precision onco-surgery" in pancreatic adenocarcinoma. Br J Cancer 2022; 126:1676-1683. [PMID: 35197581 PMCID: PMC9174156 DOI: 10.1038/s41416-022-01745-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 12/13/2021] [Accepted: 02/04/2022] [Indexed: 12/20/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is predicted to become the third leading cause of cancer-related mortality within the next decade. Management of PDAC remains challenging with limited effective treatment options and a dismal long-term prognosis. Liquid biopsy and circulating biomarkers seem to be promising to improve the multidisciplinary approach in PDAC treatment. Circulating tumour DNA (ctDNA) is the most studied blood liquid biopsy analyte and can provide insight into the molecular profile and individual characteristics of the tumour in real-time and in advance of standard imaging modalities. This could pave the way for identifying new therapeutic targets and markers of tumour response to supplement diagnostic and provide enhanced stratified treatment. Although its specificity seems excellent, the current sensitivity of ctDNA remains a limitation for clinical use, especially in patients with a low tumour burden. Increasing evidence suggests that ctDNA is a pertinent candidate biomarker to assess minimal residual disease after surgery but also a strong independent prognostic biomarker. This review explores the current knowledge and recent developments in ctDNA as a screening, diagnostic, prognostic and predictive biomarker in the management of resectable PDAC but also technical and analytical challenges that must be overcome to move toward "precision onco-surgery."
Collapse
Affiliation(s)
- Daniel Pietrasz
- APHP Hôpital Paul-Brousse, Centre Hépato-Biliaire, Université Paris-Saclay, 94800, Villejuif, France.
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy.
| | - Elisabetta Sereni
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Francesco Lancelotti
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Antonio Pea
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Giulio Innamorati
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Roberto Salvia
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| | - Claudio Bassi
- Unit of General and Pancreatic Surgery, Department of Surgery and Oncology, University of Verona Hospital Trust, Verona, Italy
| |
Collapse
|
35
|
Alese OB, Cook N, Ortega-Franco A, Ulanja MB, Tan L, Tie J. Circulating Tumor DNA: An Emerging Tool in Gastrointestinal Cancers. Am Soc Clin Oncol Educ Book 2022; 42:1-20. [PMID: 35471832 DOI: 10.1200/edbk_349143] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Circulating tumor DNA (ctDNA) is tumor-derived fragmented DNA in the bloodstream that has come from primary or metastatic cancer sites. Neoplasm-specific genetic and epigenetic abnormalities are increasingly being identified through liquid biopsy: a novel, minimally invasive technique used to isolate and analyze ctDNA in the peripheral circulation. Liquid biopsy and other emerging ctDNA technologies represent a paradigm shift in cancer diagnostics because they allow for the detection of minimal residual disease in patients with early-stage disease, improve risk stratification, capture tumor heterogeneity and genomic evolution, and enhance ctDNA-guided adjuvant and palliative cancer therapy. Moreover, ctDNA can be used to monitor the tumor response to neoadjuvant and postoperative therapy in patients with metastatic disease. Using clearance of ctDNA as an endpoint for escalation/de-escalation of adjuvant chemotherapy for patients considered to have high-risk disease has become an important area of research. The possibility of using ctDNA as a surrogate for treatment response-including for overall survival, progression-free survival, and disease-free survival-is an attractive concept; this surrogate will arguably reduce study duration and expedite the development of new therapies. In this review, we summarize the current evidence on the applications of ctDNA for the diagnosis and management of gastrointestinal tumors. Gastrointestinal cancers-including tumors of the esophagus, stomach, colon, liver, and pancreas-account for one-quarter of global cancer diagnoses and contribute to more than one-third of cancer-related deaths. Given the prevalence of gastrointestinal malignancies, ctDNA technology represents a powerful tool to reduce the global burden of disease.
Collapse
Affiliation(s)
- Olatunji B Alese
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, GA
| | - Natalie Cook
- Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester, United Kingdom.,Division of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Ana Ortega-Franco
- Experimental Cancer Medicine Team, The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Mark B Ulanja
- Christus Ochsner St. Patrick Hospital, Lake Charles, LA
| | - Lavinia Tan
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Jeanne Tie
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia.,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia.,Division of Personalized Oncology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| |
Collapse
|
36
|
Dopico PJ, Le MCN, Burgess B, Yang Z, Zhao Y, Wang Y, George TJ, Fan ZH. Longitudinal Study of Circulating Biomarkers in Patients with Resectable Pancreatic Ductal Adenocarcinoma. BIOSENSORS 2022; 12:206. [PMID: 35448266 PMCID: PMC9028387 DOI: 10.3390/bios12040206] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 05/12/2023]
Abstract
While patients with resectable pancreatic ductal adenocarcinoma (PDAC) show improved survival compared to their non-resectable counterparts, survival remains low owing to occult metastatic disease and treatment resistance. Liquid biopsy based on circulating tumor cells (CTCs) and cell-free DNA (cfDNA) has been shown to predict recurrence and treatment resistance in various types of cancers, but their utility has not been fully demonstrated in resectable PDAC. We have simultaneously tracked three circulating biomarkers, including CTCs, cfDNA, and circulating tumor DNA (ctDNA), over a period of cancer treatment using a microfluidic device and droplet digital PCR (ddPCR). The microfluidic device is based on the combination of filtration and immunoaffinity mechanisms. We have measured CTCs, cfDNA, and ctDNA in a cohort of seven resectable PDAC patients undergoing neoadjuvant therapy followed by surgery, and each patient was followed up to 10 time points over a period of 4 months. CTCs were detectable in all patients (100%) at some point during treatment but were detectable in only three out of six patients (50%) prior to the start of treatment. Median cfDNA concentrations remained comparable to negative controls throughout treatment. ddPCR was able to find KRAS mutations in six of seven patients (86%); however, these mutations were present in only two of seven patients (29%) prior to treatment. Overall, the majority of circulating biomarkers (81% for CTCs and 91% for cfDNA/ctDNA) were detected after the start of neoadjuvant therapy but before surgery. This study suggests that a longitudinal study of circulating biomarkers throughout treatment provides more useful information than those single time-point tests for resectable PDAC patients.
