1
|
Cardone C, Facchini S, de Stefano A, Rachiglio AM, Avallone A. Transient 'NeoBRAF wild type' state in a patient with BRAF V600E mutant metastatic colorectal cancer. Cytopathology 2024; 35:671-673. [PMID: 39086078 DOI: 10.1111/cyt.13429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
We report the case of a patient with a BRAFV600E mutant mCRC, with evidence of acquired 'NeoBRAF wild-type' (wt) state. The patient, longitudinally assessed by liquid biopsy, obtained a remarkable clinical outcome with a multimodal approach including surgery, systemic treatment and targeted therapy. In patients with newly diagnosed RAS and BRAFV600E mutant mCRC, longitudinal assessment with liquid biopsy is not routinely used in clinical practice. We report the case of a patient with a BRAFV600E mutant mCRC, with evidence of acquired 'neoBRAF wild-type' (wt) state. The patient obtained a remarkable clinical outcome and has been longitudinally assessed by liquid biopsy.
Collapse
Affiliation(s)
- Claudia Cardone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Sergio Facchini
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Alfonso de Stefano
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Anna Maria Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| | - Antonio Avallone
- Experimental Clinical Abdominal Oncology Unit, Istituto Nazionale Tumori-IRCCS 'Fondazione G. Pascale', Naples, Italy
| |
Collapse
|
2
|
Qin P, Li Q, Zu Q, Dong R, Qi Y. Natural products targeting autophagy and apoptosis in NSCLC: a novel therapeutic strategy. Front Oncol 2024; 14:1379698. [PMID: 38628670 PMCID: PMC11019012 DOI: 10.3389/fonc.2024.1379698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related mortality worldwide, with non-small cell lung cancer (NSCLC) being the predominant type. The roles of autophagy and apoptosis in NSCLC present a dual and intricate nature. Additionally, autophagy and apoptosis interconnect through diverse crosstalk molecules. Owing to their multitargeting nature, safety, and efficacy, natural products have emerged as principal sources for NSCLC therapeutic candidates. This review begins with an exploration of the mechanisms of autophagy and apoptosis, proceeds to examine the crosstalk molecules between these processes, and outlines their implications and interactions in NSCLC. Finally, the paper reviews natural products that have been intensively studied against NSCLC targeting autophagy and apoptosis, and summarizes in detail the four most retrieved representative drugs. This paper clarifies good therapeutic effects of natural products in NSCLC by targeting autophagy and apoptosis and aims to promote greater consideration by researchers of natural products as candidates for anti-NSCLC drug discovery.
Collapse
Affiliation(s)
- Peiyi Qin
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong College of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Qingchen Li
- Department of Orthopedics, First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qi Zu
- Shandong College of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Ruxue Dong
- Shandong College of Traditional Chinese Medicine, Yantai, Shandong, China
| | - Yuanfu Qi
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
3
|
Stenzinger A, Vogel A, Lehmann U, Lamarca A, Hofman P, Terracciano L, Normanno N. Molecular profiling in cholangiocarcinoma: A practical guide to next-generation sequencing. Cancer Treat Rev 2024; 122:102649. [PMID: 37984132 DOI: 10.1016/j.ctrv.2023.102649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 10/29/2023] [Indexed: 11/22/2023]
Abstract
Cholangiocarcinomas (CCA) are a heterogeneous group of tumors that are classified as intrahepatic, perihilar, or distal according to the anatomic location within the biliary tract. Each CCA subtype is associated with distinct genomic alterations, including single nucleotide variants, copy number variants, and chromosomal rearrangements or gene fusions, each of which can influence disease prognosis and/or treatment outcomes. Molecular profiling using next-generation sequencing (NGS) is a powerful technique for identifying unique gene variants carried by an individual tumor, which can facilitate their accurate diagnosis as well as promote the optimal selection of gene variant-matched targeted treatments. NGS is particularly useful in patients with CCA because between one-third and one-half of these patients have genomic alterations that can be targeted by drugs that are either approved or in clinical development. NGS can also provide information about disease evolution and secondary resistance alterations that can develop during targeted therapy, and thus facilitate assessment of prognosis and choice of alternative targeted treatments. Pathologists play a critical role in assessing the viability of biopsy samples for NGS, and advising treating clinicians whether NGS can be performed and which of the available platforms should be used to optimize testing outcomes. This review aims to provide clinical pathologists and other healthcare professionals with practical step-by-step guidance on the use of NGS for molecular profiling of patients with CCA, with respect to tumor biopsy techniques, pre-analytic sample preparation, selecting the appropriate NGS panel, and understanding and interpreting results of the NGS test.
Collapse
Affiliation(s)
- Albrecht Stenzinger
- Institute of Pathology Heidelberg (IPH), Center for Molecular Pathology, University Hospital Heidelberg, In Neuenheimer Feld 224, 69120 Heidelberg, Building 6224, Germany.
| | - Arndt Vogel
- Division of Gastroenterology and Hepatology, Toronto General Hospital Medical Oncology, Princess Margaret Cancer Centre, Schwartz Reisman Liver Research Centre, 200 Elizabeth Street, Office: 9 EB 236 Toronto, ON, M5G 2C4, Canada.
| | - Ulrich Lehmann
- Institute for Pathology, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany.
| | - Angela Lamarca
- Department of Medical Oncology, Oncohealth Institute, Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Fundación Jiménez Díaz University Hospital, Av. de los Reyes Católicos, 2, 28040 Madrid, Spain; Department of Medical Oncology, The Christie NHS Foundation Trust, Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, FHU OncoAge, IHU RespirERA, Siège de l'Université: Grand Château, 28 Avenue de Valrose, 06103 Nice CEDEX 2, France.
| | - Luigi Terracciano
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, 4, 20072 Pieve Emanuele, Milan, Italy; IRCCS Humanitas Research Hospital, Via Alessandro Manzoni, 56, 20089 Rozzano, Milan, Italy.
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori - IRCCS - Fondazione G. Pascale, Napoli, Italy.
| |
Collapse
|
4
|
Franzi S, Seresini G, Borella P, Raviele PR, Bonitta G, Croci GA, Bareggi C, Tosi D, Nosotti M, Tabano S. Liquid biopsy in non-small cell lung cancer: a meta-analysis of state-of-the-art and future perspectives. Front Genet 2023; 14:1254839. [PMID: 38116291 PMCID: PMC10728669 DOI: 10.3389/fgene.2023.1254839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/20/2023] [Indexed: 12/21/2023] Open
Abstract
Introduction: To date, tissue biopsy represents the gold standard for characterizing non-small-cell lung cancer (NSCLC), however, the complex architecture of the disease has introduced the need for new investigative approaches, such as liquid biopsy. Indeed, DNA analyzed in liquid biopsy is much more representative of tumour heterogeneity. Materials and methods: We performed a meta-analysis of 17 selected papers, to attest to the diagnostic performance of liquid biopsy in identifying EGFR mutations in NSCLC. Results: In the overall studies, we found a sensitivity of 0.59, specificity of 0.96 and diagnostic odds ratio of 24,69. Since we noticed a high heterogeneity among different papers, we also performed the meta-analysis in separate subsets of papers, divided by 1) stage of disease, 2) experimental design and 3) method of mutation detection. Liquid biopsy has the highest sensitivity/specificity in high-stage tumours, and prospective studies are more reliable than retrospective ones in terms of sensitivity and specificity, both NGS and PCR-based techniques can be used to detect tumour DNA in liquid biopsy. Discussion: Overall, liquid biopsy has the potential to help the management of NSCLC, but at present the non-homogeneous literature data, lack of optimal detection methods, together with relatively high costs make its applicability in routine diagnostics still challenging.
Collapse
Affiliation(s)
- Sara Franzi
- Thoracic Surgery and Lung Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Gabriele Seresini
- Laboratory of Medical Genetics, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Paolo Borella
- Thoracic Surgery and Lung Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Gianluca Bonitta
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Giorgio Alberto Croci
- Division of Pathology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Claudia Bareggi
- Medical Oncology Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Davide Tosi
- Thoracic Surgery and Lung Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mario Nosotti
- Thoracic Surgery and Lung Transplantation, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Silvia Tabano
- Laboratory of Medical Genetics, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| |
Collapse
|
5
|
Takano S, Fukasawa M, Enomoto N. Molecular assessment of endoscopically collected pancreatic juice and duodenal fluid from patients with pancreatic diseases. Dig Endosc 2023; 35:19-32. [PMID: 35665966 DOI: 10.1111/den.14371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 06/01/2022] [Indexed: 01/17/2023]
Abstract
One concern associated with pancreatic diseases is the poor prognosis of pancreatic cancer. Even with advances in diagnostic modalities, risk stratification of premalignant lesions and differentiation of pancreatic cysts are challenging. Pancreatic lesions of concern include intraductal papillary mucinous neoplasms, mucinous cystic neoplasms, serous cystadenomas, pseudocysts, and retention cysts, as well as cystic degeneration of solid tumors such as solid pseudopapillary neoplasms and pancreatic neuroendocrine neoplasms. Pancreatic juice obtained during endoscopic retrograde cholangiopancreatography has previously been used for the detection of KRAS mutation. Recently, duodenal fluid, which can be obtained during the relatively minimally invasive procedures of endoscopic ultrasound (EUS) and esophagogastroduodenoscopy, and cyst fluid collected by EUS-guided fine-needle aspiration (FNA) were used for molecular biological analysis. Furthermore, advanced analytic methods with high sensitivity were used for the detection of single and multiple markers. Early detection of malignant pancreatic tumors and risk stratification of premalignant tumors can be performed using duodenal fluid samples with a single marker with high sensitivity. Technological advances in simultaneous detection of multiple markers allow for the differentiation of cystic pancreatic tumors. One thing to note is that the clinical guidelines do not recommend pancreatic cyst fluid and pancreatic juice (PJ) sampling by EUS-FNA and endoscopic retrograde cholangiopancreatography, respectively, in actual clinical practice, but state that they be performed at experienced facilities, and duodenal fluid sampling is not mentioned in the guidelines. With improved specimen handling and the combination of markers, molecular markers in PJ samples may be used in clinical practice in the near future.
Collapse
Affiliation(s)
- Shinichi Takano
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Mitsuharu Fukasawa
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Nobuyuki Enomoto
- First Department of Internal Medicine, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
6
|
Husain A, Mishra S, Hadi R, Sahu A, Kumari S, Rastogi M, Khurana R, Shukla S, Siddiqui MH, Husain N. Dynamics of cell-free DNA in predicting response in adult diffuse glioma on chemoradiotherapy. Cancer Genet 2022; 268-269:55-63. [PMID: 36166960 DOI: 10.1016/j.cancergen.2022.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 06/25/2022] [Accepted: 09/17/2022] [Indexed: 01/25/2023]
Abstract
BACKGROUND Adult diffuse glioma (ADG) is a heterogeneous primary brain tumor with a variable prognosis and treatment response. Tissue biomarkers and molecular genetic profiling form an integral part of diagnosis and prognostication. However, obtaining tissue in inoperable locations and diagnosis of recurrence can be an issue. Cell-free DNA (cfDNA) may help to meet these challenges in the management of ADG. OBJECTIVES The study aimed to serially quantify cfDNA in ADG on chemoradiation and to correlate mutational profiling of the cfDNA with biopsy. MATERIAL AND METHODS The study group comprised of histopathological confirmed ADG (n = 50), including grade II, III and IV glioma, and controls (n = 25). Serum cfDNA was extracted using ChargeSwitch gDNA 1 mL Serum Kit (Invitrogen, USA) and quantified using SYBR based quantitative polymerase chain reaction (qPCR). Next-generation sequencing (NGS) was performed in 07 pre-operative and 05 post-operative cfDNA and tumor biopsy DNA on an Ion personal genome machine (IonPGM) with an in-house designed NGS panel (including TP53, ATRX, and IDH1 and IDH2). RESULTS In patients with ADG, the pre-radiotherapy cfDNA level was significantly higher (Median; 113.46 ng/mL) than normal controls (Median; 74.37 ng/mL), (p = 0.048). Non-responders had significantly higher cfDNA levels (Median; 184.4 ng/mL), than responders (Median; 68.12 ng/mL), (p = 0.023). TP53 gene mutation was most common in both pre-operative and post-operative cfDNA samples. CONCLUSION Pre-radiotherapy cfDNA levels are associated with clinical outcomes independent of other prognostic factors. Targeted NGS in pre-operative cfDNA matches the results of IHC analysis with high concordance, and it may be helpful in inoperable cases or ADG recurrence.
Collapse
Affiliation(s)
- Adil Husain
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India; Department of Biosciences, Integral University, Lucknow, India
| | - Sridhar Mishra
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - Rahat Hadi
- Department of Radiation Oncology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - Avnish Sahu
- Department of Radiation Oncology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - Swati Kumari
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - Madhup Rastogi
- Department of Radiation Oncology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - Rohini Khurana
- Department of Radiation Oncology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | - Saumya Shukla
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| | | | - Nuzhat Husain
- Department of Pathology, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India.
| |
Collapse
|
7
|
Roma C, Sacco A, Forgione L, Esposito Abate R, Lambiase M, Dotolo S, Maiello MR, Frezzetti D, Nasti G, Morabito A, De Luca A, Normanno N. Low Impact of Clonal Hematopoiesis on the Determination of RAS Mutations by Cell-Free DNA Testing in Routine Clinical Diagnostics. Diagnostics (Basel) 2022; 12:diagnostics12081956. [PMID: 36010306 PMCID: PMC9406879 DOI: 10.3390/diagnostics12081956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/04/2022] [Accepted: 08/09/2022] [Indexed: 11/23/2022] Open
Abstract
Targeted sequencing of circulating cell-free DNA (cfDNA) is used in routine clinical diagnostics for the identification of predictive biomarkers in cancer patients in an advanced stage. The presence of KRAS mutations associated with clonal hematopoiesis of indeterminate potential (CHIP) might represent a confounding factor. We used an amplicon-based targeted sequencing panel, covering selected regions of 52 genes, for circulating cell-free total nucleic acid (cfTNA) analysis of 495 plasma samples from cancer patients. The cfDNA test failed in 4 cases, while circulating cell-free RNA (cfRNA) sequencing was invalid in 48 cases. In the 491 samples successfully tested on cfDNA, at least one genomic alteration was found in 222 cases (45.21%). We identified 316 single nucleotide variants (SNVs) in 21 genes. The most frequently mutated gene was TP53 (74 variants), followed by KRAS (71), EGFR (56), PIK3CA (33) and BRAF (19). Copy number variations (CNVs) were detected in 36 cases, while sequencing of cfRNA revealed 6 alterations. Analysis with droplet digital PCR (ddPCR) of peripheral blood leukocyte (PBL)-derived genomic DNA did not identify any KRAS mutations in 39 cases that showed KRAS mutations at cfDNA analysis. These findings suggest that the incidence of CHIP-associated KRAS mutations is relatively rare in routine clinical diagnostics.
