1
|
Jin KM, Bao Q, Zhao TT, Wang HW, Huang LF, Wang K, Xing BC. Comparing baseline VAF in circulating tumor DNA and tumor tissues predicting prognosis of patients with colorectal cancer liver metastases after curative resection. Clin Res Hepatol Gastroenterol 2024; 48:102464. [PMID: 39276854 DOI: 10.1016/j.clinre.2024.102464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 07/12/2024] [Accepted: 09/12/2024] [Indexed: 09/17/2024]
Abstract
INTRODUCTION The prognostic value of baseline variant allele frequency (VAF) in circulating tumor DNA (ctDNA) of colorectal cancer liver metastases (CRLM) patients after curative resection was rarely investigated. METHODS A single-center prospective study was performed to investigate the prognostic impact of baseline VAF in ctDNA and matched tumor tissues of CRLM patients after curative resection between May 2019 and May 2021 by the Illumina NovoSeq 6000 platform. The relationship of the tumor burden score (TBS) and the VAF in ctDNA and matched tumor tissues was evaluated by the Pearson correlation method. The survival curves of recurrence-free survival (RFS) and overall survival (OS) were plotted. Factors associated with RFS were calculated using Cox regression analysis, and an integrated prognostic model using significant baseline variables was proposed. RESULTS There were 121 patients with baseline ctDNA and matched tumor tissues enrolled in the study. A total of 417 mutations spanning 20 genes were identified in baseline tumor tissues of 119/121 (98.3 %) cases. The overall mutations in tumor tissues were completely covered by ctDNA in 52 of 121(43.0 %) patients. Baseline VAF in ctDNA but not in tumor tissues was significantly correlated to TBS of CRLM (R = 0.36, p < 0.001). Significantly longer RFS but not OS was observed in patients with lower VAF in ctDNA compared to those with higher one (p < 0.001 and p = 0.33 respectively). Multivariate Cox regression analysis showed higher VAF in baseline ctDNA was an independent risk factor for RFS. An integrated prognostic model including baseline metastasis location and VAF in ctDNA outperformed the traditional CRS model in predicting RFS. CONCLUSION Baseline VAF in ctDNA but not in tumor tissues influenced RFS of CRLM patients after curative resection.
Collapse
Affiliation(s)
- Ke-Min Jin
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China
| | - Quan Bao
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China
| | - Ting-Ting Zhao
- Research Institute, GloriousMed Clinical Laboratory Co., Ltd., Shanghai, PR China
| | - Hong-Wei Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China
| | - Long-Fei Huang
- Research Institute, GloriousMed Clinical Laboratory Co., Ltd., Shanghai, PR China
| | - Kun Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China.
| | - Bao-Cai Xing
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Hepatobiliary and Pancreatic Surgery Unit I, Peking University Cancer Hospital & Institute, Haidian District, Beijing, PR China.
| |
Collapse
|
2
|
Ye WL, Huang L, Yang XQ, Wan S, Gan WJ, Yang Y, He XS, Liu F, Guo X, Liu YX, Hu G, Li XM, Shi WY, He K, Wu YY, Wu WX, Lu JH, Song Y, Qu CJ, Wu H. TRIM21 induces selective autophagic degradation of c-Myc and sensitizes regorafenib therapy in colorectal cancer. Proc Natl Acad Sci U S A 2024; 121:e2406936121. [PMID: 39388269 PMCID: PMC11494295 DOI: 10.1073/pnas.2406936121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 08/27/2024] [Indexed: 10/12/2024] Open
Abstract
Kirsten rat sarcoma virus (KRAS) mutation is associated with malignant tumor transformation and drug resistance. However, the development of clinically effective targeted therapies for KRAS-mutant cancer has proven to be a formidable challenge. Here, we report that tripartite motif-containing protein 21 (TRIM21) functions as a target of extracellular signal-regulated kinase 2 (ERK2) in KRAS-mutant colorectal cancer (CRC), contributing to regorafenib therapy resistance. Mechanistically, TRIM21 directly interacts with and ubiquitinates v-myc avian myelocytomatosis viral oncogene homolog (c-Myc) at lysine 148 (K148) via K63-linkage, enabling c-Myc to be targeted to the autophagy machinery for degradation, ultimately resulting in the downregulation of enolase 2 expression and inhibition of glycolysis. However, mutant KRAS (KRAS/MT)-driven mitogen-activated protein kinase (MAPK) signaling leads to the phosphorylation of TRIM21 (p-TRIM21) at Threonine 396 (T396) by ERK2, disrupting the interaction between TRIM21 and c-Myc and thereby preventing c-Myc from targeting autophagy for degradation. This enhances glycolysis and contributes to regorafenib resistance. Clinically, high p-TRIM21 (T396) is associated with an unfavorable prognosis. Targeting TRIM21 to disrupt KRAS/MT-driven phosphorylation using the antidepressant vilazodone shows potential for enhancing the efficacy of regorafenib in treating KRAS-mutant CRC in preclinical models. These findings are instrumental for KRAS-mutant CRC treatment aiming at activating TRIM21-mediated selective autophagic degradation of c-Myc.
Collapse
Affiliation(s)
- Wen-Long Ye
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
- Department of Pathology, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou215000, China
| | - Long Huang
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow University, Suzhou215600, China
| | - Xiao-Qin Yang
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Shan Wan
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow University, Suzhou215600, China
| | - Wen-Juan Gan
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
- Department of Pathology, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou215000, China
| | - Yun Yang
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Xiao-Shun He
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Feng Liu
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Xin Guo
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Yi-Xuan Liu
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Guang Hu
- Department of Bioinformatics, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Xiu-Ming Li
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Wei-Yi Shi
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Kuang He
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
- Department of Pathology, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou215000, China
| | - Yue-Yue Wu
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Wen-Xin Wu
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Jun-Hou Lu
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| | - Yu Song
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow University, Suzhou215600, China
| | - Chen-Jiang Qu
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow University, Suzhou215600, China
| | - Hua Wu
- Department of Pathology, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
- Department of Pathology, The Fourth Affiliated Hospital of Soochow University, Soochow University, Suzhou215000, China
- Department of General Surgery, The Affiliated Zhangjiagang Hospital of Soochow University, Soochow University, Suzhou215600, China
- Cancer Institute, Suzhou Medical College of Soochow University, Soochow University, Suzhou215123, China
| |
Collapse
|
3
|
Zhang P, Li X, Wang X, Yang Y, Wang J, Cao D. SHR-8068 combined with adebrelimab and bevacizumab in the treatment of refractory advanced colorectal cancer: study protocol for a single-arm, phase Ib/II study. Front Immunol 2024; 15:1450533. [PMID: 39445023 PMCID: PMC11496094 DOI: 10.3389/fimmu.2024.1450533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 09/13/2024] [Indexed: 10/25/2024] Open
Abstract
Background The third-line treatment for refractory colorectal cancer (CRC) has limited efficacy. This study aimed to evaluate the safety and efficacy of SHR-8068 (an anti-CTLA-4 antibody), combined with adebrelimab (an anti-PD-L1 antibody), and bevacizumab in refractory non-microsatellite instability-high (MSI-H) or proficient mismatch repair (pMMR) CRC. Method This study is a prospective, open-label, single-center phase Ib/II clinical trial. Patients with pathologically confirmed pMMR/non-MSI-H metastatic colorectal adenocarcinoma who have failed ≥2 lines prior standard systemic treatments will be enrolled (n=36). The Ib phase will evaluate two dosing regimens of SHR-8068 in combination therapy (n=9 each dosage): SHR-8068 (1 mg per kilogram, every six weeks, intravenously) or SHR-8068 (4 mg per kilogram, every twelve weeks, intravenously) combined with adebrelimab (1200 mg, every three weeks, intravenously) and bevacizumab (7.5 mg per kilogram, every three weeks, intravenously). The efficacy and adverse events (AEs) of these regimens will be assessed to determine the recommended phase II dose (RP2D) of SHR-8068. Those of RP2D group from the phase Ib will be included in the phase II. The study will go to include 18 additional patients according to the one-sample log-rank test design in the phase II. The primary endpoint of the Ib phase is safety, with secondary endpoints including the objective response rate (ORR), progression-free survival (PFS), overall survival (OS), disease control rate (DCR), and quality of life (QOL). The primary endpoint for phase II was PFS, with secondary endpoints including ORR, OS, DCR, safety, and QOL. Identifying biomarkers to predict the efficacy of this regimen is the exploratory study endpoint. Discussion This proof-of-concept study would provide safety and efficacy signals of this novel combination treatment for the MSS CRCs in the late-line setting. And it may offer new insights on the clinical application of dual immunotherapy combined with anti-angiogenic therapy in the MSS CRC.
Collapse
Affiliation(s)
- Pei Zhang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaofen Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jianfei Wang
- Jiangsu Hengrui Pharmaceuticals Co., Ltd,
Shanghai, China
| | - Dan Cao
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
4
|
Pavlakis N, Shitara K, Sjoquist K, Martin A, Jaworski A, Tebbutt N, Bang YJ, Alcindor T, O'Callaghan C, Strickland A, Rha SY, Lee KW, Kim JS, Bai LY, Hara H, Oh DY, Yip S, Zalcberg J, Price T, Simes J, Goldstein D. INTEGRATE IIa Phase III Study: Regorafenib for Refractory Advanced Gastric Cancer. J Clin Oncol 2024:JCO2400055. [PMID: 39365958 DOI: 10.1200/jco.24.00055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 06/07/2024] [Accepted: 07/17/2024] [Indexed: 10/06/2024] Open
Abstract
PURPOSE Treatment options for refractory advanced gastric and esophagogastric junction cancer (AGOC) are limited. Regorafenib, an oral multikinase inhibitor, prolonged progression-free survival (PFS) versus placebo in the INTEGRATE I phase II trial. INTEGRATE IIa was designed to examine whether regorafenib improved overall survival (OS). METHODS A double-blind placebo-controlled phase III trial compared regorafenib and best supportive care (BSC) versus placebo and BSC for participants with confirmed evaluable metastatic/advanced AGOC who failed ≥two prior therapies on a 2:1 random assignment, stratified by tumor location, geographic region (Asia v rest of world), and prior vascular endothelial growth factor inhibitors. The primary end point was OS. Treatment efficacy on OS was first tested in the pooled INTEGRATE I + INTEGRATE IIa cohort and, if significant, then in the INTEGRATE IIa cohort. Secondary end points were PFS, objective response rate, safety, and quality of life (QoL). RESULTS INTEGRATE IIa enrolled 251 participants: 157 from Asia and 94 from rest of world and 169 received regorafenib and 82 received placebo. No significant heterogeneity was observed between INTEGRATE I and INTEGRATE IIa studies on OS. Pooled OS analysis hazard ratio (HR) was 0.70 (95% CI, 0.56 to 0.87; P = .001; 361 events). INTEGRATE IIa alone OS HR was 0.68 (95% CI, 0.52 to 0.90; P = .006; 238 events), the median OS was 4.5 months versus 4.0 months, and 12-month survival rates were 19% and 6%, for regorafenib versus placebo, respectively. After a preplanned adjustment for multiplicity, there were no statistically significant differences across regions or other prespecified subgroups. Regorafenib improved PFS (HR, 0.53 [95% CI, 0.40 to 0.70]; P < .0001) and delayed deterioration in global QoL (HR, 0.68 [95% CI, 0.52 to 0.89]; P = .0043). The toxicity profile was consistent with that of previous reports. CONCLUSION Regorafenib improves survival compared with placebo in refractory AGOC.
Collapse
Affiliation(s)
- Nick Pavlakis
- Department of Medical Oncology, Royal North Shore Hospital, Sydney, NSW, Australia
- University of Sydney, Sydney, NSW, Australia
| | - Kohei Shitara
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa City, Japan
| | - Katrin Sjoquist
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
- Cancer Care Centre, St George Hospital, Kogarah, NSW, Australia
| | - Andrew Martin
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Anthony Jaworski
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - Niall Tebbutt
- Olivia Newton-John Cancer Wellness & Research Centre, Melbourne, VIC, Australia
| | - Yung-Jue Bang
- Seoul National University College of Medicine, Seoul, South Korea
| | | | - Chris O'Callaghan
- Canadian Cancer Trials Group, Queens University, Kingston, ON, Canada
| | - Andrew Strickland
- Department of Medical Oncology, Monash Health, Monash University, Melbourne, VIC, Australia
| | - Sun Young Rha
- Yonsei Cancer Centre, Yonsei University Health System, Seoul, South Korea
| | - Keun-Wook Lee
- Seoul National University College of Medicine, Seoul, South Korea
- Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Jin-Soo Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Li-Yuan Bai
- Division of Hematology and Oncology, China Medical University Hospital, Taichung, Taiwan
- China Medical University, Taichung, Taiwan
| | | | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Jongno-gu, Seoul National University College of Medicine, South Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, South Korea
| | - Sonia Yip
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - John Zalcberg
- Department of Medical Oncology, Alfred Health, Melbourne, VIC, Australia
- School of Public Health, Faculty of Medicine Monash University, Melbourne, VIC, Australia
| | - Tim Price
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
| | - John Simes
- NHMRC Clinical Trials Centre, University of Sydney, Sydney, NSW, Australia
| | - David Goldstein
- Nelune Cancer Centre, Prince of Wales Hospital, Sydney, NSW, Australia
| |
Collapse
|
5
|
Shanthamallu US, Kilpatrick C, Jones A, Rubin J, Saleh A, Barabási AL, Akmaev VR, Ghiassian SD. A Network-Based Framework to Discover Treatment-Response-Predicting Biomarkers for Complex Diseases. J Mol Diagn 2024; 26:917-930. [PMID: 39067570 DOI: 10.1016/j.jmoldx.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 06/10/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
The potential of precision medicine to transform complex autoimmune disease treatment is often challenged by limited data availability and inadequate sample size when compared with the number of molecular features found in high-throughput multi-omics data sets. To address this issue, the novel framework PRoBeNet (Predictive Response Biomarkers using Network medicine) was developed. PRoBeNet operates under the hypothesis that the therapeutic effect of a drug propagates through a protein-protein interaction network to reverse disease states. PRoBeNet prioritizes biomarkers by considering i) therapy-targeted proteins, ii) disease-specific molecular signatures, and iii) an underlying network of interactions among cellular components (the human interactome). PRoBeNet helped discover biomarkers predicting patient responses to both an established autoimmune therapy (infliximab) and an investigational compound (a mitogen-activated protein kinase 3/1 inhibitor). The predictive power of PRoBeNet biomarkers was validated with retrospective gene-expression data from patients with ulcerative colitis and rheumatoid arthritis and prospective data from tissues from patients with ulcerative colitis and Crohn disease. Machine-learning models using PRoBeNet biomarkers significantly outperformed models using either all genes or randomly selected genes, especially when data were limited. These results illustrate the value of PRoBeNet in reducing features and for constructing robust machine-learning models when data are limited. PRoBeNet may be used to develop companion and complementary diagnostic assays, which may help stratify suitable patient subgroups in clinical trials and improve patient outcomes.