Collapse
Affiliation(s)
- Pablo J. Dopico
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (P.J.D.); (M.-C.N.L.)
| | - Minh-Chau N. Le
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (P.J.D.); (M.-C.N.L.)
| | - Benjamin Burgess
- UF Health Cancer Center, University of Florida, 2033 Mowry Rd., Gainesville, FL 32610, USA;
| | - Zhijie Yang
- Atila Biosystems, 740 Sierra Vista Ave., Unit E, Mountain View, CA 94043, USA; (Z.Y.); (Y.Z.); (Y.W.)
| | - Yu Zhao
- Atila Biosystems, 740 Sierra Vista Ave., Unit E, Mountain View, CA 94043, USA; (Z.Y.); (Y.Z.); (Y.W.)
| | - Youxiang Wang
- Atila Biosystems, 740 Sierra Vista Ave., Unit E, Mountain View, CA 94043, USA; (Z.Y.); (Y.Z.); (Y.W.)
| | - Thomas J. George
- UF Health Cancer Center, University of Florida, 2033 Mowry Rd., Gainesville, FL 32610, USA;
- Department of Medicine, University of Florida, 1600 SW Archer Rd., Gainesville, FL 32610, USA
| | - Z. Hugh Fan
- Interdisciplinary Microsystems Group, Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611, USA; (P.J.D.); (M.-C.N.L.)
- UF Health Cancer Center, University of Florida, 2033 Mowry Rd., Gainesville, FL 32610, USA;
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
37
|
Kato S, Li B, Adashek JJ, Cha SW, Bianchi-Frias D, Qian D, Kim L, so TW, Mitchell M, Kamei N, Hoiness R, Hoo J, Gray PN, Iyama T, Kashiwagi M, Lu HM, Kurzrock R. Serial changes in liquid biopsy-derived variant allele frequency predict immune checkpoint inhibitor responsiveness in the pan-cancer setting. Oncoimmunology 2022; 11:2052410. [PMID: 35371621 PMCID: PMC8966985 DOI: 10.1080/2162402x.2022.2052410] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Major immunotherapy challenges include a limited number of predictive biomarkers and the unusual imaging features post-therapy, such as pseudo-progression, which denote immune infiltrate-mediated tumor enlargement. Such phenomena confound clinical decision-making, since the cancer may eventually regress, and the patient should stay on treatment. We prospectively evaluated serial, blood-derived cell-free DNA (cfDNA) (baseline and 2–3 weeks post-immune checkpoint inhibitors [ICIs]) for variant allele frequency (VAF) and blood tumor mutation burden (bTMB) changes (next-generation sequencing) (N = 84 evaluable patients, diverse cancers). Low vs. high cfDNA-derived average adjusted ΔVAF (calculated by a machine-learning model) was an independent predictor of higher clinical benefit rate (stable disease ≥6 months/complete/partial response) (69.2% vs. 22.5%), and longer median progression-free (10.1 vs. 2.25 months) and overall survival (not reached vs. 6.1 months) (all P < .001, multivariate). bTMB changes did not correlate with outcomes. Therefore, early dynamic changes in cfDNA-derived VAF were a powerful predictor of pan-cancer immunotherapy outcomes. Liquid biopsy to predict immunotherapy response.
Collapse
Affiliation(s)
- Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Bing Li
- Ambry Genetics, Aliso Viejo, CA, USA
| | - Jacob J. Adashek
- Department of Internal Medicine, University of South Florida, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, USA
| | | | | | | | - Lisa Kim
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Tiffany W so
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Marcus Mitchell
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Naoki Kamei
- Bio Advanced Technology Division, Corporate R&D Headquarters, Konica Minolta, Inc, Tokyo, Japan
| | | | - Jayne Hoo
- Ambry Genetics, Aliso Viejo, CA, USA
| | | | - Teruaki Iyama
- Bio Advanced Technology Division, Corporate R&D Headquarters, Konica Minolta, Inc, Tokyo, Japan
| | - Masahide Kashiwagi
- Bio Advanced Technology Division, Corporate R&D Headquarters, Konica Minolta, Inc, Tokyo, Japan
| | | | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| |
Collapse
|
38
|
Hirahata T, ul Quraish R, Quraish AU, ul Quraish S, Naz M, Razzaq MA. Liquid Biopsy: A Distinctive Approach to the Diagnosis and Prognosis of Cancer. Cancer Inform 2022; 21:11769351221076062. [PMID: 35153470 PMCID: PMC8832574 DOI: 10.1177/11769351221076062] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/07/2022] [Indexed: 12/23/2022] Open
Abstract
Cancer is a leading cause of morbidity and mortality worldwide. Over the past decades, the concept of precision cancer medicine has emerged as a novel approach in the field of oncology that aims to tailor the most effective treatment options to each individual cancer patient based on the genetic profile of the tumor of each individual patient. Recently, tissue biopsy has become an essential part of cancer care and is widely used to characterize the tumor. However, tissue biopsy techniques face different challenges due to their invasiveness, cost, time, and adversity in potential sampling due to tissue heterogeneity. To overcome these issues, a non-invasive approach has developed, which is known as liquid biopsy. It is a simple, fast, and worthwhile technique based on the analysis of circulating tumor DNA (which is a fraction of cfDNA), circulating tumor cells (CTCs), and other tumor-derived material in blood plasma. This review provides an overview of the concept of liquid biopsy and briefly discusses the role of ctDNA and CTC analysis as tools for early diagnosis and prognosis of cancer. In this review, we also speculate on the advantages of liquid biopsy as opposed to tissue biopsy and postulate that liquid biopsy may be a comprehensive approach to overcome the current limitations associated with costly, invasive, and time-consuming tissue biopsy.
