1
|
Zhang R, Su C, Jia Y, Xing M, Jin S, Zong H. Molecular mechanisms of HER2-targeted therapy and strategies to overcome the drug resistance in colorectal cancer. Biomed Pharmacother 2024; 179:117363. [PMID: 39236476 DOI: 10.1016/j.biopha.2024.117363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/15/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024] Open
Abstract
HER2 amplification is one of the mechanisms that induce drug resistance to anti-EGFR therapy in colorectal cancer. In recent years, data from several randomized clinical trials show that anti-HER2 therapies improved the prognosis of patients with HER2-positive colorectal cancer. These results indicate that HER2 is a promising therapeutic target in advanced colorectal cancer. Despite the anti-HER2 therapies including monoclonal antibodies, tyrosine kinase inhibitors, and antibody-drug conjugates improving the outcomes, less than 30 % of the patients achieve objective response and eventually have drug resistance. It is necessary to explore the primary and secondary mechanisms for the resistance to anti-HER2 therapies, which will pave the way to overcome the drug resistance. Several studies have reported the potential mechanisms for the resistance to anti-HER2 therapies. In this review, we present a comprehensive overview of the recent advances in clinical research, mechanisms of treatment resistance, and strategies for reversing resistance in HER2-positive colorectal cancer patients.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Chang Su
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Yongliang Jia
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Menglu Xing
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Shuiling Jin
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Hong Zong
- Department of Oncology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| |
Collapse
|
2
|
Ratti M, Orlandi E, Toscani I, Vecchia S, Anselmi E, Hahne JC, Ghidini M, Citterio C. Emerging Therapeutic Targets and Future Directions in Advanced Gastric Cancer: A Comprehensive Review. Cancers (Basel) 2024; 16:2692. [PMID: 39123420 PMCID: PMC11311890 DOI: 10.3390/cancers16152692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024] Open
Abstract
Metastatic gastric cancer (GC) still represents a critical clinical challenge, with limited treatment options and a poor prognosis. Most patients are diagnosed at advanced stages, limiting the chances of surgery and cure. The identification of molecular targets and the possibility of combining immune checkpoint inhibitors with chemotherapy have recently reshaped the therapeutic landscape of metastatic gastric cancer. The new classification of gastric cancer, mainly based on immunologic and molecular criteria such as programmed cell death 1 (PD-1), microsatellite instability (MSI), and human epidermal growth factor receptor 2 (HER2), has made it possible to identify and differentiate patients who may benefit from immunotherapy, targeted therapy, or chemotherapy alone. All relevant and available molecular and immunological targets in clinical practice for the systemic treatment of this disease are presented. Particular attention is given to possible future approaches, including circulating tumor DNA (ctDNA) for therapeutic monitoring, new targeting agents against molecular pathways such as fibroblast growth factor receptor (FGFR) and MET, chimeric antigen receptor (CAR)-T cells, and cancer vaccines. This review aims to provide a comprehensive understanding of current targets in advanced gastric cancer and to offer valuable insights into future directions of research and clinical practice in this challenging disease.
Collapse
Affiliation(s)
- Margherita Ratti
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Elena Orlandi
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Ilaria Toscani
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Stefano Vecchia
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Elisa Anselmi
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| | - Jens Claus Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, Sutton, London SM2 5NG, UK;
| | - Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Chiara Citterio
- Oncology and Hematology Department, Piacenza General Hospital, Via Taverna 49, 29121 Piacenza, Italy; (E.O.); (I.T.); (S.V.); (E.A.); (C.C.)
| |
Collapse
|
3
|
Tao XY, Li QQ, Zeng Y. Clinical application of liquid biopsy in colorectal cancer: detection, prediction, and treatment monitoring. Mol Cancer 2024; 23:145. [PMID: 39014366 PMCID: PMC11250976 DOI: 10.1186/s12943-024-02063-2] [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: 03/08/2024] [Accepted: 07/09/2024] [Indexed: 07/18/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignancies affecting the gastrointestinal tract and is ranked third among cancers with the highest incidence and second-highest mortality rate worldwide. CRC exhibits a slow progression providing a wide treatment window. The currently employed CRC screening methods have shown great potential to prevent CRC and reduce CRC-related morbidity and mortality. The diagnosis of CRC is achieved by colonoscopy and tissue biopsy, with studies showing that liquid biopsy is more effective in detecting and diagnosing early CRC patients. Increasing number of studies have shown that the tumor components shed into circulating blood can be detected in liquid form, and can be applied in the clinical management of CRC. Analysis of circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), or tumor-associated platelets (TEPs) in the blood can be used for early screening and diagnosis of CRC, aid tumor staging, treatment response monitoring, and prediction of CRC recurrence and metastasis in a minimally invasive manner. This chapter provides an updated review of CTCs, ctDNA, and TEPs as novel biomarkers for CRC, highlighting their strengths and limitations.
Collapse
Affiliation(s)
- Xiang-Yuan Tao
- Translational Medicine Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- School of Pharmacy, University of South China, Hengyang, China
| | - Qian-Qian Li
- Translational Medicine Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- School of Pharmacy, University of South China, Hengyang, China
| | - Yong Zeng
- Translational Medicine Center, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.
- School of Pharmacy, University of South China, Hengyang, China.
| |
Collapse
|
4
|
Hedayat S, Cascione L, Cunningham D, Schirripa M, Lampis A, Hahne JC, Tunariu N, Hong SP, Marchetti S, Khan K, Fontana E, Angerilli V, Delrieux M, Nava Rodrigues D, Procaccio L, Rao S, Watkins D, Starling N, Chau I, Braconi C, Fotiadis N, Begum R, Guppy N, Howell L, Valenti M, Cribbes S, Kolozsvari B, Kirkin V, Lonardi S, Ghidini M, Passalacqua R, Elghadi R, Magnani L, Pinato DJ, Di Maggio F, Ghelardi F, Sottotetti E, Vetere G, Ciracì P, Vlachogiannis G, Pietrantonio F, Cremolini C, Cortellini A, Loupakis F, Fassan M, Valeri N. Circulating microRNA Analysis in a Prospective Co-clinical Trial Identifies MIR652-3p as a Response Biomarker and Driver of Regorafenib Resistance Mechanisms in Colorectal Cancer. Clin Cancer Res 2024; 30:2140-2159. [PMID: 38376926 DOI: 10.1158/1078-0432.ccr-23-2748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE The multi-kinase inhibitor (mKi) regorafenib has demonstrated efficacy in chemorefractory patients with metastatic colorectal cancer (mCRC). However, lack of predictive biomarkers and concerns over significant toxicities hamper the use of regorafenib in clinical practice. EXPERIMENTAL DESIGN Serial liquid biopsies were obtained at baseline and monthly until disease progression in chemorefractory patients with mCRC treated with regorafenib in a phase II clinical trial (PROSPECT-R n = 40; NCT03010722) and in a multicentric validation cohort (n = 241). Tissue biopsies collected at baseline, after 2 months and at progression in the PROSPECT-R trial were used to establish patient-derived organoids (PDO) and for molecular analyses. MicroRNA profiling was performed on baseline bloods using the NanoString nCounter platform and results were validated by digital-droplet PCR and/or ISH in paired liquid and tissue biopsies. PDOs co-cultures and PDO-xenotransplants were generated for functional analyses. RESULTS Large-scale microRNA expression analysis in longitudinal matched liquid and tissue biopsies from the PROSPECT-R trial identified MIR652-3p as a biomarker of clinical benefit to regorafenib. These findings were confirmed in an independent validation cohort and in a "control" group of 100 patients treated with lonsurf. Using ex vivo co-culture assays paired with single-cell RNA-sequencing of PDO established pre- and post-treatment, we modeled regorafenib response observed in vivo and in patients, and showed that MIR652-3p controls resistance to regorafenib by impairing regorafenib-induced lethal autophagy and by orchestrating the switch from neo-angiogenesis to vessel co-option. CONCLUSIONS Our results identify MIR652-3p as a potential biomarker and as a driver of cell and non-cell-autonomous mechanisms of resistance to regorafenib.
Collapse
Affiliation(s)
- Somaieh Hedayat
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Luciano Cascione
- Bioinformatics Core Unit, Institute of Oncology Research (IOR), Faculty of Biomedical Sciences, Università della Svizzera italiana, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Bellinzona, Switzerland
| | - David Cunningham
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Marta Schirripa
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Andrea Lampis
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Jens C Hahne
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Nina Tunariu
- Department of Radiology, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Sung Pil Hong
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Silvia Marchetti
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Khurum Khan
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Elisa Fontana
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Valentina Angerilli
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Mia Delrieux
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Daniel Nava Rodrigues
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Letizia Procaccio
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Sheela Rao
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Chiara Braconi
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nicos Fotiadis
- Department of Interventional Radiology, The Royal Marsden Hospital, London, United Kingdom
| | - Ruwaida Begum
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Naomy Guppy
- Breast Cancer Now Nina Barough Pathology Core Facility, The Institute of Cancer Research, London, United Kingdom
| | - Louise Howell
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Melanie Valenti
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | | | | | - Vladimir Kirkin
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Sara Lonardi
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Raghad Elghadi
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Luca Magnani
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - David J Pinato
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Federica Di Maggio
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate Francesco Salvatore, Via Gaetano Salvatore, Naples, Italy
| | - Filippo Ghelardi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Guglielmo Vetere
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Paolo Ciracì
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Georgios Vlachogiannis
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessio Cortellini
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Fotios Loupakis
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Matteo Fassan
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Nicola Valeri
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Lamichhane A, Tavana H. Three-Dimensional Tumor Models to Study Cancer Stemness-Mediated Drug Resistance. Cell Mol Bioeng 2024; 17:107-119. [PMID: 38737455 PMCID: PMC11082110 DOI: 10.1007/s12195-024-00798-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 02/01/2024] [Indexed: 05/14/2024] Open
Abstract
Solid tumors often contain genetically different populations of cancer cells, stromal cells, various structural and soluble proteins, and other soluble signaling molecules. The American Cancer society estimated 1,958,310 new cancer cases and 609,820 cancer deaths in the United States in 2023. A major barrier against successful treatment of cancer patients is drug resistance. Gain of stem cell-like states by cancer cells under drug pressure or due to interactions with the tumor microenvironment is a major mechanism that renders therapies ineffective. Identifying approaches to target cancer stem cells is expected to improve treatment outcomes for patients. Most of our understanding of drug resistance and the role of cancer stemness is from monolayer cell cultures. Recent advances in cell culture technologies have enabled developing sophisticated three-dimensional tumor models that facilitate mechanistic studies of cancer drug resistance. This review summarizes the role of cancer stemness in drug resistance and highlights the various tumor models that are used to discover the underlying mechanisms and test potentially novel therapeutics.
Collapse
Affiliation(s)
- Astha Lamichhane
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325 USA
| | - Hossein Tavana
- Department of Biomedical Engineering, The University of Akron, Akron, OH 44325 USA
| |
Collapse
|
6
|
Batool SM, Yekula A, Khanna P, Hsia T, Gamblin AS, Ekanayake E, Escobedo AK, You DG, Castro CM, Im H, Kilic T, Garlin MA, Skog J, Dinulescu DM, Dudley J, Agrawal N, Cheng J, Abtin F, Aberle DR, Chia D, Elashoff D, Grognan T, Krysan K, Oh SS, Strom C, Tu M, Wei F, Xian RR, Skates SJ, Zhang DY, Trinh T, Watson M, Aft R, Rawal S, Agarwal A, Kesmodel SB, Yang C, Shen C, Hochberg FH, Wong DTW, Patel AA, Papadopoulos N, Bettegowda C, Cote RJ, Srivastava S, Lee H, Carter BS, Balaj L. The Liquid Biopsy Consortium: Challenges and opportunities for early cancer detection and monitoring. Cell Rep Med 2023; 4:101198. [PMID: 37716353 PMCID: PMC10591039 DOI: 10.1016/j.xcrm.2023.101198] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 12/01/2022] [Accepted: 08/22/2023] [Indexed: 09/18/2023]
Abstract
The emerging field of liquid biopsy stands at the forefront of novel diagnostic strategies for cancer and other diseases. Liquid biopsy allows minimally invasive molecular characterization of cancers for diagnosis, patient stratification to therapy, and longitudinal monitoring. Liquid biopsy strategies include detection and monitoring of circulating tumor cells, cell-free DNA, and extracellular vesicles. In this review, we address the current understanding and the role of existing liquid-biopsy-based modalities in cancer diagnostics and monitoring. We specifically focus on the technical and clinical challenges associated with liquid biopsy and biomarker development being addressed by the Liquid Biopsy Consortium, established through the National Cancer Institute. The Liquid Biopsy Consortium has developed new methods/assays and validated existing methods/technologies to capture and characterize tumor-derived circulating cargo, as well as addressed existing challenges and provided recommendations for advancing biomarker assays.