Collapse
Affiliation(s)
- Uday S Shanthamallu
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts
| | - Casey Kilpatrick
- Department of Therapeutics, Scipher Medicine, Waltham, Massachusetts
| | - Alex Jones
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts
| | | | - Alif Saleh
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts
| | - Albert-László Barabási
- Center for Complex Network Research, Northeastern University, Boston, Massachusetts; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts; Department of Network and Data Science, Central European University, Budapest, Hungary
| | - Viatcheslav R Akmaev
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts
| | - Susan D Ghiassian
- Department of Data Science and Network Medicine, Scipher Medicine, Waltham, Massachusetts.
| |
Collapse
|
6
|
Ishizaki T, Sugimoto M, Kuboyama Y, Mazaki J, Kasahara K, Tago T, Udo R, Iwasaki K, Hayashi Y, Nagakawa Y. Stage-Specific Plasma Metabolomic Profiles in Colorectal Cancer. J Clin Med 2024; 13:5202. [PMID: 39274416 PMCID: PMC11396754 DOI: 10.3390/jcm13175202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Background/Objectives: The objective of this study was to investigate the metabolomic profiles of patients with colorectal cancer (CRC) across various stages of the disease. Methods: The plasma samples were obtained from 255 subjects, including patients with CRC in stages I-IV, polyps, and controls. We employed capillary electrophoresis time-of-flight mass spectrometry and liquid chromatography triple quadrupole mass spectrometry to analyze hydrophilic metabolites comprehensively. The data were randomly divided into two groups, and consistent differences observed in both groups were analyzed. Results: Acetylated polyamines, such as N1-acetylspermine and N1, N12-diacetylspermine, consistently showed elevated concentrations in stage IV compared to stages I-III. Non-acetylated polyamines, including spermine and spermidine, exhibited increasing trends from polyp to stage IV. Other metabolites, such as histidine and o-acetylcarnitine, showed decreasing trends across stages. While acetylated polyamines have been reported as CRC detection markers, our findings suggest that they also possess diagnostic potential for distinguishing stage IV from other stages. Conclusions: This study showed stage-specific changes in metabolic profiles, including polyamines, of colorectal cancer.
Collapse
Affiliation(s)
- Tetsuo Ishizaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Masahiro Sugimoto
- Institute of Medical Science, Tokyo Medical University, Shinjuku 160-8402, Tokyo, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka 997-0052, Yamagata, Japan
| | - Yu Kuboyama
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Junichi Mazaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Kenta Kasahara
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Tomoya Tago
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Ryutaro Udo
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Kenichi Iwasaki
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Yutaka Hayashi
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| | - Yuichi Nagakawa
- Department of Gastrointestinal and Pediatric Surgery, Tokyo Medical University, 6-7-1 Nishi-Shinjuku, Shinjuku 160-0023, Tokyo, Japan
| |
Collapse
|
7
|
Adenis A, Ghiringhelli F, Gauthier L, Mazard T, Evesque L, Evrard A, Chalbos P, Moussion A, Gourgou S, Ychou M. Regorafenib plus FOLFIRINOX as first-line treatment for patients with RAS-mutant metastatic colorectal cancer (FOLFIRINOX-R trial): a dose-escalation study. Cancer Chemother Pharmacol 2024; 94:443-452. [PMID: 38987363 PMCID: PMC11420384 DOI: 10.1007/s00280-024-04682-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 05/29/2024] [Indexed: 07/12/2024]
Abstract
PURPOSE The combination of bevacizumab and FOLFIRINOX is used in patients with RAS-mutant metastatic colorectal cancer (RASm-mCRC). Regorafenib, an oral multi-tyrosine kinase inhibitor, has antiangiogenic properties, cytostatic effects and also true cytotoxic effects, unlike bevacizumab. The aim of this study was to determine the maximum tolerated dose (MTD) and the recommended phase 2 dose (RP2D) of the regorafenib-FOLFIRINOX combination in patients with RASm-mCRC. METHODS The FOLFIRINOX-R trial was a phase 1/2 study where the dose-escalation part (3 + 3 design with three dose levels, DLs) was completed before its early termination. FOLFIRINOX (14-day cycle) included oxaliplatin (standard dose), folinic acid, fluorouracil and irinotecan (150 or 180 mg/m²). Regorafenib (120 or 160 mg daily) was given from day 4 to day 10 of each cycle. Dose-limiting toxicity (DLT) was studied in the first three cycles. Eligibility criteria included ECOG performance status ≤ 1 and not previously treated RASm-mCRC. RESULTS Thirteen patients (median age: 65 years; min-max: 40-76) were enrolled. DLT could not be evaluated in one patient (DL3) due to poor observance. The median treatment duration and median follow-up were 6.2 (min-max: 2.3-10) and 13.4 (min-max: 3.8-18.0) months, respectively. Dose was modified in 12/13 (92%) patients. One grade 3 hypokalemia occurred at DL2. MTD was not reached at DL3. Grade 3 diarrhea was recorded in 7/13 patients (13 events) equally distributed in all DLs. CONCLUSION The RP2D for this regorafenib-FFX combination could not be determined due to a high prevalence of grade 3 diarrhea related to treatment as advised by our Independent Data Monitoring Committee. TRIAL REGISTRATION NUMBERS ClinicalTrials.gov : NCT03828799.
Collapse
Affiliation(s)
- Antoine Adenis
- Medical Oncology Department, Montpellier Cancer Institute (ICM), INSERM U1194, Montpellier University, Montpellier, 34298, France.
| | | | - Ludovic Gauthier
- Biometrics Unit, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | - Thibault Mazard
- Medical Oncology Department, Montpellier Cancer Institute (ICM), INSERM U1194, Montpellier University, Montpellier, 34298, France
| | - Ludovic Evesque
- Medical Oncology Department, Centre Antoine Lacassagne, Nice, France
| | - Alexandre Evrard
- Montpellier Cancer Research Institute (IRCM), INSERM U1194, Montpellier University, Montpellier, France
- Laboratory of Biochemistry and Molecular Biology, Nimes University Hospital, IRCM, University of Montpellier, INSERM, Montpellier, France
| | - Patrick Chalbos
- Clinical Research and Innovation Department, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | - Aurore Moussion
- Clinical Research and Innovation Department, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | - Sophie Gourgou
- Biometrics Unit, Montpellier Cancer Institute (ICM), Montpellier University, Montpellier, France
| | - Marc Ychou
- Medical Oncology Department, Montpellier Cancer Institute (ICM), INSERM U1194, Montpellier University, Montpellier, 34298, France
| |
Collapse
|
8
|
Suenaga M, Mashima T, Kawata N, Dan S, Seimiya H, Yamaguchi K. Exploratory Study Identifies Matrix Metalloproteinase-14 and -9 as Potential Biomarkers of Regorafenib Efficacy in Metastatic Colorectal Cancer. Cancers (Basel) 2024; 16:2855. [PMID: 39199626 PMCID: PMC11352555 DOI: 10.3390/cancers16162855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/11/2024] [Accepted: 08/14/2024] [Indexed: 09/01/2024] Open
Abstract
In identifying biomarkers for anticancer drugs, the lack of objectivity in selecting candidate factors makes interpretation difficult. We performed preclinical analysis and a translational validation study to identify candidate biomarkers for regorafenib efficacy in metastatic colorectal cancer (mCRC). Using in silico COMPARE analysis with a human cancer cell line panel, JFCR39, we selected candidate biomarkers whose expression correlates with regorafenib sensitivity. We validated predictive values in mCRC patients receiving regorafenib (discovery, n = 53) and FTD/TPI (control, n = 16). Blood samples were obtained at baseline (BL), before the second cycle (2nd), and at progressive disease (PD), and biomarker levels were measured using ELISA. Our analysis showed that high matrix metalloproteinase (MMP)-14 expression was associated with a high sensitivity to regorafenib. In the discovery cohort, high MMP-14 levels at BL and PD were correlated with tumor shrinkage and longer progression-free survival (PFS). A subsequent analysis of other related factors further indicated that the patients with decreased MMP-9 levels at the 2nd had higher disease control rates, tumor shrinkage, longer PFS, and overall survival than those with increased changes. These findings were not observed in the control cohort. Our study suggests MMP-14 and MMP-9 may serve as prognostic markers for regorafenib and provide insights into novel combination therapies with anti-MMP-9 agents or FTD/TPI.
Collapse
Affiliation(s)
- Mitsukuni Suenaga
- Gastroenterology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (N.K.); (K.Y.)
- Department of Clinical Oncology, Tokyo Medical and Dental University (TMDU), 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | - Tetsuo Mashima
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (T.M.); (H.S.)
| | - Naomi Kawata
- Gastroenterology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (N.K.); (K.Y.)
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (T.M.); (H.S.)
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan;
| | - Hiroyuki Seimiya
- Division of Molecular Biotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (T.M.); (H.S.)
| | - Kensei Yamaguchi
- Gastroenterology Center, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31 Ariake, Koto-ku, Tokyo 135-8550, Japan; (N.K.); (K.Y.)
| |
Collapse
|
9
|
Cervantes A, Tabernero J, Garcia-Carbonero R, Sastre J, Feliu J, Carmen Guillén-Ponce, Paredes BG, Carral A, Muñoz J. Regorafenib in patients with metastatic colorectal cancer in Spain: from clinical trials to real-world evidence. Future Oncol 2024; 20:1401-1413. [PMID: 38861286 PMCID: PMC11385448 DOI: 10.1080/14796694.2024.2340422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 04/04/2024] [Indexed: 06/12/2024] Open
Abstract
Aim: To describe the evolution of regorafenib use, since its approval, in patients with previously treated metastatic colorectal cancer (mCRC) in routine clinical practice in Spain.Methods: We extracted patient characteristics, dosing, safety and efficacy data for the Spanish cohorts of the CORRECT and CONSIGN trials, and the real-world CORRELATE study.Results: The Spanish cohorts represented 10.7-13.8% of the global cohorts. Efficacy and safety in the Spanish cohorts reflected findings from the global cohorts, with evidence of a flexible dosing approach being adopted in routine clinical practice.Conclusion: Regorafenib use in patients with mCRC has evolved in the real-world setting, emphasizing the need for further research evaluating dosing patterns that can optimize clinical outcomes in these patients.Clinical trial registration: The CORRECT trial is registered at ClinicalTrials.gov, number NCT01103323; the CONSIGN trial is registered at ClinicalTrials.gov, number NCT01538680; the CORRELATE study is registered at ClinicalTrials.gov, number NCT02042144.
Collapse
Affiliation(s)
- Andres Cervantes
- Medical Oncology Department, Biomedical Research Institute INCLIVA, Hospital Clínico de Valencia, Universidad de Valencia, Valencia, Spain
- CIBERONC, Instituto de Salud Carlos III, Madrid, Spain
| | - Josep Tabernero
- Medical Oncology Department, Vall d'Hebron Hospital Campus and Institute of Oncology (VHIO), UVic-UCC, IOB-Quiron, Barcelona, Spain
| | - Rocio Garcia-Carbonero
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Imas12, UCM, CIBERONC, Madrid, Spain
| | - Javier Sastre
- Medical Oncology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - Jaime Feliu
- Medical Oncology Department, Hospital Universitario La Paz, IDIPAZ, CIBERONC, Cátedra UAM-AMGEN, Madrid, Spain
| | - Carmen Guillén-Ponce
- Medical Oncology Department, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | | | - Alberto Carral
- Medical Oncology Department, Hospital Universitario Lucus Augusti, Lugo, Spain
| | - Jorge Muñoz
- Medical Oncology Department, Hospital San Pedro de Alcántara, Cáceres, Spain
| |
Collapse
|
10
|
Hedayat S, Cascione L, Cunningham D, Schirripa M, Lampis A, Hahne JC, Tunariu N, Hong SP, Marchetti S, Khan K, Fontana E, Angerilli V, Delrieux M, Nava Rodrigues D, Procaccio L, Rao S, Watkins D, Starling N, Chau I, Braconi C, Fotiadis N, Begum R, Guppy N, Howell L, Valenti M, Cribbes S, Kolozsvari B, Kirkin V, Lonardi S, Ghidini M, Passalacqua R, Elghadi R, Magnani L, Pinato DJ, Di Maggio F, Ghelardi F, Sottotetti E, Vetere G, Ciracì P, Vlachogiannis G, Pietrantonio F, Cremolini C, Cortellini A, Loupakis F, Fassan M, Valeri N. Circulating microRNA Analysis in a Prospective Co-clinical Trial Identifies MIR652-3p as a Response Biomarker and Driver of Regorafenib Resistance Mechanisms in Colorectal Cancer. Clin Cancer Res 2024; 30:2140-2159. [PMID: 38376926 DOI: 10.1158/1078-0432.ccr-23-2748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 02/15/2024] [Accepted: 02/16/2024] [Indexed: 02/21/2024]
Abstract
PURPOSE The multi-kinase inhibitor (mKi) regorafenib has demonstrated efficacy in chemorefractory patients with metastatic colorectal cancer (mCRC). However, lack of predictive biomarkers and concerns over significant toxicities hamper the use of regorafenib in clinical practice. EXPERIMENTAL DESIGN Serial liquid biopsies were obtained at baseline and monthly until disease progression in chemorefractory patients with mCRC treated with regorafenib in a phase II clinical trial (PROSPECT-R n = 40; NCT03010722) and in a multicentric validation cohort (n = 241). Tissue biopsies collected at baseline, after 2 months and at progression in the PROSPECT-R trial were used to establish patient-derived organoids (PDO) and for molecular analyses. MicroRNA profiling was performed on baseline bloods using the NanoString nCounter platform and results were validated by digital-droplet PCR and/or ISH in paired liquid and tissue biopsies. PDOs co-cultures and PDO-xenotransplants were generated for functional analyses. RESULTS Large-scale microRNA expression analysis in longitudinal matched liquid and tissue biopsies from the PROSPECT-R trial identified MIR652-3p as a biomarker of clinical benefit to regorafenib. These findings were confirmed in an independent validation cohort and in a "control" group of 100 patients treated with lonsurf. Using ex vivo co-culture assays paired with single-cell RNA-sequencing of PDO established pre- and post-treatment, we modeled regorafenib response observed in vivo and in patients, and showed that MIR652-3p controls resistance to regorafenib by impairing regorafenib-induced lethal autophagy and by orchestrating the switch from neo-angiogenesis to vessel co-option. CONCLUSIONS Our results identify MIR652-3p as a potential biomarker and as a driver of cell and non-cell-autonomous mechanisms of resistance to regorafenib.
Collapse
Affiliation(s)
- Somaieh Hedayat
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Luciano Cascione
- Bioinformatics Core Unit, Institute of Oncology Research (IOR), Faculty of Biomedical Sciences, Università della Svizzera italiana, Bellinzona, Switzerland
- Swiss Institute of Bioinformatics, Bellinzona, Switzerland
| | - David Cunningham
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Marta Schirripa
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Andrea Lampis
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Jens C Hahne
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Nina Tunariu
- Department of Radiology, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Sung Pil Hong
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Silvia Marchetti
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Khurum Khan
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Elisa Fontana
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Valentina Angerilli
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Mia Delrieux
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Daniel Nava Rodrigues
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Letizia Procaccio
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Sheela Rao
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - David Watkins
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Naureen Starling
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Ian Chau
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Chiara Braconi
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
- Institute of Cancer Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Nicos Fotiadis
- Department of Interventional Radiology, The Royal Marsden Hospital, London, United Kingdom
| | - Ruwaida Begum
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
| | - Naomy Guppy
- Breast Cancer Now Nina Barough Pathology Core Facility, The Institute of Cancer Research, London, United Kingdom
| | - Louise Howell
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
| | - Melanie Valenti
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | | | | | - Vladimir Kirkin
- Division of Cancer Therapeutics, The Institute of Cancer Research, London, United Kingdom
| | - Sara Lonardi
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Michele Ghidini
- Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Raghad Elghadi
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Luca Magnani
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - David J Pinato
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Federica Di Maggio
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy
- CEINGE-Biotecnologie Avanzate Francesco Salvatore, Via Gaetano Salvatore, Naples, Italy
| | - Filippo Ghelardi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Guglielmo Vetere
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Paolo Ciracì
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Georgios Vlachogiannis
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Chiara Cremolini
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Alessio Cortellini
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
- Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Fotios Loupakis
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
| | - Matteo Fassan
- Istituto Oncologico Veneto, Istituto di Ricovero e Cura a Carattere Scientifico, Padua, Italy
- Department of Medicine, Surgical Pathology Unit, University of Padua, Padua, Italy
| | - Nicola Valeri
- Division of Molecular Pathology and Centre for Evolution and Cancer, The Institute of Cancer Research, London, United Kingdom
- Department of Medicine, The Royal Marsden Hospital, London and Sutton, United Kingdom
- Division of Surgery and Cancer, Imperial College London, London, United Kingdom
| |
Collapse
|
11
|
Corrias G, Lai E, Ziranu P, Mariani S, Donisi C, Liscia N, Saba G, Pretta A, Persano M, Fanni D, Spanu D, Balconi F, Loi F, Deidda S, Restivo A, Pusceddu V, Puzzoni M, Solinas C, Massa E, Madeddu C, Gerosa C, Zorcolo L, Faa G, Saba L, Scartozzi M. Prediction of Response to Anti-Angiogenic Treatment for Advanced Colorectal Cancer Patients: From Biological Factors to Functional Imaging. Cancers (Basel) 2024; 16:1364. [PMID: 38611042 PMCID: PMC11011199 DOI: 10.3390/cancers16071364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Colorectal cancer (CRC) is a leading tumor worldwide. In CRC, the angiogenic pathway plays a crucial role in cancer development and the process of metastasis. Thus, anti-angiogenic drugs represent a milestone for metastatic CRC (mCRC) treatment and lead to significant improvement of clinical outcomes. Nevertheless, not all patients respond to treatment and some develop resistance. Therefore, the identification of predictive factors able to predict response to angiogenesis pathway blockade is required in order to identify the best candidates to receive these agents. Unfortunately, no predictive biomarkers have been prospectively validated to date. Over the years, research has focused on biologic factors such as genetic polymorphisms, circulating biomarkers, circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and microRNA. Moreover, research efforts have evaluated the potential correlation of molecular biomarkers with imaging techniques used for tumor assessment as well as the application of imaging tools in clinical practice. In addition to functional imaging, radiomics, a relatively newer technique, shows real promise in the setting of correlating molecular medicine to radiological phenotypes.