Collapse
Affiliation(s)
- Cristin Roma
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Alessandra Sacco
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Laura Forgione
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Matilde Lambiase
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Serena Dotolo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Monica Rosaria Maiello
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Daniela Frezzetti
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Guglielmo Nasti
- SSD Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Alessandro Morabito
- Thoracic Medical Oncology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy
- Correspondence:
| |
Collapse
|
8
|
Gitogenin suppresses lung cancer progression by inducing apoptosis and autophagy initiation through the activation of AMPK signaling. Int Immunopharmacol 2022; 111:108806. [PMID: 35914447 DOI: 10.1016/j.intimp.2022.108806] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 04/18/2022] [Accepted: 04/24/2022] [Indexed: 11/22/2022]
Abstract
Lung cancer is a leading cause of tumor-associated death worldwide. Autophagy plays a key role in regulating lung cancer progression, and is a promising option for lung cancer treatment. Saponins are a group of naturally occurring plant glycosides, characterized by their strong foam-forming properties in aqueous solution, and exert various biological properties, such as anti-inflammation and anti-cancer. In the present study, we for the first time explored the effects of gitogenin (GIT), an important saponin derived from Tribulus longipetalus, on lung cancer progression both in vitro and in vivo. We found that GIT markedly reduced the proliferation and induced apoptosis in lung cancer cells through increasing the cleavage of Caspase-3 and poly (ADP-ribose) polymerases (PARPs). In addition, GIT-incubated lung cancer cells exhibited clear accumulation of autophagosome, which was essential for GIT-suppressed lung cancer. Mechanistically, GIT-induced autophagy initiation was mainly through activating AMP-activated protein kinase (AMPK) and blocking protein kinase B (AKT) signaling pathways, respectively. Moreover, the autophagic flux was disrupted in GIT-treated lung cancer cells, contributing to the accumulation of impaired autophagolysosomes. Importantly, we found that suppressing autophagy initiation could abolish GIT-induced cell death; however, autophagosomes accumulation sensitized lung cancer cells to cell death upon GIT treatment. More in vitro experiments showed that GIT led to reactive oxygen species (ROS) production in lung cancer cells, which was also involved in the modulation of apoptosis. The in vivo findings confirmed the effects of GIT against lung cancer progression with undetectable toxicity to organs. In conclusion, we provided new insights into the treatment of lung cancer, and GIT might be an effective strategy for future clinical application.
Collapse
|
9
|
Stasik S, Mende M, Schuster C, Mahler S, Aust D, Tannapfel A, Reinacher-Schick A, Baretton G, Krippendorf C, Bornhäuser M, Ehninger G, Folprecht G, Thiede C. Sensitive Quantification of Cell-Free Tumor DNA for Early Detection of Recurrence in Colorectal Cancer. Front Genet 2022; 12:811291. [PMID: 35069704 PMCID: PMC8766716 DOI: 10.3389/fgene.2021.811291] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/13/2021] [Indexed: 12/12/2022] Open
Abstract
The detection of plasma cell–free tumor DNA (ctDNA) is prognostic in colorectal cancer (CRC) and has potential for early prediction of disease recurrence. In clinical routine, ctDNA-based diagnostics are limited by the low concentration of ctDNA and error rates of standard next-generation sequencing (NGS) approaches. We evaluated the potential to increase the stability and yield of plasma cell–free DNA (cfDNA) for routine diagnostic purposes using different blood collection tubes and various manual or automated cfDNA extraction protocols. Sensitivity for low-level ctDNA was measured in KRAS-mutant cfDNA using an error-reduced NGS procedure. To test the applicability of rapid evaluation of ctDNA persistence in clinical routine, we prospectively analyzed postoperative samples of 67 CRC (stage II) patients. ctDNA detection was linear between 0.0045 and 45%, with high sensitivity (94%) and specificity (100%) for mutations at 0.1% VAF. The stability and yield of cfDNA were superior when using Streck BCT tubes and a protocol by Zymo Research. Sensitivity for ctDNA increased 1.5-fold by the integration of variant reads from triplicate PCRs and with PCR template concentration. In clinical samples, ctDNA persistence was found in ∼9% of samples, drawn 2 weeks after surgery. Moreover, in a retrospective analysis of 14 CRC patients with relapse during adjuvant therapy, we successfully detected ctDNA (median 0.38% VAF; range 0.18–5.04% VAF) in 92.85% of patients significantly prior (median 112 days) to imaging-based surveillance. Using optimized pre-analytical conditions, the detection of postoperative ctDNA is feasible with excellent sensitivity and allows the prediction of CRC recurrence in routine oncology testing.
Collapse
Affiliation(s)
- Sebastian Stasik
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Heidelberg, Germany
| | - Marika Mende
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | | | - Sandra Mahler
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Daniela Aust
- Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | | | - Anke Reinacher-Schick
- Department of Hematology, Oncology and Palliative Care, St. Josef Hospital, Ruhr University, Bochum, Germany
| | - Gustavo Baretton
- Institute of Pathology, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | | | - Martin Bornhäuser
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany.,National Center for Tumor Diseases (NCT), Partner Site Dresden, Heidelberg, Germany
| | - Gerhard Ehninger
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Gunnar Folprecht
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| | - Christian Thiede
- Medical Department I, University Hospital Carl Gustav Carus, Technische Universität, Dresden, Germany
| |
Collapse
|
10
|
Csoma SL, Bedekovics J, Veres G, Árokszállási A, András C, Méhes G, Mokánszki A. Circulating Cell-Free DNA-Based Comprehensive Molecular Analysis of Biliary Tract Cancers Using Next-Generation Sequencing. Cancers (Basel) 2022; 14:cancers14010233. [PMID: 35008396 PMCID: PMC8750273 DOI: 10.3390/cancers14010233] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 12/21/2021] [Accepted: 01/01/2022] [Indexed: 01/06/2023] Open
Abstract
Biliary tract cancer (BTC) is a rare malignancy with a long disease course and an overall poor prognosis. Despite multiple chemotherapy agents, there is no defined second-line treatment opportunity for advanced BTCs. In the era of precision oncology, NGS plays an important role in identifying mutations that may predict the molecular pathomechanism and manage the BTC therapy. The peripheral blood liquid biopsy (LB) of cancer patients represents variable amounts of cell-free DNA (cfDNA) released from tumor foci of any anatomical location. Our study aimed to identify somatic mutations and tumor variant burden (TVB) in cell-free and matched tumor DNA. We found a positive correlation between the estimated tumor volume and cfDNA yield (r = 0.9326, p < 0.0001). Comparing tissue and LB results, similar TVB was observed. SNVs were proven in 84% of the cases, while in two cases, only the LB sample was informative for molecular analysis. The most important aberrations in BTCs, such as FGFR2, IDH1, IDH2, KRAS, and TP53, could be detected in matched LB samples. Our prospective study demonstrates a minimally invasive testing approach to identify molecular genetic alterations in cholangiocarcinoma and gallbladder cancers. Clinical applications of cfDNA reflect by capturing the outstanding spatial tumor heterogeneity and guarantee novel aspects for the precision oncology treatment.
Collapse
Affiliation(s)
- Szilvia Lilla Csoma
- Department of Pathology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (S.L.C.); (J.B.); (G.M.)
| | - Judit Bedekovics
- Department of Pathology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (S.L.C.); (J.B.); (G.M.)
| | - Gergő Veres
- Division of Radiology and Imaging Science, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Anita Árokszállási
- Department of Oncology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.Á.); (C.A.)
| | - Csilla András
- Department of Oncology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.Á.); (C.A.)
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (S.L.C.); (J.B.); (G.M.)
| | - Attila Mokánszki
- Department of Pathology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (S.L.C.); (J.B.); (G.M.)
- Correspondence: ; Tel.: +36-52-411-600
| |
Collapse
|
11
|
Ris F, Hellan M, Douissard J, Nieva JJ, Triponez F, Woo Y, Geller D, Buchs NC, Buehler L, Moenig S, Iselin CE, Karenovics W, Petignat P, Lam GT, Undurraga Malinervo M, Tuttle R, Ouellette J, Bose D, Ismail N, Toso C. Blood-Based Multi-Cancer Detection Using a Novel Variant Calling Assay (DEEPGEN TM): Early Clinical Results. Cancers (Basel) 2021; 13:4104. [PMID: 34439258 PMCID: PMC8392437 DOI: 10.3390/cancers13164104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 01/22/2023] Open
Abstract
This is an early clinical analysis of the DEEPGENTM platform for cancer detection. Newly diagnosed cancer patients and individuals with no known malignancy were included in a prospective open-label case-controlled study (NCT03517332). Plasma cfDNA that was extracted from peripheral blood was sequenced and data were processed using machine-learning algorithms to derive cancer prediction scores. A total of 260 cancer patients and 415 controls were included in the study. Overall, sensitivity for all cancers was 57% (95% CI: 52, 64) at 95% specificity, and 43% (95% CI: 37, 49) at 99% specificity. With 51% sensitivity and 95% specificity for all stage 1 cancers, the stage-specific sensitivities trended to improve with higher stages. Early results from this preliminary clinical, prospective evaluation of the DEEPGENTM liquid biopsy platform suggests the platform offers a clinically relevant ability to differentiate individuals with and without known cancer, even at early stages of cancer.
Collapse
Affiliation(s)
- Frederic Ris
- Division of Visceral Surgery, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (J.D.); (N.C.B.); (S.M.); (N.I.); (C.T.)
| | - Minia Hellan
- Surgical Oncology, Wright State University, Dayton, OH 45435, USA; (M.H.); (R.T.); (J.O.)
| | - Jonathan Douissard
- Division of Visceral Surgery, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (J.D.); (N.C.B.); (S.M.); (N.I.); (C.T.)
| | - Jorge J. Nieva
- Norris Cancer Center, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA 90033, USA;
| | - Frederic Triponez
- Division of Thoracic Surgery, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (F.T.); (W.K.)
| | - Yanghee Woo
- Division of Surgical Oncology, Department of Surgery and Cancer Immunotherapeutics Program, City of Hope, Duarte, CA 91010, USA;
| | - David Geller
- Department of Surgery, University of Pittsburgh, Pittsburg, PA 15260, USA;
| | - Nicolas C. Buchs
- Division of Visceral Surgery, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (J.D.); (N.C.B.); (S.M.); (N.I.); (C.T.)
| | - Leo Buehler
- Division of Visceral Surgery, Department of Surgery, University Hospital Fribourg, 1700 Fribourg, Switzerland;
| | - Stefan Moenig
- Division of Visceral Surgery, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (J.D.); (N.C.B.); (S.M.); (N.I.); (C.T.)
| | - Christophe E. Iselin
- Division of Urology, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland;
| | - Wolfram Karenovics
- Division of Thoracic Surgery, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (F.T.); (W.K.)
| | - Patrick Petignat
- Divison of Gynecology, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (P.P.); (G.T.L.); (M.U.M.)
| | - Giang Thanh Lam
- Divison of Gynecology, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (P.P.); (G.T.L.); (M.U.M.)
| | - Manuela Undurraga Malinervo
- Divison of Gynecology, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (P.P.); (G.T.L.); (M.U.M.)
| | - Rebecca Tuttle
- Surgical Oncology, Wright State University, Dayton, OH 45435, USA; (M.H.); (R.T.); (J.O.)
| | - James Ouellette
- Surgical Oncology, Wright State University, Dayton, OH 45435, USA; (M.H.); (R.T.); (J.O.)
| | - Debashish Bose
- The Center for Hepatobiliary Disease, Mercy, Baltimore, MD 21202, USA;
| | - Nael Ismail
- Division of Visceral Surgery, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (J.D.); (N.C.B.); (S.M.); (N.I.); (C.T.)
| | - Christian Toso
- Division of Visceral Surgery, Department of Surgery, University Hospital Geneva and Medical School, 1211 Geneva, Switzerland; (J.D.); (N.C.B.); (S.M.); (N.I.); (C.T.)
| |
Collapse
|
12
|
Underhill HR. Leveraging the Fragment Length of Circulating Tumour DNA to Improve Molecular Profiling of Solid Tumour Malignancies with Next-Generation Sequencing: A Pathway to Advanced Non-invasive Diagnostics in Precision Oncology? Mol Diagn Ther 2021; 25:389-408. [PMID: 34018157 PMCID: PMC8249304 DOI: 10.1007/s40291-021-00534-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2021] [Indexed: 12/20/2022]
Abstract
Circulating cell-free DNA (ccfDNA) has emerged as a promising diagnostic tool in oncology. Identification of tumour-derived ccfDNA (i.e. circulating tumour DNA [ctDNA]) provides non-invasive access to a malignancy’s molecular landscape to diagnose, inform therapeutic strategies, and monitor treatment efficacy. Current applications of ccfDNA to detect somatic mutations, however, have been largely constrained to tumour-informed searches and identification of common mutations because of the interaction between ctDNA signal and next-generation sequencing (NGS) noise. Specifically, the low allele frequency of ctDNA associated with non-metastatic and early-stage lesions may be indistinguishable from artifacts that accrue during sample preparation and NGS. Thus, using ccfDNA to achieve non-invasive and personalized molecular profiling to optimize individual patient care is a highly sought goal that remains limited in clinical practice. There is growing evidence, however, that further advances in the field of ccfDNA diagnostics may be achieved by improving detection of somatic mutations through leveraging the inherently shorter fragment lengths of ctDNA compared to non-neoplastic ccfDNA. Here, the origins and rationale for seeking to improve the mutation-based detection of ctDNA by using ccfDNA size profiling are reviewed. Subsequently, in vitro and in silico methods to enrich for a target ccfDNA fragment length are detailed to identify current practices and provide perspective into the potential of using ccfDNA size profiling to impact clinical applications in oncology.