Collapse
Affiliation(s)
| | | | | | | | - Munazzah Naz
- Hirahata Gene Therapy Laboratory, HIC Clinic, Tokyo, Japan
| | | |
Collapse
|
39
|
Exploring the Clinical Utility of Pancreatic Cancer Circulating Tumor Cells. Int J Mol Sci 2022; 23:ijms23031671. [PMID: 35163592 PMCID: PMC8836025 DOI: 10.3390/ijms23031671] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/27/2022] [Accepted: 01/28/2022] [Indexed: 01/27/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the most frequent pancreatic cancer type, characterized by a dismal prognosis due to late diagnosis, frequent metastases, and limited therapeutic response to standard chemotherapy. Circulating tumor cells (CTCs) are a rare subset of tumor cells found in the blood of cancer patients. CTCs has the potential utility for screening, early and definitive diagnosis, prognostic and predictive assessment, and offers the potential for personalized management. However, a gold-standard CTC detection and enrichment method remains elusive, hindering comprehensive comparisons between studies. In this review, we summarize data regarding the utility of CTCs at different stages of PDAC from early to metastatic disease and discuss the molecular profiling and culture of CTCs. The characterization of CTCs brings us closer to defining the specific CTC subpopulation responsible for metastasis with the potential to uncover new therapies and more effective management options for PDAC.
Collapse
|
40
|
Botrus G, Uson Junior PLS, Raman P, Kaufman AE, Kosiorek H, Yin J, Fu Y, Majeed U, Sonbol MB, Ahn DH, Chang IW, Drusbosky LM, Dada H, Starr J, Borad M, Mody K, Bekaii-Saab TS. Circulating Cell-Free Tumor DNA in Advanced Pancreatic Adenocarcinoma Identifies Patients With Worse Overall Survival. Front Oncol 2022; 11:794009. [PMID: 35083150 PMCID: PMC8784799 DOI: 10.3389/fonc.2021.794009] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 12/20/2021] [Indexed: 12/24/2022] Open
Abstract
Background Plasma-based circulating cell-free tumor DNA (ctDNA) genomic profiling by next-generation sequencing (NGS)is an emerging diagnostic tool for pancreatic cancer (PC). The impact of detected genomic alterations and variant allele fraction (VAF) in tumor response to systemic treatments and outcomes is under investigation. Methods Patients with advanced PC who had ctDNA profiled at time of initial diagnosis were retrospectively evaluated. We considered the somatic alteration with the highest VAF as the dominant clone allele frequency (DCAF). ctDNA NGS results were related to clinical demographics, progression-free survival (PFS) and overall survival (OS). Results A total of 104 patients were evaluated. Somatic alterations were detected in 84.6% of the patients. Patients with ≥ 2 detectable genomic alterations had worse median PFS (p < 0.001) and worse median OS (p = 0.001). KRAS was associated with disease progression to systemic treatments (80.4% vs 19.6%, p = 0.006), worse median PFS (p < 0.001) and worse median OS (p = 0.002). TP53 was associated with worse median PFS (p = 0.02) and worse median OS (p = 0.001). The median DCAF was 0.45% (range 0-55%). DCAF >0.45% was associated with worse median PFS (p<0.0001) and median OS (p=0.0003). Patients that achieved clearance of KRAS had better PFS (p=0.047), while patients that achieved clearance of TP53 had better PFS (p=0.0056) and OS (p=0.037). Conclusions Initial detection of ctDNA in advanced PC can identify somatic alterations that may help predict clinical outcomes. The dynamics of ctDNA are prognostic of outcomes and should be evaluated in prospective studies.
Collapse
Affiliation(s)
- Gehan Botrus
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Pedro Luiz Serrano Uson Junior
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States.,Center for Personalized Medicine, Hospital Israelita Albert Einstein, Sao Paulo, Brazil
| | - Puneet Raman
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Adrienne E Kaufman
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Heidi Kosiorek
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Jun Yin
- Division of Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Yu Fu
- Guardant Health, Inc., Redwood City, CA, United States
| | - Umair Majeed
- Division of Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Mohamad Bassam Sonbol
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Daniel H Ahn
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | - Isabela W Chang
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States
| | | | - Hiba Dada
- Guardant Health, Inc., Redwood City, CA, United States
| | - Jason Starr
- Division of Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Mitesh Borad
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States.,Center of individualized Medicine, Mayo Clinic, Rochester, MN, United States.,Mayo Clinic Cancer Center, Phoenix, AZ, United States
| | - Kabir Mody
- Division of Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Tanios S Bekaii-Saab
- Division of Hematology and Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, United States
| |
Collapse
|
41
|