Collapse
Affiliation(s)
- Giuseppe Corrias
- Department of Radiology, University of Cagliari, 09042 Cagliari, Italy;
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clelia Donisi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Nicole Liscia
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy;
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Mara Persano
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Daniela Fanni
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Francesca Balconi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Francesco Loi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Simona Deidda
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Angelo Restivo
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Cinzia Solinas
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Elena Massa
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clelia Madeddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clara Gerosa
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Luigi Zorcolo
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Gavino Faa
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Luca Saba
- Department of Radiology, University of Cagliari, 09042 Cagliari, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| |
Collapse
|
12
|
Najafi S, Majidpoor J, Mortezaee K. Liquid biopsy in colorectal cancer. Clin Chim Acta 2024; 553:117674. [PMID: 38007059 DOI: 10.1016/j.cca.2023.117674] [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: 09/09/2023] [Revised: 11/16/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
Liquid biopsy refers to a set of pathological samples retrieved from non-solid sources, such as blood, cerebrospinal fluid, urine, and saliva through non-invasive or minimally invasive approaches. In the recent decades, an increasing number of studies have focused on clinical applications and improving technological investigation of liquid biopsy biosources for diagnostic goals particularly in cancer. Materials extracted from these sources and used for medical evaluations include cells like circulating tumor cells (CTCs), tumor-educated platelets (TEPs), cell-free nucleic acids released by cells, such as circulating tumor DNA (ctDNA), cell-free DNA (cfDNA), cell-free RNA (cfRNA), and exosomes. Playing significant roles in the pathogenesis of human malignancies, analysis of these sources can provide easier access to genetic and transcriptomic information of the cancer tissue even better than the conventional tissue biopsy. Notably, they can represent the inter- and intra-tumoral heterogeneity and accordingly, liquid biopsies demonstrate strengths for improving diagnosis in early detection and screening, monitoring and follow-up after therapies, and personalization of therapeutical strategies in various types of human malignancies. In this review, we aim to discuss the roles, functions, and analysis approaches of liquid biopsy sources and their clinical implications in human malignancies with a focus on colorectal cancer.
Collapse
Affiliation(s)
- Sajad Najafi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamal Majidpoor
- Department of Anatomy, School of Medicine, Infectious Diseases Research Center, Gonabad University of Medical Sciences, Gonabad, Iran
| | - Keywan Mortezaee
- Department of Anatomy, School of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
13
|
Parigger T, Gassner FJ, Drothler S, Scherhäufl C, Hödlmoser A, Schultheis L, Bakar AA, Huemer F, Greil R, Geisberger R, Weiss L, Zaborsky N. Combined DNA Analysis from Stool and Blood Samples Improves Tumor Tracking and Assessment of Clonal Heterogeneity in Localized Rectal Cancer Patients. Technol Cancer Res Treat 2024; 23:15330338241252706. [PMID: 38766867 PMCID: PMC11104029 DOI: 10.1177/15330338241252706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 03/20/2024] [Accepted: 04/08/2024] [Indexed: 05/22/2024] Open
Abstract
Objectives: In this study, stool samples were evaluated for tumor mutation analysis via a targeted next generation sequencing (NGS) approach in a small patient cohort suffering from localized rectal cancer. Introduction: Colorectal cancer (CRC) causes the second highest cancer-related death rate worldwide. Thus, improvements in disease assessment and monitoring that may facilitate treatment allocation and allow organ-sparing "watch-and-wait" treatment strategies are highly relevant for a significant number of CRC patients. Methods: Stool-based results were compared with mutation profiles derived from liquid biopsies and the gold standard procedure of tumor biopsy from the same patients. A workflow was established that enables the detection of de-novo tumor mutations in stool samples of CRC patients via ultra-sensitive cell-free tumor DNA target enrichment. Results: Notably, only a 19% overall concordance was found in mutational profiles across the compared sample specimens of stool, tumor, and liquid biopsies. Conclusion: Based on these results, the analysis of stool and liquid biopsy samples can provide important additional information on tumor heterogeneity and potentially on the assessment of minimal residual disease and clonal tumor evolution.
Collapse
Affiliation(s)
- Thomas Parigger
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Franz Josef Gassner
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Stephan Drothler
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Department of Biosciences, Paris-Lodron-University Salzburg, Salzburg, Austria
| | - Christian Scherhäufl
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Department of Biosciences, Paris-Lodron-University Salzburg, Salzburg, Austria
| | - Alexandra Hödlmoser
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Lena Schultheis
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Aryunni Abu Bakar
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Department of Biosciences, Paris-Lodron-University Salzburg, Salzburg, Austria
| | - Florian Huemer
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Richard Greil
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Roland Geisberger
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Lukas Weiss
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Nadja Zaborsky
- Department Laboratory of Immunological and Molecular Cancer Research-Salzburg Cancer Research Institute, Cancer Cluster Salzburg, Salzburg, Austria
- Department of Internal Medicine III with Haematology, Medical Oncology, Haemostaseology, Infectiology and Rheumatology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| |
Collapse
|
14
|
Sunakawa Y, Kuboki Y, Watanabe J, Terazawa T, Kawakami H, Yokota M, Nakamura M, Kotaka M, Sugimoto N, Ojima H, Oki E, Kajiwara T, Yamamoto Y, Tsuji Y, Denda T, Tamura T, Ishihara S, Taniguchi H, Nakajima TE, Morita S, Shirao K, Takenaka N, Ozawa D, Yoshino T. Exploratory Biomarker Analysis Using Plasma Angiogenesis-Related Factors and Cell-Free DNA in the TRUSTY Study: A Randomized, Phase II/III Study of Trifluridine/Tipiracil Plus Bevacizumab as Second-Line Treatment for Metastatic Colorectal Cancer. Target Oncol 2024; 19:59-69. [PMID: 38194163 PMCID: PMC10830797 DOI: 10.1007/s11523-023-01027-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/02/2023] [Indexed: 01/10/2024]
Abstract
BACKGROUND The TRUSTY study evaluated the efficacy of second-line trifluridine/tipiracil (FTD/TPI) plus bevacizumab in metastatic colorectal cancer (mCRC). OBJECTIVE This exploratory biomarker analysis of TRUSTY investigated the relationship between baseline plasma concentrations of angiogenesis-related factors and cell-free DNA (cfDNA), and the efficacy of FTD/TPI plus bevacizumab in patients with mCRC. PATIENTS AND METHODS The disease control rate (DCR) and progression-free survival (PFS) were compared between baseline plasma samples of patients with high and low plasma concentrations (based on the median value) of angiogenesis-related factors. Correlations between cfDNA concentrations and PFS were assessed. RESULTS Baseline characteristics (n = 65) were as follows: male/female, 35/30; median age, 64 (range 25-84) years; and RAS status wild-type/mutant, 29/36. Patients in the hepatocyte growth factor (HGF)-low and interleukin (IL)-8-low groups had a significantly higher DCR (risk ratio [95% confidence intervals {CIs}]) than patients in the HGF-high (1.83 [1.12-2.98]) and IL-8-high (1.70 [1.02-2.82]) groups. PFS (hazard ratio {HR} [95% CI]) was significantly longer in patients in the HGF-low (0.33 [0.14-0.79]), IL-8-low (0.31 [0.14-0.70]), IL-6-low (0.19 [0.07-0.50]), osteopontin-low (0.39 [0.17-0.88]), thrombospondin-2-low (0.42 [0.18-0.98]), and tissue inhibitor of metalloproteinase-1-low (0.26 [0.10-0.67]) groups versus those having corresponding high plasma concentrations of these angiogenesis-related factors. No correlation was observed between cfDNA concentration and PFS. CONCLUSION Low baseline plasma concentrations of HGF and IL-8 may predict better DCR and PFS in patients with mCRC receiving FTD/TPI plus bevacizumab, however further studies are warranted. CLINICAL TRIAL REGISTRATION NUMBER jRCTs031180122.
Collapse
Affiliation(s)
- Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki, Kanagawa, 216-8511, Japan.
| | - Yasutoshi Kuboki
- Department of Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Jun Watanabe
- Department of Surgery, Gastroenterological Center, Yokohama City University Medical Center, Yokohama, Japan
| | - Tetsuji Terazawa
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University, Takatsuki, Japan
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of Medicine Hospital, Osaka-Sayama, Japan
| | - Mitsuru Yokota
- Department of General Surgery, Kurashiki Central Hospital, Kurashiki, Japan
| | - Masato Nakamura
- Aizawa Comprehensive Cancer Center, Aizawa Hospital, Matsumoto, Japan
| | | | - Naotoshi Sugimoto
- Department of Genetic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Hitoshi Ojima
- Department of Gastroenterological Surgery, Gunma Prefectural Cancer Center, Ota, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Science, Kyushu University, Fukuoka, Japan
| | - Takeshi Kajiwara
- Department of Gastrointestinal Medical Oncology, National Hospital Organization Shikoku Cancer Center, Matsuyama, Japan
| | - Yoshiyuki Yamamoto
- Department of Gastroenterology, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yasushi Tsuji
- Department of Medical Oncology, Tonan Hospital, Sapporo, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Takao Tamura
- Department of Medical Oncology, Kindai University Nara Hospital, Ikoma, Japan
| | - Soichiro Ishihara
- Department of Surgical Oncology, The University of Tokyo, Tokyo, Japan
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Takako Eguchi Nakajima
- Department of Early Clinical Development, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | | | - Naruhito Takenaka
- Clinical Development and Medical Affairs Division, Taiho Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Daisuke Ozawa
- Clinical Development and Medical Affairs Division, Taiho Pharmaceutical Co., Ltd, Tokyo, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| |
Collapse
|
15
|
Matsuoka T, Yashiro M. Current status and perspectives of genetic testing in gastrointestinal cancer (Review). Oncol Lett 2024; 27:21. [PMID: 38058469 PMCID: PMC10696628 DOI: 10.3892/ol.2023.14155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/30/2023] [Indexed: 12/08/2023] Open
Abstract
Genetic testing has become widespread in daily medical care for gastrointestinal (GI) cancers. However, unlike breast cancer and non-small cell lung cancer, in which personalized medicine targeting various driver genes is standardized, the incidence of targeted gene abnormalities in GI cancers is low. Nevertheless, such abnormalities may be linked to therapeutic agents and the further development of therapeutic agents for personalized medicine for GI cancers is desired. A liquid biopsy is of great benefit in offering clinical decision support, in applications such as GI cancer screening, surgical interventions, monitoring disease status and enhancing patient survival outcomes, all of which would contribute to personalized medicine. Germline genetic testing is required for several types of GI cancer, which shows clinical indications of hereditary predisposition. The increasing use of multigene panel testing has redefined gene-cancer associations, and consequently the estimate of cancer risk that vary from low to high penetrance. Comprehensive genetic testing can enable the detection of novel treatment targets and the discovery of undefined multiple diagnostic/predictive markers, which may enhance the molecular-level understanding of GI cancers. Genetic testing can also aid the design of more appropriate and adequate genomic-driven therapies for patients who may benefit from other standardized therapeutic methods.
Collapse
Affiliation(s)
- Tasuku Matsuoka
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, Osaka 5458585, Japan
- Institute of Medical Genetics, Osaka Metropolitan University, Osaka 5458585, Japan
| |
Collapse
|
16
|
Gambaro K, Marques M, McNamara S, Couetoux du Tertre M, Hoffert C, Srivastava A, Schab A, Alcindor T, Langleben A, Sideris L, Abdelsalam M, Tehfe M, Couture F, Batist G, Kavan P. A Phase II Exploratory Study to Identify Biomarkers Predictive of Clinical Response to Regorafenib in Patients with Metastatic Colorectal Cancer Who Have Failed First-Line Therapy. Int J Mol Sci 2023; 25:43. [PMID: 38203214 PMCID: PMC10778949 DOI: 10.3390/ijms25010043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
Single-agent regorafenib is approved in Canada for metastatic colorectal cancer (mCRC) patients who have failed previous lines of therapy. Identifying prognostic biomarkers is key to optimizing therapeutic strategies for these patients. In this clinical study (NCT01949194), we evaluated the safety and efficacy of single-agent regorafenib as a second-line therapy for mCRC patients who received it after failing first-line therapy with an oxaliplatin or irinotecan regimen with or without bevacizumab. Using various omics approaches, we also investigated putative biomarkers of response and resistance to regorafenib in metastatic lesions and blood samples in the same cohort. Overall, the safety profile of regorafenib seemed similar to the CORRECT trial, where regorafenib was administered as ≥ 2 lines of therapy. While the mutational landscape showed typical mutation rates for the top five driver genes (APC, KRAS, BRAF, PIK3CA, and TP53), KRAS mutations were enriched in intrinsically resistant lesions. Additional exploration of genomic-phenotype associations revealed several biomarker candidates linked to unfavorable prognoses in patients with mCRC using various approaches, including pathway analysis, cfDNA profiling, and copy number analysis. However, further research endeavors are necessary to validate the potential utility of these promising genes in understanding patients' responses to regorafenib treatment.
Collapse
Affiliation(s)
- Karen Gambaro
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Maud Marques
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Suzan McNamara
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
| | - Mathilde Couetoux du Tertre
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
| | - Cyrla Hoffert
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Archana Srivastava
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Anna Schab
- Canadian National Centres of Excellence-Exactis Innovations, Montreal, QC H3T 1Y6, Canada; (K.G.); (M.M.); (S.M.)