Collapse
Affiliation(s)
| | - Anudeep Yekula
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Prerna Khanna
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tiffaney Hsia
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Austin S Gamblin
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Emil Ekanayake
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ana K Escobedo
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Dong Gil You
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Cesar M Castro
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hyungsoon Im
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tugba Kilic
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Johan Skog
- Exosome Diagnostics Inc., Waltham, MA, USA
| | | | - Jonathan Dudley
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Jordan Cheng
- University of California Los Angeles, Los Angeles, CA, USA
| | | | | | - David Chia
- University of California Los Angeles, Los Angeles, CA, USA
| | - David Elashoff
- University of California Los Angeles, Los Angeles, CA, USA
| | | | | | - Scott S Oh
- University of California Los Angeles, Los Angeles, CA, USA
| | - Charles Strom
- University of California Los Angeles, Los Angeles, CA, USA
| | - Michael Tu
- Liquid Diagnostics LLC., Los Angeles, CA, USA
| | - Fang Wei
- University of California Los Angeles, Los Angeles, CA, USA
| | - Rena R Xian
- Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven J Skates
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Thi Trinh
- Yale University School of Medicine, New Haven, CT, USA
| | - Mark Watson
- Washington University School of Medicine, St. Louis, MO, USA
| | - Rebecca Aft
- Washington University School of Medicine, St. Louis, MO, USA
| | - Siddarth Rawal
- Washington University School of Medicine, St. Louis, MO, USA; Circulogix Inc., St. Louis, MO, USA
| | | | | | | | - Cheng Shen
- California Institute of Technology, Pasadena, CA, USA
| | | | - David T W Wong
- University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | | - Richard J Cote
- Washington University School of Medicine, St. Louis, MO, USA; Circulogix Inc., St. Louis, MO, USA
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Hakho Lee
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Bob S Carter
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leonora Balaj
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Verner EL, Jackson JB, Severson E, Valkenburg KC, Greer AE, Riley DR, Sausen M, Maddox C, McGregor PM, Karandikar A, Hastings SB, Previs RA, Reddy VP, Jensen TJ, Ramkissoon SH. Validation of the Labcorp Plasma Focus Test to Facilitate Precision Oncology Through Cell-Free DNA Genomic Profiling of Solid Tumors. J Mol Diagn 2023; 25:477-489. [PMID: 37068734 DOI: 10.1016/j.jmoldx.2023.03.008] [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: 12/24/2022] [Revised: 02/28/2023] [Accepted: 03/30/2023] [Indexed: 04/19/2023] Open
Abstract
Genomic profiling is critical for precision oncology to guide treatment decisions. Liquid biopsy testing is a complementary approach to tissue testing, particularly when tissue is not readily available. The Labcorp Plasma Focus test is a circulating cell-free DNA genomic profiling test that identifies actionable variants in solid cancers, including non-small-cell lung, colorectal, melanoma, breast, esophageal, gastroesophageal junction, and gastric cancers. This study highlights the analytical validation of the test, including accuracy compared with orthogonal methods, as well as sensitivity, specificity, precision, reproducibility, and repeatability. Concordance with orthogonal methods showed percent positive agreement of 98.7%, 89.3%, and 96.2% for single nucleotide variants (SNVs), insertion/deletions (indels), and copy number amplifications (CNAs), respectively, and 100.0% for translocations and microsatellite instability (MSI). Analytical sensitivity revealed a median limit of detection of 0.7% and 0.6% for SNVs and indels, 1.4-fold for CNAs, 0.5% variant allele frequency for translocations, and 0.6% for MSI. Specificity was >99% for SNVs/indels and 100% for CNAs, translocations, and MSI. Average positive agreement from precision, reproducibility, and repeatability experiments was 97.5% and 88.9% for SNVs/indels and CNAs, and 100% for translocations and MSI. Taken together, these data show that the Labcorp Plasma Focus test is a highly accurate, sensitive, and specific approach for cell-free DNA genomic profiling to supplement tissue testing and inform treatment decisions.
Collapse
Affiliation(s)
- Ellen L Verner
- Personal Genome Diagnostics (PGDx), Baltimore, Maryland.
| | | | - Eric Severson
- Enterprise Oncology, Labcorp, Durham, North Carolina
| | | | - Amy E Greer
- Personal Genome Diagnostics (PGDx), Baltimore, Maryland
| | - David R Riley
- Personal Genome Diagnostics (PGDx), Baltimore, Maryland
| | - Mark Sausen
- Personal Genome Diagnostics (PGDx), Baltimore, Maryland
| | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Ye H, Wang Y, Yao S, Liu Z, Liang C, Zhu Y, Cui Y, Zhao K. Necrosis score as a prognostic factor in stage I-III colorectal cancer: a retrospective multicenter study. Discov Oncol 2023; 14:61. [PMID: 37155090 PMCID: PMC10167085 DOI: 10.1007/s12672-023-00655-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 04/12/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND Tumor necrosis results from failure to meet the requirement for rapid proliferation of tumor, related to unfavorable prognosis in colorectal cancer (CRC). However, previous studies used traditional microscopes to evaluate necrosis on slides, lacking a simultaneous phase and panoramic view for assessment. Therefore, we proposed a whole-slide images (WSIs)-based method to develop a necrosis score and validated its prognostic value in multicenter cohorts. METHODS Necrosis score was defined as the proportion of necrosis in the tumor area, semi-quantitatively classified into 3-level score groups by the cut-off of 10% and 30% on HE-stained WSIs. 768 patients from two centers were enrolled in this study, divided into a discovery (N = 445) and a validation (N = 323) cohort. The prognostic value of necrosis score was evaluated by Kaplan-Meier curves and the Cox model. RESULT Necrosis score was associated with overall survival, with hazard ratio for high vs. low in discovery and validation cohorts being 2.62 (95% confidence interval 1.59-4.32) and 2.51 (1.39-4.52), respectively. The 3-year disease free survival rates of necrosis-low, middle, and high were 83.6%, 80.2%, and 59.8% in discovery cohort, and 86.5%, 84.2%, and 66.5% in validation cohort. In necrosis middle plus high subgroup, there was a trend but no significant difference in overall survival between surgery alone and adjuvant chemotherapy group in stage II CRC (P = .075). CONCLUSION As a stable prognostic factor, high-level necrosis evaluated by the proposed method on WSIs was associated with unfavorable outcomes. Additionally, adjuvant chemotherapy provide survival benefits for patients with high necrosis in stage II CRC.
Collapse
Affiliation(s)
- Huifen Ye
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Yiting Wang
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-Sen University, 26 Yuan Cun 2 Cross Road, TianHe District, Guangzhou, 510655, China
| | - Su Yao
- Department of Pathology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, 106 Zhongshan Er Road, Guangzhou, 510080, China
| | - Yaxi Zhu
- Department of Pathology, The Sixth Affiliated Hospital of Sun Yat-Sen University, 26 Yuan Cun 2 Cross Road, TianHe District, Guangzhou, 510655, China.
| | - Yanfen Cui
- Department of Radiology, Shanxi Cancer Hospital, Shanxi Medical University, No.3, Xinjie West Alley, Taiyuan, 030013, China.
| | - Ke Zhao
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, 106 Zhongshan Er Road, Guangzhou, 510080, China.
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| |
Collapse
|
9
|
Bai JW, Qiu SQ, Zhang GJ. Molecular and functional imaging in cancer-targeted therapy: current applications and future directions. Signal Transduct Target Ther 2023; 8:89. [PMID: 36849435 PMCID: PMC9971190 DOI: 10.1038/s41392-023-01366-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 01/19/2023] [Accepted: 02/14/2023] [Indexed: 03/01/2023] Open
Abstract
Targeted anticancer drugs block cancer cell growth by interfering with specific signaling pathways vital to carcinogenesis and tumor growth rather than harming all rapidly dividing cells as in cytotoxic chemotherapy. The Response Evaluation Criteria in Solid Tumor (RECIST) system has been used to assess tumor response to therapy via changes in the size of target lesions as measured by calipers, conventional anatomically based imaging modalities such as computed tomography (CT), and magnetic resonance imaging (MRI), and other imaging methods. However, RECIST is sometimes inaccurate in assessing the efficacy of targeted therapy drugs because of the poor correlation between tumor size and treatment-induced tumor necrosis or shrinkage. This approach might also result in delayed identification of response when the therapy does confer a reduction in tumor size. Innovative molecular imaging techniques have rapidly gained importance in the dawning era of targeted therapy as they can visualize, characterize, and quantify biological processes at the cellular, subcellular, or even molecular level rather than at the anatomical level. This review summarizes different targeted cell signaling pathways, various molecular imaging techniques, and developed probes. Moreover, the application of molecular imaging for evaluating treatment response and related clinical outcome is also systematically outlined. In the future, more attention should be paid to promoting the clinical translation of molecular imaging in evaluating the sensitivity to targeted therapy with biocompatible probes. In particular, multimodal imaging technologies incorporating advanced artificial intelligence should be developed to comprehensively and accurately assess cancer-targeted therapy, in addition to RECIST-based methods.
Collapse
Affiliation(s)
- Jing-Wen Bai
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Department of Medical Oncology, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China
| | - Si-Qi Qiu
- Diagnosis and Treatment Center of Breast Diseases, Clinical Research Center, Shantou Central Hospital, 515041, Shantou, China
- Guangdong Provincial Key Laboratory for Breast Cancer Diagnosis and Treatment, Shantou University Medical College, 515041, Shantou, China
| | - Guo-Jun Zhang
- Fujian Key Laboratory of Precision Diagnosis and Treatment in Breast Cancer, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Department of Breast-Thyroid-Surgery and Cancer Center, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
- Cancer Research Center of Xiamen University, School of Medicine, Xiamen University, 361100, Xiamen, China.
| |
Collapse
|
10
|
Liu B, Hu Z, Ran J, Xie N, Tian C, Tang Y, Ouyang Q. The circulating tumor DNA (ctDNA) alteration level predicts therapeutic response in metastatic breast cancer: Novel prognostic indexes based on ctDNA. Breast 2022; 65:116-123. [PMID: 35926241 PMCID: PMC9356206 DOI: 10.1016/j.breast.2022.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/13/2022] [Accepted: 07/14/2022] [Indexed: 11/18/2022] Open
Abstract
Purpose Circulating tumor DNA (ctDNA) has good clinical guiding value for metastatic breast cancer (MBC) patients. This study aimed to apply a novel genetic analysis approach for therapeutic prediction based on ctDNA alterations. Method This nonrandomized, multicenter study recruited 223 MBC patients (NCT05079074). Plasma samples were collected for target-capture deep sequencing of ctDNA at baseline, after the 2nd cycle of treatment, and when progressive disease (PD) was evaluated. Samples were categorized into four levels according to the number of ctDNA alterations: level 1 (no alterations), level 2 (1–2 alterations), level 3 (3–4 alterations) and level 4 (≥5 alterations). According to ctDNA alteration level and variant allele frequency (VAF), a novel ctDNA-level Response Evaluation Criterion in Solid Tumors (ctle-RECIST) was established to assess treatment response and predict progression-free survival (PFS). Results The median PFS in level 1 (6.63 months) patients was significantly longer than that in level 2–4 patients (level 2: 5.70 months; level 3–4: 4.90 months, p < 0.05). After 2 cycles of treatment, based on ctle-RECIST, the median PFS of level-based disease control rate (lev-DCR) patients was significantly longer than that of level-based PD (lev-PD) patients [HR 2.42 (1.52–3.85), p < 0.001]. In addition, we found that ctDNA level assessment could be a good supplement to radiologic assessment. The median PFS in the dual-DCR group tended to be longer than that in the single-DCR group [HR 1.41 (0.93–2.13), p = 0.107]. Conclusion The ctDNA alteration level and ctle-RECIST could be novel biomarkers of prognosis and could complement radiologic assessment in MBC. Based on the number of ctDNA alterations, samples were categorized into four levels: level 1 to level 4. ctDNA alterations differed in different alteration level groups. Higher ctDNA alteration levels (levels 3–4) were associated with a higher probability of liver metastasis. According to ctDNA alteration level and variant allele frequency, a novel ctDNA-level RECIST (ctle-RECIST) was established to assess treatment response. ctle-RECIST can not only independently predict PFS, but also assist radiologic assessment and improve the clinical application value of prediction.
Collapse
Affiliation(s)
- Binliang Liu
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Zheyu Hu
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Jialu Ran
- Department of Biostatistics and Bioinformatics, Rollins School of Public Heath, Emory University, Atlanta, GA 30322, USA
| | - Ning Xie
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Can Tian
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China
| | - Yu Tang
- Department of Gastroenterology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, China
| | - Quchang Ouyang
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital/the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
11
|
Callesen LB, Hamfjord J, Boysen AK, Pallisgaard N, Guren TK, Kure EH, Spindler KLG. Circulating tumour DNA and its clinical utility in predicting treatment response or survival in patients with metastatic colorectal cancer: a systematic review and meta-analysis. Br J Cancer 2022; 127:500-513. [PMID: 35440666 PMCID: PMC9345951 DOI: 10.1038/s41416-022-01816-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/29/2022] [Accepted: 03/31/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We investigate the current knowledge on circulating tumour DNA (ctDNA) and its clinical utility in predicting outcomes in patients with metastatic colorectal cancer (mCRC). METHODS PubMed, Embase, Cochrane Database of Systematic Reviews and Cochrane Central Register of Controlled Trials were searched. Last search 16/12/2020. We included studies on patients with mCRC reporting the predictive or prognostic value of ctDNA. We performed separate random-effects meta-analyses to investigate if baseline ctDNA and early changes in ctDNA levels during treatment were associated with survival. The risk of bias was assessed according to the Quality in Prognosis Studies tool. RESULTS Seventy-one studies were included with 6930 patients. Twenty-four studies were included in meta-analyses. High baseline ctDNA level was associated with short progression-free survival (PFS) (HR = 2.2; 95% CI 1.8-2.8; n = 509) and overall survival (OS) (HR = 2.4; 95% CI 1.9-3.1; n = 1336). A small or no early decrease in ctDNA levels during treatment was associated with short PFS (HR = 3.0; 95% CI 2.2-4.2; n = 479) and OS (HR = 2.8; 95% CI 2.1-3.9; n = 583). Results on clonal evolution and lead-time were inconsistent. A majority of included studies (n = 50/71) had high risk of bias in at least one domain. CONCLUSIONS Plasma ctDNA is a strong prognostic biomarker in mCRC. However, true clinical utility is lacking.
Collapse
Affiliation(s)
- Louise B Callesen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark.
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Julian Hamfjord
- Department of Oncology, Oslo University Hospital, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anders K Boysen
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Niels Pallisgaard
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Tormod K Guren
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Elin H Kure
- Department of Cancer Genetics, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Department of Natural Sciences and Environmental Health, Faculty of Technology, Natural Sciences and Maritime Sciences, University of South-Eastern Norway, Campus Bø, Bø, Norway
| | - Karen-Lise G Spindler
- Department of Experimental Clinical Oncology, Aarhus University Hospital, Aarhus, Denmark
- Institute of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
12
|
Huang M, Lin Y, Wang C, Deng L, Chen M, Assaraf YG, Chen ZS, Ye W, Zhang D. New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects. Drug Resist Updat 2022; 64:100849. [PMID: 35842983 DOI: 10.1016/j.drup.2022.100849] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiogenesis is a hallmark of cancer and is required for tumor growth and progression. Antiangiogenic therapy has been revolutionarily developing and was approved for the treatment of various types of cancer for nearly two decades, among which bevacizumab and sorafenib continue to be the two most frequently used antiangiogenic drugs. Although antiangiogenic therapy has brought substantial survival benefits to many cancer patients, resistance to antiangiogenic drugs frequently occurs during clinical treatment, leading to poor outcomes and treatment failure. Cumulative evidence has demonstrated that the intricate interplay among tumor cells, bone marrow-derived cells, and local stromal cells critically allows for tumor escape from antiangiogenic therapy. Currently, drug resistance has become the main challenge that hinders the therapeutic efficacies of antiangiogenic therapy. In this review, we describe and summarize the cellular and molecular mechanisms conferring tumor drug resistance to antiangiogenic therapy, which was predominantly associated with redundancy in angiogenic signaling molecules (e.g., VEGFs, GM-CSF, G-CSF, and IL17), alterations in biological processes of tumor cells (e.g., tumor invasiveness and metastasis, stemness, autophagy, metabolic reprogramming, vessel co-option, and vasculogenic mimicry), increased recruitment of bone marrow-derived cells (e.g., myeloid-derived suppressive cells, tumor-associated macrophages, and tumor-associated neutrophils), and changes in the biological functions and features of local stromal cells (e.g., pericytes, cancer-associated fibroblasts, and endothelial cells). We also review potential biomarkers to predict the response to antiangiogenic therapy in cancer patients, which mainly consist of imaging biomarkers, cellular and extracellular proteins, a certain type of bone marrow-derived cells, local stromal cell content (e.g., pericyte coverage) as well as serum or plasma biomarkers (e.g., non-coding RNAs). Finally, we highlight the recent advances in combination strategies with the aim of enhancing the response to antiangiogenic therapy in cancer patients and mouse models. This review introduces a comprehensive understanding of the mechanisms and biomarkers associated with the evasion of antiangiogenic therapy in cancer, providing an outlook for developing more effective approaches to promote the therapeutic efficacy of antiangiogenic therapy.