Collapse
Affiliation(s)
- Hunter R Underhill
- Division of Medical Genetics, Department of Pediatrics, University of Utah, 295 Chipeta Way, Salt Lake City, UT, 84108, USA. .,Department of Radiology, University of Utah, Salt Lake City, UT, USA. .,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
13
|
Bekaii-Saab TS, Bridgewater J, Normanno N. Practical considerations in screening for genetic alterations in cholangiocarcinoma. Ann Oncol 2021; 32:1111-1126. [PMID: 33932504 DOI: 10.1016/j.annonc.2021.04.012] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 04/06/2021] [Accepted: 04/18/2021] [Indexed: 12/14/2022] Open
Abstract
Cholangiocarcinoma (CCA) encompasses diverse epithelial tumors historically associated with poor outcomes due to an aggressive disease course, late diagnosis, and limited benefit of standard chemotherapy for advanced disease. Comprehensive molecular profiling has revealed a diverse landscape of genomic alterations as oncogenic drivers in CCA. TP53 mutations, CDKN2A/B loss, and KRAS mutations are the most common genetic alterations in CCA. However, intrahepatic CCA (iCCA) and extrahepatic CCA (eCCA) differ substantially in the frequency of many alterations. This includes actionable alterations, such as isocitrate dehydrogenase 1 (IDH1) mutations and a large variety of FGFR2 rearrangements, which are found in up to 29% and ∼10% of patients with iCCA, respectively, but are rare in eCCA. FGFR2 rearrangements are currently the only genetic alteration in CCA for which a targeted therapy, the fibroblast growth factor receptor 1-3 inhibitor pemigatinib, has been approved. However, favorable phase III results for IDH1-targeted therapy with ivosidenib in iCCA have been published, and numerous other alterations are actionable by targeted therapies approved in other indications. Recent advances in next-generation sequencing (NGS) have led to the development of assays that allow comprehensive genomic profiling of large gene panels within 2-3 weeks, including in vitro diagnostic tests approved in the United States. These assays vary regarding acceptable source material (tumor tissue or peripheral whole blood), genetic source for library construction (DNA or RNA), target selection technology, gene panel size, and type of detectable genomic alterations. While some large commercial laboratories offer rapid and comprehensive genomic profiling services based on proprietary assay platforms, clinical centers may use commercial genomic profiling kits designed for clinical research to develop their own customized laboratory-developed tests. Large-scale genomic profiling based on NGS allows for a detailed and precise molecular diagnosis of CCA and provides an important opportunity for improved targeted treatment plans tailored to the individual patient's genetic signature.
Collapse
Affiliation(s)
| | - J Bridgewater
- University College London Cancer Institute, London, UK
| | - N Normanno
- Istituto Nazionale Tumori 'Fondazione Giovanni Pascale' IRCCS, Naples, Italy
| |
Collapse
|
14
|
Ye P, Cai P, Xie J, Wei Y. The diagnostic accuracy of digital PCR, ARMS and NGS for detecting KRAS mutation in cell-free DNA of patients with colorectal cancer: A systematic review and meta-analysis. PLoS One 2021; 16:e0248775. [PMID: 33770081 PMCID: PMC7997033 DOI: 10.1371/journal.pone.0248775] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 03/04/2021] [Indexed: 02/06/2023] Open
Abstract
Introduction Before anti-EGFR therapy is given to patients with colorectal cancer, it is required to determine KRAS mutation status in tumor. When tumor tissue is not available, cell-free DNA (liquid biopsy) is commonly used as an alternative. Due to the low abundance of tumor-derived DNA in cell-free DNA samples, methods with high sensitivity were preferred, including digital polymerase chain reaction, amplification refractory mutation system and next-generation sequencing. The aim of this systemic review and meta-analysis was to investigate the accuracy of those methods in detecting KRAS mutation in cell-free DNA sample from patients with colorectal cancer. Methods Literature search was performed in Pubmed, Embase, and Cochrane Library. After removing duplicates from the 170 publications found by literature search, eligible studies were identified using pre-defined criteria. Quality of the publications and relevant data were assessed and extracted thereafter. Meta-DiSc and STATA softwares were used to pool the accuracy parameters from the extracted data. Results A total of 33 eligible studies were identified for this systemic review and meta-analysis. After pooling, the overall sensitivity, specificity, and diagnostic odds ratio were 0.77 (95%CI: 0.74–0.79), 0.87 (95%CI: 0.85–0.89), and 23.96 (95%CI: 13.72–41.84), respectively. The overall positive and negative likelihood ratios were 5.55 (95%CI: 3.76–8.19) and 0.29 (95%CI: 0.21–0.38), respectively. Area under curve of the summarized ROC curve was 0.8992. Conclusion Digital polymerase chain reaction, amplification refractory mutation system, and next-generation sequencing had overall high accuracy in detecting KRAS mutation in cell-free DNA sample. Large prospective randomized clinical trials are needed to further convince the accuracy and usefulness of KRAS mutation detection using cfDNA/liquid biopsy samples in clinical practice. Trial registration PROSPERO CRD42020176682; https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=176682.
Collapse
Affiliation(s)
- Peng Ye
- Department of Anatomy and Histology, School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan Province, People’s Republic of China
- * E-mail: (PY); (YW)
| | - Peiling Cai
- Department of Anatomy and Histology, School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan Province, People’s Republic of China
| | - Jing Xie
- Department of Pathology and Clinical Laboratory, Sichuan Provincial Fourth People’s Hospital, Chengdu, Sichuan Province, People’s Republic of China
| | - Yuanyuan Wei
- Department of Physiology, School of Preclinical Medicine, Chengdu University, Chengdu, Sichuan Province, People’s Republic of China
- * E-mail: (PY); (YW)
| |
Collapse
|
15
|
Takano S, Fukasawa M, Shindo H, Takahashi E, Fukasawa Y, Kawakami S, Hayakawa H, Kuratomi N, Kadokura M, Maekawa S, Enomoto N. Digital next-generation sequencing of cell-free DNA for pancreatic cancer. JGH OPEN 2021; 5:508-516. [PMID: 33860102 PMCID: PMC8035455 DOI: 10.1002/jgh3.12530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/28/2021] [Accepted: 03/08/2021] [Indexed: 12/26/2022]
Abstract
Background and Aim The clinical applicability of digital next‐generation sequencing (dNGS), which eliminates polymerase chain reaction (PCR) and sequencing error‐derived noise by using molecular barcodes (MBs), has not been fully evaluated. We evaluated the utility of dNGS of cell‐free DNA (cfDNA) in liquid biopsies obtained from patients with pancreatic cancer. Methods Fifty‐eight patients with pancreatic cancer undergoing endoscopic ultrasound‐guided fine‐needle aspiration (EUS‐FNA) were included. Samples were subjected to sequencing of 50 cancer‐related genes using next‐generation sequencing (NGS). The results were used as reference gene alterations. NGS of cfDNA from plasma was performed for patients with a mutant allele frequency (MAF) >1% and an absolute mutant number > 10 copies/plasma mL in KRAS or GNAS by digital PCR. Sequence readings with and without MBs were compared with reference to EUS‐FNA‐derived gene alterations. Results The concordance rate between dNGS of cfDNA and EUS‐FNA‐derived gene alterations was higher with than without MBs (p = 0.039), and MAF cut‐off values in dNGS could be decreased to 0.2%. dNGS using MBs eliminated PCR and sequencing error by 74% and 68% for TP53 and all genes, respectively. Overall, dNGS detected mutations in KRAS (45%) and TP53 (26%) and copy number alterations in CCND2, CCND3, CDK4, FGFR1, and MYC, which are targets of molecular‐targeted drugs. Conclusions dNGS of cfDNA using MBs is useful for accurate detection of gene alterations even with low levels of MAFs. These results may be used to inform the development of diagnostics and therapeutics that can improve the prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Shinichi Takano
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Mitsuharu Fukasawa
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Hiroko Shindo
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Ei Takahashi
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Yoshimitsu Fukasawa
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Satoshi Kawakami
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Hiroshi Hayakawa
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Natsuhiko Kuratomi
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Makoto Kadokura
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Shinya Maekawa
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| | - Nobuyuki Enomoto
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Chuo Japan
| |
Collapse
|
16
|
Huang ZB, Zhang HT, Yu B, Yu DH. Cell-free DNA as a liquid biopsy for early detection of gastric cancer. Oncol Lett 2021; 21:3. [PMID: 33240409 PMCID: PMC7681206 DOI: 10.3892/ol.2020.12264] [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: 04/17/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most common malignant tumors with poor prognosis worldwide, mainly due to the lack of suitable modalities for population-based screening and early detection of this disease. Therefore, novel and less invasive tests with improved clinical utility are urgently required. The remarkable advances in genomics and proteomics, along with emerging new technologies for highly sensitive detection of genetic alterations, have shown the potential to map the genomic makeup of a tumor in liquid biopsies, in order to assist with early detection and clinical management. The present review summarize the current status in the identification and development of cell-free DNA (cfDNA)-based biomarkers in GC, and also discusses their potential utility and the technical challenges in developing practical cfDNA-based liquid biopsy for early detection of GC.
Collapse
Affiliation(s)
- Zheng-Bin Huang
- Department of Surgery, Hanchuan Renmin Hospital, Hanchuan, Hubei 431600, P.R. China
| | - Hai-Tao Zhang
- Department of Gastrointestinal Surgery, The Second People's Hospital of Shenzhen, Shenzhen, Guangdong 518037, P.R. China
| | - Benjamin Yu
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - De-Hua Yu
- Shenzhen USK Bioscience Co., Ltd., Shenzhen, Guangdong 518110, P.R. China
| |
Collapse
|
17
|
Esposito Abate R, Frezzetti D, Maiello MR, Gallo M, Camerlingo R, De Luca A, De Cecio R, Morabito A, Normanno N. Next Generation Sequencing-Based Profiling of Cell Free DNA in Patients with Advanced Non-Small Cell Lung Cancer: Advantages and Pitfalls. Cancers (Basel) 2020; 12:E3804. [PMID: 33348595 PMCID: PMC7766403 DOI: 10.3390/cancers12123804] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022] Open
Abstract
Lung cancer (LC) is the main cause of death for cancer worldwide and non-small cell lung cancer (NSCLC) represents the most common histology. The discovery of genomic alterations in driver genes that offer the possibility of therapeutic intervention has completely changed the approach to the diagnosis and therapy of advanced NSCLC patients, and tumor molecular profiling has become mandatory for the choice of the most appropriate therapeutic strategy. However, in approximately 30% of NSCLC patients tumor tissue is inadequate for biomarker analysis. The development of highly sensitive next generation sequencing (NGS) technologies for the analysis of circulating cell-free DNA (cfDNA) is emerging as a valuable alternative to assess tumor molecular landscape in case of tissue unavailability. Additionally, cfDNA NGS testing can better recapitulate NSCLC heterogeneity as compared with tissue testing. In this review we describe the main advantages and limits of using NGS-based cfDNA analysis to guide the therapeutic decision-making process in advanced NSCLC patients, to monitor the response to therapy and to identify mechanisms of resistance early. Therefore, we provide evidence that the implementation of cfDNA NGS testing in clinical research and in the clinical practice can significantly improve precision medicine approaches in patients with advanced NSCLC.
Collapse
Affiliation(s)
- Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Daniela Frezzetti
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Monica Rosaria Maiello
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Marianna Gallo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Rosa Camerlingo
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Antonella De Luca
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| | - Rossella De Cecio
- Department of Pathology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy;
| | - Alessandro Morabito
- Department of Thoracic Medical Oncology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy;
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (R.E.A.); (D.F.); (M.R.M.); (M.G.); (R.C.); (A.D.L.)
| |
Collapse
|
18
|
Sebastião MM, Ho RS, de Carvalho JPV, Nussbaum M. Diagnostic Accuracy of Next Generation Sequencing Panel using Circulating Tumor DNA in Patients with Advanced Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. JOURNAL OF HEALTH ECONOMICS AND OUTCOMES RESEARCH 2020; 7:158-163. [PMID: 33043062 PMCID: PMC7539761 DOI: 10.36469/jheor.2020.17088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND/OBJECTIVES Until now, no meta-analysis has been published to evaluate the diagnostic performance of next-generation sequencing (NGS) panel using circulating tumor (ctDNA) in patients with advanced non-small cell lung cancer (aNSCLC). The aim of the study was to carry out a systematic review and a meta-analysis in order to determine the accuracy of NGS of ctDNA to detect six oncogenic driver alterations: epidermal growth factor receptor (EGFR); anaplastic lymphoma kinase (ALK); ROS proto-oncogene 1, receptor tyrosine kinase (ROS-1); serine/threonine-protein kinase B-RAF (BRAF); RET proto-oncogene (RET); and MET proto-oncogene, receptor tyrosine kinase (MET) exon 14 in patients with aNSCLC. METHODS MEDLINE/PubMed, Cochrane Library, Latin American and Caribbean Health Sciences Literature (LILACS), and Centre for Reviews and Dissemination databases and articles obtained from other sources were searched for relevant studies that evaluate the accuracy (sensitivity and specificity) of NGS using ctDNA in patients with aNSCLC. The studies were eligible when NGS of ctDNA was compared with tissue tests to detect at least one of the six oncogenic driver alterations. Diagnostic measures (sensitivity and specificity) were pooled with a bivariate diagnostic random effect. All statistical analyses were performed with software R, v.4.0.0. RESULTS Ten studies were eligible for data extraction. The overall pooled estimates of sensitivity and specificity were 0.766 (95% CI: 0.678-0.835); 0.999 (95% CI: 0.990-1.000), respectively. CONCLUSIONS The analysis has demonstrated that the NGS panel using ctDNA has a high accuracy to identify the six actionable oncogenic driver alterations in patients with aNSCLC. Therefore, it can be considered a reliable alternative to guide the patients with aNSCLC to the right treatment who cannot undergo an invasive procedure or have insufficient tissue material for molecular tests.
Collapse
|
19
|
Clinical biomarkers directing the management of patients with colon and lung cancer (beyond oncogene-addicted NSCLC). FORUM OF CLINICAL ONCOLOGY 2020. [DOI: 10.2478/fco-2019-0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Abstract
Treatment personalisation plays a key role in the current management of patients with cancer. Several biomarkers have shown clinical utility and may guide therapeutic decisions. Amongst patients with lung cancer, the level of expression of programmed death ligand 1 (PD-L1) has both prognostic and predictive values in terms of response to the inhibition of programmed cell death protein 1 (PD-1). Depending on the clinical setting, the expression of PD-L1 ≥1% or ≥50% has been associated with improved outcomes amongst patients receiving pembrolizumab. Regarding patients with colorectal carcinoma, mutations in the KRAS oncogene predict the responsiveness to the inhibition of epidermal growth factor receptor (EGFR). Only patients with wild-type KRAS tumours derive benefit from cetuximab and panitumumab in terms of response and survival. In conclusion, future research should aim in the optimisation of the use of biomarker in the clinical practice in order to provide the optimal drug combination to each individual patient.