- Consortium de Recherche en Oncologie Clinique du Québec (Q-CROC), Quebec, QC G1V 3X8, Canada
| | | | | | - Lucas Sideris
- Hôpital Maisonneuve Rosemont, Montreal, QC H1T 2M4, Canada
| | | | - Mustapha Tehfe
- Hematology-Oncology, Oncology Center-Centre Hospitalier de l’Université de Montreal, Montreal, QC H2X 0C1, Canada
| | | | - Gerald Batist
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| | - Petr Kavan
- Segal Cancer Centre-Jewish General Hospital, Montreal, QC H3T 1E2, Canada
| |
Collapse
|
17
|
Quan M, Chen J, Chen Z, Hai Y, Zhou Y, Chao Q, Chen C, Li H, Wang M, Gao Y. China special issue on gastrointestinal tumors-Cetuximab retreatment plus camrelizumab and liposomal irinotecan in patients with RAS wild-type metastatic colorectal cancer: Cohort B of the phase II CRACK study. Int J Cancer 2023; 153:1877-1884. [PMID: 37163613 DOI: 10.1002/ijc.34531] [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: 01/05/2023] [Revised: 03/22/2023] [Accepted: 03/23/2023] [Indexed: 05/12/2023]
Abstract
Patients with metastatic colorectal cancer (mCRC) have poor long-term survival. Rechallenge with anti-epidermal growth factor receptor (anti-EGFR) based therapy has shown certain activity as late-line therapy. To further improve clinical outcomes, we evaluated the antitumor efficacy and safety of cetuximab in combination with camrelizumab and liposomal irinotecan in patients with RASwt mCRC pretreated with anti-EGFR-based therapy. Patients with RASwt mCRC who had received at least two prior systemic therapies, including anti-EGFR-based treatment in the metastatic or unresectable disease setting, were enrolled in cohort B. Patients were treated with cetuximab (500 mg/m2 ) and camrelizumab (200 mg) plus liposomal irinotecan (HR070803, 60 mg/m2 ) intravenously once every 2 weeks. The primary endpoint was the objective response rate (ORR) by RECIST v1.1. The secondary endpoints included disease control rate (DCR), progression-free survival (PFS), overall survival (OS) and safety. At the data cutoff (23 November 2022), 19 patients were enrolled in the two stages, and 16 were evaluable for efficacy analyses. The ORR was 25% (95% confidence interval [CI]: 10.2%-49.5%), and DCR was 75% (95% CI: 50.5%-89.8%). The median PFS and OS were 6.9 (95% CI: 2.6-11.2) and 15.1 (95% CI: 6.1-24.0) months, respectively. Grade 3 treatment-related adverse events (TRAEs) occurred in 15.8% (3/19) of patients. No grade ≥4 TRAEs were found in the safety population. Our study suggests that anti-EGFR retreatment therapy with cetuximab plus camrelizumab and liposomal irinotecan (HR070803) is a promising late-line treatment option with good antitumor activity and well-tolerated toxicity in RASwt mCRC patients.
Collapse
Affiliation(s)
- Ming Quan
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingde Chen
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiqin Chen
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yannan Hai
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Zhou
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qian Chao
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Chen
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Huajun Li
- Jiangsu Hengrui Pharmaceuticals Co., Ltd, Shanghai, China
| | - Mei Wang
- Department of Oncology, Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yong Gao
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
18
|
Nguyen MTN, Rajavuori A, Huhtinen K, Hietanen S, Hynninen J, Oikkonen J, Hautaniemi S. Circulating tumor DNA-based copy-number profiles enable monitoring treatment effects during therapy in high-grade serous carcinoma. Biomed Pharmacother 2023; 168:115630. [PMID: 37806091 DOI: 10.1016/j.biopha.2023.115630] [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: 07/25/2023] [Revised: 09/23/2023] [Accepted: 10/03/2023] [Indexed: 10/10/2023] Open
Abstract
Circulating tumor DNA (ctDNA) analysis has emerged as a promising tool for detecting and profiling longitudinal genomics changes in cancer. While copy-number alterations (CNAs) play a major role in cancers, treatment effect monitoring using copy-number profiles has received limited attention as compared to mutations. A major reason for this is the insensitivity of CNA analysis for the real-life tumor-fraction ctDNA samples. We performed copy-number analysis on 152 plasma samples obtained from 29 patients with high-grade serous ovarian cancer (HGSC) using a sequencing panel targeting over 500 genes. Twenty-one patients had temporally matched tissue and plasma sample pairs, which enabled assessing concordance with tissues sequenced with the same panel or whole-genome sequencing and to evaluate sensitivity. Our approach could detect concordant CNA profiles in most plasma samples with as low as 5% tumor content and highly amplified regions in samples with ∼1% of tumor content. Longitudinal profiles showed changes in the CNA profiles in seven out of 11 patients with high tumor-content plasma samples at relapse. These changes included focal acquired or lost copy-numbers, even though most of the genome remained stable. Two patients displayed major copy-number profile changes during therapy. Our analysis revealed ctDNA-detectable subclonal selection resulting from both surgical operations and chemotherapy. Overall, longitudinal ctDNA data showed acquired and diminished CNAs at relapse when compared to pre-treatment samples. These results highlight the importance of genomic profiling during treatment as well as underline the usability of ctDNA.
Collapse
Affiliation(s)
- Mai T N Nguyen
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki 00291, Finland
| | - Anna Rajavuori
- Department of Obstetrics and Gynecology, Turku University Hospital, Kiinamyllynkatu 4, Turku 20521, Finland
| | - Kaisa Huhtinen
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki 00291, Finland; Institute of Biomedicine, University of Turku, Kiinamyllynkatu 10, Turku 20014, Finland
| | - Sakari Hietanen
- Department of Obstetrics and Gynecology, Turku University Hospital, Kiinamyllynkatu 4, Turku 20521, Finland
| | - Johanna Hynninen
- Department of Obstetrics and Gynecology, Turku University Hospital, Kiinamyllynkatu 4, Turku 20521, Finland
| | - Jaana Oikkonen
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki 00291, Finland.
| | - Sampsa Hautaniemi
- Research Program in Systems Oncology, Faculty of Medicine, University of Helsinki, Helsinki 00291, Finland.
| |
Collapse
|
19
|
Aydin SG, Kavak EE, Topcu A, Bayramgil A, Akgul F, Kahraman S, Aykan MB, Altıntas YE, Helvaci K, Urun Y, Bilici A, Seker M, Nahit Sendur MA, Olmez OF, Acikgoz O, Cicin I. Prognostic factors for regorafenib treatment in patients with refractory metastatic colorectal cancer: A real-life retrospective multi-center study. BIOMOLECULES & BIOMEDICINE 2023; 23:1089-1095. [PMID: 37289436 PMCID: PMC10655877 DOI: 10.17305/bb.2023.9253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/02/2023] [Accepted: 06/02/2023] [Indexed: 06/09/2023]
Abstract
Regorafenib, an oral multikinase inhibitor, has improved survival in metastatic colorectal cancer (mCRC) patients who have progressed on standard therapies. Our study aimed to evaluate prognostic factors influencing regorafenib treatment and assess the optimal dosing regimen in a real-life setting. We retrospectively analysed 263 patients with mCRC from multiple medical oncology clinics in Turkey. Treatment responses and prognostic factors for survival were evaluated using univariate and multivariate analysis. Of the patients, 120 were male, and 143 were female; 28.9% of tumors were located in the rectum. RAS mutations were present in 3.0% of tumors, while BRAF, K-RAS, and N-RAS mutations were found in 3.0%, 29.7%, and 25.9% of tumor tissues, respectively. Dose escalation was preferred in 105 (39.9%) patients. The median treatment duration was 3.0 months, with an objective response rate (ORR) of 4.9%. Grade ≥ 3 treatment-related toxicity occurred in 133 patients, leading to discontinuation, interruption, and modification rates of 50.6%, 43.7%, and 79.0%, respectively. Median progression-free survival (PFS) and overall survival (OS) were 3.0 and 8.1 months, respectively. RAS/RAF mutation (hazard ratio [HR] 1.5, 95% confidence interval [CI] 1.1-2.3; P = 0.01), pretreatment carcinoembryonic antigen (CEA) levels (HR 1.6, 95% CI 1.1-2.3; P = 0.008), and toxicity-related treatment interruption or dose adjustment (HR 1.6, 95% CI 1.1-2.4; P = 0.01) were identified as independent prognostic factors for PFS. Dose escalation had no significant effect on PFS but was associated with improved OS (P < 0.001). Independent prognostic factors for OS were the initial TNM stage (HR 1.3, 95% CI 1.0-1.9; P = 0.04) and dose interruption/adjustment (HR 0.4, 95% CI 0.2-0.9; P = 0.03). Our findings demonstrate the efficacy and safety of regorafenib. Treatment line influences the response, with dose escalation being more favorable than adjustment or interruption, thus impacting survival.
Collapse
Affiliation(s)
- Sabin Goktas Aydin
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Engin Eren Kavak
- Department of Medical Oncology, Medical Faculty, Ankara University, Ankara, Turkey
| | - Atakan Topcu
- Department of Medical Oncology, Medical Faculty, Bezmialem Vakif University, Istanbul, Turkey
| | - Ayberk Bayramgil
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Fahri Akgul
- Department of Medical Oncology, Medical Faculty, Trakya University, Edirne, Turkey
| | - Seda Kahraman
- Department of Medical Oncology, Medical Faculty, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Musa Baris Aykan
- Department of Medical Oncology, Gulhane Education and Research Hospital, Ankara, Turkey
| | - Yunus Emre Altıntas
- Department of Medical Oncology, Medical Faculty, Koc University, Istanbul, Turkey
| | - Kaan Helvaci
- Department of Medical Oncology, Memorial Ankara Hospital, Ankara, Turkey
| | - Yuksel Urun
- Department of Medical Oncology, Medical Faculty, Ankara University, Ankara, Turkey
| | - Ahmet Bilici
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Mesut Seker
- Department of Medical Oncology, Medical Faculty, Bezmialem Vakif University, Istanbul, Turkey
| | - Mehmet Ali Nahit Sendur
- Department of Medical Oncology, Medical Faculty, Ankara Yildirim Beyazit University, Ankara, Turkey
| | - Omer Fatih Olmez
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Ozgur Acikgoz
- Department of Medical Oncology, Medical Faculty, Medipol University, Istanbul, Turkey
| | - Irfan Cicin
- Department of Medical Oncology, Medical Faculty, Trakya University, Edirne, Turkey
| |
Collapse
|
20
|
Genco E, Modena F, Sarcina L, Björkström K, Brunetti C, Caironi M, Caputo M, Demartis VM, Di Franco C, Frusconi G, Haeberle L, Larizza P, Mancini MT, Österbacka R, Reeves W, Scamarcio G, Scandurra C, Wheeler M, Cantatore E, Esposito I, Macchia E, Torricelli F, Viola FA, Torsi L. A Single-Molecule Bioelectronic Portable Array for Early Diagnosis of Pancreatic Cancer Precursors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2304102. [PMID: 37452695 DOI: 10.1002/adma.202304102] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
A cohort of 47 patients is screened for pancreatic cancer precursors with a portable 96-well bioelectronic sensing-array for single-molecule assay in cysts fluid and blood plasma, deployable at point-of-care (POC). Pancreatic cancer precursors are mucinous cysts diagnosed with a sensitivity of at most 80% by state-of-the-art cytopathological molecular analyses (e.g., KRASmut DNA). Adding the simultaneous assay of proteins related to malignant transformation (e.g., MUC1 and CD55) is deemed essential to enhance diagnostic accuracy. The bioelectronic array proposed here, based on single-molecule-with-a-large-transistor (SiMoT) technology, can assay both nucleic acids and proteins at the single-molecule limit-of-identification (LOI) (1% of false-positives and false-negatives). It comprises an enzyme-linked immunosorbent assay (ELISA)-like 8 × 12-array organic-electronics disposable cartridge with an electrolyte-gated organic transistor sensor array, and a reusable reader, integrating a custom Si-IC chip, operating via software installed on a USB-connected smart device. The cartridge is complemented by a 3D-printed sensing gate cover plate. KRASmut , MUC1, and CD55 biomarkers either in plasma or cysts-fluid from 5 to 6 patients at a time, are multiplexed at single-molecule LOI in 1.5 h. The pancreatic cancer precursors are classified via a machine-learning analysis resulting in at least 96% diagnostic-sensitivity and 100% diagnostic-specificity. This preliminary study opens the way to POC liquid-biopsy-based early diagnosis of pancreatic-cancer precursors in plasma.
Collapse
Affiliation(s)
- Enrico Genco
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Francesco Modena
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Rubattino 81, Milan, 20134, Italy
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Piazza Leonardo da Vinci 32, Milan, 20133, Italy
| | - Lucia Sarcina
- Dipartimento di Chimica and Centre for Colloid and Surface Science, Università degli Studi di Bari Aldo Moro, Bari, 20125, Italy
| | - Kim Björkström
- The Faculty of Science and Engineering, Åbo Akademi University, Turku, 20500, Finland
| | | | - Mario Caironi
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Rubattino 81, Milan, 20134, Italy
| | - Mariapia Caputo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, 70125, Italy
| | - Virginia Maria Demartis
- Dipartimento Ingegneria dell'Informazione, Università degli Studi di Brescia, Brescia, 25123, Italy
| | | | - Giulia Frusconi
- Dipartimento Ingegneria dell'Informazione, Università degli Studi di Brescia, Brescia, 25123, Italy
| | - Lena Haeberle
- Institute of Pathology, Heinrich-Heine University and University Hospital of Düsseldorf, 40225, Duesseldorf, Germany
| | - Piero Larizza
- Masmec Biomed - Masmec SpA division, Modugno (BA), 70026, Italy
| | | | - Ronald Österbacka
- The Faculty of Science and Engineering, Åbo Akademi University, Turku, 20500, Finland
| | | | - Gaetano Scamarcio
- CNR IFN, Bari, 70126, Italy
- Dipartimento Interateneo di Fisica, Università degli Studi di Bari Aldo Moro, Bari, 70125, Italy
| | - Cecilia Scandurra
- Dipartimento di Chimica and Centre for Colloid and Surface Science, Università degli Studi di Bari Aldo Moro, Bari, 20125, Italy
| | - May Wheeler
- FlexEnable Technology Ltd, Cambridge, CB4 0FX, UK
| | - Eugenio Cantatore
- Department of Electrical Engineering, Eindhoven University of Technology, Eindhoven, 5600 MB, The Netherlands
| | - Irene Esposito
- Institute of Pathology, Heinrich-Heine University and University Hospital of Düsseldorf, 40225, Duesseldorf, Germany
| | - Eleonora Macchia
- The Faculty of Science and Engineering, Åbo Akademi University, Turku, 20500, Finland
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari "Aldo Moro", Bari, 70125, Italy
| | - Fabrizio Torricelli
- Dipartimento Ingegneria dell'Informazione, Università degli Studi di Brescia, Brescia, 25123, Italy
| | - Fabrizio Antonio Viola
- Center for Nano Science and Technology, Istituto Italiano di Tecnologia, Via Rubattino 81, Milan, 20134, Italy
| | - Luisa Torsi
- Dipartimento di Chimica and Centre for Colloid and Surface Science, Università degli Studi di Bari Aldo Moro, Bari, 20125, Italy
| |
Collapse
|
21
|
Udagawa S, Ooki A, Shinozaki E, Fukuda K, Yamaguchi K, Osumi H. Circulating Tumor DNA: The Dawn of a New Era in the Optimization of Chemotherapeutic Strategies for Metastatic Colo-Rectal Cancer Focusing on RAS Mutation. Cancers (Basel) 2023; 15:1473. [PMID: 36900264 PMCID: PMC10001242 DOI: 10.3390/cancers15051473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/10/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Genotyping of tumor tissues to assess RAS and BRAF V600E mutations enables us to select optimal molecularly targeted therapies when considering treatment strategies for patients with metastatic colorectal cancer. Tissue-based genetic testing is limited by the difficulty of performing repeated tests, due to the invasive nature of tissue biopsy, and by tumor heterogeneity, which can limit the usefulness of the information it yields. Liquid biopsy, represented by circulating tumor DNA (ctDNA), has attracted attention as a novel method for detecting genetic alterations. Liquid biopsies are more convenient and much less invasive than tissue biopsies and are useful for obtaining comprehensive genomic information on primary and metastatic tumors. Assessing ctDNA can help track genomic evolution and the status of alterations in genes such as RAS, which are sometimes altered following chemotherapy. In this review, we discuss the potential clinical applications of ctDNA, summarize clinical trials focusing on RAS, and present the future prospects of ctDNA analysis that could change daily clinical practice.