Collapse
Affiliation(s)
- Maohua Huang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yuning Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Chenran Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Minfeng Chen
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John's University, NY 11439, USA.
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
13
|
Rata M, Khan K, Collins DJ, Koh DM, Tunariu N, Bali MA, d'Arcy J, Winfield JM, Picchia S, Valeri N, Chau I, Cunningham D, Fassan M, Leach MO, Orton MR. DCE-MRI is more sensitive than IVIM-DWI for assessing anti-angiogenic treatment-induced changes in colorectal liver metastases. Cancer Imaging 2021; 21:67. [PMID: 34924031 PMCID: PMC8684660 DOI: 10.1186/s40644-021-00436-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Diffusion weighted imaging (DWI) with intravoxel incoherent motion (IVIM) modelling can inform on tissue perfusion without exogenous contrast administration. Dynamic-contrast-enhanced (DCE) MRI can also characterise tissue perfusion, but requires a bolus injection of a Gadolinium-based contrast agent. This study compares the use of DCE-MRI and IVIM-DWI methods in assessing response to anti-angiogenic treatment in patients with colorectal liver metastases in a cohort with confirmed treatment response. METHODS This prospective imaging study enrolled 25 participants with colorectal liver metastases to receive Regorafenib treatment. A target metastasis > 2 cm in each patient was imaged before and at 15 days after treatment on a 1.5T MR scanner using slice-matched IVIM-DWI and DCE-MRI protocols. MRI data were motion-corrected and tumour volumes of interest drawn on b=900 s/mm2 diffusion-weighted images were transferred to DCE-MRI data for further analysis. The median value of four IVIM-DWI parameters [diffusion coefficient D (10-3 mm2/s), perfusion fraction f (ml/ml), pseudodiffusion coefficient D* (10-3 mm2/s), and their product fD* (mm2/s)] and three DCE-MRI parameters [volume transfer constant Ktrans (min-1), enhancement fraction EF (%), and their product KEF (min-1)] were recorded at each visit, before and after treatment. Changes in pre- and post-treatment measurements of all MR parameters were assessed using Wilcoxon signed-rank tests (P<0.05 was considered significant). DCE-MRI and IVIM-DWI parameter correlations were evaluated with Spearman rank tests. Functional MR parameters were also compared against Response Evaluation Criteria In Solid Tumours v.1.1 (RECIST) evaluations. RESULTS Significant treatment-induced reductions of DCE-MRI parameters across the cohort were observed for EF (91.2 to 50.8%, P<0.001), KEF (0.095 to 0.045 min-1, P<0.001) and Ktrans (0.109 to 0.078 min-1, P=0.002). For IVIM-DWI, only D (a non-perfusion parameter) increased significantly post treatment (0.83 to 0.97 × 10-3 mm2/s, P<0.001), while perfusion-related parameters showed no change. No strong correlations were found between DCE-MRI and IVIM-DWI parameters. A moderate correlation was found, after treatment, between Ktrans and D* (r=0.60; P=0.002) and fD* (r=0.67; P<0.001). When compared to RECIST v.1.1 evaluations, KEF and D correctly identified most clinical responders, whilst non-responders were incorrectly identified. CONCLUSION IVIM-DWI perfusion-related parameters showed limited sensitivity to the anti-angiogenic effects of Regorafenib treatment in colorectal liver metastases and showed low correlation with DCE-MRI parameters, despite profound and significant post-treatment reductions in DCE-MRI measurements. TRIAL REGISTRATION NCT03010722 clinicaltrials.gov; registration date 6th January 2015.
Collapse
Affiliation(s)
- Mihaela Rata
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom.
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom.
- Royal Marsden NHS Foundation Trust & Institute of Cancer Research, Downs Road, SM2 5PT, Sutton, London, UK.
| | - Khurum Khan
- Department of Medicine, GI and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, United Kingdom
| | - David J Collins
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Dow-Mu Koh
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Nina Tunariu
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Maria Antonietta Bali
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - James d'Arcy
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
- Cancer Research UK National Cancer Imaging Translational Accelerator (NCITA), London, United Kingdom
| | - Jessica M Winfield
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Simona Picchia
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
| | - Nicola Valeri
- Department of Medicine, GI and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, United Kingdom
- Centre for Evolution and Cancer, The Institute of Cancer Research, London and Sutton, United Kingdom
- Division of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ian Chau
- Department of Medicine, GI and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, United Kingdom
| | - David Cunningham
- Department of Medicine, GI and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, United Kingdom
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Martin O Leach
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| | - Matthew R Orton
- Department of Radiology, MRI Unit, The Royal Marsden NHS Foundation Trust, London, United Kingdom
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
14
|
Jiang Y, Xu S, Fan H, Qian J, Luo W, Zhen S, Tao Y, Sun J, Lin H. ALA-Net: Adaptive Lesion-Aware Attention Network for 3D Colorectal Tumor Segmentation. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:3627-3640. [PMID: 34197319 DOI: 10.1109/tmi.2021.3093982] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Accurate and reliable segmentation of colorectal tumors and surrounding colorectal tissues on 3D magnetic resonance images has critical importance in preoperative prediction, staging, and radiotherapy. Previous works simply combine multilevel features without aggregating representative semantic information and without compensating for the loss of spatial information caused by down-sampling. Therefore, they are vulnerable to noise from complex backgrounds and suffer from misclassification and target incompleteness-related failures. In this paper, we address these limitations with a novel adaptive lesion-aware attention network (ALA-Net) which explicitly integrates useful contextual information with spatial details and captures richer feature dependencies based on 3D attention mechanisms. The model comprises two parallel encoding paths. One of these is designed to explore global contextual features and enlarge the receptive field using a recurrent strategy. The other captures sharper object boundaries and the details of small objects that are lost in repeated down-sampling layers. Our lesion-aware attention module adaptively captures long-range semantic dependencies and highlights the most discriminative features, improving semantic consistency and completeness. Furthermore, we introduce a prediction aggregation module to combine multiscale feature maps and to further filter out irrelevant information for precise voxel-wise prediction. Experimental results show that ALA-Net outperforms state-of-the-art methods and inherently generalizes well to other 3D medical images segmentation tasks, providing multiple benefits in terms of target completeness, reduction of false positives, and accurate detection of ambiguous lesion regions.
Collapse
|
15
|
Olmedillas-López S, Olivera-Salazar R, García-Arranz M, García-Olmo D. Current and Emerging Applications of Droplet Digital PCR in Oncology: An Updated Review. Mol Diagn Ther 2021; 26:61-87. [PMID: 34773243 DOI: 10.1007/s40291-021-00562-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2021] [Indexed: 12/14/2022]
Abstract
In the era of personalized medicine and targeted therapies for the management of patients with cancer, ultrasensitive detection methods for tumor genotyping, such as next-generation sequencing or droplet digital polymerase chain reaction (ddPCR), play a significant role. In the search for less invasive strategies for diagnosis, prognosis and disease monitoring, the number of publications regarding liquid biopsy approaches using ddPCR has increased substantially in recent years. There is a long list of malignancies in which ddPCR provides a reliable and accurate tool for detection of nucleic acid-based markers derived from cell-free DNA, cell-free RNA, circulating tumor cells, extracellular vesicles or exosomes when isolated from whole blood, plasma and serum, helping to anticipate tumor relapse or unveil intratumor heterogeneity and clonal evolution in response to treatment. This updated review describes recent developments in ddPCR platforms and provides a general overview about the major applications of liquid biopsy in blood, including its utility for molecular response and minimal residual disease monitoring in hematological malignancies or the therapeutic management of patients with colorectal or lung cancer, particularly for the selection and monitoring of treatment with tyrosine kinase inhibitors. Although plasma is the main source of genetic material for tumor genomic profiling, liquid biopsy by ddPCR is being investigated in a wide variety of biologic fluids, such as cerebrospinal fluid, urine, stool, ocular fluids, sputum, saliva, bronchoalveolar lavage, pleural effusion, mucin, peritoneal fluid, fine needle aspirate, bile or pancreatic juice. The present review focuses on these "alternative" sources of genetic material and their analysis by ddPCR in different kinds of cancers.
Collapse
Affiliation(s)
- Susana Olmedillas-López
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain.
| | - Rocío Olivera-Salazar
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain
| | - Mariano García-Arranz
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain.,Department of Surgery, School of Medicine, Universidad Autónoma de Madrid (UAM), 28029, Madrid, Spain
| | - Damián García-Olmo
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Avda. Reyes Católicos, 2, 28040, Madrid, Spain.,Department of Surgery, School of Medicine, Universidad Autónoma de Madrid (UAM), 28029, Madrid, Spain.,Department of Surgery, Fundación Jiménez Díaz University Hospital (FJD), 28040, Madrid, Spain
| |
Collapse
|
16
|
Bhat SM, Badiger VA, Vasishta S, Chakraborty J, Prasad S, Ghosh S, Joshi MB. 3D tumor angiogenesis models: recent advances and challenges. J Cancer Res Clin Oncol 2021; 147:3477-3494. [PMID: 34613483 PMCID: PMC8557138 DOI: 10.1007/s00432-021-03814-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/21/2021] [Indexed: 01/02/2023]
Abstract
The development of blood vessels, referred to as angiogenesis, is an intricate process regulated spatially and temporally through a delicate balance between the qualitative and quantitative expression of pro and anti-angiogenic molecules. As angiogenesis is a prerequisite for solid tumors to grow and metastasize, a variety of tumor angiogenesis models have been formulated to better understand the underlying mechanisms and associated clinical applications. Studies have demonstrated independent mechanisms inducing angiogenesis in tumors such as (a) HIF-1/VEGF mediated paracrine interactions between a cancer cell and endothelial cells, (b) recruitment of progenitor endothelial cells, and (c) vasculogenic mimicry. Moreover, single-cell sequencing technologies have indicated endothelial cell heterogeneity among organ systems including tumor tissues. However, existing angiogenesis models often rely upon normal endothelial cells which significantly differ from tumor endothelial cells exhibiting distinct (epi)genetic and metabolic signatures. Besides, the existence of intra-individual variations necessitates the development of improved tumor vascular model systems for personalized medicine. In the present review, we summarize recent advancements of 3D tumor vascular model systems which include (a) tissue engineering-based tumor models; (b) vascular organoid models, and (c) organ-on-chips and their importance in replicating the tumor angiogenesis along with the associated challenges to design improved models.
Collapse
Affiliation(s)
- Sharath M Bhat
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Vaishnavi A Badiger
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Sampara Vasishta
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Juhi Chakraborty
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Seetharam Prasad
- Department of Surgery, Kasturba Medical College, Manipal Academy of Higher Education, Manipal, 576104, India
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile and Fibre Engineering, Indian Institute of Technology, Delhi, 110016, India
| | - Manjunath B Joshi
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
| |
Collapse
|
17
|
Personalized Therapy and Liquid Biopsy-A Focus on Colorectal Cancer. J Pers Med 2021; 11:jpm11070630. [PMID: 34357097 PMCID: PMC8305103 DOI: 10.3390/jpm11070630] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/21/2021] [Accepted: 06/29/2021] [Indexed: 12/24/2022] Open
Abstract
(1) Background: Resistance mechanisms represent a barrier to anti-cancer therapies. Liquid biopsies would allow obtaining additional information in order to develop targeted therapies to thwart the resistance phenomena but also to follow in time real response to treatment and be able to adapt it the most quickly possible way in case of resistance. (2) Methods: herein we summarize the different liquid biopsies which are currently under research; we then review the literature and focalize on one of their potential roles: the theranostic one and especially in the cases of colorectal cancers. (3) Results: few studies targeting liquid biopsy as a potential tool to adapt cancer treatments are present in the literature and encompass few patients. (4) Conclusions: further research is needed to prove the efficiency of LB. Indeed, it seems a promising tool to guide treatment by targeting actionable mutations with detection of resistant mutations.
Collapse
|
18
|
Pastor B, André T, Henriques J, Trouilloud I, Tournigand C, Jary M, Mazard T, Louvet C, Azan S, Bauer A, Roch B, Sanchez C, Vernerey D, Thierry AR, Adenis A. Monitoring levels of circulating cell-free DNA in patients with metastatic colorectal cancer as a potential biomarker of responses to regorafenib treatment. Mol Oncol 2021; 15:2401-2411. [PMID: 33934494 PMCID: PMC8410523 DOI: 10.1002/1878-0261.12972] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/09/2021] [Accepted: 04/20/2021] [Indexed: 12/25/2022] Open
Abstract
Circulating cell‐free DNA (cfDNA) contains circulating tumor DNA (ctDNA), which can be obtained from serial liquid biopsies to enable tumor genome analysis throughout the course of treatment. We investigated cfDNA and mutant ctDNA as potential biomarkers to predict the best outcomes of regorafenib‐treated metastatic colorectal cancer (mCRC) patients. We analyzed longitudinally collected plasma cfDNA of 43 mCRC patients prospectively enrolled in the phase II TEXCAN trial by IntPlex qPCR. Qualitative (KRAS, NRAS, BRAFV600E mutations) and quantitative (total cfDNA concentration, mutant ctDNA concentration, mutant ctDNA fraction) parameters were correlated with overall survival (OS) and progression‐free survival (PFS). When examined as classes or continuous variables, the concentrations of total cfDNA, mutant ctDNA, and, partly, mutant ctDNA fraction prior to regorafenib treatment correlated with OS. Patients with baseline cfDNA > 26 ng·mL−1 had shorter OS than those with cfDNA value below this threshold (4.0 vs 6.9 months; log‐rank P = 0.0366). Patients with baseline mutant ctDNA > 2 ng·mL−1 had shorter OS than those with mutant ctDNA below this threshold (log‐rank P = 0.0154). We show that pretreatment cfDNA and mutant ctDNA levels may identify mCRC patients that may benefit from regorafenib treatment.