Nikas IP, Mountzios G, Sydney GI, Ioakim KJ, Won JK, Papageorgis P. Evaluating Pancreatic and Biliary Neoplasms with Small Biopsy-Based Next Generation Sequencing (NGS): Doing More with Less. Cancers (Basel) 2022; 14:cancers14020397. [PMID: 35053560 PMCID: PMC8773813 DOI: 10.3390/cancers14020397] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/05/2022] [Accepted: 01/10/2022] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Pancreatic cancer and cholangiocarcinoma are aggressive diseases mostly diagnosed at an advanced and inoperable stage. This review presents the value of next-generation sequencing (NGS) when performed on small biopsies—including fine-needle aspiration/biopsy samples, brushings, pancreatic juice and bile, and also blood—in the field of pancreatobiliary neoplasia. NGS could guide physicians while evaluating pancreatic solid and cystic lesions or suspicious biliary strictures, performing surveillance in high-risk individuals, or monitoring the disease and assessing prognosis in already diagnosed cancer patients. Evidence suggests that NGS performed on small biopsies is a robust tool for the diagnosis and pre-operative risk stratification of pancreatic and biliary lesions, whereas it also carries significant prognostic and therapeutic value. However, effective standardization of the pre-analytical and analytical assay parameters used for each clinical scenario is needed to fully implement NGS into routine practice and provide more personalized management in patients with suspected or established pancreatobiliary neoplasia. Abstract Pancreatic cancer and cholangiocarcinoma are lethal diseases mainly diagnosed at an inoperable stage. As pancreatobiliary surgical specimens are often unavailable for further molecular testing, this review aimed to highlight the diagnostic, prognostic, and therapeutic impact of next-generation sequencing (NGS) performed on distinct small biopsies, including endoscopic ultrasound fine-needle aspirations and biopsies of pancreatic solid and cystic lesions, biliary duct brushings, and also “liquid biopsies” such as the pancreatic juice, bile, and blood. NGS could clarify indeterminate pancreatic lesions or biliary strictures, for instance by identifying TP53 or SMAD4 mutations indicating high-grade dysplasia or cancer. It could also stratify pancreatic cystic lesions, by distinguishing mucinous from non-mucinous cysts and identifying high-risk cysts that should be excised in surgically fit patients, whereas the combination of cytology, elevated cystic CEA levels and NGS could improve the overall diagnostic accuracy. When NGS is performed on the pancreatic juice, it could stratify high-risk patients under surveillance. On the plasma, it could dynamically monitor the disease course and response to therapy. Notably, the circulating tumor DNA (ctDNA) levels have been associated with staging, grading, and survival. Lastly, NGS has shown potential in identifying potentially actionable molecular alterations. In conclusion, NGS applied on small biopsies could carry significant diagnostic, prognostic, and therapeutic value.
Collapse
Affiliation(s)
- Ilias P. Nikas
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus; (G.I.S.); (K.J.I.)
- Correspondence:
| | - Giannis Mountzios
- Fourth Department of Medical Oncology and Clinical Trials Unit, Henry Dunant Hospital Center, 11526 Athens, Greece;
| | - Guy I. Sydney
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus; (G.I.S.); (K.J.I.)
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Kalliopi J. Ioakim
- School of Medicine, European University Cyprus, Nicosia 2404, Cyprus; (G.I.S.); (K.J.I.)
- Department of Internal Medicine, Limassol General Hospital, Limassol 4131, Cyprus
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital and College of Medicine, Seoul 03080, Korea;
| | - Panagiotis Papageorgis
- Tumor Microenvironment, Metastasis and Experimental Therapeutics Laboratory, Basic and Translational Cancer Research Center, Department of Life Sciences, European University Cyprus, Nicosia 2404, Cyprus;
| |
Collapse
|
42
|
Bunduc S, Gede N, Váncsa S, Lillik V, Kiss S, Dembrovszky F, Eróss B, Szakács Z, Gheorghe C, Mikó A, Hegyi P. Prognostic role of cell-free DNA biomarkers in pancreatic adenocarcinoma: A systematic review and meta-analysis. Crit Rev Oncol Hematol 2021; 169:103548. [PMID: 34843928 DOI: 10.1016/j.critrevonc.2021.103548] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/22/2021] [Accepted: 11/23/2021] [Indexed: 12/16/2022] Open
Abstract
This systematic review and meta-analysis evaluated the prognostic role of cell-free DNA (cfDNA) in pancreatic ductal adenocarcinoma (PDAC). Eligible studies reported differences in overall (OS) and progression-free survival (PFS) by cfDNA status. The random effect model yielded the pooled hazard ratios (HRs) and 95 % confidence intervals (CI). Detection of circulant-tumor DNA (ctDNA), KRAS mutations and other cfDNA alterations constitute detectable cfDNA biomarkers. Altogether, 38 studies (3,318 patients) were eligible. Progression-free and overall survival were decreased with detectable ctDNA (HR = 1.92, 95 %CI:(1.29,2.86); HR = 2.25, 95 %CI:(1.73,2.92)) and KRAS mutations (HR = 1.88, CI:1.22,2.92,); HR = 1.52, 95 %CI:(1.22,1.90)) respectively, across various stages. In unresectable cases, ctDNA (HR = 2.50, 95 %CI:(1.94,3.23)), but not KRAS mutations (HR = 1.16, 95 %CI:(0.46,2.94)) signaled risk for progression. Detectable cfDNA biomarkers correlated with worse prognosis in resectable cases and if detected during treatment. In conclusion, cfDNA biomarkers indicate accelerated progression and decreased survival in PDAC. Significance of KRAS mutations detection in unresectable cases is to be determined.