Collapse
|
20
|
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
|
21
|
Chang WJ, Sung JS, Lee SY, Kang EJ, Kwon NJ, Kim HM, Shin SW, Choi JY, Choi YJ, Kim JW, Park KH, Kim YH. The Clinical Significance of RAS, PIK3CA, and PTEN Mutations in Non-Small Cell Lung Cancer Using Cell-Free DNA. J Clin Med 2020; 9:jcm9082642. [PMID: 32823871 PMCID: PMC7465200 DOI: 10.3390/jcm9082642] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/22/2022] Open
Abstract
Mutations in the EGFR gene downstream signaling pathways may cause receptor-independent pathway activation, making tumors unresponsive to EGFR inhibitors. However, the clinical significance of RAS, PIK3CA or PTEN mutations in NSCLC is unclear. In this study, patients who were initially diagnosed with NSCLC or experienced recurrence after surgical resection were enrolled, and blood samples was collected. Ultra-deep sequencing analysis of cfDNA using Ion AmpliSeq Cancer Hotspot Panel v2 with Proton platforms was conducted. RAS/PIK3CA/PTEN mutations were frequently detected in cfDNA in stage IV NSCLC (58.1%), and a high proportion of the patients (47.8%) with mutations had bone metastases at diagnosis. The frequency of RAS/PIK3CA/PTEN mutations in patients with activating EGFR mutation was 61.7%. The median PFS for EGFR-TKIs was 15.1 months in patients without RAS/PIK3CA/PTEN mutations, and 19.9 months in patients with mutations (p = 0.549). For patients with activating EGFR mutations, the overall survival was longer in patients without RAS/PIK3CA/PTEN mutations (53.8 months vs. 27.4 months). For the multivariate analysis, RAS/PIK3CA/PTEN mutations were independent predictors of poor prognosis in patients with activating EGFR mutations. In conclusion, RAS, PIK3CA and PTEN mutations do not hamper EGFR-TKI treatment outcome; however, they predict a poor OS when activating EGFR mutations coexist.
Collapse
Affiliation(s)
- Won Jin Chang
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (W.J.C.); (S.W.S.); (J.Y.C.); (Y.J.C.); (J.W.K.); (K.H.P.)
| | - Jae Sook Sung
- Cancer Research Institute, Korea University, Seoul 02841, Korea;
| | - Sung Yong Lee
- Department of Internal Medicine, Korea University Guro Hospital, Seoul 08308, Korea; (S.Y.L.); (E.J.K.)
| | - Eun Joo Kang
- Department of Internal Medicine, Korea University Guro Hospital, Seoul 08308, Korea; (S.Y.L.); (E.J.K.)
| | - Nak-Jung Kwon
- Macrogen, 254, Beotkkot-ro, Geumcheon-gu, Seoul 08511, Korea; (N.-J.K.); (H.M.K.)
| | - Hae Mi Kim
- Macrogen, 254, Beotkkot-ro, Geumcheon-gu, Seoul 08511, Korea; (N.-J.K.); (H.M.K.)
| | - Sang Won Shin
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (W.J.C.); (S.W.S.); (J.Y.C.); (Y.J.C.); (J.W.K.); (K.H.P.)
| | - Jung Yoon Choi
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (W.J.C.); (S.W.S.); (J.Y.C.); (Y.J.C.); (J.W.K.); (K.H.P.)
| | - Yoon Ji Choi
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (W.J.C.); (S.W.S.); (J.Y.C.); (Y.J.C.); (J.W.K.); (K.H.P.)
| | - Ju Won Kim
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (W.J.C.); (S.W.S.); (J.Y.C.); (Y.J.C.); (J.W.K.); (K.H.P.)
| | - Kyong Hwa Park
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (W.J.C.); (S.W.S.); (J.Y.C.); (Y.J.C.); (J.W.K.); (K.H.P.)
- Cancer Research Institute, Korea University, Seoul 02841, Korea;
| | - Yeul Hong Kim
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Anam Hospital, Korea University College of Medicine, Seoul 02841, Korea; (W.J.C.); (S.W.S.); (J.Y.C.); (Y.J.C.); (J.W.K.); (K.H.P.)
- Cancer Research Institute, Korea University, Seoul 02841, Korea;
- Correspondence: ; Tel.: +82-2-920-5569; Fax: +82-2-920-6622
| |
Collapse
|
22
|
García-Olmo DC, Peiró-Pastor R, Picazo MG, Olmedillas-López S, García-Arranz M, Aguado B, García-Olmo D. Liquid biopsy by NGS: Differential presence of exons (DPE) is related to metastatic potential in a colon-cancer model in the rat. Transl Oncol 2020; 13:100837. [PMID: 32736333 PMCID: PMC7393324 DOI: 10.1016/j.tranon.2020.100837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/09/2020] [Accepted: 07/16/2020] [Indexed: 11/17/2022] Open
Abstract
Differential presence of exons (DPE) is a method of interpretation of exome sequencing, which has been proposed to design a predictive algorithm with clinical value in patients with colorectal cancer (CRC). The goal of the present study was to examine the reproducibility in a rat model of metastatic colon cancer. DHD/K12-TRb cells were injected in syngenic immunocompetent BD-IX rats. Cells were from two stocks with low and normal metastatic potential, and injected into two separate groups of rats. Five to ten weeks after injection, blood samples were taken prior euthanasia and whole exome sequencing performed. Through DPE analysis, we identified a set of exons whose differential presence in plasma allowed us to compare both groups of tumor-bearing animals. A verification test was performed to confirm that the algorithm was able to classify extracted samples into their corresponding groups of origin. The highest mean probability was 0.8954. In conclusion, the DPE analysis in tumor-bearing animals was able to discriminate between different disease status, which fully supports previous results in CRC patients.
Collapse
Affiliation(s)
- Dolores C García-Olmo
- Centre de Recerca Experimental Biomédica Aplicada (CREBA), IRBLleida, Lleida, Spain; Experimental Research Unit, University Hospital of Albacete, Spain
| | - Ramón Peiró-Pastor
- Genomics and NGS Service, Centro de Biología Molecular Severo Ochoa (CBMSO), CSIC-UAM, Madrid, Spain
| | - María G Picazo
- Experimental Research Unit, University Hospital of Albacete, Spain
| | - Susana Olmedillas-López
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid, Spain
| | - Mariano García-Arranz
- New Therapies Laboratory, Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid, Spain
| | - Begoña Aguado
- Genomics and NGS Service, Centro de Biología Molecular Severo Ochoa (CBMSO), CSIC-UAM, Madrid, Spain
| | - Damián García-Olmo
- Department of Surgery, Fundación Jiménez Díaz University Hospital, Madrid, Spain.
| |
Collapse
|
23
|
Esagian SM, Grigoriadou GΙ, Nikas IP, Boikou V, Sadow PM, Won JK, Economopoulos KP. Comparison of liquid-based to tissue-based biopsy analysis by targeted next generation sequencing in advanced non-small cell lung cancer: a comprehensive systematic review. J Cancer Res Clin Oncol 2020; 146:2051-2066. [PMID: 32462295 PMCID: PMC7456570 DOI: 10.1007/s00432-020-03267-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/14/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE To explore whether targeted next generation sequencing (NGS) of liquid biopsy in advanced non-small cell lung cancer (NSCLC) could potentially overcome the innate problems that arise with standard tissue biopsy, like intratumoral heterogeneity and the inability to obtain adequate samples for analysis. METHODS The Scopus, Cochrane Library, and MEDLINE (via PubMed) databases were searched for studies with matched tissue and liquid biopsies from advanced NSCLC patients, analyzed with targeted NGS. The number of mutations detected in tissue biopsy only, liquid biopsy only, or both was assessed and the positive percent agreement (PPA) of the two methods was calculated for every clinically relevant gene. RESULTS A total of 644 unique relevant articles were retrieved and data were extracted from 38 studies fulfilling the inclusion criteria. The sample size was composed of 2000 mutations tested in matched tissue and liquid biopsies derived from 1141 patients. No studies analyzed circulating tumor cells. The calculated PPA rates were 53.6% (45/84) for ALK, 53.9% (14/26) for BRAF, 56.5% (13/23) for ERBB2, 67.8% (428/631) for EGFR, 64.2% (122/190) for KRAS, 58.6% (17/29) for MET, 54.6% (12/22) for RET, and 53.3% (8/15) for ROS1. We additionally recorded data for 65 genes that are not recommended by current guidelines for mutational testing. An extra category containing results of unspecified genes was added, with a PPA rate of 55.7% (122/219). CONCLUSION Despite many advantages, liquid biopsy might be unable to fully substitute its tissue counterpart in detecting clinically relevant mutations in advanced NSCLC patients. However, it may serve as a helpful tool when making therapeutic decisions. More studies are needed to evaluate its role in everyday clinical practice.
Collapse
Affiliation(s)
- Stepan M Esagian
- Oncology Working Group, Society of Junior Doctors, Athens, Greece
| | - Georgia Ι Grigoriadou
- Oncology Working Group, Society of Junior Doctors, Athens, Greece
- 1st Department of Medical Oncology, Theageneio Anticancer Hospital, Thessaloníki, Greece
| | - Ilias P Nikas
- School of Medicine, European University of Cyprus, Nicosia, Cyprus
| | - Vasileios Boikou
- Oncology Working Group, Society of Junior Doctors, Athens, Greece
- Athens University of Economics and Business, Athens, Greece
| | - Peter M Sadow
- Department of Pathology, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Konstantinos P Economopoulos
- Oncology Working Group, Society of Junior Doctors, Athens, Greece.
- Department of Surgery, Duke University Medical Center, 2301 Erwin Rd, Durham, NC, 27710, USA.
| |
Collapse
|
24
|
Ishida Y, Takano S, Maekawa S, Yamaguchi T, Yoshida T, Kobayashi S, Iwamoto F, Kuno T, Hayakawa H, Matsuda S, Fukasawa M, Shindo H, Inoue T, Nakayama Y, Ichikawa D, Sato T, Enomoto N. Fractionated small cell-free DNA increases possibility to detect cancer-related gene mutations in advanced colorectal cancer. JGH OPEN 2020; 4:978-986. [PMID: 33102773 PMCID: PMC7578331 DOI: 10.1002/jgh3.12379] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/11/2020] [Indexed: 12/19/2022]
Abstract
Background and Aim Liquid biopsy is a method that can efficiently detect tumor genetic abnormalities from body fluids such as blood and urine. Detection sensitivity and the available number of mutations in cell-free DNA (cfDNA) are limited. In this study, we develop a highly sensitive and comprehensive method to detect mutations from cfDNA by concentrating tumor fractions of small cfDNA in advanced colorectal cancers. Methods Biopsied specimens and 37 serum samples were collected from 27 patients with advanced colorectal carcinoma. A serum-extracted cfDNA was divided into enriched fractionated small cfDNA and unfractionated cfDNA. Both cfDNAs were subjected to digital polymerase chain reaction (PCR) to evaluate their KRAS, BRAF, CDKN2A, and TP53 status. Consequently, their mutant allele frequencies (MAFs) were compared and analyzed by next-generation sequencing (NGS) in conjunction with tissue-derived DNA. Results NGS analyses revealed mutations in TP53 (63%), KRAS (63%), APC (30%), and PIK3CA (22%). Digital PCR could detect mutations in 25 of 27 samples (93%) of unfractionated cfDNA, a rate that increased to 100% when samples were enriched with fractionated small cfDNA (6.8 vs 10.7%, P < 0.001). NGS also showed increased MAFs in fractionated small cfDNA compared to unfractionated cfDNA (16.3 vs 18.8%, P = 0.012) and a tendency to detect a greater number of cancer-related genes in fractionated cfDNA. Conclusions Fractionated small cfDNA increased MAFs of gene mutations and increases the possibilities to detect cancer-related genes even in advanced cancer patients from whom it is difficult to obtain tissue samples.
Collapse
Affiliation(s)
- Yasuaki Ishida
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Shinichi Takano
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Shinya Maekawa
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Tatsuya Yamaguchi
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Takashi Yoshida
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Shoji Kobayashi
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Fumihiko Iwamoto
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Toru Kuno
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Hiroshi Hayakawa
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Shuya Matsuda
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Mitsuharu Fukasawa
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Hiroko Shindo
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Taisuke Inoue
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Yasuhiro Nakayama
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Daisuke Ichikawa
- First Department of Surgery, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Tadashi Sato
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| | - Nobuyuki Enomoto
- First Department of Internal Medicine, Faculty of Medicine University of Yamanashi Yamanashi Japan
| |
Collapse
|
25
|
Allegretti M, Cottone G, Carboni F, Cotroneo E, Casini B, Giordani E, Amoreo CA, Buglioni S, Diodoro M, Pescarmona E, Zazza S, Federici O, Zeuli M, Conti L, Cigliana G, Fiorentino F, Valle M, Giacomini P, Spinella F. Cross-sectional analysis of circulating tumor DNA in primary colorectal cancer at surgery and during post-surgery follow-up by liquid biopsy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2020; 39:69. [PMID: 32312295 PMCID: PMC7168847 DOI: 10.1186/s13046-020-01569-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Liquid biopsy (LB) in early-stage, non-metastatic colorectal cancer (CRC) must be sensitive enough to detect extremely low circulating tumor DNA (ctDNA) levels. This challenge has been seldom and non-systematically investigated. METHODS Next generation sequencing (NGS) and digital PCR (dPCR) were combined to test tumor DNAs (tDNAs) and paired ctDNAs collected at surgery from 39 patients, 12 of whom were also monitored during the immediate post-surgery follow up. Patients treated for metastatic disease (n = 14) were included as controls. RESULTS NGS and dPCR concordantly (100% agreement) called at least one single nucleotide variant (SNV) in 34 tDNAs, estimated differences in allelic frequencies being negligible (±1.4%). However, despite dPCR testing, SNVs were only detectable in 15/34 (44.1%) ctDNAs from patients at surgery, as opposed to 14/14 (100%) metastatic patients. This was likely due to striking differences (average 10 times, up to 500) in ctDNA levels between groups. NGS revealed blood-only SNVs, suggesting spatial heterogeneity since pre-surgery disease stages, and raising the combined NGS/dPCR sensitivity to 58.8%. ctDNA levels at surgery correlated with neither tumor size, stage, grade, or nodal status, nor with variant abundance in paired tDNA. LB sensitivity reached 63.6% when ctDNA was combined with CEA. Finally, persistence and absence of ctDNA on the first conventional (month 3) post-surgery follow-up were associated with fast relapse and a disease-free status in 3 and 7 patients, respectively. CONCLUSIONS A simple clinical NGS/dPCR/CEA combination effectively addresses the LB challenge in a fraction of non-metastatic CRC patients.