Collapse
Affiliation(s)
- Brice Pastor
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Montpellier Cancer Institute (ICM), France
| | - Thierry André
- Department of Medical Oncology, Saint-Antoine University Hospital, Sorbonne University, Paris, France.,Oncology Multidisciplinary Research Group (GERCOR), Paris, France
| | - Julie Henriques
- Methodology and Quality of Life Unit in Oncology, Besançon University Hospital, France
| | - Isabelle Trouilloud
- Department of Medical Oncology, Saint-Antoine University Hospital, Sorbonne University, Paris, France.,Oncology Multidisciplinary Research Group (GERCOR), Paris, France
| | - Christophe Tournigand
- Oncology Multidisciplinary Research Group (GERCOR), Paris, France.,Medical Oncology Service, Henri Mondor Hospital, AP-HP, Université Paris Est Créteil Créteil, France
| | - Marine Jary
- Oncology Multidisciplinary Research Group (GERCOR), Paris, France.,INSERM, Etablissement Français du Sang Bourgogne Franche-Comté, UMR1098, Interactions Hôte-Greffon-Tumeur/Ingénierie Cellulaire et Génique, Bourgogne Franche-Comté University, Besançon, France.,Department of Medical Oncology, Besançon University Hospital, France
| | - Thibault Mazard
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Montpellier Cancer Institute (ICM), France.,Department of Medical Oncology, Montpellier Cancer Institute (ICM), France
| | - Christophe Louvet
- Oncology Multidisciplinary Research Group (GERCOR), Paris, France.,Department of Medical Oncology, Institut Monsouris, Paris, France
| | - Simon Azan
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Montpellier Cancer Institute (ICM), France
| | - Audrey Bauer
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Montpellier Cancer Institute (ICM), France
| | - Benoit Roch
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Montpellier Cancer Institute (ICM), France.,Department of Thoracic Oncology, Montpellier University Hospital, Université de Montpellier, France
| | - Cynthia Sanchez
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Montpellier Cancer Institute (ICM), France
| | - Dewi Vernerey
- Oncology Multidisciplinary Research Group (GERCOR), Paris, France.,Methodology and Quality of Life Unit in Oncology, Besançon University Hospital, France
| | - Alain R Thierry
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Montpellier Cancer Institute (ICM), France.,Department of Medical Oncology, Montpellier Cancer Institute (ICM), France
| | - Antoine Adenis
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Montpellier Cancer Institute (ICM), France.,Department of Medical Oncology, Montpellier Cancer Institute (ICM), France
| |
Collapse
|
19
|
Wang C, Li H, Wu L, Jiao X, Jin Z, Zhu Y, Fang Z, Zhang X, Huang H, Zhao L. Coiled-Coil Domain-Containing 68 Downregulation Promotes Colorectal Cancer Cell Growth by Inhibiting ITCH-Mediated CDK4 Degradation. Front Oncol 2021; 11:668743. [PMID: 33968776 PMCID: PMC8100586 DOI: 10.3389/fonc.2021.668743] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/30/2021] [Indexed: 12/31/2022] Open
Abstract
Coiled-coil domain-containing 68 (CCDC68) plays different roles in cancer and is predicted as a tumor suppressor in human colorectal cancer (CRC). However, the specific role of CCDC68 in CRC and the underlying mechanisms remain unknown. Here, we showed that CCDC68 expression was lower in CRC than that in corresponding normal tissues, and CCDC68 level was positively correlated with disease-free survival. Ectopic expression of CCDC68 decreased CRC cell proliferation in vitro and suppressed the growth of CRC xenograft tumors in vivo. CCDC68 caused G0/G1 cell cycle arrest, downregulated CDK4, and upregulated ITCH, the E3 ubiquitin ligase responsible for CDK4 protein degradation. This increased CDK4 degradation, which decreased CDK4 protein levels and inhibited CRC tumor growth. Collectively, the present results identify a novel CDK4 regulatory axis consisting of CCDC68 and ITCH, which suggest that CCDC68 is a promising target for the treatment of CRC.
Collapse
Affiliation(s)
- Cong Wang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongyan Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lei Wu
- Department of General Surgery, Heze Municipal Hospital, Heze, China
| | - Xueli Jiao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zihui Jin
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yujie Zhu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ziling Fang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaodong Zhang
- Department of Colorectal anal surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haishan Huang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lingling Zhao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
20
|
Ruiz-Bañobre J, Goel A. Genomic and epigenomic biomarkers in colorectal cancer: From diagnosis to therapy. Adv Cancer Res 2021; 151:231-304. [PMID: 34148615 PMCID: PMC10338180 DOI: 10.1016/bs.acr.2021.02.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the second leading cause of cancer-related deaths in the United States. Despite ongoing efforts aimed at increasing screening for CRC and early detection, and development of more effective therapeutic regimens, the overall morbidity and mortality from this malignancy remains a clinical challenge. Therefore, identifying and developing genomic and epigenomic biomarkers that can improve CRC diagnosis and help predict response to current therapies are of paramount importance for improving survival outcomes in CRC patients, sparing patients from toxicity associated with current regimens, and reducing the economic burden associated with these treatments. Although efforts to develop biomarkers over the past decades have achieved some success, the recent availability of high-throughput analytical tools, together with the use of machine learning algorithms, will likely hasten the development of more robust diagnostic biomarkers and improved guidance for clinical decision-making in the coming years. In this chapter, we provide a systematic and comprehensive overview on the current status of genomic and epigenomic biomarkers in CRC, and comment on their potential clinical significance in the management of patients with this fatal malignancy, including in the context of precision medicine.
Collapse
Affiliation(s)
- Juan Ruiz-Bañobre
- Medical Oncology Department, University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), CIBERONC, Santiago de Compostela, Spain; Translational Medical Oncology Group (Oncomet), Health Research Institute of Santiago (IDIS), University Clinical Hospital of Santiago de Compostela, University of Santiago de Compostela (USC), CIBERONC, Santiago de Compostela, Spain
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope Comprehensive Cancer Center, Duarte, CA, United States.
| |
Collapse
|
21
|
Parisi A, Porzio G, Pulcini F, Cannita K, Ficorella C, Mattei V, Delle Monache S. What Is Known about Theragnostic Strategies in Colorectal Cancer. Biomedicines 2021; 9:biomedicines9020140. [PMID: 33535557 PMCID: PMC7912746 DOI: 10.3390/biomedicines9020140] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 01/22/2021] [Accepted: 01/25/2021] [Indexed: 02/08/2023] Open
Abstract
Despite the paradigmatic shift occurred in recent years for defined molecular subtypes in the metastatic setting treatment, colorectal cancer (CRC) still remains an incurable disease in most of the cases. Therefore, there is an urgent need for new tools and biomarkers for both early tumor diagnosis and to improve personalized treatment. Thus, liquid biopsy has emerged as a minimally invasive tool that is capable of detecting genomic alterations from primary or metastatic tumors, allowing the prognostic stratification of patients, the detection of the minimal residual disease after surgical or systemic treatments, the monitoring of therapeutic response, and the development of resistance, establishing an opportunity for early intervention before imaging detection or worsening of clinical symptoms. On the other hand, preclinical and clinical evidence demonstrated the role of gut microbiota dysbiosis in promoting inflammatory responses and cancer initiation. Altered gut microbiota is associated with resistance to chemo drugs and immune checkpoint inhibitors, whereas the use of microbe-targeted therapies including antibiotics, pre-probiotics, and fecal microbiota transplantation can restore response to anticancer drugs, promote immune response, and therefore support current treatment strategies in CRC. In this review, we aim to summarize preclinical and clinical evidence for the utilization of liquid biopsy and gut microbiota in CRC.
Collapse
Affiliation(s)
- Alessandro Parisi
- Department of Life, Health and Environmental Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy; (G.P.); (K.C.); (C.F.)
| | - Giampiero Porzio
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy; (G.P.); (K.C.); (C.F.)
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Fanny Pulcini
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Katia Cannita
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy; (G.P.); (K.C.); (C.F.)
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Corrado Ficorella
- Medical Oncology Unit, St. Salvatore Hospital, 67100 L’Aquila, Italy; (G.P.); (K.C.); (C.F.)
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, via Angelo Maria Ricci 35A, 02100 Rieti, Italy;
| | - Simona Delle Monache
- Department of Biotechnology and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
- Correspondence: ; Tel.: +39-086-243-3569
| |
Collapse
|
22
|
Abstract
Response evaluation for cancer treatment consists primarily of clinical and radiological assessments. In addition, a limited number of serum biomarkers that assess treatment response are available for a small subset of malignancies. Through recent technological innovations, new methods for measuring tumor burden and treatment response are becoming available. By utilization of highly sensitive techniques, tumor-specific mutations in circulating DNA can be detected and circulating tumor DNA (ctDNA) can be quantified. These so-called liquid biopsies provide both molecular information about the genomic composition of the tumor and opportunities to evaluate tumor response during therapy. Quantification of tumor-specific mutations in plasma correlates well with tumor burden. Moreover, with liquid biopsies, it is also possible to detect mutations causing secondary resistance during treatment. This review focuses on the clinical utility of ctDNA as a response and follow-up marker in patients with non-small cell lung cancer, melanoma, colorectal cancer, and breast cancer. Relevant studies were retrieved from a literature search using PubMed database. An overview of the available literature is provided and the relevance of ctDNA as a response marker in anti-cancer therapy for clinical practice is discussed. We conclude that the use of plasma-derived ctDNA is a promising tool for treatment decision-making based on predictive testing, detection of resistance mechanisms, and monitoring tumor response. Necessary steps for translation to daily practice and future perspectives are discussed.
Collapse
|
23
|
Jones RP, Pugh SA, Graham J, Primrose JN, Barriuso J. Circulating tumour DNA as a biomarker in resectable and irresectable stage IV colorectal cancer; a systematic review and meta-analysis. Eur J Cancer 2021; 144:368-381. [PMID: 33422803 DOI: 10.1016/j.ejca.2020.11.025] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 11/01/2020] [Accepted: 11/15/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND For patients with metastatic colorectal cancer, stratification for treatment (surgery or chemotherapy) is often based on crude clinicopathological characteristics like tumour size and number of lesions. Circulating tumour DNA (ctDNA) acts as a potential biomarker of disease trajectory and biology, allowing better stratification. This study aims to systematically review ctDNA in stage IV colorectal cancer to assess its potential role as a prospective biomarker to guide management decisions. METHODS A literature search was performed to identify studies where the measurement of ctDNA in stage IV colorectal cancer was correlated with a clinical outcome (radiological response, secondary resection rate, PFS, DFS or OS). RESULTS Twenty-eight studies were included, reporting on 2823 patients. Circulating tumour DNA was detectable in between 80% and 90% of patients prior to treatment. Meta-analysis identified a strong correlation between detectable ctDNA after treatment (surgery or chemotherapy) and overall survival (HR 2.2, 95% CI 1.79-2.69, p < 0.00001), as well as progression-free survival (HR 3.15, 95% CI 2.10-4.73, p < 0.00001). ctDNA consistently offered an early marker of long-term prognosis in irresectable disease, with changes after one cycle of systemic therapy demonstrating prognostic value. In resectable disease treated with curative intent, detection of ctDNA offered a lead time over radiological recurrence of 10 months. CONCLUSION Circulating tumour DNA is detectable in the majority of resectable and irresectable patients. The presence of ctDNA is clearly associated with shorter overall survival, with changes in ctDNA an early biomarker of adverse disease behaviour. Prospective trials are essential to test its clinical efficacy.
Collapse
Affiliation(s)
- Robert P Jones
- School of Cancer Studies, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Hepatobiliary Surgery, Liverpool University Teaching Hospitals NHS Foundation Trust, Liverpool, UK.
| | | | - Janet Graham
- Beatson West of Scotland Cancer Centre, Glasgow, UK; University of Glasgow, UK
| | | | - Jorge Barriuso
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, UK; Department of Medical Oncology, The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|
24
|
Khan K, Gonzalez-Exposito R, Cunningham D, Koh DM, Woolston A, Barber L, Griffiths B, Kouvelakis K, Calamai V, Bali M, Khan N, Bryant A, Saffery C, Dearman C, Begum R, Rao S, Starling N, Watkins D, Chau I, Braconi C, Valeri N, Gerlinger M, Fotiadis N. Diagnostic Accuracy and Safety of Coaxial System in Oncology Patients Treated in a Specialist Cancer Center With Prospective Validation Within Clinical Trial Data. Front Oncol 2020; 10:1634. [PMID: 33014822 PMCID: PMC7500492 DOI: 10.3389/fonc.2020.01634] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/27/2020] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND Image-guided tissue biopsies are critically important in the diagnosis and management of cancer patients. High-yield samples are also vital for biomarker and resistance mechanism discovery through molecular/genomic analyses. PATIENTS AND METHODS All consecutive patients who underwent plugged image-guided biopsy at Royal Marsden from June 2013 until September 2016 were included in the analysis. In the next step, a second cohort of patients prospectively treated within two clinical trials (PROSPECT-C and PROSPECT-R) were assessed for the DNA yield from biopsies assessed for complex genomic analysis. RESULTS A total of 522 plugged core biopsies were performed in 457 patients [men, 52%; median age, 63 years (range, 17-93)]. Histological diagnosis was achieved in 501 of 522 (96%) performed biopsies. Age, gender, modality, metastatic site, and seniority of the interventionist were not found to be significant factors associated with odds of failure on a logistic regression. Seventeen (3.3%) were admitted due to biopsy-related complications; nine, three, two, one, one, and one were admitted for grade I/II pain control, sepsis, vasovagal syncope, thrombosis, hematuria, and deranged liver functions, respectively; two patients with right upper quadrant pain after liver biopsy were found to have radiologically confirmed subcapsular hematoma requiring conservative treatment. One patient (0.2%) developed grade III hemorrhage following biopsy of a gastric gastrointestinal stromal tumor (GIST). Overall molecular analysis was successful in 89% (197/222 biopsies). Prospective validation in 62 biopsies gave success rates of 92.06 and 79.03% for DNA extraction of >1 μm and tmour content of >20%, respectively. CONCLUSION The probability of diagnostic success for complex molecular analysis is increased with plugged large coaxial needle biopsy technique, which also minimizes complications and reduces hospital stay. High-yield DNA acquisition allows genomic molecular characterization for personalized medicine.