Collapse
Affiliation(s)
- Stefania Bunduc
- Carol Davila University of Medicine and Pharmacy, 020021, Bucharest, Dionisie Lupu street 37, Romania; Fundeni Clinical Institute, 022328, Fundeni street 258, Bucharest, Romania; Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; Centre for Translational Medicine, Semmelweis University, 1085, Budapest, Üllői út. 26, Hungary; Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085, Budapest, Baross út. 8, Hungary.
| | - Noémi Gede
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary.
| | - Szilárd Váncsa
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; Centre for Translational Medicine, Semmelweis University, 1085, Budapest, Üllői út. 26, Hungary; Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085, Budapest, Baross út. 8, Hungary.
| | - Veronika Lillik
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; University of Pécs, Medical School, Department of Medical Genetics, 7624, Pécs, Szigeti út 12, Hungary.
| | - Szabolcs Kiss
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; Doctoral School of Clinical Medicine, University of Szeged, 6720, Szeged, Szeged, Dugonics tér 1, Hungary.
| | - Fanni Dembrovszky
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; Centre for Translational Medicine, Semmelweis University, 1085, Budapest, Üllői út. 26, Hungary.
| | - Bálint Eróss
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; Centre for Translational Medicine, Semmelweis University, 1085, Budapest, Üllői út. 26, Hungary; Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085, Budapest, Baross út. 8, Hungary.
| | - Zsolt Szakács
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; First Department of Medicine, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary.
| | - Cristian Gheorghe
- Carol Davila University of Medicine and Pharmacy, 020021, Bucharest, Dionisie Lupu street 37, Romania; Fundeni Clinical Institute, 022328, Fundeni street 258, Bucharest, Romania.
| | - Alexandra Mikó
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; University of Pécs, Medical School, Department of Medical Genetics, 7624, Pécs, Szigeti út 12, Hungary.
| | - Péter Hegyi
- Institute for Translational Medicine, Szentágothai Research Centre, Medical School, University of Pécs, 7624, Pécs, Szigeti út 12, Hungary; Centre for Translational Medicine, Semmelweis University, 1085, Budapest, Üllői út. 26, Hungary; Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, 1085, Budapest, Baross út. 8, Hungary.
| |
Collapse
|
43
|
Pietrasz D, Wang-Renault S, Taieb J, Dahan L, Postel M, Durand-Labrunie J, Le Malicot K, Mulot C, Rinaldi Y, Phelip JM, Doat S, Blons H, de Reynies A, Bachet JB, Taly V, Laurent-Puig P. Prognostic value of circulating tumour DNA in metastatic pancreatic cancer patients: post-hoc analyses of two clinical trials. Br J Cancer 2021; 126:440-448. [PMID: 34811505 DOI: 10.1038/s41416-021-01624-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 10/20/2021] [Accepted: 10/29/2021] [Indexed: 11/09/2022] Open
Abstract
OBJECTIVE The prognostication of metastatic pancreatic adenocarcinoma (mPDAC) patients remains uncertain, mainly based on carbohydrate antigen 19-9 (CA19-9), with limited utility. Circulating tumour DNA (ctDNA) has been suggested as a prognostic factor, but its added value has been poorly explored. The objective was to determine whether ctDNA is an independent factor for the prognostication of mPDAC. DESIGN Translational study based on two prospective collections of plasma samples of mPDAC patients naïve for chemotherapy. One used as a test series and the other as validation series coming from two randomised trials (Prodige 35 and Prodige 37). CtDNA was assessed by digital droplet PCR targeting two methylated markers (HOXD8 and POU4F1) according to a newly developed and validated method. Univariate and multivariate analyses were performed according to ctDNA status. RESULTS Of 372 plasma samples available, 354 patients were analyzed for survival. In the validation series, 145 of 255 patients were found ctDNA positive (56.8%), Median PFS and OS were 5.3 and 8.2 months in ctDNA-positive and 6.2 and 12.6 months in ctDNA-negative patients, respectively. ctDNA positivity was more often associated with young age, high CA19-9 level and neutrophils lymphocytes ratio. In multivariate analysis including these previous markers, ctDNA was confirmed as an independent prognostic marker for PFS (adjusted hazard ratio (HR) 1.5, CI 95% [1.03-2.18], p = 0.034) and OS (HR 1.62, CI 95% [1.05-2.5], p = 0.029). CONCLUSIONS In this first ctDNA assessment in a large series of mPDAC derived from clinical trials, ctDNA was detectable in 56.8% of patients and confirmed as an independent prognostic marker.
Collapse
Affiliation(s)
- Daniel Pietrasz
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France.,Assistance Publique-Hopitaux de Paris Hôpital Paul-Brousse, Centre Hépato-Biliaire, Université Paris-Saclay, 94800, Villejuif, France
| | - Shufang Wang-Renault
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France
| | - Julien Taieb
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France.,Institut du Cancer Paris Carpem, APHP.Centre -Université de Paris, Hopital Européen Georges Pompidou, Assistance Publique Hopitaux de Paris, Paris, France
| | - Laetitia Dahan
- Hepato-Gastroenterology and Oncology Department, University Hospital la Timone, Marseille, France
| | - Mathilde Postel
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France
| | - Jerome Durand-Labrunie
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France
| | - Karine Le Malicot
- Fédération Francophone de Cancérologie Digestive (FFCD); EPICAD INSERM LNC-UMR 1231, University of Burgundy and Franche Comté, Dijon, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France.,Biological resources center-EPIGENETEC BB-0033-00055, Paris, France
| | - Yves Rinaldi
- Gastroenterology Departement, Hôpital Européen, Marseille, France
| | | | - Solene Doat
- Gastroenterology and Digestive Oncology Department, Pitié-Salpêtrière Hospital, Sorbonne University, UPMC University, Paris, France
| | - Hélène Blons
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France.,Institut du cancer Paris Carpem; APHP.Centre-Université de Paris, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Aurelien de Reynies
- Programme Cartes d'Identité des Tumeurs, Ligue Nationale Contre le Cancer, Paris, France
| | - Jean-Baptiste Bachet
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France.,Gastroenterology and Digestive Oncology Department, Pitié-Salpêtrière Hospital, Sorbonne University, UPMC University, Paris, France
| | - Valérie Taly
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France.