Collapse
Affiliation(s)
- Matteo Allegretti
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Giuliano Cottone
- Oncogenomics Division, Eurofins Genoma Group, Via Castel Giubileo, 11, 00138, Rome, Italy
| | - Fabio Carboni
- Digestive Surgery, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Ettore Cotroneo
- Oncogenomics Division, Eurofins Genoma Group, Via Castel Giubileo, 11, 00138, Rome, Italy
| | - Beatrice Casini
- Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Elena Giordani
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | | | | | - Maria Diodoro
- Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Settimio Zazza
- Digestive Surgery, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Orietta Federici
- Digestive Surgery, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Massimo Zeuli
- Medical Oncology 1, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Conti
- Clinical Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Cigliana
- Clinical Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Francesco Fiorentino
- Oncogenomics Division, Eurofins Genoma Group, Via Castel Giubileo, 11, 00138, Rome, Italy
| | - Mario Valle
- Digestive Surgery, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Patrizio Giacomini
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy.
| | - Francesca Spinella
- Oncogenomics Division, Eurofins Genoma Group, Via Castel Giubileo, 11, 00138, Rome, Italy.
| |
Collapse
|
26
|
Pasquale R, Forgione L, Roma C, Fenizia F, Bergantino F, Rachiglio AM, De Luca A, Gallo M, Maiello MR, Palumbo G, Morabito A, Azzaro R, Normanno N. Targeted sequencing analysis of cell-free DNA from metastatic non-small-cell lung cancer patients: clinical and biological implications. Transl Lung Cancer Res 2020; 9:61-70. [PMID: 32206554 PMCID: PMC7082281 DOI: 10.21037/tlcr.2020.01.01] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 10/12/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Sequencing artifacts, clonal hematopoietic mutations of indeterminate potential (CHIP) and tumor heterogeneity have been hypothesized to contribute to the low concordance between tissue and cell-free DNA (cfDNA) molecular profiling with targeted sequencing. METHODS We analyzed by targeted sequencing cfDNA from 30 healthy individuals, and cfDNA and matched tumor samples from 30 EGFR-mutant and 77 EGFR wild-type metastatic non-small-cell lung cancer (mNSCLC) patients. Discordant cases were solved by droplet digital PCR (ddPCR). RESULTS By testing cfDNA from healthy donors, we developed an algorithm to recognize sequencing artifacts. Applying this method to cfDNA from mNSCLC patients, EGFR mutations were detected with a good sensitivity (76.7%) and specificity (97.4%). In contrast, sensitivity and specificity for KRAS variants were 61.5% and 93.8%, respectively. All EGFR and KRAS variants detected in plasma but not in tissue were confirmed by ddPCR, thus excluding sequencing artifacts. In a fraction of cases, KRAS mutations found in plasma samples were confirmed in tumor tissue suggesting tumor heterogeneity. KRAS variants were found to be more likely sub-clonal as compared with EGFR mutations, and a correlation between clonal origin and frequency of detection in plasma was found. In a case with both EGFR and KRAS variants in cfDNA, we could demonstrate the presence of the KRAS variant in tumor tissue associated with lack of response to tyrosine kinase inhibitors (TKIs). CONCLUSIONS Although sequencing artifacts can be identified in targeted sequencing of cfDNA, tumor heterogeneity and CHIP are likely to influence the concordance between plasma and tissue testing.
Collapse
Affiliation(s)
- Raffaella Pasquale
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Laura Forgione
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Cristin Roma
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Francesca Fenizia
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Francesca Bergantino
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Anna M. Rachiglio
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Antonella De Luca
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Marianna Gallo
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Monica R. Maiello
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Giuliano Palumbo
- Medical Oncology, Ospedale Santa Maria della Pietà, Casoria, Italy
| | - Alessandro Morabito
- Thoracic Medical Oncology, Department of Hemathology, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Rosa Azzaro
- Transfusion Service, Department of Hemathology, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy, Istituto Nazionale Tumori “Fondazione G. Pascale” - IRCCS, Via M. Semmola, Naples, Italy
| |
Collapse
|
27
|
Jahangiri L, Hurst T. Assessing the Concordance of Genomic Alterations between Circulating-Free DNA and Tumour Tissue in Cancer Patients. Cancers (Basel) 2019; 11:cancers11121938. [PMID: 31817150 PMCID: PMC6966532 DOI: 10.3390/cancers11121938] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 11/28/2019] [Accepted: 11/29/2019] [Indexed: 12/23/2022] Open
Abstract
Somatic alterations to the genomes of solid tumours, which in some cases represent actionable drivers, provide diagnostic and prognostic insight into these complex diseases. Spatial and longitudinal tracking of somatic genomic alterations (SGAs) in patient tumours has emerged as a new avenue of investigation, not only as a disease monitoring strategy, but also to improve our understanding of heterogeneity and clonal evolution from diagnosis through disease progression. Furthermore, analysis of circulating-free DNA (cfDNA) in the so-called "liquid biopsy" has emerged as a non-invasive method to identify genomic information to inform targeted therapy and may also capture the heterogeneity of the primary and metastatic tumours. Considering the potential of cfDNA analysis as a translational laboratory tool in clinical practice, establishing the extent to which cfDNA represents the SGAs of tumours, particularly actionable driver alterations, becomes a matter of importance, warranting standardisation of methods and practices. Here, we assess the utilisation of cfDNA for molecular profiling of SGAs in tumour tissue across a broad range of solid tumours. Moreover, we examine the underlying factors contributing to discordance of detected SGAs between cfDNA and tumour tissue.
Collapse
Affiliation(s)
- Leila Jahangiri
- Department of Life Sciences, Birmingham City University, Birmingham B15 3TN, UK;
- Division of Cellular and Molecular Pathology, Department of Pathology, University of Cambridge, Lab blocks level 3, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Correspondence:
| | - Tara Hurst
- Department of Life Sciences, Birmingham City University, Birmingham B15 3TN, UK;
| |
Collapse
|
28
|
Canale M, Pasini L, Bronte G, Delmonte A, Cravero P, Crinò L, Ulivi P. Role of liquid biopsy in oncogene-addicted non-small cell lung cancer. Transl Lung Cancer Res 2019; 8:S265-S279. [PMID: 31857950 DOI: 10.21037/tlcr.2019.09.15] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The discovery of actionable oncogene in non-small cell lung cancer (NSCLC) allowed the identification of a subgroup of patients who benefit from targeted tyrosine kinase inhibitors more than others. Mutations in the epidermal growth factor receptor (EGFR), translocations in the anaplastic lymphoma kinase (ALK) and rearrangements in the ROS proto-oncogene 1 (ROS1) must be identified in tumor tissue to guide the proper treatment choice. Liquid biopsy is based on the analysis of tumor materials released in the circulation. Liquid biopsy can be complementary to tissue biopsy, both at baseline and at progression, especially in the detection of somatic gene alterations emerging during the treatment with tyrosine kinase inhibitors (TKIs). Particularly, circulating DNA is used to find mutations in driver oncogenes, while circulating tumor cells, extracellular vesicles (EVs) and cell-free microRNAs (cfmiRNAs) are still under investigation. To help the unbiased use of liquid biopsy in the choice of the appropriate therapy, some recommendations were delivered by expert panels. Currently, analysis of EGFR mutations in cell-free DNA (cfDNA) is recommended at baseline when tissue biopsy harbors scarce tumor cells, and at progression before performing tissue biopsy; liquid biopsy analysis for other oncogenic drivers is not indicated in the clinical practice.
Collapse
Affiliation(s)
- Matteo Canale
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Luigi Pasini
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Giuseppe Bronte
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Angelo Delmonte
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Cravero
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Lucio Crinò
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Paola Ulivi
- Biosciences Laboratory, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| |
Collapse
|
29
|
Kastrisiou M, Zarkavelis G, Pentheroudakis G, Magklara A. Clinical Application of Next-Generation Sequencing as A Liquid Biopsy Technique in Advanced Colorectal Cancer: A Trick or A Treat? Cancers (Basel) 2019; 11:E1573. [PMID: 31623125 PMCID: PMC6826585 DOI: 10.3390/cancers11101573] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/01/2019] [Accepted: 10/13/2019] [Indexed: 12/24/2022] Open
Abstract
Owing to its advantages over prior relevant technologies, massive parallel or next-generation sequencing (NGS) is rapidly evolving, with growing applications in a wide range of human diseases. The burst in actionable molecular alterations in many cancer types advocates for the practicality of using NGS in the clinical setting, as it permits the parallel characterization of multiple genes in a cost- and time-effective way, starting from low-input DNA. In advanced clinical practice, the oncological management of colorectal cancer requires prior knowledge of KRAS, NRAS, and BRAF status, for the design of appropriate therapeutic strategies, with more gene mutations still surfacing as potential biomarkers. Tumor heterogeneity, as well as the need for serial gene profiling due to tumor evolution and the emergence of novel genetic alterations, have promoted the use of liquid biopsies-especially in the form of circulating tumor DNA (ctDNA)-as a promising alternative to tissue molecular analysis. This review discusses recent studies that have used plasma NGS in advanced colorectal cancer and summarizes the clinical applications, as well as the technical challenges involved in adopting this technique in a clinically beneficial oncological practice.
Collapse
Affiliation(s)
- Myrto Kastrisiou
- Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece.
- Department of Medical Oncology, University General Hospital of Ioannina, 45500 Ioannina, Greece.
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45444 Ioannina, Greece.
| | - George Zarkavelis
- Department of Medical Oncology, University General Hospital of Ioannina, 45500 Ioannina, Greece.
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45444 Ioannina, Greece.
| | - George Pentheroudakis
- Department of Medical Oncology, University General Hospital of Ioannina, 45500 Ioannina, Greece.
- Society for Study of Clonal Heterogeneity of Neoplasia (EMEKEN), 45444 Ioannina, Greece.
| | - Angeliki Magklara
- Laboratory of Clinical Chemistry, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece.
- Department of Biomedical Research, Institute of Molecular Biology & Biotechnology, Foundation for Research & Technology-Hellas, 45110 Ioannina, Greece.
| |
Collapse
|
30
|
Liquid Biopsy Testing Can Improve Selection of Advanced Non-Small-Cell Lung Cancer Patients to Rechallenge with Gefitinib. Cancers (Basel) 2019; 11:cancers11101431. [PMID: 31557965 PMCID: PMC6826724 DOI: 10.3390/cancers11101431] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 09/19/2019] [Accepted: 09/20/2019] [Indexed: 01/02/2023] Open
Abstract
The ICARUS trial is a phase II, open label, multicenter, single arm study conducted to investigate the efficacy, safety, and tolerability of a rechallenge treatment with the first-generation tyrosine kinase inhibitor (TKI) gefitinib in advanced non-small-cell lung cancer (NSCLC) patients carrying activating mutations of the epidermal growth factor receptor (EGFR). The ICARUS trial enrolled 61 patients who were rechallenged with gefitinib at progression after second-line chemotherapy. Serum-derived circulating cell-free DNA (cfDNA) collected before the rechallenge from a cohort of 29 patients, was retrospectively analyzed for the EGFR exon 19 deletions and for the p.L858R and p.T790M single nucleotide variants (SNV). The analysis of cfDNA detected the same EGFR activating mutation reported in the tumor tissue in 20/29 patients, with a sensitivity of 69%. Moreover, a p.T790M variant was found in 14/29 patients (48.3%). The median progression-free survival (PFS) was 2.7 months for p.T790M positive patients (CI 95% 1.4–3.1 months) versus 3.5 months for the p.T790M negative patients (CI 95% 1.6–5.3 months), resulting in a statistically significant difference (Long rank test p = 0.0180). These findings confirmed the role of the p.T790M mutation in the resistance to first-generation TKIs. More importantly, our data suggest that TKI rechallenge should be guided by biomarker testing.
Collapse
|
31
|
Franceschini T, Giunchi F, Montironi R, Scarpelli M, Lopez-Beltran A, Cheng L, Fiorentino M. Liquid biopsies in urological cancers: what we need to know before starting using them. Expert Rev Mol Diagn 2019; 20:135-139. [PMID: 31509027 DOI: 10.1080/14737159.2019.1665508] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
| | | | | | | | | | - Liang Cheng
- Department of Pathology, Indiana University, Indianapolis, IN, USA
| | | |
Collapse
|
32
|
Galvano A, Taverna S, Badalamenti G, Incorvaia L, Castiglia M, Barraco N, Passiglia F, Fulfaro F, Beretta G, Duro G, Vincenzi B, Tagliaferri P, Bazan V, Russo A. Detection of RAS mutations in circulating tumor DNA: a new weapon in an old war against colorectal cancer. A systematic review of literature and meta-analysis. Ther Adv Med Oncol 2019; 11:1758835919874653. [PMID: 31534493 PMCID: PMC6737868 DOI: 10.1177/1758835919874653] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/12/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Tissue evaluation for RAS (KRAS or NRAS) gene status in metastatic colorectal cancer (mCRC) patients represent the standard of care to establish the optimal therapeutic strategy. Unfortunately, tissue biopsy is hampered by several critical limitations due to its invasiveness, difficulty to access to disease site, patient’s compliance and, more recently, neoplastic tissue spatial and temporal heterogeneity. Methods: The authors performed a systematic literature review to identify available trials with paired matched tissue and ctDNA RAS gene status evaluation. The authors searched EMBASE, MEDLINE, Cochrane, www.ClinicalTrials.gov, and abstracts from international meetings. In total, 19 trials comparing standard tissue RAS mutational status matched paired ctDNA evaluated through polymerase chain reaction (PCR), next generation sequencing (NGS) or beads, emulsions, amplification and magnetics (BEAMing) were identified. Results: The pooled sensitivity and specificity of ctDNA were 0.83 (95% CI: 0.80–0.85) and 0.91 (95% CI: 0.89–0.93) respectively. The pooled positive predictive value (PPV) and negative predictive value (NPV) of the ctDNA were 0.87 (95% CI: 0.81–0.92) and 0.87 (95% CI: 0.82–0.92), respectively. Positive likelihood ratio (PLR) was 8.20 (95% CI: 5.16–13.02) and the negative likelihood ratio (NLR) was 0.22 (95% CI: 0.16–0.30). The pooled diagnostic odds ratio (DOR) was 50.86 (95% CI: 26.15–98.76), and the area under the curve (AUC) of the summary receiver operational characteristics (sROC) curve was 0.94. Conclusion: The authors’ meta-analysis produced a complete and updated overview of ctDNA diagnostic accuracy to test RAS mutation in mCRC. Results provide a strong rationale to include the RAS ctDNA test into randomized clinical trials to validate it prospectively.