Collapse
Affiliation(s)
- Khurum Khan
- Department of Gastrointestinal Oncology, UCL Cancer Institute, University College NHS Foundation Trust, London, United Kingdom
| | | | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Dow-Mu Koh
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, United Kingdom
| | - Andrew Woolston
- Translational Oncogenomics Laboratory, Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Louise Barber
- Translational Oncogenomics Laboratory, Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Beatrice Griffiths
- Translational Oncogenomics Laboratory, Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | | | - Vanessa Calamai
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Monia Bali
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, United Kingdom
| | - Nasir Khan
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, United Kingdom
| | - Annette Bryant
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Claire Saffery
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Charles Dearman
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Ruwaida Begum
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Sheela Rao
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Chiara Braconi
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
| | - Nicola Valeri
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London, United Kingdom
| | - Marco Gerlinger
- Department of Medicine, The Royal Marsden NHS Trust, London, United Kingdom
- Translational Oncogenomics Laboratory, Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Nicos Fotiadis
- Cancer Research UK Cancer Imaging Centre, Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, United Kingdom
| |
Collapse
|
25
|
Vacante M, Ciuni R, Basile F, Biondi A. The Liquid Biopsy in the Management of Colorectal Cancer: An Overview. Biomedicines 2020; 8:E308. [PMID: 32858879 PMCID: PMC7555636 DOI: 10.3390/biomedicines8090308] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 08/19/2020] [Accepted: 08/24/2020] [Indexed: 02/07/2023] Open
Abstract
Currently, there is a crucial need for novel diagnostic and prognostic biomarkers with high specificity and sensitivity in patients with colorectal cancer. A "liquid biopsy" is characterized by the isolation of cancer-derived components, such as circulating tumor cells, circulating tumor DNA, microRNAs, long non-coding RNAs, and proteins, from peripheral blood or other body fluids and their genomic or proteomic assessment. The liquid biopsy is a minimally invasive and repeatable technique that could play a significant role in screening and diagnosis, and predict relapse and metastasis, as well as monitoring minimal residual disease and chemotherapy resistance in colorectal cancer patients. However, there are still some practical issues that need to be addressed before liquid biopsy can be widely used in clinical practice. Potential challenges may include low amounts of circulating tumor cells and circulating tumor DNA in samples, lack of pre-analytical and analytical consensus, clinical validation, and regulatory endorsement. The aim of this review was to summarize the current knowledge of the role of liquid biopsy in the management of colorectal cancer.
Collapse
Affiliation(s)
- Marco Vacante
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Via S. Sofia 78, 95123 Catania, Italy; (R.C.); (F.B.); (A.B.)
| | | | | | | |
Collapse
|
26
|
Xie H, Kim RD. The Application of Circulating Tumor DNA in the Screening, Surveillance, and Treatment Monitoring of Colorectal Cancer. Ann Surg Oncol 2020; 28:1845-1858. [PMID: 32776184 DOI: 10.1245/s10434-020-09002-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
Precision medicine with genetic profiling of tumor tissue has become an essential part of routine clinical practice in colorectal cancer. However, tissue genetic profiling suffers from clonal evolution, tumor heterogeneity, and time needed to deliver critical information for prompt clinical decision making. In contrast, liquid biopsy with plasma circulating tumor DNA provides genetic and epigenetic information from both the primary and metastatic colorectal cancer, which can potentially capture tumor heterogeneity and evolution with time and treatment. In addition, liquid biopsy with circulating tumor DNA is minimally invasive, quicker, and easily repeatable with high patient compliance to provide both qualitative and quantitative molecular information in real-time. We provide an overview on the potential clinical applications of circulating tumor DNA in the screening, surveillance, and treatment monitoring of colorectal cancer.
Collapse
Affiliation(s)
- Hao Xie
- Division of Medical Oncology, Mayo Clinic, Rochester, USA
| | - Richard D Kim
- Department of Gastrointestinal Oncology, Moffitt Cancer Center, Tampa, FL, USA.
| |
Collapse
|
27
|
Liu Y, Lyu J, Bell Burdett K, Sibley AB, Hatch AJ, Starr MD, Brady JC, Hammond K, Marmorino F, Rossini D, Goldberg RM, Falcone A, Cremolini C, Owzar K, Ivanova A, Moore DT, Lee MS, Sanoff HK, Innocenti F, Nixon AB. Prognostic and Predictive Biomarkers in Patients with Metastatic Colorectal Cancer Receiving Regorafenib. Mol Cancer Ther 2020; 19:2146-2154. [PMID: 32747417 DOI: 10.1158/1535-7163.mct-20-0249] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/08/2020] [Accepted: 07/20/2020] [Indexed: 11/16/2022]
Abstract
Regorafenib is a tyrosine kinase inhibitor approved by the FDA for the treatment of patients with chemotherapy refractory metastatic colorectal cancer (mCRC). Regorafenib inhibits signaling through multiple receptors associated with angiogenesis, metastasis, and tumor immunity. Here, we report biomarker results from LCCC1029, a randomized, placebo-controlled, phase II trial of chemotherapy ± regorafenib in patients with second-line mCRC. A panel of 20 soluble protein biomarkers (termed the Angiome) was assessed in the plasma of 149 patients from the LCCC1029 trial both at baseline and along the treatment continuum. Baseline protein levels were analyzed for prognostic and predictive value for progression-free survival (PFS) and overall survival (OS). Changes in protein levels during treatment were analyzed for potential pharmacodynamic effects. Six markers (HGF, IL6, PlGF, VEGF-R1, OPN, and IL6R) were found to be prognostic for PFS. Nine markers (IL6, TIMP-1, PlGF, VCAM-1, ICAM-1, OPN, TSP-2, HGF, and VEGF-R1) were prognostic for OS. Higher baseline levels of OPN (P intx = 0.0167), VCAM-1 (P intx = 0.0216), and PDGF-AA (P intx = 0.0435) appeared to predict for PFS benefit from regorafenib compared with placebo. VCAM-1 was also potentially predictive of OS benefit from regorafenib compared with placebo (P intx = 0.0124). On-treatment changes of six markers reflected potential on-target effect of regorafenib. Consistent results were observed in an Italian cohort where 105 patients with late-stage mCRC received regorafenib monotherapy. The key findings of this study suggest that VCAM-1 may be a predictive biomarker for regorafenib benefit, while multiple protein markers may be prognostic of outcome in patients with mCRC.
Collapse
Affiliation(s)
- Yingmiao Liu
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Jing Lyu
- Duke Cancer Institute, Durham, North Carolina
| | | | | | - Ace J Hatch
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Mark D Starr
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - John C Brady
- Department of Medicine, Duke University Medical Center, Durham, North Carolina
| | - Kelli Hammond
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Federica Marmorino
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology, Azienda Ospedaliero- Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Daniele Rossini
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology, Azienda Ospedaliero- Universitaria Pisana, University of Pisa, Pisa, Italy
| | | | - Alfredo Falcone
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology, Azienda Ospedaliero- Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, Unit of Medical Oncology, Azienda Ospedaliero- Universitaria Pisana, University of Pisa, Pisa, Italy
| | - Kouros Owzar
- Duke Cancer Institute, Durham, North Carolina.,Duke Department of Biostatistics & Bioinformatics, Durham, North Carolina
| | - Anastasia Ivanova
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Dominic T Moore
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Michael S Lee
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Hanna K Sanoff
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Federico Innocenti
- The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Andrew B Nixon
- Department of Medicine, Duke University Medical Center, Durham, North Carolina.
| |
Collapse
|
28
|
Wu Y, Fan Y, Dong D, Dong X, Hu Y, Shi Y, Jing J, Li E. Efficacy and safety of regorafenib as beyond second-line therapy in patients with metastatic colorectal cancer: an adjusted indirect meta-analysis and systematic review. Ther Adv Med Oncol 2020; 12:1758835920940932. [PMID: 32728393 PMCID: PMC7364795 DOI: 10.1177/1758835920940932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 06/12/2020] [Indexed: 12/12/2022] Open
Abstract
Background: The evidence base for optimum third-line therapy for metastatic colorectal cancer (mCRC) is not conclusive. Recent studies have demonstrated the efficacy of regorafenib as third-line therapy in mCRC. This indirect meta-analysis compared the efficacy and safety of regorafenib with other available third-line therapies for mCRC. Methods: A literature search for randomized controlled trials (RCTs) was conducted in PubMed, Embase, and Cochrane Library for studies evaluating the efficacy and safety of fruquintinib, regorafenib, TAS-102, and nintedanib as third-line therapies in patients with mCRC. Overall survival (OS) and progression-free survival (PFS) were the primary outcomes, while objective response rate (ORR) and safety were the secondary outcomes. Hazard ratio (HR) and relative risk (RR) with their respective 95% confidence interval (CI) were used for analysis of survival, clinical response, and safety data. An adjusted indirect meta-analysis with placebo as the common comparator was performed. Results: We identified eight RCTs comparing regorafenib (two studies), fruquintinib (two studies), TAS-102 (three studies), and nintedanib (one study) against placebo. The OS with regorafenib was significantly better when compared with nintedanib (HR = 0.66; 95% CI: 0.45, 0.95, p = 0.02) but was similar to that of fruquintinib (HR = 1.01; 95% CI: 0.67, 1.52, p = 0.94) and TAS-102 (HR = 0.97; 95% CI: 0.68, 1.38, p = 0.88). The PFS and ORR for regorafenib were slightly better than those of TAS-102 (PFS: HR = 0.86, 95% CI: 0.54, 1.37, p = 0.5; ORR: RR = 1.13, 95% CI: 0.11, 11.05, p = 0.92) and nintedanib (PFS: HR = 0.68, 95% CI: 0.42, 1.10, p = 0.12; ORR: not reported) but were lower than those for fruquintinib (PFS: HR = 1.53, 95% CI: 0.93, 2.52, p = 0.08; ORR: RR = 0.68269, 95% CI: 0.045, 10.32, p = 0.79). Safety analysis showed that the RR of adverse events (AEs) was lesser in patients treated with regorafenib in comparison with that in patients treated with fruquintinib, but was similar to that in patients treated with nintedanib and TAS-102. Conclusion: Regorafenib has efficacy similar to that of TAS-102 and better safety when compared with fruquintinib. Considering the mechanism of action of regorafenib, which targets multiple factors in the angiogenic pathway, it could be an ideal option for treatment in the beyond second-line setting.
Collapse
Affiliation(s)
- Yinying Wu
- Department of Medical Oncology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yangwei Fan
- Department of Medical Oncology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Danfeng Dong
- Department of Medical Oncology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xuyuan Dong
- Department of Medical Oncology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yuan Hu
- Department of Medical Oncology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yu Shi
- Department of Medical Oncology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiayu Jing
- Department of Medical Oncology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Enxiao Li
- Department of Medical Oncology, Xi'an Jiaotong University, 277 Yanta West Road, Xi'an, Shaanxi 710061, China
| |
Collapse
|
29
|
Circulating Tumour DNAs and Non-Coding RNAs as Liquid Biopsies for the Management of Colorectal Cancer Patients. GASTROINTESTINAL DISORDERS 2020. [DOI: 10.3390/gidisord2030022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Circulating tumour DNAs and non-coding RNAs present in body fluids have been under investigation as tools for cancer diagnosis, disease monitoring, and prognosis for many years. These so-called liquid biopsies offer the opportunity to obtain information about the molecular make-up of a cancer in a minimal invasive way and offer the possibility to implement theranostics for precision oncology. Furthermore, liquid biopsies could overcome the limitations of tissue biopsies in capturing the complexity of tumour heterogeneity within the primary cancer and among different metastatic sites. Liquid biopsies may also be implemented to detect early tumour formation or to monitor cancer relapse of response to therapy with greater sensitivity compared with the currently available protein-based blood biomarkers. Most colorectal cancers are often diagnosed at late stages and have a high mortality rate. Hence, biomolecules as nucleic acids present in liquid biopsies might have prognostic potential and could serve as predictive biomarkers for chemotherapeutic regimens. This review will focus on the role of circulating tumour DNAs and non-coding RNAs as diagnostic, prognostic, and predictive biomarkers in the context of colorectal cancer.
Collapse
|
30
|
Fotiadis N, De Paepe KN, Bonne L, Khan N, Riddell A, Turner N, Starling N, Gerlinger M, Rao S, Chau I, Cunningham D, Koh DM. Comparison of a coaxial versus non-coaxial liver biopsy technique in an oncological setting: diagnostic yield, complications and seeding risk. Eur Radiol 2020; 30:6702-6708. [PMID: 32666317 PMCID: PMC7599171 DOI: 10.1007/s00330-020-07038-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 05/04/2020] [Accepted: 06/16/2020] [Indexed: 11/24/2022]
Abstract
Objectives Percutaneous liver biopsy (PLB) poses specific challenges in oncological patients such as bleeding and tumour seeding. This study’s aim was to compare a coaxial (C-PLB) and non-coaxial (NC-PLB) biopsy technique in terms of diagnostic yield, safety and seeding risk of image-guided PLB techniques in an oncological setting. Methods Local research committee approval was obtained for this single-site retrospective study. Patients who underwent a PLB between November 2011 and December 2017 were consecutively included. Medical records were reviewed to determine diagnostic yield and complications. Follow-up imaging was re-reviewed for seeding, defined as visible tumour deposits along the PLB track. Mann-Whitney U and chi-squared tests were performed to investigate differences between biopsy techniques in sample number, complications and seeding rate. Results In total, 741 patients (62 ± 13 years, 378 women) underwent 932 PLB (C-PLB 72.9% (679/932); NC-PLB 27.1% (253/932)). More tissue cores (p < 0.001) were obtained with C-PLB (median 4 cores; range 1–12) compared with NC-PLB (2 cores; range 1–4) and diagnostic yield was similar for both techniques (C-PLB 92.6% (629/679); NC-PLB 92.5% (234/253); p = 0.940). Complication rate (9.3%; 87/932) using C-PLB (8.2% (56/679)) was lower compared with NC-PLB (12.3% (31/253); p = 0.024). Major complications were uncommon (C-PLB 2.7% (18/679); NC-PLB 2.8% (7/253)); bleeding developed in 1.2% (11/932; C-PLB 1.2% (8/679); NC-PLB 1.2% (3/253)). Seeding was a rare event, occurring significantly less in C-PLB cases (C-PLB 1.3% (7/544); NC-PLB 3.1% (6/197); p = 0.021). Conclusions C-PLB allows for high diagnostic tissue yield with a lower complication and seeding rate than a NC-PLB and should be the preferred method in an oncological setting. Key Points • A coaxial percutaneous liver biopsy achieves a significant higher number of cores and fewer complications than a non-coaxial biopsy technique. • The risk of tumour seeding is very low and is significantly lower using the coaxial biopsy technique. • In this study, a larger number of cores (median = 4) could be safely acquired using the coaxial technique, providing sufficient material for advanced molecular analysis.