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, CNRS, Sorbonne Université, USPC, Université de Paris, Equipe labellisée Ligue Nationale contre le cancer, CNRS SNC 5096, Paris, France. .,Institut du cancer Paris Carpem; APHP.Centre-Université de Paris, Assistance Publique - Hopitaux de Paris, Paris, France.
| |
Collapse
|
44
|
Bratlie SO, Wennerblom J, Vilhav C, Persson J, Rangelova E. Resectable, borderline, and locally advanced pancreatic cancer-"the good, the bad, and the ugly" candidates for surgery? J Gastrointest Oncol 2021; 12:2450-2460. [PMID: 34790406 DOI: 10.21037/jgo-2020-slapc-04] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/10/2020] [Indexed: 12/31/2022] Open
Abstract
The possibility of surgical resection strongly overrules medical oncologic treatment and is the only modality, causa sine qua non, long-term survival can be achieved in patients with pancreatic cancer. For this reason, the clinical classification of local resectability, subdividing tumors into resectable, borderline resectable, and locally advanced cancer, that is very technical in nature, is the one most widely used and accepted. As multimodality treatment with potent agents, particularly in the neoadjuvant setting, seems to be stepping forward as the new standard of treatment of pancreatic cancer, the established technical surgical landmarks tend to get challenged. This review aims to highlight the grey zones in the current classifications for local tumor involvement with respect to the observed patient outcome in the current multimodality treatment era. It summarizes the latest reported series on the outcome of resected primary resectable, borderline and locally advanced pancreatic cancer, and particularly vascular resections during pancreatectomy, in the background of different types of neoadjuvant therapy. It also hints what the new horizons of cancer biology tend to reveal whenever the technical hinders start being pushed aside. The current calls for the necessity of re-classification of the clinical categories of pancreatic cancer, from technically oriented to biology-focused individualized approach, are being elucidated.
Collapse
Affiliation(s)
- Svein Olav Bratlie
- Section for Upper Abdominal Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Surgery, The Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Johanna Wennerblom
- Section for Upper Abdominal Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Surgery, The Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Caroline Vilhav
- Section for Upper Abdominal Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Surgery, The Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Jan Persson
- Section for Upper Abdominal Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Surgery, The Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Elena Rangelova
- Section for Upper Abdominal Surgery, Department of Surgery, Sahlgrenska University Hospital, Gothenburg, Sweden.,Department of Surgery, The Institute of Clinical Sciences, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
45
|
Xu Z, Wang X, Zeng S, Ren X, Yan Y, Gong Z. Applying artificial intelligence for cancer immunotherapy. Acta Pharm Sin B 2021; 11:3393-3405. [PMID: 34900525 PMCID: PMC8642413 DOI: 10.1016/j.apsb.2021.02.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/07/2020] [Accepted: 12/21/2020] [Indexed: 02/07/2023] Open
Abstract
Artificial intelligence (AI) is a general term that refers to the use of a machine to imitate intelligent behavior for performing complex tasks with minimal human intervention, such as machine learning; this technology is revolutionizing and reshaping medicine. AI has considerable potential to perfect health-care systems in areas such as diagnostics, risk analysis, health information administration, lifestyle supervision, and virtual health assistance. In terms of immunotherapy, AI has been applied to the prediction of immunotherapy responses based on immune signatures, medical imaging and histological analysis. These features could also be highly useful in the management of cancer immunotherapy given their ever-increasing performance in improving diagnostic accuracy, optimizing treatment planning, predicting outcomes of care and reducing human resource costs. In this review, we present the details of AI and the current progression and state of the art in employing AI for cancer immunotherapy. Furthermore, we discuss the challenges, opportunities and corresponding strategies in applying the technology for widespread clinical deployment. Finally, we summarize the impact of AI on cancer immunotherapy and provide our perspectives about underlying applications of AI in the future.
Collapse
Key Words
- AI, artificial intelligence
- Artificial intelligence
- CT, computed tomography
- CTLA-4, cytotoxic T lymphocyte-associated antigen 4
- Cancer immunotherapy
- DL, deep learning
- Diagnostics
- ICB, immune checkpoint blockade
- MHC-I, major histocompatibility complex class I
- ML, machine learning
- MMR, mismatch repair
- MRI, magnetic resonance imaging
- Machine learning
- PD-1, programmed cell death protein 1
- PD-L1, PD-1 ligand1
- TNBC, triple-negative breast cancer
- US, ultrasonography
- irAEs, immune-related adverse events
Collapse
Affiliation(s)
- Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xiang Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Xinxin Ren
- Center for Molecular Medicine, Xiangya Hospital, Key Laboratory of Molecular Radiation Oncology of Hunan Province, Central South University, Changsha 410008, China
| | - Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
46
|
Jensen TJ, Goodman AM, Ellison CK, Holden KA, Kato S, Kim L, Daniels GA, Fitzgerald K, McCarthy E, Nakashe P, Mazloom AR, Almasri E, McLennan G, Grosu DS, Eisenberg M, Kurzrock R. Genome-wide Sequencing of Cell-free DNA Enables Detection of Copy-number Alterations in Patients with Cancer Where Tissue Biopsy is Not Feasible. Mol Cancer Ther 2021; 20:2274-2279. [PMID: 34465593 PMCID: PMC9398131 DOI: 10.1158/1535-7163.mct-20-1066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/03/2021] [Accepted: 08/20/2021] [Indexed: 01/07/2023]
Abstract
When tissue biopsy is not medically prudent or tissue is insufficient for molecular testing, alternative methods are needed. Because cell-free DNA (cfDNA) has been shown to provide a representative surrogate for tumor tissue, we sought to evaluate its utility in this clinical scenario. cfDNA was isolated from the plasma of patients and assayed with low-coverage (∼0.3×), genome-wide sequencing. Copy-number alterations (CNA) were identified and characterized using analytic methods originally developed for noninvasive prenatal testing (NIPT) and quantified using the genomic instability number (GIN), a metric that reflects the quantity and magnitude of CNAs across the genome. The technical variability of the GIN was first evaluated in an independent cohort comprising genome-wide sequencing results from 27,754 women who consented to have their samples used for research and whose NIPT results yielded no detected CNAs to establish a detection threshold. Subsequently, cfDNA sequencing data from 96 patients with known cancers but for whom a tissue biopsy could not be obtained are presented. An elevated GIN was detected in 35% of patients and detection rates varied by tumor origin. Collectively, CNAs covered 96.6% of all autosomes. Survival was significantly reduced in patients with an elevated GIN relative to those without. Overall, these data provide a proof of concept for the use of low-coverage, genome-wide sequencing of cfDNA from patients with cancer to obtain relevant molecular information in instances where tissue is difficult to access. These data may ultimately serve as an informative complement to other molecular tests.