Collapse
Affiliation(s)
- Antonio Galvano
- Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Simona Taverna
- Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Giuseppe Badalamenti
- Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Lorena Incorvaia
- Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Marta Castiglia
- Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Nadia Barraco
- Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Francesco Passiglia
- Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | - Fabio Fulfaro
- Medical Oncology, Department of Surgical, Oncological and Stomatological Sciences, University of Palermo, Palermo, Italy
| | | | - Giovanni Duro
- Institute of Biomedicine and Molecular Immunology 'A. Monroy', National Research Council, Palermo, Italy
| | - Bruno Vincenzi
- Medical Oncology Department, Campus Bio-Medico University of Rome, Rome, Italy
| | - Pierosandro Tagliaferri
- Medical Oncology Unit, AUO 'Materdomini and Department of Experimental and Clinical Medicine', Magna Grecia University, Catanzaro, Italy
| | - Viviana Bazan
- Department of Experimental Biomedicine and Clinical Neurosciences, School of Medicine, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Medical Oncology Director, Department of Oncology, A.O.U.P. P. Giaccone University Hospital, 2013 ESMO Designated Centers of Integrated Oncology and Palliative Care, Via del Vespro 129, Palermo, 90127, Italy
| |
Collapse
|
33
|
Nappi A, Nasti G, Romano C, Berretta M, Ottaiano A. Metastatic Colorectal Cancer: Prognostic and Predictive Factors. Curr Med Chem 2019; 27:2779-2791. [PMID: 31218949 DOI: 10.2174/0929867326666190620110732] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 04/18/2019] [Accepted: 05/31/2019] [Indexed: 12/15/2022]
Abstract
Colorectal cancer represents the third most frequently occurring cancer worldwide. In the last decade, the survival of patients affected by metastatic colorectal cancer (mCRC) has improved through the introduction of biological drugs. However, in this new and dynamic therapeutic context, research about prognostic and predictive factors is important to guide the oncologists to effective therapies as well as to improve the understanding of colorectal cancer biology. Their identification is an intensive area of research and our future goal will be to depict tumour-specific "molecular signatures" in order to predict the clinical course of the disease and the best treatments. In this report, we describe clinical, pathological and molecular biomarkers that can play a role as prognostic or predictive factors in mCRC.
Collapse
Affiliation(s)
- Anna Nappi
- Abdominal medical oncology, National Cancer Institute, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | - Guglielmo Nasti
- SSD-Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | - Carmela Romano
- Abdominal medical oncology, National Cancer Institute, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| | | | - Alessandro Ottaiano
- SSD-Innovative Therapies for Abdominal Metastases, Istituto Nazionale Tumori di Napoli, IRCCS "G. Pascale", Naples, Italy
| |
Collapse
|
34
|
Tang Y, Liu X, Ou Z, He Z, Zhu Q, Wang Y, Yang M, Ye J, Han-Zhang H, Qiao G. Maximum allele frequency observed in plasma: A potential indicator of liquid biopsy sensitivity. Oncol Lett 2019; 18:2118-2124. [PMID: 31423285 DOI: 10.3892/ol.2019.10490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 05/22/2019] [Indexed: 02/05/2023] Open
Abstract
Personalized medicine is revolutionizing the diagnosis and treatment of cancer; however, for personalized medicine to be used accurately, patient information is essential to determine the appropriate diagnosis, prognosis and treatment. The detection of genomic mutations in liquid biopsy samples is a non-invasive method of characterizing the genotype of a tumor. However, next generation sequencing-based plasma genotyping only has a sensitivity of ~70%. Identifying potential indicators that may reflect the sensitivity of a liquid biopsy analysis could offer important information for its clinical application. In the present study, 47 pairs of patient-matched plasma and tumor tissue samples obtained from patients with advanced lung cancer were sequenced using a panel of 56 cancer-associated genes. The plasma maximum allele frequency (Max AF) was identified as a novel biomarker to indicate the sensitivity of plasma genotyping. Using the identified somatic mutations in patient tissue biopsy samples as a reference, the sensitivity of the corresponding patient plasma test was investigated. The by-variant sensitivity of the plasma test was 68.1%, with 79 matched and 37 missed genetic aberrances. The by-patient sensitivity was calculated as 83%. Patients with a high plasma Max AF value (>2.2%) demonstrated a higher concordance with the range of mutations identified in the patient-matched tissue samples. The Max AF observed in patient plasma samples was positively correlated with liquid biopsy sensitivity and could be used as a potential indicator of liquid biopsy sensitivity. Therefore, patients with a low plasma Max AF (≤2.2%) may need to undergo further tissue biopsy to allow personalized oncology treatment. In summary, the present study may offer a non-invasive testing method for a sub-group of patients with advanced lung cancer.
Collapse
Affiliation(s)
- Yong Tang
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, Guangzhou, Guangdong 510010, P.R. China
| | - Xianling Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Zhu'An Ou
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, Guangzhou, Guangdong 510010, P.R. China
| | - Zhe He
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, Guangzhou, Guangdong 510010, P.R. China
| | - Qihang Zhu
- Department of Thoracic Surgery, General Hospital of Southern Theater Command, Guangzhou, Guangdong 510010, P.R. China
| | - Ye Wang
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P.R. China
| | - Mei Yang
- Department of Cadre Treatment, The Third Affiliated Hospital of Kunming Medical University (Yunnan Province Tumor Hospital), Kunming, Yunnan 650118, P.R. China
| | - Junyi Ye
- Burning Rock Biotech, Guangzhou, Guangdong 510300, P.R. China
| | - Han Han-Zhang
- Burning Rock Biotech, Guangzhou, Guangdong 510300, P.R. China
| | - Guibin Qiao
- Department of Thoracic Surgery, Guangdong General Hospital, Guangzhou, Guangdong 510245, P.R. China
| |
Collapse
|
35
|
Lyu M, Zhou J, Ning K, Ying B. The diagnostic value of circulating tumor cells and ctDNA for gene mutations in lung cancer. Onco Targets Ther 2019; 12:2539-2552. [PMID: 31040697 PMCID: PMC6454989 DOI: 10.2147/ott.s195342] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
PURPOSE Detecting gene mutations by two competing biomarkers, circulating tumor cells (CTCs) and ctDNA has gradually paved a new diagnostic avenue for personalized medicine. We performed a comprehensive analysis to compare the diagnostic value of CTCs and ctDNA for gene mutations in lung cancer. METHODS Publications were electronically searched in PubMed, Embase, and Web of Science as of July 2018. Pooled sensitivity, specificity, and AUC, each with a 95% CI, were yielded. Subgroup analyses and sensitivity analyses were conducted. Quality assessment of included studies was also performed. RESULTS From 4,283 candidate articles, we identified 47 articles with a total of 7,244 patients for qualitative review and meta-analysis. When detecting EGFR, the CTC and ctDNA groups had pooled sensitivity of 75.4% (95% CI 0.683-0.817) and 67.1% (95% CI 0.647-0.695), respectively. When testing KRAS, pooled sensitivity was 38.7% (95% CI 0.266-0.519) in the CTC group and 65.1% (95% CI 0.558-0.736) in the ctDNA group. The diagnostic performance of ctDNA in testing ALK and BRAF was also evaluated. Heterogeneity among the 47 articles was acceptable. CONCLUSION ctDNA might be a more promising biomarker with equivalent performance to CTCs when detecting EGFR and its detailed subtypes, and superior diagnostic capacity when testing KRAS and ALK. In addition, the diagnostic performance of ctDNA and CTCs depends on the detection methods greatly, and this warrants further studies to explore more sensitive methods.
Collapse
Affiliation(s)
- Mengyuan Lyu
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China,
| | - Jian Zhou
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Kang Ning
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Binwu Ying
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China,
| |
Collapse
|
36
|
Zeng C, Stroup EK, Zhang Z, Chiu BCH, Zhang W. Towards precision medicine: advances in 5-hydroxymethylcytosine cancer biomarker discovery in liquid biopsy. Cancer Commun (Lond) 2019; 39:12. [PMID: 30922396 PMCID: PMC6440138 DOI: 10.1186/s40880-019-0356-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 03/19/2019] [Indexed: 12/13/2022] Open
Abstract
Robust and clinically convenient biomarkers for cancer diagnosis, early detection, and prognosis have great potential to improve patient survival and are the key to precision medicine. The advent of next-generation sequencing technologies enables a more sensitive and comprehensive profiling of genetic and epigenetic information in tumor-derived materials. Researchers are now able to monitor the dynamics of tumorigenesis in new dimensions, such as using circulating cell-free DNA (cfDNA) and tumor DNA (ctDNA). Mutation-based assays in liquid biopsy cannot always provide consistent results across studies due partly to intra- and inter-tumoral heterogeneity as well as technical limitations. In contrast, epigenetic analysis of patient-derived cfDNA is a promising alternative, especially for early detection and disease surveillance, because epigenetic modifications are tissue-specific and reflect the dynamic process of cancer progression. Therefore, cfDNA-based epigenetic assays are emerging to be a highly sensitive, minimally invasive tool for cancer diagnosis and prognosis with great potential in future precise care of cancer patients. The major obstacle for applying epigenetic analysis of cfDNA, however, has been the lack of enabling techniques with high sensitivity and technical robustness. In this review, we summarized the advances in epigenome-wide profiling of 5-hydroxymethylcytosine (5hmC) in cfDNA, focusing on the detection approaches and potential role as biomarkers in different cancer types.
Collapse
Affiliation(s)
- Chang Zeng
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Emily Kunce Stroup
- Driskill Graduate Program in Life Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA
| | - Zhou Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lake Shore Dr., Suite 1400, Chicago, IL, 60611, USA
| | - Brian C-H Chiu
- Department of Public Health Sciences, University of Chicago, Chicago, IL, 60637, USA
| | - Wei Zhang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, 680 N. Lake Shore Dr., Suite 1400, Chicago, IL, 60611, USA. .,The Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, 60611, USA. .,Institute of Precision Medicine, Jining Medical University, Jining, 272067, Shandong, P. R. China.
| |
Collapse
|
37
|
Normanno N, Esposito Abate R, Lambiase M, Forgione L, Cardone C, Iannaccone A, Sacco A, Rachiglio AM, Martinelli E, Rizzi D, Pisconti S, Biglietto M, Bordonaro R, Troiani T, Latiano TP, Giuliani F, Leo S, Rinaldi A, Maiello E, Ciardiello F. RAS testing of liquid biopsy correlates with the outcome of metastatic colorectal cancer patients treated with first-line FOLFIRI plus cetuximab in the CAPRI-GOIM trial. Ann Oncol 2019; 29:112-118. [PMID: 28950295 DOI: 10.1093/annonc/mdx417] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Background Liquid biopsy is an alternative to tissue for RAS testing in metastatic colorectal carcinoma (mCRC) patients. Little information is available on the predictive role of liquid biopsy RAS testing in patients treated with first-line anti-EGFR monoclonal antibody-based therapy. Patients and methods In the CAPRI-GOIM trial, 340 KRAS exon-2 wild-type mCRC patients received first-line cetuximab plus FOLFIRI. Tumor samples were retrospectively assessed by next generation sequencing (NGS). Baseline plasma samples were analyzed for KRAS and NRAS mutations using beads, emulsion, amplification, and magnetics digital PCR (BEAMing). Discordant cases were solved by droplet digital PCR (ddPCR) or deep-sequencing. Results A subgroup of 92 patients with available both NGS data on tumor samples and baseline plasma samples were included in this study. Both NGS analysis of tumor tissue and plasma testing with BEAMing identified RAS mutations in 33/92 patients (35.9%). However, 10 cases were RAS tissue mutant and plasma wild-type, and additional 10 cases were tissue wild-type and plasma mutant, resulting in a concordance rate of 78.3%. Analysis of plasma samples with ddPCR detected RAS mutations in 2/10 tissue mutant, plasma wild-type patients. In contrast, in all tissue wild-type and plasma mutant cases, ddPCR or deep-sequencing analysis of tumor tissue confirmed the presence of RAS mutations at allelic frequencies ranging between 0.15% and 1.15%. The median progression-free survival of RAS mutant and wild-type patients according to tissue (7.9 versus 12.6 months; P = 0.004) and liquid biopsy testing (7.8 versus 13.8 moths; P < 0.001) were comparable. Similar findings were observed for the median overall survival of RAS mutant and wild-type patients based on tissue (22.1 versus 35.8 months; P = 0.016) and plasma (19.9 versus 35.8 months; P = 0.013) analysis. Conclusion This study indicates that RAS testing of liquid biopsy results in a similar outcome when compared with tissue testing in mCRC patients receiving first-line anti-EGFR monoclonal antibodies.