Collapse
Affiliation(s)
- Nicos Fotiadis
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Rd, Sutton, London, SW2 5PT, UK.
| | - Katja N De Paepe
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Rd, Sutton, London, SW2 5PT, UK.
| | - Lawrence Bonne
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Rd, Sutton, London, SW2 5PT, UK
| | - Nasir Khan
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Rd, Sutton, London, SW2 5PT, UK
| | - Angela Riddell
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Rd, Sutton, London, SW2 5PT, UK
| | - Nicholas Turner
- Department of Medical, The Royal Marsden NHS Foundation Trust, London, UK
| | - Naureen Starling
- Department of Medical, The Royal Marsden NHS Foundation Trust, London, UK
| | - Marco Gerlinger
- Translational Oncogenomics, Institute of Cancer Research, London, UK
| | - Sheela Rao
- Department of Medical, The Royal Marsden NHS Foundation Trust, London, UK
| | - Ian Chau
- Department of Medical, The Royal Marsden NHS Foundation Trust, London, UK
| | - David Cunningham
- Department of Medical, The Royal Marsden NHS Foundation Trust, London, UK
| | - Dow-Mu Koh
- Department of Radiology, The Royal Marsden NHS Foundation Trust, Downs Rd, Sutton, London, SW2 5PT, UK
| |
Collapse
|
31
|
Grothey A, Blay JY, Pavlakis N, Yoshino T, Bruix J. Evolving role of regorafenib for the treatment of advanced cancers. Cancer Treat Rev 2020; 86:101993. [DOI: 10.1016/j.ctrv.2020.101993] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 02/14/2020] [Accepted: 02/17/2020] [Indexed: 12/13/2022]
|
32
|
Xie N, Tian C, Wu H, Yang X, Liu L, Li J, Xiao H, Gao J, Lu J, Hu X, Cao M, Shui Z, Tang Y, Wang X, Yang J, Hu ZY, Ouyang Q. FGFR aberrations increase the risk of brain metastases and predict poor prognosis in metastatic breast cancer patients. Ther Adv Med Oncol 2020; 12:1758835920915305. [PMID: 32499836 PMCID: PMC7243401 DOI: 10.1177/1758835920915305] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 02/05/2020] [Indexed: 02/06/2023] Open
Abstract
Background: The survival status of patients with breast cancer and brain metastasis (BCBM) receiving current treatments is poor. Method: We designed a real-world study to investigate using patients’ clinical and genetic aberrations to forecast the prognoses of BCBM patients. We recruited 146 BCBM patients and analyzed their clinical features to evaluate the overall survival (OS). For genetic testing, 30 BCBM and 165 non-brain-metastatic (BM) metastatic breast cancer (MBC) patients from Hunan Cancer Hospital, and 86 BCBM and 1416 non-BM MBC patients from the Geneplus database who received circulating tumor DNA testing, were compared and analyzed. Results: Ki67 >14% and >3 metastatic brain tumors were significant risk factors associated with poor OS, while chemotherapy and brain radiotherapy were beneficial factors for better OS. Compared with non-BM MBC patients, BCBM patients had more fibroblast growth factor receptor (FGFR) aberrations. The combination of FGFR, TP53 and FLT1 aberrations plus immunohistochemistry HER2-positive were associated with an increased risk of brain metastasis (AUC = 77.13%). FGFR aberration alone was not only a predictive factor (AUC = 67.90%), but also a significant risk factor for poor progression-free survival (Logrank p = 0.029). FGFR1 aberration was more frequent than other FGFR family genes in BCBM patients, and FGFR1 aberration was significantly higher in BCBM patients than non-BM MBC patients. Most FGFR1-amplified MBC patients progressed within 3 months of the late-line (>2 lines) treatment. Conclusion: A group of genetic events, including FGFR, TP53 and FLT1 genetic aberrations, and HER2-positivity, forecasted the occurrence of BM in breast cancers. FGFR genetic aberration alone predicted poor prognosis.
Collapse
Affiliation(s)
- Ning Xie
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Can Tian
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Hui Wu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Xiaohong Yang
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Liping Liu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Jing Li
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Huawu Xiao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Jianxiang Gao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Jun Lu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Xuming Hu
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Min Cao
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Zhengrong Shui
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Yu Tang
- Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha, China
| | - Xiao Wang
- ICF, 3 Corporate Square NE., Atlanta, GA, USA
| | - Jianbo Yang
- Department of Otolaryngology, Medical School, University of Minnesota, Minneapolis, Minnesota, USA
| | - Zhe-Yu Hu
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, No. 283, Tongzipo Road, Changsha, 410013, P.R. China
| | - Quchang Ouyang
- Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, No. 283, Tongzipo Road, Changsha, 410013, P.R. China
| |
Collapse
|
33
|
Imaging and clinical correlates with regorafenib in metastatic colorectal cancer. Cancer Treat Rev 2020; 86:102020. [PMID: 32278232 DOI: 10.1016/j.ctrv.2020.102020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 12/11/2022]
Abstract
In colorectal cancer (CRC), imaging is important in determining tumor stage, selecting treatment strategies, and in assessing response to therapy. However, some challenges remain with established imaging techniques, such as computed tomography, and with some commonly used response criteria, such as Response Evaluation Criteria in Solid Tumors, which measures change in size of several target lesions instead of change in tumor morphology or metabolic function. In addition, these assessments are not typically conducted until after 8 weeks of treatment, meaning that potential non-responders are often not identified in a timely manner. Regorafenib, an oral tyrosine kinase inhibitor indicated for the treatment of metastatic CRC, blocks the activity of several protein kinases involved in angiogenesis, oncogenesis, metastasis, and tumor immunity. Timely differentiation of regorafenib responders from non-responders using appropriate imaging techniques that recognize not only changes in tumor size but also changes in tumor density or vasculature, may reduce unnecessary drug-related toxicity in patients who are unlikely to respond to treatment. This review discusses the latest developments in computed tomography, magnetic resonance imaging, and positron emission tomography tumor imaging modalities, and how these aid in identifying patients with metastatic CRC who are responders or non-responders to regorafenib treatment.
Collapse
|
34
|
Oki E, Kawahira M, Kusumoto T, Yuki S, Hatanaka K, Kobayashi Y, Nishie A, Kawanami S, Makiyama A, Saeki H, Sakamoto S, Komatsu Y, Shimokawa M, Mori M, Esaki T. Multicenter Cohort Study to Assess the Association between Changes on Imaging and Outcome after Regorafenib Treatment (KSCC1603). Oncology 2020; 98:719-726. [DOI: 10.1159/000507814] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 03/25/2020] [Indexed: 11/19/2022]
Abstract
<b><i>Background:</i></b> Molecular targeted drugs having angiogenesis-inhibiting properties allow the induction of necrosis inside tumors. We retrospectively investigated the relationship between changes on imaging associated with regorafenib (REGO) and treatment outcomes using real-world data. <b><i>Patients and Methods:</i></b> The eligibility criteria included an ECOG PS of 0–1, a starting dose of 120 or 160 mg/day of REGO, and a duration of treatment of at least 35 days. Regarding changes on imaging, cavitation in lung lesions (CLL), morphologic response of liver lesions (MRL), and change of liver metastasis density (CLD) were evaluated. <b><i>Results:</i></b> We finally screened 671 cases, and 226 cases were eligible. In total, 172 and 145 patients had lung and liver metastases, respectively. Among the patients with lung metastasis, CLL was found in 69 patients (40.0%). The median progression-free survival (PFS) of the patients with and those without CLL was 3.2 and 2.4 months, respectively (hazard ratio [HR] = 0.758; 95% confidence interval [CI]: 0.529–1.087), and the median overall survival (OS) of these groups was 10.5 and 8.9 months, respectively (HR = 0.862; 95% CI: 0.579–1.285). MRL and CLD of liver metastasis were analyzed in 145 and 90 patients, respectively. The median OS with and without MRL was 8.9 and 8.2 months, respectively, whereas the median OS with and without CLD was 11.6 and 7.7 months, respectively (HR = 0.523; 95% CI: 0.275–0.992). <b><i>Conclusion:</i></b> CLL may predict PFS but not OS among patients with lung metastasis. CLD was predictive of favorable outcomes for REGO in patients with liver metastasis.
Collapse
|
35
|
Conteduca V, Scarpi E, Matteucci F, Caroli P, Ravaglia G, Fantini L, Gurioli G, Schepisi G, Wetterskog D, Menna C, Burgio SL, Lolli C, Paganelli G, Attard G, De Giorgi U. Multimodal Approach to Outcome Prediction in Metastatic Castration-Resistant Prostate Cancer by Integrating Functional Imaging and Plasma DNA Analysis. JCO Precis Oncol 2019; 3:1-13. [DOI: 10.1200/po.18.00302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
PURPOSE Biomarkers for treatment personalization in metastatic castration-resistant prostate cancer (mCRPC) could help improve patient outcomes. Multiple tests on blood have reported associations with poorer outcome, including serum lactate dehydrogenase (LDH), chromogranin A (CGA), neutrophil:lymphocyte ratio (NLR), and, recently, copy number (CN) of androgen receptor (AR) in plasma DNA. Biologic data suggest an association between choline uptake and AR signaling. We aimed to integrate 18F-fluorocholine (FCH) uptake on positron emission tomography/computed tomography (PET/CT) scanning with plasma AR CN and other routinely obtained circulating biomarkers to evaluate their association with outcome. MATERIALS AND METHODS We determined plasma AR CN by digital droplet polymerase chain reaction from 105 mCRPC samples collected before abiraterone (n = 65) or enzalutamide (n = 40) therapy in the before (n = 26) and after (n = 79) chemotherapy settings. Pretreatment serum LDH, CGA, and NLR were also measured. FCH-PET/CT scan was performed at baseline, and maximum standardized uptake value (SUVmax), total lesion activity (TLA), and metabolic tumor volume (MTV) were calculated. Main end points were the correlation of FCH-PET/CT parameters with circulating biomarkers and their impact on outcome. RESULTS Plasma AR CN gain was observed in 27 patients (25.7%), and it correlated significantly with higher median SUVmax, TLA, and MTV values ( P < .001). Kaplan-Meier curves showed significantly worse progression-free survival and overall survival in patients with plasma AR gain and higher SUVmax, TLA, and MTV values ( P < .001 in each prognostic group). Conversely, no association was reported for prostate-specific antigen response. On multivariable analysis of overall survival, we showed as independent factors AR gain (hazard ratio [HR], 1.92; 95% CI, 1.07 to 3.47; P = .029), presence of visceral metastasis (HR, 3.04; 95% CI, 1.66 to 5.58; P = < .001), LDH (HR, 2.95; 95% CI, 1.72 to 5.05; P < .001), NLR (HR, 3.51; 95% CI, 2.14 to 5.74; P < .001), serum CGA (HR, 3.36; 95% CI, 1.99 to 5.67; P < .001), and MTV (HR, 2.09; 95% CI, 1.25 to 3.50; P = .005). CONCLUSION Our results indicate the potential usefulness of integrating functional imaging with plasma DNA analysis and other noninvasive biomarkers as a tool to improve treatment selection for CRPC. A larger prospective evaluation is warranted.
Collapse
Affiliation(s)
- Vincenza Conteduca
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Emanuela Scarpi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Federica Matteucci
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Caroli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgia Ravaglia
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Lorenzo Fantini
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giorgia Gurioli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giuseppe Schepisi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Cecilia Menna
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Salvatore Luca Burgio
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Cristian Lolli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giovanni Paganelli
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | | | - Ugo De Giorgi
- Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
36
|
Arai H, Battaglin F, Wang J, Lo JH, Soni S, Zhang W, Lenz HJ. Molecular insight of regorafenib treatment for colorectal cancer. Cancer Treat Rev 2019; 81:101912. [PMID: 31715423 PMCID: PMC7491975 DOI: 10.1016/j.ctrv.2019.101912] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 12/22/2022]
Abstract
Regorafenib is a multi-targeting kinase inhibitor approved for the treatment of metastatic colorectal cancer patients in refractory to standard chemotherapy. Similarly to sorafenib, this agent was originally developed as a RAF1 inhibitor. However, the kinase inhibitory profile is distinct from sorafenib. A broad-spectrum of kinase inhibition induces wide-range drug sensitivity, irrespective of mutation status of major oncogenes. This agent's main therapeutic effects are anti-angiogenesis and the remodeling of tumor microenvironment through several mechanisms of action. The dual blockade of VEGF receptors and TIE2 can lead to both additive anti-angiogenesis effects and the suggestive unique regulation of vessel stability. Additionally, it inhibits molecular escape pathways to VEGF inhibition (e.g., FGF, PIGF, and PDGF signaling), enabling its continuous antiangiogenic effect even in tumors resistant to VEGF inhibitors. Furthermore, regorafenib has the important effect of enhancing anti-tumor immunity via macrophage modulation. Based on this concept, clinical trials have been recently launched for the development of a combination strategy with immune checkpoint inhibitors. Contrary to regorafenib induced clinical benefits and advances in the novel strategy, currently no predictive biomarkers have been identified. In the present review, we revisit and summarize regorafenib's unique mechanisms of action. The review could highlight molecular insights and provide some perspective for the search of predictive biomarkers used in metastatic colorectal cancer patients treated with regorafenib.