Collapse
Affiliation(s)
- Taylor J. Jensen
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California.,Laboratory Corporation of America, Durham, North Carolina.,Corresponding Author: Taylor J. Jensen, Research and Development, Laboratory Corporation of America, 1912 TW Alexander, Durham, NC 27703. Phone: 858-242-6842; E-mail:
| | - Aaron M. Goodman
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego.,Department of Medicine, Division of Blood and Marrow Transplantation, Moores Cancer Center, University of California, San Diego
| | - Christopher K. Ellison
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Kimberly A. Holden
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Shumei Kato
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego.,Department of Medicine, Division of Precision Medicine, Moores Cancer Center, University of California, San Diego
| | - Lisa Kim
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego
| | - Gregory A. Daniels
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego
| | - Kerry Fitzgerald
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Erin McCarthy
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Prachi Nakashe
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Amin R. Mazloom
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Eyad Almasri
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Graham McLennan
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | - Daniel S. Grosu
- Sequenom, Inc, a wholly owned subsidiary of Laboratory Corporation of America Holdings, San Diego, California
| | | | - Razelle Kurzrock
- Department of Medicine, Division of Hematology/Oncology, and Center for Personalized Cancer Therapy, Moores Cancer Center, University of California, San Diego.,Department of Medicine, Division of Precision Medicine, Moores Cancer Center, University of California, San Diego
| |
Collapse
|
47
|
Roosan MR, Mambetsariev I, Pharaon R, Fricke J, Husain H, Reckamp KL, Koczywas M, Massarelli E, Bild AH, Salgia R. Usefulness of Circulating Tumor DNA in Identifying Somatic Mutations and Tracking Tumor Evolution in Patients With Non-small Cell Lung Cancer. Chest 2021; 160:1095-1107. [PMID: 33878340 PMCID: PMC8449001 DOI: 10.1016/j.chest.2021.04.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 03/21/2021] [Accepted: 04/01/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The usefulness of circulating tumor DNA (ctDNA) in detecting mutations and monitoring treatment response has not been well studied beyond a few actionable biomarkers in non-small cell lung cancer (NSCLC). RESEARCH QUESTION How does the usefulness of ctDNA analysis compare with that of solid tumor biopsy analysis in patients with NSCLC? METHODS We retrospectively evaluated 370 adult patients with NSCLC treated at the City of Hope between November 2015 and August 2019 to assess the usefulness of ctDNA in mutation identification, survival, concordance with matched tissue samples in 32 genes, and tumor evolution. RESULTS A total of 1,688 somatic mutations were detected in 473 ctDNA samples from 370 patients with NSCLC. Of the 473 samples, 177 showed at least one actionable mutation with currently available Food and Drug Administration-approved NSCLC therapies. MET and CDK6 amplifications co-occurred with BRAF amplifications (false discovery rate [FDR], < 0.01), and gene-level mutations were mutually exclusive in KRAS and EGFR (FDR, 0.0009). Low cumulative percent ctDNA levels were associated with longer progression-free survival (hazard ratio [HR], 0.56; 95% CI, 0.37-0.85; P = .006). Overall survival was shorter in patients harboring BRAF mutations (HR, 2.35; 95% CI, 1.24-4.6; P = .009), PIK3CA mutations (HR, 2.77; 95% CI, 1.56-4.9; P < .001) and KRAS mutations (HR, 2.32; 95% CI, 1.30-4.1; P = .004). Gene-level concordance was 93.8%, whereas the positive concordance rate was 41.6%. More mutations in targetable genes were found in ctDNA than in tissue biopsy samples. Treatment response and tumor evolution over time were detected in repeated ctDNA samples. INTERPRETATION Although ctDNA analysis exhibited similar usefulness to tissue biopsy analysis, more mutations in targetable genes were missed in tissue biopsy analyses. Therefore, the evaluation of ctDNA in conjunction with tissue biopsy samples may help to detect additional targetable mutations to improve clinical outcomes in advanced NSCLC.
Collapse
Affiliation(s)
| | | | | | - Jeremy Fricke
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Hatim Husain
- UC San Diego Health Moores Cancer Center, La Jolla, CA
| | - Karen L Reckamp
- City of Hope Comprehensive Cancer Center, Duarte, CA; Division of Medical Oncology, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | | | - Andrea H Bild
- Division of Molecular Pharmacology, Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA
| | - Ravi Salgia
- City of Hope Comprehensive Cancer Center, Duarte, CA.