Collapse
Affiliation(s)
- N Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - R Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - M Lambiase
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - L Forgione
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - C Cardone
- Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi," Università della Campania "L. Vanvitelli," Napoli, Italy
| | - A Iannaccone
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - A Sacco
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - A M Rachiglio
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori "Fondazione Giovanni Pascale" IRCCS, Napoli, Italy
| | - E Martinelli
- Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi," Università della Campania "L. Vanvitelli," Napoli, Italy
| | - D Rizzi
- Gruppo Oncologico dell'Italia Meridionale (GOIM), Bari, Italy
| | - S Pisconti
- Medical Oncology, Hospital SS. Annunziata, Taranto, Italy
| | - M Biglietto
- Medical Oncology, Hospital "A. Cardarelli," Napoli, Italy
| | - R Bordonaro
- Medical Oncology, Hospital Garibaldi-Nesima, Catania, Italy
| | - T Troiani
- Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi," Università della Campania "L. Vanvitelli," Napoli, Italy
| | - T P Latiano
- Medical Oncology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - F Giuliani
- Medical Oncology, National Cancer Institute Giovanni Paolo II, Bari, Italy
| | - S Leo
- Medical Oncology, Hospital Vito Fazzi, Lecce, Italy
| | - A Rinaldi
- Medical Oncology, Presidio Ospedaliero Polo Occidentale, Castellaneta, Bari, Italy
| | - E Maiello
- Medical Oncology, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - F Ciardiello
- Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi," Università della Campania "L. Vanvitelli," Napoli, Italy
| | | |
Collapse
|
38
|
Kienle DL, Dietrich D, Ribi K, Wicki A, Quagliata L, Winterhalder RC, Koeberle D, Horber D, Bastian S, Kueng M, Saletti P, Helbling D, Baertschi D, Lugli A, Bernhard J, Andrieu C, von Moos R. Cetuximab monotherapy and cetuximab plus capecitabine as first-line treatment in older patients with RAS- and BRAF wild-type metastatic colorectal cancer. Results of the multicenter phase II trial SAKK 41/10. J Geriatr Oncol 2019; 10:304-310. [DOI: 10.1016/j.jgo.2018.11.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 10/23/2018] [Accepted: 11/30/2018] [Indexed: 02/07/2023]
|
39
|
Xie W, Xie L, Song X. The diagnostic accuracy of circulating free DNA for the detection of KRAS mutation status in colorectal cancer: A meta-analysis. Cancer Med 2019; 8:1218-1231. [PMID: 30791218 PMCID: PMC6434340 DOI: 10.1002/cam4.1989] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/17/2018] [Accepted: 01/04/2019] [Indexed: 02/06/2023] Open
Abstract
KRAS mutations have been reported as a reliable biomarker for epidermal growth factor receptor (EGFR) targeted therapy and are also associated with poor prognosis in colorectal cancer (CRC) patients. However, limitations of detecting KRAS mutations in tissues are obvious. KRAS mutations in the peripheral blood can be detected as an alternative to tissue analysis. The objective of this meta‐analysis was to evaluate the diagnostic value of cfDNA (circulating free DNA) compared with tissues and to investigate the prognostic potential of cfDNA KRAS mutations in CRC patients. Searches were performed in PubMed, Embase, and Cochrane Library for published studies. We extracted true‐positive (TP), false‐positive (FP), false‐negative (FN), true‐negative (TN) values, survival rate of CRC patients with mutant and wild‐type KRAS and calculated pooled sensitivity and specificity, positive/negative likelihood ratios [PLRs/NLRs], diagnostic odds ratios [DORs], and corresponding 95% confidence intervals [95% CIs]. We also generated a summary receiver operating characteristic (SROC) curve to evaluate the overall diagnostic potential. Totally, 31 relevant studies were recruited and used for the meta‐analysis on the efficacy of cfDNA testing in detecting KRAS mutations. The pooled sensitivity, specificity, PLR, NLR, and DOR were 0.637 (95% CI: 0.607‐0.666), 0.943 (95% CI: 0.930‐0.954), 10.024 (95% CI: 6.912‐14.535), 0.347 (95% CI: 0.269‐0.447), and 37.882 (95% CI: 22.473‐63.857), respectively. The area under the SROC curve was 0.9392. Together, the results suggest that detecting KRAS mutations in cfDNA has adequate diagnostic efficacy in terms of specificity. There is a promising role for cfDNA in the detection of KRAS mutations in CRC patients. However, prospective studies with larger patient cohorts are still required before definitive conclusions of the prognostic potential of cfDNA KRAS mutations in CRC patients were drawn.
Collapse
Affiliation(s)
- Wenli Xie
- Shandong Cancer Hospital Affiliated to Shandong University, Shandong University, Shandong Province, Jinan, P.R. China
| | - Li Xie
- Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Shandong Province, Jinan, P.R. China
| | - Xianrang Song
- Shandong Cancer Hospital Affiliated to Shandong University, Shandong University, Shandong Province, Jinan, P.R. China.,Shandong Cancer Hospital Affiliated to Shandong University, Shandong Academy of Medical Sciences, Shandong Province, Jinan, P.R. China
| |
Collapse
|
40
|
Herbreteau G, Vallée A, Charpentier S, Normanno N, Hofman P, Denis MG. Circulating free tumor DNA in non-small cell lung cancer (NSCLC): clinical application and future perspectives. J Thorac Dis 2019; 11:S113-S126. [PMID: 30775034 DOI: 10.21037/jtd.2018.12.18] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Major advances in the treatment of non-small cell lung cancer (NSCLC) patients have been obtained during the last decade. Molecular testing of tumor samples is therefore mandatory in routine clinical practice. Tumor DNA is also present as cell-free molecules in blood, which is therefore a very useful and convenient source of tumor DNA. In this review, we discuss pre-analytical and analytical aspects of circulating tumor DNA (ctDNA) analysis. We also describe the use of ctDNA analysis in routine clinical practice, and discuss the potential use of ctDNA monitoring both to identify minimal residual disease and as a potential tool to early identify patients' response to treatment.
Collapse
Affiliation(s)
- Guillaume Herbreteau
- Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, F-44093 Nantes Cedex, France
| | - Audrey Vallée
- Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, F-44093 Nantes Cedex, France
| | - Sandrine Charpentier
- Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, F-44093 Nantes Cedex, France
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori, IRCCS, "Fondazione G. Pascale", Naples, Italy
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Inserm U1081/CNRS 7284, Université Côte d'Azur, CHU Nice and FHU OncoAge, and Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, Nice, France
| | - Marc G Denis
- Department of Biochemistry, Nantes University Hospital, 9 quai Moncousu, F-44093 Nantes Cedex, France
| |
Collapse
|
41
|
Babji D, Nayak R, Bhat K, Kotrashetti V. Cell-free tumor DNA: Emerging reality in oral squamous cell carcinoma. J Oral Maxillofac Pathol 2019; 23:273-279. [PMID: 31516235 PMCID: PMC6714275 DOI: 10.4103/jomfp.jomfp_36_19] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
The intention of this review was to condense ongoing findings on the use of circulating DNAs from bodily fluids (blood, serum and plasma) as cancer biomarkers in patients with oral squamous cell carcinoma. Studies were collected after searching databases: PubMed and Google library. Additional search was performed through cross-check on the bibliography of selected articles. After the selection process made by two of the authors, articles which met the inclusion criteria were included in the review. Results revealed that circulating DNAs from blood, serum or plasma appear as favorable candidates as cancer biomarkers in patients suffering from oral cancer. The possibility to forecast recurrences and metastases through follow-up by quantification of candidate DNAs serve as another possible characteristic to be directed in forthcoming studies. However, methodological standardization and even sampling are required to increase the power and accuracy of results.
Collapse
Affiliation(s)
- Deepa Babji
- Department of Oral Pathology and Microbiology, Maratha Mandal's NG Halgekar Institute of Dental Sciences and Research Centre, Belgaum, Karnataka, India
| | - Ramakant Nayak
- Department of Oral Pathology and Microbiology, Maratha Mandal's NG Halgekar Institute of Dental Sciences and Research Centre, Belgaum, Karnataka, India
| | - Kishore Bhat
- Department of Microbiology, Maratha Mandal's NGH Institute of Dental Sciences, Belgaum, Karnataka, India
| | - Vijayalakshmi Kotrashetti
- Department of Oral Pathology and Microbiology, Maratha Mandal's NG Halgekar Institute of Dental Sciences and Research Centre, Belgaum, Karnataka, India
| |
Collapse
|
42
|
Galbiati S, Damin F, Burgio V, Brisci A, Soriani N, Belcastro B, Di Resta C, Gianni L, Chiari M, Ronzoni M, Ferrari M. Evaluation of three advanced methodologies, COLD-PCR, microarray and ddPCR, for identifying the mutational status by liquid biopsies in metastatic colorectal cancer patients. Clin Chim Acta 2018; 489:136-143. [PMID: 30550935 DOI: 10.1016/j.cca.2018.12.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/27/2018] [Accepted: 12/07/2018] [Indexed: 01/22/2023]
Abstract
A major effort has been focused on the detection of oncogenes' mutations in diverse types of clinical specimens including formalin-fixed and paraffin embedded tissues, presently the gold-standard samples, up to plasma, that constitute a noninvasive alternative source of tumor DNA. The reliable detection of mutations in circulating tumor DNA requires a high analytical sensitivity. Here, we applied three different highly sensitive methodologies (COLD-PCR, a microarray-based approach and the droplet digital PCR, ddPCR) to identify mutations in the plasma of 30 metastatic colorectal cancer patients previously genotyped on tissue biopsy. The methods showed a modest concordance rate with respect to the results obtained on tissue biopsies: 63.3% by ddPCR, 63% by microarray and 55.6% by COLD-PCR. This could be ascribed either to the different timing between tissue and liquid biopsy collection, which could reflect a different stage of disease progression or to the diverse sensitivity of the methodologies applied. Indeed, if we compare the results obtained on plasma samples, the concordance rates were higher especially by comparing ddPCR versus COLD-PCR (92.6%). Thus, we consider both methodologies as useful procedures easily transferable in a clinical setting. Notably, the ddPCR allows a quantitative assessment of the fractional abundance of the mutation.
Collapse
Affiliation(s)
- Silvia Galbiati
- Unit of Genomic for the Diagnosis of Human Pathologies, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Francesco Damin
- Consiglio Nazionale delle Ricerche, Istituto di Chimica del Riconoscimento Molecolare, Milano, Italy
| | - Valentina Burgio
- Dipartimento di Oncologia Medica, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Angela Brisci
- Unit of Genomic for the Diagnosis of Human Pathologies, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Nadia Soriani
- Unit of Genomic for the Diagnosis of Human Pathologies, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Bernadette Belcastro
- Unit of Genomic for the Diagnosis of Human Pathologies, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Di Resta
- Unit of Genomic for the Diagnosis of Human Pathologies, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Gianni
- Dipartimento di Oncologia Medica, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Marcella Chiari
- Consiglio Nazionale delle Ricerche, Istituto di Chimica del Riconoscimento Molecolare, Milano, Italy
| | - Monica Ronzoni
- Dipartimento di Oncologia Medica, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maurizio Ferrari
- Unit of Genomic for the Diagnosis of Human Pathologies, Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy; Laboratory of Clinical Molecular Biology, IRCCS Ospedale San Raffaele, Milan, Italy; Università Vita-Salute San Raffaele, Milano, Italy
| |
Collapse
|
43
|
Esposito Abate R, Pasquale R, Fenizia F, Rachiglio AM, Roma C, Bergantino F, Forgione L, Lambiase M, Sacco A, Piccirillo MC, Morabito A, Normanno N. The role of circulating free DNA in the management of NSCLC. Expert Rev Anticancer Ther 2018; 19:19-28. [PMID: 30462523 DOI: 10.1080/14737140.2019.1548938] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Circulating cell-free DNA (cfDNA) testing has emerged as an alternative to tumor tissue analyses for the management of metastatic non-small-cell lung cancer (NSCLC) patients. Analysis of cfDNA is a minimally invasive procedure that might better reflect tumor heterogeneity and allows repeated testing over the time. Areas covered: This review article covers the different applications of cfDNA testing in NSCLC: early diagnosis of the disease; detection of minimal residual disease in early lung cancer; identification of predictive and prognostic markers in advanced NSCLC patients; monitoring the response to therapy; assessment of tumor mutation burden. Expert commentary: The use of liquid biopsy is rapidly expanding to different applications. The combination of different circulating biomarkers (cfDNA, protein, miRNA) might improve the sensitivity and specificity of this approach in patients with low tumor burden. cfDNA testing is representing a valid source for molecular profiling in management of metastatic NSCLC patients and is providing important knowledge on tumor heterogeneity. Clinical trials are needed in order to transfer the information deriving from liquid biopsy testing in new therapeutic strategies.
Collapse
Affiliation(s)
- Riziero Esposito Abate
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Raffaella Pasquale
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Francesca Fenizia
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Anna Maria Rachiglio
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Cristin Roma
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Francesca Bergantino
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Laura Forgione
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Matilde Lambiase
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Alessandra Sacco
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| | - Maria Carmela Piccirillo
- b Clinical Trials Unit , Istituto Nazionale Tumori - IRCSS - Fondazione G. Pascale , Napoli , Italy
| | - Alessandro Morabito
- c Department of Thoracic Medical Oncology , Istituto Nazionale Tumori - IRCSS - Fondazione G. Pascale , Napoli , Italy
| | - Nicola Normanno
- a Cell Biology and Biotherapy Unit , Istituto Nazionale Tumori, IRCSS, Fondazione G. Pascale , Napoli , Italy
| |
Collapse
|
44
|
Development of a Fluorinated Analogue of Erlotinib for PET Imaging of EGFR Mutation–Positive NSCLC. Mol Imaging Biol 2018; 21:696-704. [DOI: 10.1007/s11307-018-1286-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
45
|
Gabriel E, Bagaria SP. Assessing the Impact of Circulating Tumor DNA (ctDNA) in Patients With Colorectal Cancer: Separating Fact From Fiction. Front Oncol 2018; 8:297. [PMID: 30128304 PMCID: PMC6088154 DOI: 10.3389/fonc.2018.00297] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 07/16/2018] [Indexed: 12/16/2022] Open
Abstract
Significant advances and increased awareness have been in made in the field of non-invasive liquid biopsies for cancer, spanning several malignancies from gastrointestinal, pulmonary, and other etiologies. Broadly, the genetic source material for liquid biopsies includes circulating tumor cells, cell-free circulating tumor DNA (ctDNA), or cell-free circulating tumor microRNA (mRNA). In this review, we specifically focus on ctDNA and its current role in colorectal cancer. While there are several commercially available assays that detect ctDNA, the utility of these products is still variable and therefore the clinical applications of ctDNA in the management of patients with cancer has yet to be determined. This is reflected by the recent joint review set forth by the American Society of Clinical Oncology (ASCO) and the College of American Pathologists (CAP), clarifying and somewhat tempering the present role of ctDNA in patients with cancer. This review provides additional detail regarding ctDNA in the limited setting of colorectal cancer. The increasing importance and promise of ctDNA remains an area of active research, and further prospective studies may enhance the clinical utility of ctDNA in the future.
Collapse
Affiliation(s)
- Emmanuel Gabriel
- Section of Surgical Oncology, Department of Surgery, Mayo Clinic Florida, Jacksonville, FL, United States
| | - Sanjay P Bagaria
- Section of Surgical Oncology, Department of Surgery, Mayo Clinic Florida, Jacksonville, FL, United States
| |
Collapse
|
46
|
Pavic M, Bogowicz M, Würms X, Glatz S, Finazzi T, Riesterer O, Roesch J, Rudofsky L, Friess M, Veit-Haibach P, Huellner M, Opitz I, Weder W, Frauenfelder T, Guckenberger M, Tanadini-Lang S. Influence of inter-observer delineation variability on radiomics stability in different tumor sites. Acta Oncol 2018. [PMID: 29513054 DOI: 10.1080/0284186x.2018.1445283] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Radiomics is a promising methodology for quantitative analysis and description of radiological images using advanced mathematics and statistics. Tumor delineation, which is still often done manually, is an essential step in radiomics, however, inter-observer variability is a well-known uncertainty in radiation oncology. This study investigated the impact of inter-observer variability (IOV) in manual tumor delineation on the reliability of radiomic features (RF). METHODS Three different tumor types (head and neck squamous cell carcinoma (HNSCC), malignant pleural mesothelioma (MPM) and non-small cell lung cancer (NSCLC)) were included. For each site, eleven individual tumors were contoured on CT scans by three experienced radiation oncologists. Dice coefficients (DC) were calculated for quantification of delineation variability. RF were calculated with an in-house developed software implementation, which comprises 1404 features: shape (n = 18), histogram (n = 17), texture (n = 137) and wavelet (n = 1232). The IOV of RF was studied using the intraclass correlation coefficient (ICC). An ICC >0.8 indicates a good reproducibility. For the stable RF, an average linkage hierarchical clustering was performed to identify classes of uncorrelated features. RESULTS Median DC was high for NSCLC (0.86, range 0.57-0.90) and HNSCC (0.72, 0.21-0.89), whereas it was low for MPM (0.26, 0-0.9) indicating substantial IOV. Stability rate of RF correlated with DC and depended on tumor site, showing a high stability in NSCLC (90% of total parameters), acceptable stability in HNSCC (59% of total parameters) and low stability in MPM (36% of total parameters). Shape features showed the weakest stability across all tumor types. Hierarchical clustering revealed 14 groups of correlated and stable features for NSCLC and 6 groups for both HNSCC and MPM. CONCLUSION Inter-observer delineation variability has a relevant influence on radiomics analysis and is strongly influenced by tumor type. This leads to a reduced number of suitable imaging features.