Collapse
Affiliation(s)
| | | | | | | | | | - Hiroki Osumi
- Department of Gastroenterology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| |
Collapse
|
22
|
Poad H, Khan S, Wheaton L, Thomas A, Sweeting M, Bujkiewicz S. The Validity of Surrogate Endpoints in Sub Groups of Metastatic Colorectal Cancer Patients Defined by Treatment Class and KRAS Status. Cancers (Basel) 2022; 14:5391. [PMID: 36358810 PMCID: PMC9654686 DOI: 10.3390/cancers14215391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Background and Aim: Findings from the literature suggest that the validity of surrogate endpoints in metastatic colorectal cancer (mCRC) may depend on a treatments' mechanism of action. We explore this and the impact of Kirsten rat sarcoma (KRAS) status on surrogacy patterns in mCRC. Methods: A systematic review was undertaken to identify randomized controlled trials (RCTs) for pharmacological therapies in mCRC. Bayesian meta-analytic methods for surrogate endpoint evaluation were used to evaluate surrogate relationships across all RCTs, by KRAS status and treatment class. Surrogate endpoints explored were progression free survival (PFS) as a surrogate endpoint for overall survival (OS), and tumour response (TR) as a surrogate for PFS and OS. Results: 66 RCTs were identified from the systematic review. PFS showed a strong surrogate relationship with OS across all data and in subgroups by KRAS status. The relationship appeared stronger within individual treatment classes compared to the overall analysis. The TR-PFS and TR-OS relationships were found to be weak overall but stronger within the Epidermal Growth Factor Receptor + Chemotherapy (EGFR + Chemo) treatment class; both overall and in the wild type (WT) patients for TR-PFS, but not in patients with the mutant (MT) KRAS status where data were limited. Conclusions: PFS appeared to be a good surrogate endpoint for OS. TR showed a moderate surrogate relationship with PFS and OS for the EGFR + Chemo treatment class. There was some evidence of impact of the mechanism of action on the strength of the surrogacy patterns in mCRC, but little evidence of the impact of KRAS status on the validity of surrogate endpoints.
Collapse
Affiliation(s)
- Heather Poad
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Sam Khan
- Leicester Cancer Research Centre, University of Leicester, Leicester LE1 7RH, UK
| | - Lorna Wheaton
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Anne Thomas
- Leicester Cancer Research Centre, University of Leicester, Leicester LE1 7RH, UK
| | - Michael Sweeting
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| | - Sylwia Bujkiewicz
- Biostatistics Research Group, Department of Health Sciences, University of Leicester, Leicester LE1 7RH, UK
| |
Collapse
|
23
|
Bazzichetto C, Milella M, Zampiva I, Simionato F, Amoreo CA, Buglioni S, Pacelli C, Le Pera L, Colombo T, Bria E, Zeuli M, Del Bufalo D, Sperduti I, Conciatori F. Interleukin-8 in Colorectal Cancer: A Systematic Review and Meta-Analysis of Its Potential Role as a Prognostic Biomarker. Biomedicines 2022; 10:biomedicines10102631. [PMID: 36289899 PMCID: PMC9599846 DOI: 10.3390/biomedicines10102631] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/09/2022] [Accepted: 10/16/2022] [Indexed: 12/05/2022] Open
Abstract
Among soluble actors that have emerged as druggable factors, the chemokine interleukin-8 (IL-8) has emerged as a possible determinant of response to immunotherapy and targeted treatment in several cancer types; however, its prognostic/predictive role in colorectal cancer (CRC) remains to be established. We: (i) conducted a systematic review of published literature on IL-8 expression in CRC; (ii) searched public transcriptomics databases; (iii) investigated IL-8 expression, by tumor and infiltrating cells, in a series of CRC samples; and (iv) carried out a meta-analysis of published literature correlating IL-8 expression and CRC prognosis. IL-8 possesses an important role as a mediator of the bidirectional crosstalk between tumor/stromal cells. Transcriptomic analysis indicated that specific IL-8 transcripts were significantly overexpressed in CRC compared to normal colon mucosa. Moreover, in our series we observed a statistically significant correlation between PTEN-loss and IL-8 expression by infiltrating mononuclear and tumor cells. In total, 12 papers met our meta-analysis inclusion criteria, demonstrating that high IL-8 levels significantly correlated with shorter overall survival and progression-free survival. Sensitivity analysis demonstrated a highly significant correlation with outcome for circulating, but not for tissue-detected, IL-8. IL-8 is overexpressed in CRC tissues and differentially produced by tumor or stromal components depending on CRC genetic background. Moreover, circulating IL-8 represents a strong prognostic factor in CRC, suggesting its use in the refining of prognostic CRC assessment and potentially the tailoring of therapeutic strategies in individual CRC patients.
Collapse
Affiliation(s)
- Chiara Bazzichetto
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Correspondence: (C.B.); (M.M.); Tel.: +39-06-52665185 (C.B.)
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona-School of Medicine and Verona University Hospital Trust, 37134 Verona, Italy
- Correspondence: (C.B.); (M.M.); Tel.: +39-06-52665185 (C.B.)
| | - Ilaria Zampiva
- Section of Oncology, Department of Medicine, University of Verona-School of Medicine and Verona University Hospital Trust, 37134 Verona, Italy
| | | | | | - Simonetta Buglioni
- Pathology Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Chiara Pacelli
- Department of Biochemical Sciences “A. Rossi Fanelli”, Sapienza University of Rome, 00185 Rome, Italy
| | - Loredana Le Pera
- Servizio Grandi Strumentazioni e Core Facilities, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Teresa Colombo
- Institute of Molecular Biology and Pathology-National Research Council (IBPM-CNR), 00185 Rome, Italy
| | - Emilio Bria
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Massimo Zeuli
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Donatella Del Bufalo
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Isabella Sperduti
- Biostatistics Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| | - Fabiana Conciatori
- Preclinical Models and New Therapeutic Agents Unit, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
- Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, 00144 Rome, Italy
| |
Collapse
|
24
|
Caliez O, Pietrasz D, Ksontini F, Doat S, Simon JM, Vaillant JC, Taly V, Laurent-Puig P, Bachet JB. Circulating tumor DNA: a help to guide therapeutic strategy in patients with borderline and locally advanced pancreatic adenocarcinoma? Dig Liver Dis 2022; 54:1428-1436. [PMID: 35120842 DOI: 10.1016/j.dld.2022.01.126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 12/16/2021] [Accepted: 01/11/2022] [Indexed: 12/29/2022]
Abstract
BACKGROUND prognostic biomarkers could be useful to better select patients with borderline resectable (BR) or locally advanced (LA) pancreatic adenocarcinoma (PA) for chemoradiotherapy (CRT) and/or secondary resection. AIMS The main objective of this work was to study characteristics, received treatments and prognostic of patients with BR or LA PA according to their baseline circulating tumor DNA status and, for secondary objective, neutrophil-to-lymphocyte Ratio (NLR). METHODS ctDNA status at baseline was determined using Next Generation Sequencing in a consecutive monocentric cohort of patients with a BR or LA PA. RESULTS 69 patients were included, 31 with BR PA and 38 with LA PA. 14 (20.3%) patients had baseline positive ctDNA. Five (7.8%) patients had NLR> 5. Patients with positive ctDNA had 3.7 months shorter progression free survival (p = 0.006). Patients with positive ctDNA had earlier progression after the beginning of CRT (4.4 vs 7.1 months; p = 0.068) and shorter relapse free survival after secondary resection (9.2 vs 22.9 months; p = 0.016). CONCLUSIONS positive ctDNA at baseline was associated with a worse prognosis in patients with BR or LA PA. These data are exploratory and must be confirmed in further prospective trials.
Collapse
Affiliation(s)
- Olivier Caliez
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Sorbonne Université, UPMC, Paris 6, France
| | - Daniel Pietrasz
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Department of Digestive Surgery, Hôpital Paul Brousse, Villejuif, France
| | - Feryel Ksontini
- Department of Oncology, Institute Salah-Azaïz, Tunis, Tunisia
| | - Solène Doat
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Marc Simon
- Department of Radiation Oncology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Jean-Christophe Vaillant
- Department of Digestive Surgery, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Valerie Taly
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France
| | - Pierre Laurent-Puig
- French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France
| | - Jean-Baptiste Bachet
- Department of Gastroenterology, La Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; French National Institute of Health and Medical Research (INSERM), Centre de Recherche des Cordeliers, Sorbonne Université, Sorbonne Paris Cité, Université Paris Descartes, Université de Paris, Paris, France; Sorbonne Université, UPMC, Paris 6, France.
| |
Collapse
|
25
|
Ebinç S, Oruç Z, Urakçi Z, Kalkan Z, Kaplan MA, Küçüköner M, Işikdoğan A. Evaluation of Factors Predicting the Effectiveness of Regorafenib in the Treatment of Metastatic Colorectal Cancer. Eurasian J Med 2022; 54:229-234. [PMID: 35950825 PMCID: PMC9797696 DOI: 10.5152/eurasianjmed.2022.21162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/27/2021] [Indexed: 01/05/2023] Open
Abstract
OBJECTIVE Regorafenib is a multikinase inhibitor, the effectiveness of which was demonstrated in metastatic colorectal cancer. This study aimed to investigate the factors that could predict the effectiveness of regorafenib. MATERIALS AND METHODS This study retrospectively reviewed the clinical characteristics, tumor characteristics, and previous therapies in 62 patients who presented to our center between 2016 and 2020 and used regorafenib for metastatic colorectal cancer. The effects of the investigated variables on the response obtained with regorafenib use were evaluated. RESULTS This study included a total of 62 patients diagnosed with metastatic colorectal cancer, of whom 30 (48.4%) were males and 32 (51.6%) were females. Patients' median age at diagnosis was 49 years (18- 68). Regorafenib therapy yielded a disease control rate of 64% [complete response=0, partial response= 14 (28%), and stable disease=18 (36%)]. Objective response was obtained in 28% of patients [complete response=0 and partial response=14 (28%)]. Progression-free survival was 4 months. The evaluation of the effects of patients' age, sex, performance status, previous treatments, metastatic sites, and RAS mutation status on the disease control rate and progression-free survival did not determine any positive or negative effects on progression-free survival. However, left-sided tumors had a positive effect on disease control rate (69.8% vs. 28.6%, P=.029). and previous use of cetuximab had a negative effect on disease control rate [76.5% vs. 37.5% (P=.007)]. CONCLUSION In our study, tumor localization and previous cetuximab use were found to be correlated with the disease control rate in patients on regorafenib. However, the need for novel biomarkers that will predict the effectiveness of regorafenib in metastatic colorectal cancer treatment persists.
Collapse
Affiliation(s)
- Senar Ebinç
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Zeynep Oruç
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Zuhat Urakçi
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Ziya Kalkan
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Muhammet Ali Kaplan
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Mehmet Küçüköner
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| | - Abdurrahman Işikdoğan
- Department of Medical Oncology, Dicle University Faculty of Medicine, Diyarbakır, Turkey
| |
Collapse
|
26
|
Ghani M, Liau J, Eskander R, Mell L, Yusufaly T, Obrzut S. Imaging Biomarkers and Liquid Biopsy in Assessment of Cervical Cancer. J Comput Assist Tomogr 2022; 46:707-715. [PMID: 35995483 PMCID: PMC9474655 DOI: 10.1097/rct.0000000000001358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT The role of imaging has been increasing in pretherapy planning and response assessment in cervical cancer, particularly in high-resource settings that provide access to computed tomography (CT), magnetic resonance imaging (MRI), and positron emission tomography (PET). In 2018, imaging was incorporated into the International Federation of Gynecology and Obstetrics staging system for cervical cancer. Magnetic resonance imaging is advantageous over CT for evaluation of the primary cervical cancer size and extent, because of superior contrast resolution. Furthermore, quantitative methods, including diffusion-weighted and dynamic contrast-enhanced MRI, show promise in improving treatment response and prognosis evaluation. Molecular imaging with fluorodeoxyglucose-PET/CT and PET/MRI can be particularly helpful in the detection of nodal disease and distant metastases. Semiautomated delineation of 3-dimensional tumor regions of interest has facilitated the development of novel PET-derived biomarkers that include metabolic volume and radiomics textural analysis features for prediction of outcomes. However, posttreatment inflammatory changes can be a confounder and lymph node evaluation is challenging, even with the use of PET/CT. Liquid biopsy has emerged as a promising tool that may be able to overcome some of the drawbacks inherent with imaging, such as limited ability to detect microscopic metastases or to distinguish between postchemoradiotherapy changes and residual tumor. Preliminary evidence suggests that liquid biopsy may be able to identify cervical cancer treatment response and resistance earlier than traditional methods. Future work should prioritize how to best synergize imaging and liquid biopsy as an integrated approach for optimal cervical cancer management.
Collapse
Affiliation(s)
- Mansur Ghani
- Department of Radiology, University of California San Diego, CA, USA
| | - Joy Liau
- Department of Radiology, University of California San Diego, CA, USA
| | - Ramez Eskander
- Division of Hematology/Oncology, University of California San Diego, CA, USA
| | - Loren Mell
- Department of Radiation Oncology, University of California San Diego, CA, USA
| | - Tahir Yusufaly
- Department of Radiology, Johns Hopkins University, MD, USA
| | - Sebastian Obrzut
- Department of Radiology, University of California San Diego, CA, USA
| |
Collapse
|
27
|
Liu Z, Gao Z, Yang W, Zhang L, Xiao N, Qu D, Su Z, Xu K, Liu G, Wang Y, Ren Q, Yu S, Cheng Y, Zhou Y, Deng Q, Zhao Y, Wang Z, Yang H. A randomized, double-blind, single-dose, parallel phase I clinical trial to compare the bioequivalence, immunogenicity and safety of bevacizumab biosimilar and bevacizumab in healthy Chinese subjects. Expert Opin Drug Metab Toxicol 2022; 18:519-527. [PMID: 35961948 DOI: 10.1080/17425255.2022.2113382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
BACKGROUND Bevacizumab, a humanized monoclonal antibody against VEGF, can be used as a target therapy for colorectal cancer. A phase I clinical trial was conducted to compare the bioequivalence, immunogenicity and safety of bevacizumab biosimilar (Chia Tai Tianqing Pharmaceutical Group Co., Ltd.) and Bevacizumab (Roche Diagnostics GmbH) in healthy Chinese males. RESEARCH DESIGN & METHOD Healthy Chinese subjects (N = 98) were randomly divided into two groups. A single-dose bevacizumab biosimilar or Bevacizumab was given for per cycle. Plasma drug concentrations were detected by liquid chromatography-tandem mass spectrometry (LC-MC/MS) assay. We detected the levels of anti-drug antibody (ADA) to evaluate drug immunogenicity and the safety of drugs throughout the study. RESULTS The geometric mean ratios (GMRs) of AUC0-t, Cmax and AUC0-∞ for bevacizumab biosimilar and Bevacizumab were 96.27%, 93.69% and 97.01%, respectively. The 90% CIs were all within 80%-125%, meeting the bioequivalence standards. The levels of ADA were similar. In addition, the two drugs both demonstrated excellent safety in the trial. CONCLUSION This study showed that bevacizumab biosimilar and Bevacizumab had similar pharmacokinetics (PK) parameters and safety in healthy Chinese subjects. CLINICAL TRIAL REGISTRATION INFORMATION This trial was registered in ClinicalTrials.gov (Number: NCT05476341, date registered: 25, Jul 2022) and Drug Clinical Trial Registration and Information Disclosure Platform (Number: CTR20171308, date registered: 16, Nov 2017).