| |
Collapse
|
48
|
Hua H, Zhang H, Chen J, Wang J, Liu J, Jiang Y. Targeting Akt in cancer for precision therapy. J Hematol Oncol 2021; 14:128. [PMID: 34419139 PMCID: PMC8379749 DOI: 10.1186/s13045-021-01137-8] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 08/03/2021] [Indexed: 02/08/2023] Open
Abstract
Biomarkers-guided precision therapeutics has revolutionized the clinical development and administration of molecular-targeted anticancer agents. Tailored precision cancer therapy exhibits better response rate compared to unselective treatment. Protein kinases have critical roles in cell signaling, metabolism, proliferation, survival and migration. Aberrant activation of protein kinases is critical for tumor growth and progression. Hence, protein kinases are key targets for molecular targeted cancer therapy. The serine/threonine kinase Akt is frequently activated in various types of cancer. Activation of Akt promotes tumor progression and drug resistance. Since the first Akt inhibitor was reported in 2000, many Akt inhibitors have been developed and evaluated in either early or late stage of clinical trials, which take advantage of liquid biopsy and genomic or molecular profiling to realize personalized cancer therapy. Two inhibitors, capivasertib and ipatasertib, are being tested in phase III clinical trials for cancer therapy. Here, we highlight recent progress of Akt signaling pathway, review the up-to-date data from clinical studies of Akt inhibitors and discuss the potential biomarkers that may help personalized treatment of cancer with Akt inhibitors. In addition, we also discuss how Akt may confer the vulnerability of cancer cells to some kinds of anticancer agents.
Collapse
Affiliation(s)
- Hui Hua
- State Key Laboratory of Biotherapy, Laboratory of Stem Cell Biology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingzhu Chen
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jieya Liu
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yangfu Jiang
- State Key Laboratory of Biotherapy, Laboratory of Oncogene, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
49
|
Haghighat Jahromi A, Zabel M, Okamura R, Hoh CK, Kurzrock R. Variant allele fraction of genomic alterations in circulating tumor DNA (%ctDNA) correlates with SUV max in PET scan. AMERICAN JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING 2021; 11:307-312. [PMID: 34513284 PMCID: PMC8414395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 07/21/2021] [Indexed: 06/13/2023]
Abstract
The relationship between higher variant allele fraction (VAF) of genomic alterations in circulating tumor DNA (%ctDNA), an indicator of poor outcome, and maximum standardized uptake value (SUVmax), the most commonly used semi-quantitative parameter in 18F-FDG PET/CT, has not been studied. Overall, 433 cancer patients had blood-based next generation sequencing. Maximum and sum of %ctDNA alterations (%ctDNAmax and %ctDNAsum, respectively) represent the maximum and sum of VAF, reported as a percentage. The subset of 46 eligible patients had treatment-naïve metastatic disease and PET/CT imaging, with median 13 days prior to ctDNA testing. We found a linear correlation between the maximum VAF (%ctDNAmax) (as well as the sum of the VAFs (%ctDNAsum)) and SUVmax of the most 18F-FDG-avid lesion (r=0.43, P=0.003; r=0.43, P=0.002; respectively). Our data suggest that SUVmax may be a non-invasive and readily available surrogate indicator for %ctDNA, a prognostic factor for patient survival. Since higher %ctDNA has been previously correlated with worse outcome, the relationship between SUVmax, %ctDNA and survival warrants further study.
Collapse
Affiliation(s)
- Amin Haghighat Jahromi
- Department of Radiology, University of CaliforniaSan Diego, La Jolla, California, USA
- Division of Nuclear Medicine, Edward Mallinckrodt Institute of Radiology, Washington University School of MedicineSt. Louis, MO, USA
| | - Matthew Zabel
- Department of Radiology, University of CaliforniaSan Diego, La Jolla, California, USA
| | - Ryosuke Okamura
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer CenterLa Jolla, CA, USA
| | - Carl K Hoh
- Department of Radiology, University of CaliforniaSan Diego, La Jolla, California, USA
| | - Razelle Kurzrock
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego Moores Cancer CenterLa Jolla, CA, USA
| |
Collapse
|
50
|
Balendran-Braun S, Kieler M, Liebmann-Reindl S, Unseld M, Bianconi D, W Prager G, Streubel B. Bead-Based Isolation of Circulating Tumor DNA from Pancreatic Cancer Patients Enables High Fidelity Next Generation Sequencing. Cancer Manag Res 2021; 13:6249-6261. [PMID: 34393517 PMCID: PMC8357621 DOI: 10.2147/cmar.s308029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/07/2021] [Indexed: 11/23/2022] Open
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDAC) is one of the most aggressive cancers and poses a challenge to the treating clinician. With the emergence of genomic profiling technologies, circulating tumor DNA (ctDNA) is increasingly recognized as a versatile biomarker for risk stratification and disease monitoring. We aimed to compare two commercially available NGS panels in a cohort of patients with advanced PDAC undergoing palliative chemotherapy. METHODS CtDNA was isolated with a magnetic bead-based protocol from two consecutive blood samples before and during chemotherapy in 21 patients with PDAC. Mutations were assessed by using a panel covering 15 (GP15) or 50 (GP50) cancer-associated genes. Results were compared to tumor tissue (GP15), if available. RESULTS Isolation of ctDNA resulted in a high mean value of 1.9 ng/µL (total volume of ~40 µL). Although the same number of patients were positive for at least one mutation (76%), the most commonly mutated oncogene in PDAC, KRAS, was detectable in an additional 25% of all patients with the GP15 panel due to a higher coverage. The genomic concordance rate between tissue DNA and ctDNA analyses was 65.22%. DISCUSSION Our study demonstrates the feasibility of an NGS-based approach for ctDNA analysis and underlines the importance of using a disease-specific panel with a sufficiently high coverage.
Collapse
Affiliation(s)
| | - Markus Kieler
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University, Vienna, Austria
| | | | - Matthias Unseld
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University, Vienna, Austria
| | - Daniela Bianconi
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University, Vienna, Austria
| | - Gerald W Prager
- Department of Medicine I, Division of Oncology, Comprehensive Cancer Center, Medical University, Vienna, Austria
| | - Berthold Streubel
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Core Facility Genomics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|