Collapse
Affiliation(s)
- Hiroyuki Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States
| | - Francesca Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| | - Jingyuan Wang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| | - Jae Ho Lo
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| | - Shivani Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States
| | - Wu Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| | - Heinz-Josef Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, 1441 Eastlate Avenue, Los Angeles, CA 90033, United States.
| |
Collapse
|
37
|
Reece M, Saluja H, Hollington P, Karapetis CS, Vatandoust S, Young GP, Symonds EL. The Use of Circulating Tumor DNA to Monitor and Predict Response to Treatment in Colorectal Cancer. Front Genet 2019; 10:1118. [PMID: 31824558 PMCID: PMC6881479 DOI: 10.3389/fgene.2019.01118] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/16/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Colorectal cancer is one of the most common cancers worldwide and has a high mortality rate following disease recurrence. Treatment efficacy is maximized by providing tailored cancer treatment, ideally involving surgical resection and personalized neoadjuvant and adjuvant therapies, including chemotherapy, radiotherapy and increasingly, targeted therapy. Early detection of recurrence or disease progression results in more treatable disease and is essential to improving survival outcomes. Recent advances in the understanding of tumor genetics have resulted in the discovery of circulating tumor DNA (ctDNA). A growing body of evidence supports the use of these sensitive biomarkers in detecting residual disease and diagnosing recurrence as well as enabling targeted and tumor-specific adjuvant therapies. Methods: A literature search in Pubmed was performed to identify all original articles preceding April 2019 that utilize ctDNA for the purpose of monitoring response to colorectal cancer treatment. Results: Ninety-two clinical studies were included. These studies demonstrate that ctDNA is a reliable measure of tumor burden. Studies show the utility of ctDNA in assessing the adequacy of surgical tumor clearance and changes in ctDNA levels reflect response to systemic treatments. ctDNA can be used in the selection of targeted treatments. The reappearance or increase in ctDNA, as well as the emergence of new mutations, correlates with disease recurrence, progression, and resistance to therapy, with ctDNA measurement allowing more sensitive monitoring than currently used clinical tools. Conclusions: ctDNA shows enormous promise as a sensitive biomarker for monitoring response to many treatment modalities and for targeting therapy. Thus, it is emerging as a new way for guiding treatment decisions-initiating, altering, and ceasing treatments, or prompting investigation into the potential for residual disease. However, many potentially useful ctDNA markers are available and more work is needed to determine which are best suited for specific purposes and for improving specific outcomes.
Collapse
Affiliation(s)
- Mifanwy Reece
- Colorectal Surgery, Division of Surgery & Perioperative Medicine, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Hariti Saluja
- Department of Medicine, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Paul Hollington
- Colorectal Surgery, Division of Surgery & Perioperative Medicine, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Christos S Karapetis
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Department of Medical Oncology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Sina Vatandoust
- Department of Medical Oncology, Flinders Medical Centre, Bedford Park, SA, Australia
| | - Graeme P Young
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia
| | - Erin L Symonds
- Flinders Centre for Innovation in Cancer, College of Medicine and Public Health, Flinders University, Bedford Park, SA, Australia.,Bowel Health Service, Flinders Medical Centre, Bedford Park, SA, Australia
| |
Collapse
|
38
|
Giulia M, Teresa T, Claudia C, Paolo VP, Davide C, Floriana M, Fortunato C, Erika M. Anti-angiogenic Treatment in Metastatic Colorectal Cancer: Current Issues and Future Aims. CURRENT CANCER THERAPY REVIEWS 2019. [DOI: 10.2174/1573394714666181119145327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
:
Blocking angiogenesis represents a fundamental process in Colorectal Cancer (CRC)
treatment. VEGF (vascular endothelial growth factor) pathway is implicated in various processes
that regulate tumor vascularization and proliferation. In the last years, great efforts have been
made thanks to the discovery of targeted drugs that block VEGF and its receptors conferring a
benefit in a variety of tumors, including CRC. To date, four drugs have been approved for the
treatment of metastatic CRC (mCRC): bevacizumab, aflibercept, ramucirumab and regorafenib.
Unfortunately, patients relapse due to the appearance of resistance. The VEGF family, its role in
the angiogenesis and complex heterogeneity of mechanisms that escape tumor blockade are not
completely understood and there is a lack of biomarkers of response to anti-angiogenic drugs. We
describe the principal mechanisms of resistance to anti-VEGF therapy and discuss potential biomarkers
to be investigated in the near future.
Collapse
Affiliation(s)
- Martini Giulia
- Medical Oncology, Department of Clinical and Experimental Medicine F. Magrassi, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Troiani Teresa
- Medical Oncology, Department of Clinical and Experimental Medicine F. Magrassi, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Cardone Claudia
- Medical Oncology, Department of Clinical and Experimental Medicine F. Magrassi, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Vitiello Pietro Paolo
- Medical Oncology, Department of Clinical and Experimental Medicine F. Magrassi, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Ciardiello Davide
- Medical Oncology, Department of Clinical and Experimental Medicine F. Magrassi, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Morgillo Floriana
- Medical Oncology, Department of Clinical and Experimental Medicine F. Magrassi, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Ciardiello Fortunato
- Medical Oncology, Department of Clinical and Experimental Medicine F. Magrassi, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| | - Martinelli Erika
- Medical Oncology, Department of Clinical and Experimental Medicine F. Magrassi, Università degli Studi della Campania L. Vanvitelli, Naples, Italy
| |
Collapse
|
39
|
Stoetzer M, Alevizakos V, Rahlf B, Gellrich NC, Kampmann A, von See C. The Impact of Different Augmentative Methods on the Expression of Inflammatory Factors. J ORAL IMPLANTOL 2019; 45:356-361. [PMID: 31536443 DOI: 10.1563/aaid-joi-d-19-00050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Many animal studies show that an intact periosteum plays an important role in osseous regeneration. The potential effect of an in vivo periosteal barrier membrane on the expression of specific proteins has not been examined sufficiently. The aim of the present study is to investigate the influence of the flap preparation method and collagen membrane on the emission of inflammatory factors. This study examines 20 patients with dental implants who had previously undergone an augmentation. A soft tissue sample was taken during augmentation and 3 months later from the same location. Samples were always taken from the margins of a previously prepared mucoperiosteal flap. The flap was raised with a conventional periosteal elevator in the control group and with a piezoelectric device in the test group. In both groups, we covered half of the augmented bone with a native collagen membrane (NCM; Geistlich Bio-Gide). This allowed us to examine the same incision area with and without a membrane. An immunohistochemical analysis was performed for collagen IV, fibronectin, and inflammatory factors such as cluster of differentiation 31 (CD31), cyclooxygenase-2 (COX-2), and interleukin 6 (IL-6). There was a clear difference in the expression of specific proteins after the piezoelectric device and the periosteal elevator were used. The expression of fibronectin, IL-6, and COX-2 was higher after preparation with the periosteal elevator than after piezoelectric periosteum dissection. The expression of collagen IV was higher after the piezoelectric procedure. No difference was observed for CD31. The membrane had no effect on the expression of collagen IV, fibronectin, IL-6, and COX-2. The type of periosteal preparation influences the expression of specific proteins. With regard to the factors examined here, NCM did not appear to influence the wound healing cascade.
Collapse
Affiliation(s)
| | - Vasilios Alevizakos
- Danube Private University, Center for Digital Technologies in Dentistry and CAD/CAM, Krems an der Donau, Austria
| | | | | | | | - Constantin von See
- Danube Private University, Center for Digital Technologies in Dentistry and CAD/CAM, Krems an der Donau, Austria
| |
Collapse
|
40
|
Andersen IR, Thorup K, Andersen MB, Olesen R, Mortensen FV, Nielsen DT, Rasmussen F. Texture in the monitoring of regorafenib therapy in patients with colorectal liver metastases. Acta Radiol 2019; 60:1084-1093. [PMID: 30612433 DOI: 10.1177/0284185118817940] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Iben R Andersen
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | - Kennet Thorup
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Rene Olesen
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Dennis T Nielsen
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | - Finn Rasmussen
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
41
|
Amatu A, Schirripa M, Tosi F, Lonardi S, Bencardino K, Bonazzina E, Palmeri L, Patanè DA, Pizzutilo EG, Mussolin B, Bergamo F, Alberti G, Intini R, Procaccio L, Arese M, Marsoni S, Nichelatti M, Zagonel V, Siena S, Bardelli A, Loupakis F, Di Nicolantonio F, Sartore-Bianchi A, Barault L. High Circulating Methylated DNA Is a Negative Predictive and Prognostic Marker in Metastatic Colorectal Cancer Patients Treated With Regorafenib. Front Oncol 2019; 9:622. [PMID: 31355139 PMCID: PMC6640154 DOI: 10.3389/fonc.2019.00622] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/24/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Regorafenib improves progression free survival (PFS) in a subset of metastatic colorectal cancer (mCRC) patients, although no biomarkers of efficacy are available. Circulating methylated DNA (cmDNA) assessed by a five-gene panel was previously associated with outcome in chemotherapy treated mCRC patients. We hypothesized that cmDNA could be used to identify cases most likely to benefit from regorafenib (i.e., patients with PFS longer than 4 months). Methods: Plasma samples from mCRC patients were collected prior to (baseline samples N = 60) and/or during regorafenib treatment (N = 62) for the assessment of cmDNA and total amount of cell free DNA (cfDNA). Results: In almost all patients, treatment with regorafenib increased the total cfDNA, but decreased cmDNA warranting the normalization of cmDNA to the total amount of circulating DNA (i.e., cmDNA/ml). We report that cmDNA/ml dynamics reflects clinical response with an increase in cmDNA/ml associated with higher risk of progression (HR for progression = 1.78 [95%CI: 1.01-3.13], p = 0.028). Taken individually, high baseline cmDNA/ml (above median) was associated with worst prognosis (HR for death = 3.471 [95%CI: 1.83-6.57], p < 0.0001) and also predicted shorter PFS (<16 weeks with PPV 86%). In addition, high cmDNA/ml values during regorafenib treatment predicted with higher accuracy shorter PFS (<16 weeks with a PPV of 96%), therefore associated with increased risk of progression (HR for progression = 2.985; [95%CI: 1.63-5.46; p < 0.0001). Conclusions: Our data highlight the predictive and prognostic value of cmDNA/ml in mCRC patients treated with regorafenib.
Collapse
Affiliation(s)
- Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Marta Schirripa
- Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Padua, Italy
| | - Federica Tosi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Sara Lonardi
- Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Padua, Italy
| | - Katia Bencardino
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Erica Bonazzina
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Laura Palmeri
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | | | | | - Francesca Bergamo
- Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Padua, Italy
| | - Giulia Alberti
- Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Rossana Intini
- Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Padua, Italy
| | - Letizia Procaccio
- Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Padua, Italy.,Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Marco Arese
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Oncology, University of Turin, Candiolo, Italy
| | - Silvia Marsoni
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Precision Oncology, FIRC Institute of Molecular Oncology (IFOM), Milan, Italy
| | - Michele Nichelatti
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Vittorina Zagonel
- Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Padua, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Dipartimento di Oncologia ed Emato-Oncologia, Università degli Studi di Milano, Milan, Italy
| | - Alberto Bardelli
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Oncology, University of Turin, Candiolo, Italy
| | - Fotios Loupakis
- Medical Oncology 1, Veneto Institute of Oncology, IRCCS, Padua, Italy
| | - Federica Di Nicolantonio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Oncology, University of Turin, Candiolo, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Dipartimento di Oncologia ed Emato-Oncologia, Università degli Studi di Milano, Milan, Italy
| | - Ludovic Barault
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.,Department of Oncology, University of Turin, Candiolo, Italy
| |
Collapse
|
42
|
Andersen IR, Olesen R, Boysen AK, Jensen LH, Mortensen FV, Nielsen DT, Rasmussen F. Dynamic contrast-enhanced computed tomography as a potential biomarker in patients with metastatic colorectal cancer treated with regorafenib. Acta Radiol 2019; 60:836-845. [PMID: 30348001 DOI: 10.1177/0284185118806652] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Iben R Andersen
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | - Rene Olesen
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Anders K Boysen
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Lars H Jensen
- Department of Oncology, Lillebaelt Hospital, Vejle, Denmark
| | | | - Dennis T Nielsen
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| | - Finn Rasmussen
- Department of Radiology, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
43
|
Ruiz-Bañobre J, Kandimalla R, Goel A. Predictive Biomarkers in Metastatic Colorectal Cancer: A Systematic Review. JCO Precis Oncol 2019; 3:PO.18.00260. [PMID: 32914007 PMCID: PMC7446314 DOI: 10.1200/po.18.00260] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2018] [Indexed: 12/13/2022] Open
Abstract
PURPOSE The development and use of predictive biomarkers to guide treatment decisions are paramount not only for improving survival in patients with metastatic colorectal cancer (mCRC), but also for sparing them from unnecessary toxicity and reducing the economic burden of expensive treatments. We conducted a systematic review of published studies and evaluated the predictive biomarker landscape in the mCRC setting from a molecular and clinical viewpoint. METHODS Studies analyzing predictive biomarkers for approved therapies in patients with mCRC were identified systematically using electronic databases. Preclinical studies and those providing no relevant information were excluded. RESULTS A total of 173 studies comprising 148 biomarkers were selected for final analysis. Of all the biomarkers analyzed, 1.4% (two of 148) were explored in a prospective manner, whereas 98.6% (146 of 148) were evaluated in retrospective studies. Of the latter group, 78.8% (115 of 146) were not tested in subsequent phases, 9.6% (14 of 146) were tested in other retrospective cohorts, 8.9% (13 of 146) were retrospectively tested in at least one or more randomized cohorts, and only 2.7% (four of 146) were prospectively tested in a clinical trial. Finally, only 1.4% (two of 148) were validated sufficiently and are recognized as biomarkers for guiding treatment decision making in patients with mCRC. These markers were RAS mutational status for anti-EGFR antibodies and microsatellite instability status for anti-programmed cell death-1 drugs. CONCLUSION Despite notable efforts to identify predictive biomarkers for various therapies used in the mCRC setting, because of a lack of data beyond retrospective studies and successful biomarker-driven approaches, only two molecular biomarkers have thus far found their translation into the clinic, highlighting the imperative need for implementing novel strategies and additional research in this clinically important field.