Collapse
Affiliation(s)
- Zhengzhi Liu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Zhenyue Gao
- Department of clinical research center, Chia Tai Tianqing Pharmaceutical Group Co.,Ltd., Nanjing, China
| | - Wei Yang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Lixiu Zhang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Nan Xiao
- Department of clinical research center, Chia Tai Tianqing Pharmaceutical Group Co.,Ltd., Nanjing, China
| | - Dongmei Qu
- Ansiterui Medical Technology Consulting Co.,Ltd., Changchun, China
| | - Zhengjie Su
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Kaibo Xu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Guangwen Liu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yanli Wang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Qing Ren
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Shuang Yu
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yang Cheng
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yannan Zhou
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Qiaohuan Deng
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| | - Yicheng Zhao
- Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, China.,Clinical Medical College, Changchun University of Chinese Medicine, Changchun, China
| | - Zeyu Wang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China.,Scientific Research Department, Changchun University of Chinese Medicine, Changchun, China
| | - Haimiao Yang
- Phase I Clinical Trial Laboratory, Affiliated Hospital to Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
28
|
Vaquero J, Pavy A, Gonzalez-Sanchez E, Meredith M, Arbelaiz A, Fouassier L. Genetic alterations shaping tumor response to anti-EGFR therapies. Drug Resist Updat 2022; 64:100863. [DOI: 10.1016/j.drup.2022.100863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
29
|
Huang M, Lin Y, Wang C, Deng L, Chen M, Assaraf YG, Chen ZS, Ye W, Zhang D. New insights into antiangiogenic therapy resistance in cancer: Mechanisms and therapeutic aspects. Drug Resist Updat 2022; 64:100849. [PMID: 35842983 DOI: 10.1016/j.drup.2022.100849] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Angiogenesis is a hallmark of cancer and is required for tumor growth and progression. Antiangiogenic therapy has been revolutionarily developing and was approved for the treatment of various types of cancer for nearly two decades, among which bevacizumab and sorafenib continue to be the two most frequently used antiangiogenic drugs. Although antiangiogenic therapy has brought substantial survival benefits to many cancer patients, resistance to antiangiogenic drugs frequently occurs during clinical treatment, leading to poor outcomes and treatment failure. Cumulative evidence has demonstrated that the intricate interplay among tumor cells, bone marrow-derived cells, and local stromal cells critically allows for tumor escape from antiangiogenic therapy. Currently, drug resistance has become the main challenge that hinders the therapeutic efficacies of antiangiogenic therapy. In this review, we describe and summarize the cellular and molecular mechanisms conferring tumor drug resistance to antiangiogenic therapy, which was predominantly associated with redundancy in angiogenic signaling molecules (e.g., VEGFs, GM-CSF, G-CSF, and IL17), alterations in biological processes of tumor cells (e.g., tumor invasiveness and metastasis, stemness, autophagy, metabolic reprogramming, vessel co-option, and vasculogenic mimicry), increased recruitment of bone marrow-derived cells (e.g., myeloid-derived suppressive cells, tumor-associated macrophages, and tumor-associated neutrophils), and changes in the biological functions and features of local stromal cells (e.g., pericytes, cancer-associated fibroblasts, and endothelial cells). We also review potential biomarkers to predict the response to antiangiogenic therapy in cancer patients, which mainly consist of imaging biomarkers, cellular and extracellular proteins, a certain type of bone marrow-derived cells, local stromal cell content (e.g., pericyte coverage) as well as serum or plasma biomarkers (e.g., non-coding RNAs). Finally, we highlight the recent advances in combination strategies with the aim of enhancing the response to antiangiogenic therapy in cancer patients and mouse models. This review introduces a comprehensive understanding of the mechanisms and biomarkers associated with the evasion of antiangiogenic therapy in cancer, providing an outlook for developing more effective approaches to promote the therapeutic efficacy of antiangiogenic therapy.
Collapse
Affiliation(s)
- Maohua Huang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China; Integrated Chinese and Western Medicine Postdoctoral Research Station, Jinan University, Guangzhou, 510632, China
| | - Yuning Lin
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Chenran Wang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Lijuan Deng
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Minfeng Chen
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Laboratory, Department of Biology, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Institute for Biotechnology, St. John's University, NY 11439, USA.
| | - Wencai Ye
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| | - Dongmei Zhang
- Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, College of Pharmacy, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
30
|
Cabezas-Camarero S, García-Barberán V, Pérez-Alfayate R, Casado-Fariñas I, Sloane H, Jones FS, Pérez-Segura P. Detection of IDH1 Mutations in Plasma Using BEAMing Technology in Patients with Gliomas. Cancers (Basel) 2022; 14:cancers14122891. [PMID: 35740557 PMCID: PMC9221506 DOI: 10.3390/cancers14122891] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/04/2022] [Accepted: 06/10/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary In contrast with other solid tumors, only a few, small studies have shown the feasibility of detecting different biomarkers in the peripheral blood (PB) of patients with gliomas. A prospective study was conducted, enrolling 10 patients with gliomas where 33 consecutive PB samples were analyzed. Among the six patients with isocitrate dehydrogenase 1 (IDH1)-mutant tumors that were surveyed, circulating tumor DNA (ctDNA) was detected in PB in three of them (50%), at timepoints at which the patients were either untreated or exhibited progressive disease. While no false positives were identified, the false-negative rate was high, reaching 86% (18/21). Finally, in one of the IDH1-mutant cases, the Beads, Emulsion, Amplification and Magnetics (BEAMing) digital PCR technology detected one of the two IDH1 mutations that had been detected in the patient’s tumor sample in plasma, 7 years prior to its detection in blood. Abstract Molecular testing using blood-based liquid biopsy approaches has not been widely investigated in patients with glioma. A prospective single-center study enrolled patients with gliomas ranging from grade II to IV. Peripheral blood (PB) was drawn at different timepoints for circulating tumour DNA (ctDNA) monitoring. Next-generation sequencing (NGS) was used for the study of isocitrate dehydrogenase 1 (IDH1) mutations in the primary tumor. Beads, Emulsion, Amplification and Magnetics (BEAMing) was used for the study of IDH1 mutations in plasma and correlated with the NGS results in the tumor. Between February 2017 and July 2018, ten patients were enrolled, six with IDH1-mutant and four with IDH1 wild-type gliomas. Among the six IDH-mutant gliomas, three had the same IDH1 mutation detected in plasma (50%), and the IDH1-positive ctDNA result was obtained in patients either at diagnosis (no treatment) or during progressive disease. While the false-negative rate reached 86% (18/21), 15 out of the 18 (83%) plasma-negative results were from PB collected from the six IDH-mutant patients at times at which there was no accompanying evidence of tumor progression, as assessed by MRI. There were no false-positive cases in plasma collected from patients with IDH1 wild-type tumors. BEAMing detected IDH1 mutations in the plasma of patients with gliomas, with a modest clinical sensitivity (true positivity rate) but with 100% clinical specificity, with complete agreement between the mutant loci detected in tumor and plasma. Larger prospective studies should be conducted to expand on these findings, and further explore the clearance of mutations in PB from IDH1-positive patients in response to therapy.
Collapse
Affiliation(s)
- Santiago Cabezas-Camarero
- Head & Neck Cancer, Neuro-Oncology and Genetic Counseling Unit, Medical Oncology Department, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico Universitario San Carlos, Paseo del Profesor Martín Lagos S/N, 28040 Madrid, Spain;
- Correspondence: ; Tel.: +34-91-330-3000
| | - Vanesa García-Barberán
- Molecular Oncology Laboratory, Medical Oncology Department, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain;
| | - Rebeca Pérez-Alfayate
- Department of Neurosurgery, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain;
| | - Isabel Casado-Fariñas
- Pathology Department, Hospital Clínico Universitario San Carlos, 28040 Madrid, Spain;
| | - Hillary Sloane
- Medical affairs Division, Sysmex Inostics, Inc., Baltimore, MD 21205, USA; (H.S.); (F.S.J.)
| | - Frederick S. Jones
- Medical affairs Division, Sysmex Inostics, Inc., Baltimore, MD 21205, USA; (H.S.); (F.S.J.)
| | - Pedro Pérez-Segura
- Head & Neck Cancer, Neuro-Oncology and Genetic Counseling Unit, Medical Oncology Department, Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico Universitario San Carlos, Paseo del Profesor Martín Lagos S/N, 28040 Madrid, Spain;
| |
Collapse
|
31
|
Caputo V, De Falco V, Ventriglia A, Famiglietti V, Martinelli E, Morgillo F, Martini G, Corte CMD, Ciardiello D, Poliero L, De Vita F, Orditura M, Fasano M, Franco R, Caraglia M, Avitabile A, Scalamogna R, Marchi B, Ciardiello F, Troiani T, Napolitano S. Comprehensive genome profiling by next generation sequencing of circulating tumor DNA in solid tumors: a single academic institution experience. Ther Adv Med Oncol 2022; 14:17588359221096878. [PMID: 35547096 PMCID: PMC9082754 DOI: 10.1177/17588359221096878] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 04/07/2022] [Indexed: 01/18/2023] Open
Abstract
Background: Recently, new evidence of the next-generation sequencing (NGS) liquid biopsy utility in clinical practice has been developed. This assay is emerging as a new promising tool to use as a noninvasive biomarker for cancer mutation profiling. Additional data supporting the clinical validity of cell free DNA (cfDNA) based testing is necessary to inform optimal use of these assays in the clinic. Materials and methods: A total of 398 cancer patients were analyzed by FoundationOne Liquid Analysis (F1LA), a genomic profiling assay and by standard NGS diagnostic ThermoFisher platform. The association between diagnostic technique was evaluated using a Poisson regression model. FoundationOne Liquid (F1L) and FoundationOne Liquid CDx (F1LCDx) detect 70 and 324 cancer-related genes alterations, respectively, including genomic signatures tumor fraction, blood tumor mutational burden (only for the 324 genes version), and microsatellite instability high status. Both assays used a single DNA extraction method to obtain cfDNA. The real-life clinical impact and feasibility of F1L and F1LCDx were evaluated across different solid tumors in our department. Results: Between 1 January 2019 and 28 February 2021, 398 samples of different tumor types from 398 patients were analyzed (overall success rate: 92%, in FoundationOne Liquid CDx Analysis success rate: 97%). Most frequent molecular alterations were TP53 (74), APC (40), DNMT3A (39), KRAS (23). The comprehensive clinical impact of F1LA compared with standard diagnostic was 64.7% versus 22.1% [risk ratio (RR) = 2.94; p < 0.001] and the potential clinical impact was 58.6% versus 11.0% (RR = 5.32; p < 0.001), respectively. Furthermore, some clinical cases were selected, in which F1LA detected actionable alterations offering an unexpected therapeutic choice. Conclusions: Although additional studies are needed to better select patients and setting, NGS F1LA is a useful, noninvasive, and repeatable assay to guide therapeutic choice in oncology. It provides a snapshot of cancer heterogeneity profile that could be incorporated in routinely clinical practice.
Collapse
Affiliation(s)
- Vincenza Caputo
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Vincenzo De Falco
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Anna Ventriglia
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Vincenzo Famiglietti
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Erika Martinelli
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Floriana Morgillo
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Giulia Martini
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Carminia Maria Della Corte
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Davide Ciardiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
- Oncology Unit, Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy
| | - Luca Poliero
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Ferdinando De Vita
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Michele Orditura
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Morena Fasano
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Renato Franco
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Michele Caraglia
- Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | | | | | | | - Fortunato Ciardiello
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Napoli, Italy
| | - Teresa Troiani
- Full Professor, Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Via S. Pansini 5, Napoli 80131, Italy
| | - Stefania Napolitano
- Medical Oncology, Department of Precision Medicine, Università degli Studi della Campania ‘Luigi Vanvitelli’, Via S. Pansini 5, Napoli 80131, Italy
| |
Collapse
|
32
|
Arisi MF, Dotan E, Fernandez SV. Circulating Tumor DNA in Precision Oncology and Its Applications in Colorectal Cancer. Int J Mol Sci 2022; 23:ijms23084441. [PMID: 35457259 PMCID: PMC9024503 DOI: 10.3390/ijms23084441] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 03/29/2022] [Accepted: 04/14/2022] [Indexed: 02/06/2023] Open
Abstract
Circulating tumor DNA (ctDNA) is a component of cell-free DNA (cfDNA) that is shed by malignant tumors into the bloodstream and other bodily fluids. ctDNA can comprise up to 10% of a patient’s cfDNA depending on their tumor type and burden. The short half-life of ctDNA ensures that its detection captures tumor burden in real-time and offers a non-invasive method of repeatedly evaluating the genomic profile of a patient’s tumor. A challenge in ctDNA detection includes clonal hematopoiesis of indeterminate potential (CHIP), which can be distinguished from tumor variants using a paired whole-blood control. Most assays for ctDNA quantification rely on measurements of somatic variant allele frequency (VAF), which is a mutation-dependent method. Patients with certain types of solid tumors, including colorectal cancer (CRC), can have levels of cfDNA 50 times higher than healthy patients. ctDNA undergoes a precipitous drop shortly after tumor resection and therapy, and rising levels can foreshadow radiologic recurrence on the order of months. The amount of tumor bulk required for ctDNA detection is lower than that for computed tomography (CT) scan detection, with ctDNA detection preceding radiologic recurrence in many cases. cfDNA/ctDNA can be used for tumor molecular profiling to identify resistance mutations when tumor biopsy is not available, to detect minimal residual disease (MRD), to monitor therapy response, and for the detection of tumor relapse. Although ctDNA is not yet implemented in clinical practice, studies are ongoing to define the appropriate way to use it as a tool in the clinic. In this review article, we examine the general aspects of ctDNA, its status as a biomarker, and its role in the management of early (II–III) and late (IV; mCRC) stage colorectal cancer (CRC).
Collapse
Affiliation(s)
- Maria F. Arisi
- Sidney Kimmel Medical School, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Efrat Dotan
- Department of Medical Oncology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA;
| | - Sandra V. Fernandez
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
- Correspondence:
| |
Collapse
|
33
|
Antoniotti C, Marmorino F, Boccaccino A, Martini S, Antista M, Rossini D, Zuco V, Prisciandaro M, Conca V, Zucchelli G, Borelli B, Cosentino P, Germani MM, Bosco MF, Carullo M, Vetere G, Moretto R, Giordano M, Masi G, Pietrantonio F, Zaffaroni N, Cremolini C. Early modulation of Angiopoietin-2 plasma levels predicts benefit from regorafenib in patients with metastatic colorectal cancer. Eur J Cancer 2022; 165:116-124. [DOI: 10.1016/j.ejca.2022.01.025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 01/07/2022] [Accepted: 01/14/2022] [Indexed: 12/12/2022]
|
34
|
Pinheiro M, Peixoto A, Rocha P, Veiga I, Pinto C, Santos C, Pinto P, Guerra J, Escudeiro C, Barbosa A, Silva J, Teixeira MR. KRAS and NRAS mutational analysis in plasma ctDNA from patients with metastatic colorectal cancer by real-time PCR and digital PCR. Int J Colorectal Dis 2022; 37:895-905. [PMID: 35303157 DOI: 10.1007/s00384-022-04126-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/02/2022] [Indexed: 02/04/2023]
Abstract
PURPOSE Mutations in the KRAS and NRAS (RAS) genes are negative predictors of response to anti-EGFR therapy in metastatic colorectal cancer (mCRC). The detection of mutations in circulating tumor DNA (ctDNA) has emerged as a less invasive strategy to assess the molecular profile of mCRC patients. We aimed to perform RAS mutational analysis in ctDNA from mCRC patients using BEAMing Digital PCR (OncoBEAM) and Idylla ctDNA qPCR and evaluate the concordance rate with RAS mutational status in tumor tissue and between these two methodologies with different limits of detection. METHODS Blood samples were collected from 47 mCRC patients previously tested for RAS mutations in tumor tissue. DNA was extracted from plasma using the QIAamp Circulating Nucleic Acid Kit, and RAS mutation analysis was conducted using OncoBEAM RAS CRC and Idylla ctRAS assays. RESULTS The overall agreement between tumor tissue and ctDNA analyses was 83% and 78.7% using the OncoBEAM and Idylla assays, respectively, with the concordance being 96.2% and 88.5% in naive treatment patients. The overall agreement between OncoBEAM and Idylla ctDNA analyses was 91.7%. CONCLUSIONS Analysis of ctDNA is a viable strategy for clinical management of mCRC patients. Although the OncoBEAM assay sensitivity is somewhat higher, the fully automated Idylla platform also has good performance, while being cheaper and much less labor-intensive, for the detection of RAS mutations in plasma, either at diagnosis or after progression when considering anti-EGFR treatment rechallenge.