Collapse
Affiliation(s)
- Matea Pavic
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Marta Bogowicz
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Xaver Würms
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Stefan Glatz
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Tobias Finazzi
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Oliver Riesterer
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Johannes Roesch
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Leonie Rudofsky
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Martina Friess
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Patrick Veit-Haibach
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Martin Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Walter Weder
- Department of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Frauenfelder
- Institute of Diagnostic and Interventional Radiology, University Hospital Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
47
|
Demuth C, Winther-Larsen A, Madsen AT, Meldgaard P, Sorensen BS. A method for treatment monitoring using circulating tumour DNA in cancer patients without targetable mutations. Oncotarget 2018; 9:31066-31076. [PMID: 30123427 PMCID: PMC6089565 DOI: 10.18632/oncotarget.25779] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Accepted: 06/12/2018] [Indexed: 02/05/2023] Open
Abstract
Background The potentials of circulating tumour DNA (ctDNA) have been studied for non-invasive disease monitoring in patients with targetable mutations. However, the majority of cancer patients harbour no targetable mutations. A workflow including targeted next-generation sequencing (NGS) and droplet digital PCR (ddPCR) could be used for monitoring treatment in these patients. Thus, our aim was to evaluate the workflow for ctDNA monitoring in a cohort of non-small cell lung cancer patients. Methods Forty patients were prospectively included. Plasma samples were collected prior to and during treatment. NGS (Ion AmpliSeq Colon and Lung Cancer panel v2) was performed on ctDNA from pre-treatment samples. The identified mutations were monitored by ddPCR in consecutively collected samples. Results Mutations were detected in 21 patients. The most commonly mutated genes were TP53 (N=20) and KRAS (N=13). Treatment was discontinued due to non-response in 18 patients. In 16 of these, a simultaneous increase in ctDNA concentration was observed. A twofold ctDNA concentration increase confirmed in a second successive sample predicted non-response on the following imaging in 83% of patients (10/12). Conclusion ctDNA monitoring can be used for early detection of non-response in patients without targetable mutations, and therefore could supplement imaging data for treatment monitoring in this subset of patients.
Collapse
Affiliation(s)
- Christina Demuth
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anne Winther-Larsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Anne Tranberg Madsen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Peter Meldgaard
- Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Boe Sandahl Sorensen
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
48
|
Mlika M, Dziri C, Zorgati MM, Ben Khelil M, Mezni F. Liquid Biopsy as Surrogate to Tissue in Lung Cancer for Molecular Profiling: A Meta-Analysis. CURRENT RESPIRATORY MEDICINE REVIEWS 2018; 14:48-60. [PMID: 30271314 PMCID: PMC6128071 DOI: 10.2174/1573398x14666180430144452] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/25/2018] [Accepted: 04/27/2018] [Indexed: 12/17/2022]
Abstract
Background: The accurate microscopic diagnosis of lung cancer has become insufficient due to the concept of personalized medicine. Tissue samples are used not only for microscopic diagnosis but also for the assessment of the different targets. Biopsies are performed in 80% of the patients and they are not sufficient for molecular diagnosis in 30% of the cases. Liquid biopsy (LB) has been reported as a possible surrogate to tissue samples and has been introduced in the management scheme of the patients since 2014. We aimed to highlight the diagnostic value of liquid biopsy in assessing the molecular profile of non small cell carcinomas in comparison with tissue biopsy. Methods: We retracted eligible articles from PubMed, Embase and Cochrane databases. We calculated the pooled sensitivity (SEN), specificity (SPE), positive likelihood ratio (PLR), negative likelihood ratio (NLR) and diagnostic odds ratio (DOR). A summary receiver operating characteristic curve (SROC) and area under curve (AUC) were used to evaluate the overall diagnostic performance using the Meta-Disc software 5.1.32. The heterogeneity was assessed using I square statistics. A meta-regression was performed in case of heterogeneity. In case of absence of covariates, a sensitivity analysis was done in order to assess publications that induced a statistical bias. Results: 39 eligible studies involving 4782 patients were included. The overall statistical studies showed heterogeneity in the SEN, SPE, PLR, NLR and DOR. No threshold effect was revealed. The meta-regression incorporating the ethnicity, the test, the technique used in tissue and plasma and the use of plasma or serum as covariates showed no impact of these factors. A sensitivity analysis allowed achieving the homogeneity in the SPE and DOR. The overall pooled SEN and SPE were 0.61 and 0.95 respectively. The PLR was 9.51, the NLR was 0.45 and DOR was 24.58. The SROC curve with AUC of 0,93 indicated that the liquid biopsy is capable of identifying wild type samples from mutated ones with a relatively high accuracy. Conclusion: This meta-analysis suggested that detection of molecular mutations by cfDNA is of adequate diagnostic accuracy in association to tissues. The high specificity and the moderate sensitivity highlight the value of LB as a screening test
Collapse
Affiliation(s)
- Mona Mlika
- 1Department of Pathology, Abderrahman Mami Hospital, Ariana, Tunisia; 2University Tunis El Manar, Faculty of Medicine of Tunis, Tunis, Tunisia; 3Department of General Surgery B, Charles Nicolle Hospital, Tunis, Tunisia; 4Medical Center of ABM, Military College, Qatar
| | - Chadli Dziri
- 1Department of Pathology, Abderrahman Mami Hospital, Ariana, Tunisia; 2University Tunis El Manar, Faculty of Medicine of Tunis, Tunis, Tunisia; 3Department of General Surgery B, Charles Nicolle Hospital, Tunis, Tunisia; 4Medical Center of ABM, Military College, Qatar
| | - Mohamed Majdi Zorgati
- 1Department of Pathology, Abderrahman Mami Hospital, Ariana, Tunisia; 2University Tunis El Manar, Faculty of Medicine of Tunis, Tunis, Tunisia; 3Department of General Surgery B, Charles Nicolle Hospital, Tunis, Tunisia; 4Medical Center of ABM, Military College, Qatar
| | - Mehdi Ben Khelil
- 1Department of Pathology, Abderrahman Mami Hospital, Ariana, Tunisia; 2University Tunis El Manar, Faculty of Medicine of Tunis, Tunis, Tunisia; 3Department of General Surgery B, Charles Nicolle Hospital, Tunis, Tunisia; 4Medical Center of ABM, Military College, Qatar
| | - Faouzi Mezni
- 1Department of Pathology, Abderrahman Mami Hospital, Ariana, Tunisia; 2University Tunis El Manar, Faculty of Medicine of Tunis, Tunis, Tunisia; 3Department of General Surgery B, Charles Nicolle Hospital, Tunis, Tunisia; 4Medical Center of ABM, Military College, Qatar
| |
Collapse
|
49
|
Hsu HC, Lapke N, Wang CW, Lin PY, You JF, Yeh CY, Tsai WS, Hung HY, Chiang SF, Chen HC, Chen SJ, Hsu A, Yang TS. Targeted Sequencing of Circulating Tumor DNA to Monitor Genetic Variants and Therapeutic Response in Metastatic Colorectal Cancer. Mol Cancer Ther 2018; 17:2238-2247. [PMID: 29997152 DOI: 10.1158/1535-7163.mct-17-1306] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/22/2018] [Accepted: 07/05/2018] [Indexed: 11/16/2022]
Abstract
Substantial improvements have been made in the management of metastatic colorectal cancer (mCRC) in the last two decades, but disease monitoring remains underdeveloped. Circulating tumor DNA (ctDNA) is a promising prognostic and predictive biomarker; however, ctDNA as a marker for mCRC patients is not well established, and there is still no consensus about how to utilize it most cost-effectively. In this study, we aim to investigate plasma ctDNA levels as a biomarker for therapeutic response of mCRC patients. We performed next-generation sequencing (NGS) by using a 12-gene panel to identify genetic variants in 136 tumor tissue and ctDNA samples from 32 mCRC patients. Genetic variants were detected in approximately 70% of samples, and there was a high concordance (85%) between tumor tissue and plasma ctDNA. We observed ctDNA changes in 18 follow-up patients, including the emergence of new variants. Changes in ctDNA levels significantly correlated with tumor shrinkage (P = 0.041), and patients with a ctDNA decrease >80% after treatment had a longer progression-free survival compared with patients with a ctDNA decrease of <80% (HR, 0.22; P = 0.015). The objective response rate among patients with a ctDNA decrease of >80% was better than those with a ctDNA decrease <80% (OR, 0.026; P = 0.007). In conclusion, this study demonstrates that monitoring of genetic ctDNA variants can serve as a valuable biomarker for therapeutic efficacy in mCRC patients, and that using a moderate-sized 12-gene NGS panel may be suitable for such clinical monitoring. Mol Cancer Ther; 17(10); 2238-47. ©2018 AACR.
Collapse
Affiliation(s)
- Hung-Chih Hsu
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan.,College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | | | - Chuang-Wei Wang
- Department of Dermatology, Drug Hypersensitivity Clinical and Research Center, Chang Gung Memorial Hospitals, Taipei, Linkou and Keelung, Taiwan.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan
| | | | - Jeng Fu You
- College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan
| | - Chien Yuh Yeh
- College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan
| | - Wen-Sy Tsai
- College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan
| | - Hsin Yuan Hung
- College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan
| | - Sum-Fu Chiang
- College of Medicine, Chang Gung University, Tao-Yuan, Taiwan.,Division of Colon and Rectal Surgery, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan
| | | | | | - An Hsu
- ACT Genomics, Co. Ltd., Taipei, Taiwan.
| | - Tsai Sheng Yang
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Tao-Yuan, Taiwan. .,College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| |
Collapse
|
50
|
Gasparello J, Allegretti M, Tremante E, Fabbri E, Amoreo CA, Romania P, Melucci E, Messana K, Borgatti M, Giacomini P, Gambari R, Finotti A. Liquid biopsy in mice bearing colorectal carcinoma xenografts: gateways regulating the levels of circulating tumor DNA (ctDNA) and miRNA (ctmiRNA). JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:124. [PMID: 29941002 PMCID: PMC6020232 DOI: 10.1186/s13046-018-0788-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 06/05/2018] [Indexed: 12/22/2022]
Abstract
Background Circulating tumor DNA (ctDNA) and miRNA (ctmiRNA) are promising biomarkers for early tumor diagnosis, prognosis and monitoring, and to predict therapeutic response. However, a clear understanding of the fine control on their circulating levels is still lacking. Methods Three human colorectal carcinoma cell lines were grown in culture and as tumor xenograft models in nude mice. Chip-based and droplet digital PCR platforms were used to systematically and quantitatively assess the levels of DNAs and miRNAs released into the culture supernatants and mouse blood plasma. Results Strikingly, mutated DNAs from the same (KRAS) and different (PIK3CA and FBWX7) genomic loci were differentially detected in culture supernatants and blood, with LS174T releasing 25 to 60 times less DNA in culture, but giving rise to 7 to 8 times more DNA in blood than LoVo cells. Greater LS174T ctDNA accumulation occurred in spite of similar CD31 immunostaining (micro-vascularization) and lesser proliferation and tissue necrosis as compared to LoVo. As to the three selected miRNAs (miR-221, miR-222 and miR-141), all of them were constitutively present in the plasma of tumor-free mice. Micro-RNA miR-141 was released into HT-29 cell supernatants 10 and 6.5 times less abundantly with respect to LoVo and LS174T, respectively; on the contrary, release of miR-141 in blood of HT-29 xenografted mice was found similar to that observed in LoVo and LS174T mice. Conclusions Taken together, our results support the existence of multiple, finely tuned (non-housekeeping) control gateways that selectively regulate the release/accumulation of distinct ctDNA and miRNA species in culture and tumor xenograft models. Different xenografts (proxies of different patients) considerably differ in gateway usage, adding several layers of complexity to the well-known idea of molecular heterogeneity. We predict that even high tissue representation of mutated DNA and miRNA may result in insufficient diagnostic analyte representation in blood. In this respect, our data show that careful modeling in mice may considerably help to alleviate complexity, for instance by pre-screening for the most abundant circulating analytes in enlarged sets of tumor xenografts. Electronic supplementary material The online version of this article (10.1186/s13046-018-0788-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jessica Gasparello
- Department of Life Sciences and Biotechnology, Biochemistry and Molecular Biology Section, Ferrara University, Via Fossato di Mortara 74, 44121, Ferrara, Italy
| | - Matteo Allegretti
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Elisa Tremante
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Enrica Fabbri
- Department of Life Sciences and Biotechnology, Biochemistry and Molecular Biology Section, Ferrara University, Via Fossato di Mortara 74, 44121, Ferrara, Italy
| | | | - Paolo Romania
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Elisa Melucci
- Pathology, IRCSS Regina Elena National Cancer Institute, Rome, Italy
| | - Katia Messana
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Monica Borgatti
- Department of Life Sciences and Biotechnology, Biochemistry and Molecular Biology Section, Ferrara University, Via Fossato di Mortara 74, 44121, Ferrara, Italy
| | - Patrizio Giacomini
- Oncogenomics and Epigenetics, IRCSS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy.
| | - Roberto Gambari
- Department of Life Sciences and Biotechnology, Biochemistry and Molecular Biology Section, Ferrara University, Via Fossato di Mortara 74, 44121, Ferrara, Italy.
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, Biochemistry and Molecular Biology Section, Ferrara University, Via Fossato di Mortara 74, 44121, Ferrara, Italy
| |
Collapse
|