Collapse
Affiliation(s)
- Juan Ruiz-Bañobre
- Arquitecto Marcide University Hospital, Ferrol, Spain
- Baylor University Medical Center, Dallas, TX
- ONCOMET, University Clinical Hospital of Santiago de Compostela, CIBERONC, Santiago de Compostela, Spain
| | | | - Ajay Goel
- Baylor University Medical Center, Dallas, TX
| |
Collapse
|
44
|
Zhou D, Ouyang Q, Liu L, Liu J, Tang Y, Xiao M, Wang Y, He Q, Hu ZY. Chemotherapy Modulates Endocrine Therapy-Related Resistance Mutations in Metastatic Breast Cancer. Transl Oncol 2019; 12:764-774. [PMID: 30893632 PMCID: PMC6423490 DOI: 10.1016/j.tranon.2019.02.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/21/2019] [Accepted: 02/21/2019] [Indexed: 01/07/2023] Open
Abstract
PURPOSE: Accumulation of PIK3CA, ESR1, and GATA3 mutations results in resistance to endocrine therapy in breast cancer patients; however, the response of these genes to chemotherapy is unclear. Therefore, we sought to evaluate the genetic response of circulating tumor DNA (ctDNA) to chemotherapy in metastatic breast cancer patients. METHODS: The mutation frequency of 1021 genes was examined prior to chemotherapy in ctDNA of 44 estrogen receptor–positive metastatic breast cancer patients. These genes were evaluated again in a subset of patients (n=24) following chemotherapy. Mutation frequency was defined as the percentage of mutations found in ctDNA compared to total cell-free DNA. RESULTS: Prior to chemotherapy, PIK3CA was the most commonly mutated gene, with mutation found in 22 of the metastatic breast cancer patients. Following chemotherapy, 16 patients exhibited progressive disease (PD), and 8 patients experienced no progression (non-PD). PIK3CA mutation frequency increased in 56.25% (9/16) of the PD patients but decreased in 62.5% (5/8) of the non-PD patients. As a result, more PD patients exhibited increased PIK3CA mutation frequency than non-PD patients (56.25% vs 0%, P=.002). Further, ESR1 and GATA3 mutations correlated with PIK3CA mutation. Interestingly, patients receiving the mTOR inhibitor everolimus exhibited a lower progression rate (0% vs 62.5%, P=.001), and the combination of everolimus and chemotherapy effectively suppressed PIK3CA, ESR1, and GATA3 gene mutations. CONCLUSION: Together, these results suggest that mTOR inhibition may be a useful chemotherapy adjuvant to suppress chemotherapy-induced gene mutations that render tumors resistant to endocrine therapy in metastatic breast cancer patients with PD.
Collapse
Affiliation(s)
- Dabo Zhou
- The Affiliated Cancer Hospital of Xiangya Medical School, Central South University / Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Quchang Ouyang
- The Affiliated Cancer Hospital of Xiangya Medical School, Central South University / Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China.
| | - Liping Liu
- The Affiliated Cancer Hospital of Xiangya Medical School, Central South University / Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Jingyu Liu
- The Affiliated Cancer Hospital of Xiangya Medical School, Central South University / Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Yu Tang
- The Affiliated Cancer Hospital of Xiangya Medical School, Central South University / Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Mengjia Xiao
- The Affiliated Cancer Hospital of Xiangya Medical School, Central South University / Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China
| | - Yikai Wang
- Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, 33022, USA
| | - Qiongzhi He
- Geneplus Beijing Institute, Beijing 102206, China
| | - Zhe-Yu Hu
- The Affiliated Cancer Hospital of Xiangya Medical School, Central South University / Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, Hunan Cancer Hospital, Changsha, 410013, China; Department of Breast Cancer Medical Oncology, the Affiliated Cancer Hospital of Xiangya Medical School, Central South University, Changsha 410013, China.
| |
Collapse
|
45
|
SUGIMOTO KIICHI, YAMADA TAKESHI, ISHIYAMA SHUN, NAKAMURA MASATO, YOSHIDA YOICHIRO, HASEGAWA SUGURU, YOKOMIZO HAJIME, NARITAKA YOSHIHIKO, SAKUYAMA NAOKI, OCHIAI TAKUMI, SONODA HIROMICHI, TANI MASAJI, ENOMOTO MASANOBU, KATSUMATA KENJI, YAMAGUCHI TATSURO, OHTA RYO, ISHIMARU KEI, WATANABE YUJI, KODA KEIJI, SAKAMOTO KAZUHIRO. Study Protocol of Regorafenib Escalation for Colorectal Cancer (RECC): A Phase II Multicenter Clinical Trial of the Efficacy and Safety of Regorafenib Dose Escalation Therapy as the Third or Fourth Line Therapy for Unresectable/Recurrent Colorectal Cancer. JUNTENDO MEDICAL JOURNAL 2019. [DOI: 10.14789/jmj.2019.65.jmj18-sp01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
Affiliation(s)
- KIICHI SUGIMOTO
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine
| | - TAKESHI YAMADA
- Department of Gastrointestinal and Hepato-Biliary-Pancreatic Surgery, Nippon Medical School
| | - SHUN ISHIYAMA
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine
| | | | - YOICHIRO YOSHIDA
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine
| | - SUGURU HASEGAWA
- Department of Gastroenterological Surgery, Fukuoka University Faculty of Medicine
| | - HAJIME YOKOMIZO
- Department of Surgery, Tokyo Women’s Medical University, Medical Center East
| | - YOSHIHIKO NARITAKA
- Department of Surgery, Tokyo Women’s Medical University, Medical Center East
| | - NAOKI SAKUYAMA
- Department of Surgery, Tobu Chiiki Hospital, Metropolitan Health and Medical Treatment Corporation
| | - TAKUMI OCHIAI
- Department of Surgery, Tobu Chiiki Hospital, Metropolitan Health and Medical Treatment Corporation
| | | | - MASAJI TANI
- Department of Surgery, Shiga University of Medical Science
| | - MASANOBU ENOMOTO
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University
| | - KENJI KATSUMATA
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University
| | - TATSURO YAMAGUCHI
- Department of Surgery, Tokyo Metropolitan Cancer and Infectious Diseases Center Komagome Hospital
| | - RYO OHTA
- Department of Surgery, Institute of Gastroenterology, Kawasaki Saiwai Hospital
| | - KEI ISHIMARU
- Department of Gastrointestinal Surgery and Surgical Oncology, Ehime University Graduate School of Medicine
| | - YUJI WATANABE
- Department of Gastrointestinal Surgery and Surgical Oncology, Ehime University Graduate School of Medicine
| | - KEIJI KODA
- Department of Surgery, Teikyo University Chiba Medical Center
| | - KAZUHIRO SAKAMOTO
- Department of Coloproctological Surgery, Juntendo University Faculty of Medicine
| |
Collapse
|
46
|
Circulating tumor DNA applications in monitoring the treatment of metastatic colorectal cancer patients. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2019. [PMCID: PMC7009314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Colorectal cancer is the third most common cancer worldwide. New cancer treatment strategies such as monoclonal antibodies against growth factor and angiogenesis receptors have improved the overall survival (OS) and progression-free survival (PFS) in metastatic colorectal cancer (mCRC) patients. However, acquired resistance could happen after these therapies. Circulating tumor DNA (ctDNA) is the DNA fraction derived from tumor cells which could be applied as a non-invasive method for detecting tumor mutations before, during, and after therapies. Here, we reviewed most of the studies examining ctDNA as treatment monitoring in mCRC patients who receive different target therapies. Also, we compared ctDNA with other existing cancer-treatment monitoring methods.
Collapse
|
47
|
Advantage of Next-Generation Sequencing in Dynamic Monitoring of Circulating Tumor DNA over Droplet Digital PCR in Cetuximab Treated Colorectal Cancer Patients. Transl Oncol 2018; 12:426-431. [PMID: 30562681 PMCID: PMC6297189 DOI: 10.1016/j.tranon.2018.11.015] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 11/27/2018] [Accepted: 11/27/2018] [Indexed: 12/24/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) blockade resistance is common in the treatment of RAS wide type colorectal cancer (CRC). During the treatment of cetuximab, acquired resistant genomic alterations always occurs earlier than disease progression observed by medical images. Identification of genomic alterations dynamically might have certain clinical significance. Because of the limitation of repeated tissue biopsy, liquid biopsy is increasingly recognized. Droplet digital polymerase chain reaction (ddPCR) is the main detection methods for circulating tumor DNA (ctDNA), however, the application of next-generation sequencing (NGS) for ctDNA detection becomes more and more popular. Here we develop a NGS-based ctDNA assay and evaluated its sensitivity and specificity while using ddPCR as control. These two technologies were both used for genomic alteration detection for the peripheral blood samples from cetuximab-treated colorectal cancer patients dynamically. Fifteen patients were enrolled in this study, including eight males and seven females. The sensitivity and specificity of our NGS assay were 87.5% and 100% respectively, and liner regression analysis comparing variant allele frequency (VAF) revealed high concordance between NGS and ddPCR (R2 = 0.98). NGS actually found more mutation information than ddPCR such as the additional dynamic changes of TP53 which were observed in the disease progression patients. Moreover, the variant allele fraction of TP53 was also found by NGS to be changed along with the clinical efficacy evaluation dynamically during the whole treatment process. In conclusion, our newly developed NGS-based ctDNA assay shows similar performance with ddPCR but have more advantages of its high throughput of multigenetic detection for the dynamic monitoring during the treatment of cetuximab in metastasis CRC patients.
Collapse
|
48
|
Khan KH, Cunningham D, Werner B, Vlachogiannis G, Spiteri I, Heide T, Mateos JF, Vatsiou A, Lampis A, Damavandi MD, Lote H, Huntingford IS, Hedayat S, Chau I, Tunariu N, Mentrasti G, Trevisani F, Rao S, Anandappa G, Watkins D, Starling N, Thomas J, Peckitt C, Khan N, Rugge M, Begum R, Hezelova B, Bryant A, Jones T, Proszek P, Fassan M, Hahne JC, Hubank M, Braconi C, Sottoriva A, Valeri N. Longitudinal Liquid Biopsy and Mathematical Modeling of Clonal Evolution Forecast Time to Treatment Failure in the PROSPECT-C Phase II Colorectal Cancer Clinical Trial. Cancer Discov 2018; 8:1270-1285. [PMID: 30166348 PMCID: PMC6380469 DOI: 10.1158/2159-8290.cd-17-0891] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 05/01/2018] [Accepted: 07/05/2018] [Indexed: 12/14/2022]
Abstract
Sequential profiling of plasma cell-free DNA (cfDNA) holds immense promise for early detection of patient progression. However, how to exploit the predictive power of cfDNA as a liquid biopsy in the clinic remains unclear. RAS pathway aberrations can be tracked in cfDNA to monitor resistance to anti-EGFR monoclonal antibodies in patients with metastatic colorectal cancer. In this prospective phase II clinical trial of single-agent cetuximab in RAS wild-type patients, we combine genomic profiling of serial cfDNA and matched sequential tissue biopsies with imaging and mathematical modeling of cancer evolution. We show that a significant proportion of patients defined as RAS wild-type based on diagnostic tissue analysis harbor aberrations in the RAS pathway in pretreatment cfDNA and, in fact, do not benefit from EGFR inhibition. We demonstrate that primary and acquired resistance to cetuximab are often of polyclonal nature, and these dynamics can be observed in tissue and plasma. Furthermore, evolutionary modeling combined with frequent serial sampling of cfDNA allows prediction of the expected time to treatment failure in individual patients. This study demonstrates how integrating frequently sampled longitudinal liquid biopsies with a mathematical framework of tumor evolution allows individualized quantitative forecasting of progression, providing novel opportunities for adaptive personalized therapies.Significance: Liquid biopsies capture spatial and temporal heterogeneity underpinning resistance to anti-EGFR monoclonal antibodies in colorectal cancer. Dense serial sampling is needed to predict the time to treatment failure and generate a window of opportunity for intervention. Cancer Discov; 8(10); 1270-85. ©2018 AACR. See related commentary by Siravegna and Corcoran, p. 1213 This article is highlighted in the In This Issue feature, p. 1195.
Collapse
Affiliation(s)
- Khurum H Khan
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - David Cunningham
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Benjamin Werner
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Georgios Vlachogiannis
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Inmaculada Spiteri
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Timon Heide
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Javier Fernandez Mateos
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Alexandra Vatsiou
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Andrea Lampis
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Mahnaz Darvish Damavandi
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Hazel Lote
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Ian Said Huntingford
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Somaieh Hedayat
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Nina Tunariu
- Department of Radiology, The Royal Marsden NHS Trust, Londonand Sutton, United Kingdom
| | - Giulia Mentrasti
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Francesco Trevisani
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Sheela Rao
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Gayathri Anandappa
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Janet Thomas
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Clare Peckitt
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Nasir Khan
- Department of Radiology, The Royal Marsden NHS Trust, Londonand Sutton, United Kingdom
| | - Massimo Rugge
- Department of Medicine and Surgical Pathology, University of Padua, Padua, Italy
| | - Ruwaida Begum
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Blanka Hezelova
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Annette Bryant
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Thomas Jones
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Paula Proszek
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Matteo Fassan
- Department of Medicine and Surgical Pathology, University of Padua, Padua, Italy
| | - Jens C Hahne
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Michael Hubank
- Clinical Genomics, The Centre for Molecular Pathology, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
| | - Chiara Braconi
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, London and Sutton, United Kingdom
| | - Andrea Sottoriva
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom.
| | - Nicola Valeri
- Department of Medicine, The Royal Marsden NHS Trust, London and Sutton, United Kingdom.
- Division of Molecular Pathology, The Institute of Cancer Research, London and Sutton, United Kingdom
| |
Collapse
|
49
|
Anandappa G, Chau I. Evolving Tissue and Circulating Biomarkers as Prognostic and Predictive Tools in Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2018. [DOI: 10.1007/s11888-018-0410-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
50
|
Magnolin promotes autophagy and cell cycle arrest via blocking LIF/Stat3/Mcl-1 axis in human colorectal cancers. Cell Death Dis 2018; 9:702. [PMID: 29899555 PMCID: PMC5999973 DOI: 10.1038/s41419-018-0660-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/28/2018] [Accepted: 05/03/2018] [Indexed: 01/07/2023]
Abstract
Magnolin is a multi-bioactive natural compound that possesses underlying anti-cancer properties. However, the mechanisms underlying remain to be elucidated. Here, we report the role of magnolin in suppressing human colorectal cancer (CRC) cells via activating autophagy and cell cycle arrest in vitro and in vivo. Pre-treatment of cells with specific autophagy inhibitor (3-methyladenine) or knockdown of endogenous LC-3B by siRNA significantly abrogates magnolin-induced cell cycle arrest. Molecular validation mechanistically shows that magnolin-induced autophagy and cell cycle arrest in CRC cells is correlated with decreased transcriptional levels of leukemia inhibitory factor (LIF), and we further find that inhibition of LIF decreases phosphorylation level of Stat3 and represses transcriptional expression of Mcl-1. Furthermore, magnolin-induced autophagy and cell cycle arrest suppress the growth of xenograft colorectal tumors without apparent toxicity. Finally, we evaluate the clinical correlation of LIF/Stat3/Mcl-1 in CRC patient tissues. As expected, LIF, p-Stat3, and Mcl-1 levels are high in CRC tissue but are scarcely found in normal colon tissue. High positive expressions of LIF or Mcl-1 are associated with poor prognosis. Doubly positive cases have shown the worst outcome. Taken together, our results have clarified a novel molecular mechanism whereby magnolin induces autophagy and cell cycle arrest through LIF/Stat3/Mcl-1 pathway in CRCs. Our results also have revealed that magnolin has a promising therapeutic potential in CRCs.
Collapse
|