Collapse
Affiliation(s)
- Manuela Pinheiro
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Ana Peixoto
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal.,Department of Genetics, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Patricia Rocha
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal.,Department of Genetics, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Isabel Veiga
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal.,Department of Genetics, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Carla Pinto
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal.,Department of Genetics, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Catarina Santos
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal.,Department of Genetics, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Pedro Pinto
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Joana Guerra
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Carla Escudeiro
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Ana Barbosa
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - João Silva
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal.,Department of Genetics, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal
| | - Manuel R Teixeira
- Cancer Genetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal. .,Department of Genetics, Portuguese Oncology Institute of Porto, Rua Dr. António Bernardino Almeida, 4200-072, Porto, Portugal. .,Institute of Biomedical Sciences Abel Salazar, University of Porto, Largo Prof. Abel Salazar, 4099-003, Porto, Portugal.
| |
Collapse
|
35
|
Cai J, Jiang H, Li S, Yan X, Wang M, Li N, Zhu C, Dong H, Wang D, Xu Y, Xie H, Wu S, Lou J, Zhao J, Li Q. The Landscape of Actionable Genomic Alterations by Next-Generation Sequencing in Tumor Tissue Versus Circulating Tumor DNA in Chinese Patients With Non-Small Cell Lung Cancer. Front Oncol 2022; 11:751106. [PMID: 35273907 PMCID: PMC8902245 DOI: 10.3389/fonc.2021.751106] [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: 07/31/2021] [Accepted: 12/30/2021] [Indexed: 12/24/2022] Open
Abstract
Background Circulating tumor DNA (ctDNA) sequence analysis shows great potential in the management of non-small cell lung cancer (NSCLC) and the prediction of drug sensitivity or resistance in many cancers. Here, we drew and compared the somatic mutational profile using ctDNA and tumor tissue sequence analysis in lung adenocarcinoma (LUAD) and squamous cell carcinoma (LUSC), and assess its potential clinical value. Methods In this study, 221 tumor tissues and 174 plasma samples from NSCLC patients were analyzed by hybridization capture-based next-generation sequencing (NGS) panel including 95 cancer-associated genes. Tumor response assessments were applied to 137 patients with advanced-stage (III and IV) NSCLC who first received targeted agents. Results Twenty significantly mutated genes were identified such as TP53, EGFR, RB1, KRAS, PIK3CA, CD3EAP, CTNNB1, ERBB2, APC, BRAF, TERT, FBXW7, and HRAS. Among them, TP53 was the most frequently mutated gene and had a higher mutation probability in male (p = 0.00124) and smoking (p < 0.0001) patients. A total of 48.35% (191/395) of NSCLC patients possessed at least one actionable alteration according to the OncoKB database. Although the sensitivity of genomic profiling from ctDNA was lower than that from tumor tissue DNA, the mutational landscape of target genes from ctDNA is similar to that from tumor tissue DNA, which led to 61.22% (30/49) of mutational concordance in NSCLC. Additionally, the mutational concordance between tissue DNA and ctDNA in LUAD differs from that in LUSC, which is 63.83% versus 46.67%, indicating that NSCLC subtypes influence the specificity of mutation detection in plasma-derived ctDNA. Lastly, patients with EGFR and TP53 co-alterations showed similar responses to Gefitinib and Icotinib, and the co-occurring TP53 mutation was most likely to be a poor prognostic factor for patients receiving Gefitinib, indicating that the distributions and types of TP53 mutations may contribute to the efficacy and prognosis of molecular targeted therapy. Conclusions As a promising alternative for tumor genomic profiling, ctDNA analysis is more credible in LUAD than in LUSC. Genomic subtyping has strong potential in prognostication and therapeutic decision-making for NSCLC patients, which indicated the necessity for the utility of target NGS in guiding clinical management.
Collapse
Affiliation(s)
- Jun Cai
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Huihui Jiang
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Shuqing Li
- Department of General Surgery, Yucheng Hospital of Traditional Chinese Medicine, Dezhou City, China
| | - Xiaoxia Yan
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Meng Wang
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Na Li
- Department of Oncology, First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Cuimin Zhu
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Hui Dong
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Dongjuan Wang
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, China
| | - Yue Xu
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Hui Xie
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Shouxin Wu
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Jingwei Lou
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Jiangman Zhao
- Zhangjiang Center for Translational Medicine, Shanghai Biotecan Pharmaceuticals Co., Ltd., Shanghai, China
| | - Qingshan Li
- Department of Oncology, Affiliated Hospital of Chengde Medical University, Chengde, China
| |
Collapse
|
36
|
The potential of liquid biopsy in the management of cancer patients. Semin Cancer Biol 2022; 84:69-79. [PMID: 35331850 DOI: 10.1016/j.semcancer.2022.03.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 03/06/2022] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
|
37
|
Larribère L, Martens UM. Advantages and Challenges of Using ctDNA NGS to Assess the Presence of Minimal Residual Disease (MRD) in Solid Tumors. Cancers (Basel) 2021; 13:5698. [PMID: 34830853 PMCID: PMC8616165 DOI: 10.3390/cancers13225698] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 11/05/2021] [Accepted: 11/10/2021] [Indexed: 12/22/2022] Open
Abstract
The ability to detect minimal residual disease (MRD) after a curative-intent surgery or treatment is of paramount importance, because it offers the possibility to help guide the clinical decisions related adjuvant therapy. Thus, the earlier MRD is detected, the earlier potentially beneficial treatment can be proposed to patients who might need it. Liquid biopsies, and in particular the next-generation sequencing of circulating tumor DNA (ctDNA) in the blood, have been the focus of an increasing amount of research in the past years. The ctDNA detection at advanced cancer stages is practicable for several solid tumors, and complements molecular information on acquired therapy resistance. In the context of MRD, it is by definition more challenging to detect ctDNA, but it is technically achievable and provides information on treatment response and probability of relapse significantly earlier than standard imaging methods. The clinical benefit of implementing this new technique in the routine is being tested in interventional clinical trials at the moment. We propose here an update of the current use of ctDNA detection by NGS as a tool to assess the presence of MRD and improve adjuvant treatment of solid tumors. We also discuss the main limitations and medium-term perspectives of this process in the clinic.
Collapse
Affiliation(s)
- Lionel Larribère
- Department of Hematology and Oncology, Cancer Center Heilbronn-Franken, SLK Clinics Heilbronn GmbH, 74078 Heilbronn, Germany;
- Skin Cancer Unit, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, 68167 Mannheim, Germany
| | - Uwe M. Martens
- Department of Hematology and Oncology, Cancer Center Heilbronn-Franken, SLK Clinics Heilbronn GmbH, 74078 Heilbronn, Germany;
- MOLIT Institute for Personalized Medicine GmbH, 74076 Heilbronn, Germany
| |
Collapse
|
38
|
De Summa S, Danza K, Pilato B, Matera G, Fasano R, Calabrese A, Lacalamita R, Silvestris N, Tommasi S, Argentiero A, Brunetti O. A Promising Role of TGF-β Pathway in Response to Regorafenib in Metastatic Colorectal Cancer: A Case Report. Medicina (B Aires) 2021; 57:medicina57111241. [PMID: 34833459 PMCID: PMC8619854 DOI: 10.3390/medicina57111241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/08/2021] [Accepted: 11/11/2021] [Indexed: 11/17/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancer types around the world. The prognosis of patients with advanced diseases is still poor in spite of currently available therapeutic options. Regorafenib is an oral tyrosine kinase inhibitor (TKI) approved to treat refractory metastatic colorectal cancer (mCRC). We investigated Somatic mutations in several genes involved in immunological response and cancer progression in both long/short responder mCRC patients who underwent third-line therapy with regorafenib to identify predictive biomarkers of response using Ion Torrent PGM sequencing and bioinformatic tools. We found Somatic mutations in TGFBR1, TGFBR2, and TGFBR3 genes in primary tumor and metastases samples of long-responder patients. Furthermore, our bioinformatic results show that they were mainly enriched in immune response, cell junction, and cell adhesion in long responder patients, particularly in primary tumor and metastatic sites. These data suggest that the TGF-b pattern could be the leading actor of a prolonged response to this drug.
Collapse
Affiliation(s)
- Simona De Summa
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Katia Danza
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Brunella Pilato
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Giuseppina Matera
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Rossella Fasano
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (R.F.); (N.S.); (A.A.)
| | - Angela Calabrese
- Radiology Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Rosanna Lacalamita
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (R.F.); (N.S.); (A.A.)
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy
| | - Stefania Tommasi
- Molecular Diagnostics and Pharmacogenetics Unit, IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy; (S.D.S.); (K.D.); (B.P.); (G.M.); (R.L.); (S.T.)
| | - Antonella Argentiero
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (R.F.); (N.S.); (A.A.)
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy; (R.F.); (N.S.); (A.A.)
- Correspondence:
| |
Collapse
|
39
|
Role of oncogenic KRAS in the prognosis, diagnosis and treatment of colorectal cancer. Mol Cancer 2021; 20:143. [PMID: 34742312 PMCID: PMC8571891 DOI: 10.1186/s12943-021-01441-4] [Citation(s) in RCA: 169] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 10/04/2021] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is a heterogeneous disease at the cellular and molecular levels. Kirsten rat sarcoma (KRAS) is a commonly mutated oncogene in CRC, with mutations in approximately 40% of all CRC cases; its mutations result in constitutive activation of the KRAS protein, which acts as a molecular switch to persistently stimulate downstream signaling pathways, including cell proliferation and survival, thereby leading to tumorigenesis. Patients whose CRC harbors KRAS mutations have a dismal prognosis. Currently, KRAS mutation testing is a routine clinical practice before treating metastatic cases, and the approaches developed to detect KRAS mutations have exhibited favorable sensitivity and accuracy. Due to the presence of KRAS mutations, this group of CRC patients requires more precise therapies. However, KRAS was historically thought to be an undruggable target until the development of KRASG12C allele-specific inhibitors. These promising inhibitors may provide novel strategies to treat KRAS-mutant CRC. Here, we provide an overview of the role of KRAS in the prognosis, diagnosis and treatment of CRC.
Collapse
|
40
|
Lafferty A, O'Farrell AC, Migliardi G, Khemka N, Lindner AU, Sassi F, Zanella ER, Salvucci M, Vanderheyden E, Modave E, Boeckx B, Halang L, Betge J, Ebert MPA, Dicker P, Argilés G, Tabernero J, Dienstmann R, Medico E, Lambrechts D, Bertotti A, Isella C, Trusolino L, Prehn JHM, Byrne AT. Molecular Subtyping Combined with Biological Pathway Analyses to Study Regorafenib Response in Clinically Relevant Mouse Models of Colorectal Cancer. Clin Cancer Res 2021; 27:5979-5992. [PMID: 34426441 DOI: 10.1158/1078-0432.ccr-21-0818] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/05/2021] [Accepted: 08/18/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Regorafenib (REG) is approved for the treatment of metastatic colorectal cancer, but has modest survival benefit and associated toxicities. Robust predictive/early response biomarkers to aid patient stratification are outstanding. We have exploited biological pathway analyses in a patient-derived xenograft (PDX) trial to study REG response mechanisms and elucidate putative biomarkers. EXPERIMENTAL DESIGN Molecularly subtyped PDXs were annotated for REG response. Subtyping was based on gene expression (CMS, consensus molecular subtype) and copy-number alteration (CNA). Baseline tumor vascularization, apoptosis, and proliferation signatures were studied to identify predictive biomarkers within subtypes. Phospho-proteomic analysis was used to identify novel classifiers. Supervised RNA sequencing analysis was performed on PDXs that progressed, or did not progress, following REG treatment. RESULTS Improved REG response was observed in CMS4, although intra-subtype response was variable. Tumor vascularity did not correlate with outcome. In CMS4 tumors, reduced proliferation and higher sensitivity to apoptosis at baseline correlated with response. Reverse phase protein array (RPPA) analysis revealed 4 phospho-proteomic clusters, one of which was enriched with non-progressor models. A classification decision tree trained on RPPA- and CMS-based assignments discriminated non-progressors from progressors with 92% overall accuracy (97% sensitivity, 67% specificity). Supervised RNA sequencing revealed that higher basal EPHA2 expression is associated with REG resistance. CONCLUSIONS Subtype classification systems represent canonical "termini a quo" (starting points) to support REG biomarker identification, and provide a platform to identify resistance mechanisms and novel contexts of vulnerability. Incorporating functional characterization of biological systems may optimize the biomarker identification process for multitargeted kinase inhibitors.
Collapse
Affiliation(s)
- Adam Lafferty
- Department of Physiology and Medical Physics, Precision Cancer Medicine Group, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Alice C O'Farrell
- Department of Physiology and Medical Physics, Precision Cancer Medicine Group, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Giorgia Migliardi
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Niraj Khemka
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Andreas U Lindner
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | | | | | - Manuela Salvucci
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Evy Vanderheyden
- Department of Human Genetics, VIB Center for Cancer Biology, Leuven, Belgium, Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium
| | - Elodie Modave
- Department of Human Genetics, VIB Center for Cancer Biology, Leuven, Belgium, Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium
| | - Bram Boeckx
- Department of Human Genetics, VIB Center for Cancer Biology, Leuven, Belgium, Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium
| | - Luise Halang
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Johannes Betge
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Junior Clinical Cooperation Unit Translational Gastrointestinal Oncology and Preclinical Models, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Matthias P A Ebert
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Patrick Dicker
- Department of Epidemiology and Public Health Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Guillem Argilés
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - Josep Tabernero
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - Rodrigo Dienstmann
- Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, CIBERONC, Barcelona, Spain
| | - Enzo Medico
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Diether Lambrechts
- Department of Human Genetics, VIB Center for Cancer Biology, Leuven, Belgium, Laboratory for Translational Genetics, KU Leuven, Leuven, Belgium
| | - Andrea Bertotti
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Claudio Isella
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino, Candiolo, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Jochen H M Prehn
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Systems Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Annette T Byrne
- Department of Physiology and Medical Physics, Precision Cancer Medicine Group, Royal College of Surgeons in Ireland, Dublin, Ireland.
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin, Ireland
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Belfield, Dublin, Ireland
| |
Collapse
|
41
|
Benhaim L, Bouché O, Normand C, Didelot A, Mulot C, Le Corre D, Garrigou S, Djadi-Prat J, Wang-Renault SF, Perez-Toralla K, Pekin D, Poulet G, Landi B, Taieb J, Selvy M, Emile JF, Lecomte T, Blons H, Chatellier G, Link DR, Taly V, Laurent-Puig P. Circulating tumor DNA is a prognostic marker of tumor recurrence in stage II and III colorectal cancer: multicentric, prospective cohort study (ALGECOLS). Eur J Cancer 2021; 159:24-33. [PMID: 34731746 DOI: 10.1016/j.ejca.2021.09.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/21/2021] [Accepted: 09/02/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND In non-metastatic colorectal cancer (CRC), we evaluated prospectively the pertinence of longitudinal detection and quantification of circulating tumor DNA (ctDNA) as a prognostic marker of recurrence. METHOD The presence of ctDNA was assessed from plasma collected before and after surgery for 184 patients classified as stage II or III and at each visit during 3-4 years of follow-up. The ctDNA analysis was performed by droplet-based digital polymerase chain reaction, targeting mutation and methylation markers, blindly from the clinical outcomes. Multivariate analyses were adjusted on age, gender, stage, and adjuvant chemotherapy. RESULTS Before surgery, 27.5% of patients were positive for ctDNA detection. The rate of recurrence was 32.7% and 11.6% in patients with or without detectable ctDNA respectively (P = 0.001). Time to recurrence (TTR) was significantly shorter in patients with detectable ctDNA before (adjusted hazard ratio [HR] = 3.58, 95% confidence interval [CI] 1.71-7.47) or immediately after surgery (adjusted HR = 3.22, 95% CI 1.32-7.89). The TTR was significantly shorter in patients with detectable ctDNA during the early postoperative follow-up (1-6 months) (adjusted HR = 5, 95% CI 1.9-12.9). Beyond this period, ctDNA remained a prognostic marker with a median anticipated diagnosis of recurrence of 13.1 weeks (interquartile range 28 weeks) when compared to imaging follow-up. The rate of ctDNA+ might be underestimated knowing that consensus pre-analytical conditions were not described at initiation of the study. CONCLUSION This prospective study confirms the relevance of ctDNA as a recurrence risk factor in stage II and III CRC before surgery and as a marker of minimal residual disease after surgery that may predict recurrence several months before imaging techniques.
Collapse
Affiliation(s)
- Leonor Benhaim
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France; Department of Visceral and Surgical Oncology, Gustave Roussy, Villejuif, France,114 Rue Edouard-Vaillant, 94805 Villejuif, France
| | - Olivier Bouché
- Department of Hepatogastroenterology and Digestive Oncology, CHU Reims, 45 Rue Cognacq-Jay, 51092 Reims Cedex, France
| | - Corinne Normand
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France
| | - Audrey Didelot
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France
| | - Claire Mulot
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France; CRB Saints-Pères - EPIGENETEC BB-0033-00055, 45 Rue des Saints Pères, 75006 Paris, France
| | - Delphine Le Corre
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France
| | - Sonia Garrigou
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France
| | - Juliette Djadi-Prat
- Assistance Publique- Hopitaux de Paris, CIC-EC4 URC, Hopital Universitaire Européen Georges-Pompidou - APHP, 20 R Leblanc, 75015 Paris, France
| | - Shu-Fang Wang-Renault
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France
| | - Karla Perez-Toralla
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France
| | - Deniz Pekin
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France
| | - Geoffroy Poulet
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France; Eurofin-Biomnis, 17/19 Avenue Tony Garnier, 69007, Lyon, France
| | - Bruno Landi
- Assistance Publique- Hopitaux de Paris, Department of Oncology, Hopital Universitaire Européen Georges-Pompidou - APHP; Paris Descartes University, 20 R Leblanc, 75015 Paris, France
| | - Julien Taieb
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France; Assistance Publique- Hopitaux de Paris, Department of Oncology, Hopital Universitaire Européen Georges-Pompidou - APHP; Paris Descartes University, 20 R Leblanc, 75015 Paris, France
| | - Marie Selvy
- CHU Clermont Ferrand, 58 Rue Montalembert, 63003 Clermont-Ferrand Cedex 1, France
| | - Jean-Francois Emile
- Assistance Publique- Hopitaux de Paris Department of Pathology, Ambroise-Paré Hospital, Université de Versailles Saint-Quentin-en-Yvelines, 7 Rue Du Parchamp 92100 Boulogne-Billancourt, France
| | - Thierry Lecomte
- Department of Hepatogastroenterology, Tours University Regional Hospital, 49 Bd Béranger, 37044 Tours Cedex 9, France
| | - Helene Blons
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France; Institut Du Cancer PARIS CARPEM, AP-HP, Department of Biology, Hopital Européen Georges-Pompidou, 20 R Leblanc, 75015 Paris, France
| | - Gilles Chatellier
- Assistance Publique- Hopitaux de Paris, CIC-EC4 URC, Hopital Universitaire Européen Georges-Pompidou - APHP, 20 R Leblanc, 75015 Paris, France
| | - Darren R Link
- Bio-Rad Laboratories, Inc., 5731 W Las Positas Blvd, Pleasanton, CA, 94588, United States
| | - Valerie Taly
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France.
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 15 Rue de L'école de Médecine, 75006 Paris, France; Institut Du Cancer PARIS CARPEM, AP-HP, Department of Biology, Hopital Européen Georges-Pompidou, 20 R Leblanc, 75015 Paris, France.
| |
Collapse
|
42
|
Procaccio L, Bergamo F, Daniel F, Rasola C, Munari G, Biason P, Crucitta S, Barsotti G, Zanella G, Angerilli V, Magro C, Paccagnella S, Di Antonio V, Loupakis F, Danesi R, Zagonel V, Del Re M, Lonardi S, Fassan M. A Real-World Application of Liquid Biopsy in Metastatic Colorectal Cancer: The Poseidon Study. Cancers (Basel) 2021; 13:cancers13205128. [PMID: 34680277 PMCID: PMC8533756 DOI: 10.3390/cancers13205128] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND First-line decision making is the key to the successful care of mCRC patients and RAS/BRAF status is crucial to select the best targeted agent. In hub centers, a relevant proportion of patients referred from small volume centers may not have standard tissue-based (STB) molecular results available at the time of the first visit (T0). Liquid biopsy (LB) may help circumvent these hurdles. METHODS A monoinstitutional prospective head-to-head comparison of LB versus (vs.) STB testing was performed in a real-world setting. Selection criteria included: mCRC diagnosis with unknown RAS/BRAF status at T0, tumoral tissue archived in external centers, no previous treatment with anti-EGFR. At T0, patients underwent plasma sampling for LB testing and procedure for tissue recovery. RAS/BRAF genotyping was carried out by droplet digital PCR on circulating-tumoral (ct) DNA. The primary endpoint was the comparison of time to LB (T1) vs. STB (T2) results using the Mann-Whitney U test. Secondary endpoints were the concordance between LB and STB defined as overall percent agreement and the accuracy of LB in terms of specificity, sensitivity, positive and negative predictive value. We also performed an exploratory analysis on urinary (u) ctDNA. RESULTS A total of 33 mCRC patients were included. Mean T1 and T2 was 7 and 22 days (d), respectively (p < 0.00001). T2 included a mean time for archival tissue recovery of 17 d. The overall percent agreement between LB and STB analysis was 83%. Compared to STB testing, LB specificity and sensitivity were 90% and 80%, respectively, with a positive predictive value of 94% and negative one of 69%. In detail, at STB and LB testing, RAS mutation was found in 45% and 42% of patients, respectively; BRAF mutation in 15%. LB results included one false positive and four false negative. False negative cases showed a significantly lower tumor burden at basal CT scan. Concordance between STB and uctDNA testing was 89%. CONCLUSIONS Faster turnaround time, high concordance and accuracy are three key points supporting the adoption of LB in routinary mCRC care, in particular when decision on first-line therapy is urgent and tissue recovery from external centers may require a long time. Results should be interpreted with caution in LB wild-type cases with low tumor burden.
Collapse
Affiliation(s)
- Letizia Procaccio
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Francesca Bergamo
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Francesca Daniel
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Cosimo Rasola
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Giada Munari
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padova, Italy
| | - Paola Biason
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Stefania Crucitta
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Giulia Barsotti
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
- Department of Surgery, Oncology, and Gastroenterology, University of Padova, 35121 Padova, Italy
| | - Giulia Zanella
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Valentina Angerilli
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
| | - Cristina Magro
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Silvia Paccagnella
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
| | - Veronica Di Antonio
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Fotios Loupakis
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Vittorina Zagonel
- Oncology Unit 1, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy; (L.P.); (F.B.); (F.D.); (C.R.); (P.B.); (G.B.); (G.Z.); (C.M.); (V.D.A.); (F.L.); (V.Z.)
| | - Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University Hospital of Pisa, 56121 Pisa, Italy; (S.C.); (R.D.); (M.D.R.)
| | - Sara Lonardi
- Oncology Unit 3, Department of Oncology, Veneto Institute of Oncology—IRCCS, 35128 Padova, Italy
- Correspondence: ; Tel.: +39-0498215953
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padova, 35121 Padova, Italy; (G.M.); (V.A.); (S.P.); (M.F.)
- Veneto Institute of Oncology (IOV-IRCCS), 35128 Padova, Italy
| |
Collapse
|
43
|
Development and Validation of a Novel Serum Prognostic Marker for Patients with Metastatic Colorectal Cancer on Regorafenib Treatment. Cancers (Basel) 2021; 13:cancers13205080. [PMID: 34680230 PMCID: PMC8533782 DOI: 10.3390/cancers13205080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/30/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Regorafenib has proven its efficacy for later-line treatment of mCRC. However, treatment often brings substantial toxicities that lead clinicians to assess the risk-to-benefit ratio in heavily pretreated patients. Thus, it is crucial to develop a prognostic factor and model for guiding patient selection. In this study, we represent a new serum biomarker to serve as an independent prognostic factor for patients receiving regorafenib. All 4 factors of the prognostic model were employed with an excellent discriminatory ability. This result should be validated in further confirmatory studies. Abstract (1) Background: To investigate the prognostic value of cancer-inflammation prognostic index (CIPI) in patients with metastatic colorectal cancer (mCRC) on regorafenib treatment; (2) Methods: Patients with mCRC who were given regorafenib as later-line treatment at Kaohsiung and Linkou Chang-Gung Memorial Hospital between November 2014 and January 2021 were consecutively enrolled. All relevant clinicopathologic, laboratory data and survival status were recorded. Independent prognostic factors were determined by the multivariate Cox regression method; (3) Results: In total, 106 patients in the training cohort and 250 in the validation cohort were enrolled. The median OS for patients with CIPI ≥ 300 and < 300 in the training cohort was 3.8 and 9.0 months, respectively (hazard ratio (HR) 2.78, 95% confidence interval (CI) 1.82–4.23; p < 0.0001). Time to regorafenib, liver metastasis and CIPI were independent factors by multivariate Cox regression analyses. A new scoring model demonstrated a good discriminatory ability to risk stratification of a patient’s survival; (4) Conclusions: We identified CIPI as a novel serum marker highly associated with overall survival in patients with mCRC receiving regorafenib treatment. Further confirmatory studies are warranted.
Collapse
|
44
|
Evaluation of a Targeted Next-Generation Sequencing Panel for the Non-Invasive Detection of Variants in Circulating DNA of Colorectal Cancer. J Clin Med 2021; 10:jcm10194487. [PMID: 34640513 PMCID: PMC8509146 DOI: 10.3390/jcm10194487] [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: 07/22/2021] [Revised: 09/20/2021] [Accepted: 09/24/2021] [Indexed: 12/17/2022] Open
Abstract
Molecular profiling of circulating cell-free DNA (cfDNA) has shown utility for the management of colorectal cancer (CRC). TruSight Tumor 170 (TST170) is a next-generation sequencing (NGS) panel that covers 170 cancer-related genes, including KRAS, which is a key driver gene in CRC. We evaluated the capacity of TST170 to detect gene variants in cfDNA from a retrospective cohort of 20 metastatic CRC patients with known KRAS variants in tumor tissue and in cfDNA previously analyzed by pyrosequencing and BEAMing, respectively. The cfDNA of most of the patients (95%) was successfully sequenced. We frequently detected variants with clinical significance in KRAS (79%, 15/19) and PIK3CA (26%, 5/19) genes. Variants with potential clinical significance were also identified in another 27 cancer genes, such as APC. The type of KRAS variant detected in cfDNA by TST170 showed high concordance with those detected in tumor tissue (77%), and very high concordance with cfDNA analyzed by BEAMing (94%). The variant allele fractions for KRAS obtained in cfDNA by TST170 and BEAMing correlated strongly. This proof-of-principle study indicates that targeted NGS analysis of cfDNA with TST170 could be useful for non-invasive detection of gene variants in metastatic CRC patients, providing an assay that could be easily implemented for detecting somatic alterations in the clinic.
Collapse
|
45
|
Moati E, Taly V, Garinet S, Didelot A, Taieb J, Laurent-Puig P, Zaanan A. Role of Circulating Tumor DNA in Gastrointestinal Cancers: Current Knowledge and Perspectives. Cancers (Basel) 2021; 13:4743. [PMID: 34638228 PMCID: PMC8507552 DOI: 10.3390/cancers13194743] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/06/2021] [Accepted: 09/18/2021] [Indexed: 12/21/2022] Open
Abstract
Gastrointestinal (GI) cancers are major health burdens worldwide and biomarkers are needed to improve the management of these diseases along their evolution. Circulating tumor DNA (ctDNA) is a promising non-invasive blood and other bodily-fluid-based biomarker in cancer management that can help clinicians in various cases for the detection, diagnosis, prognosis, monitoring and personalization of treatment in digestive oncology. In addition to the well-studied prognostic role of ctDNA, the main real-world applications appear to be the assessment of minimal residual disease to further guide adjuvant therapy and predict relapse, but also the monitoring of clonal evolution to tailor treatments in metastatic setting. Other challenges such as predicting response to treatment including immune checkpoint inhibitors could also be among the potential applications of ctDNA. Although the level of advancement of ctDNA development in the different tumor localizations is still inhomogeneous, it might be now reliable enough to be soon used in clinical routine for colorectal cancers and shows promising results in other GI cancers.
Collapse
Affiliation(s)
- Emilie Moati
- Department of Gastroenterology and Digestive Oncology, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France; (E.M.); (J.T.)
| | - Valerie Taly
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
| | - Simon Garinet
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
- Department of Biochemistry, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France
| | - Audrey Didelot
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
| | - Julien Taieb
- Department of Gastroenterology and Digestive Oncology, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France; (E.M.); (J.T.)
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
| | - Pierre Laurent-Puig
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
- Department of Biochemistry, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France
| | - Aziz Zaanan
- Department of Gastroenterology and Digestive Oncology, Institut du Cancer Paris Carpem, Assistance Publique des Hôpitaux de Paris, European Georges Pompidou Hospital, 75015 Paris, France; (E.M.); (J.T.)
- Centre de Recherche des Cordeliers, INSERM UMRS1138, Centre National de la Recherche Scientifique, Sorbonne Université, USPC, Université de Paris, Equipe Labellisée Ligue Nationale Contre le Cancer, CNRS SNC 5096, 75006 Paris, France; (V.T.); (S.G.); (A.D.); (P.L.-P.)
| |
Collapse
|
46
|
Clinical Applications of Minimal Residual Disease Assessments by Tumor-Informed and Tumor-Uninformed Circulating Tumor DNA in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13184547. [PMID: 34572774 PMCID: PMC8471730 DOI: 10.3390/cancers13184547] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/08/2021] [Accepted: 09/08/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Circulating tumor DNA, or ctDNA, are fragments of tumor DNA that can be detected in the blood of patients with colorectal cancer. Measuring ctDNA levels in the blood has shown the potential to provide important information that can be helpful in the clinical care of patients with colorectal cancer. For example, in patients with colon cancer that has been removed by surgery, measuring ctDNA in the blood can predict the likelihood of cancer recurrence, while in those with metastatic colorectal cancer, measuring ctDNA can inform the clinician whether chemotherapy is effective at earlier timepoints than currently available tests. In this review, we discuss the results from ongoing studies describing the utility of ctDNA measurements across all stages of colorectal cancer. We also discuss the various clinical scenarios that ctDNA may have the most immediate impact in colorectal cancer management. Abstract Emerging data suggest that circulating tumor DNA (ctDNA) can detect colorectal cancer (CRC)-specific signals across both non-metastatic and metastatic settings. With the development of multiple platforms, including tumor-informed and tumor-agnostic ctDNA assays and demonstration of their provocative analytic performance to detect minimal residual disease, there are now ongoing, phase III randomized clinical trials to evaluate their role in the management paradigm of CRC. In this review, we highlight landmark studies that have formed the basis for ongoing studies on the clinically applicability of plasma ctDNA assays in resected, stage I–III CRC and metastatic CRC. We discuss clinical settings by which ctDNA may have the most immediate impact in routine clinical practice. These include the potential for ctDNA to (1) guide surveillance and intensification or de-intensification strategies of adjuvant therapy in resected, stage I–III CRC, (2) predict treatment response to neoadjuvant therapy in locally advanced rectal cancer inclusive of total neoadjuvant therapy (TNT), and (3) predict response to systemic and surgical therapies in metastatic disease. We end by considering clinical variables that can influence our ability to reliably interpret ctDNA dynamics in the clinic.
Collapse
|
47
|
Personeni N, Smiroldo V, Giunta EF, Prete MG, Rimassa L, Bregni G, Sclafani F. Tackling Refractory Metastatic Colorectal Cancer: Future Perspectives. Cancers (Basel) 2021; 13:4506. [PMID: 34572729 PMCID: PMC8472765 DOI: 10.3390/cancers13184506] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/01/2021] [Accepted: 09/01/2021] [Indexed: 01/09/2023] Open
Abstract
Substantial improvements have characterized the systemic treatment of metastatic colorectal cancer (mCRC) over the past 20 years. Besides strong evidence that supports the use of RAS and BRAF status as prognostic and predictive indicators of disease and response, novel technologies have made possible the incorporation of emerging biomarkers for the management of mCRC. On one hand, the discovery of point mutations, amplifications, fusions, and gene expression profiles highlights the genomic and dynamic complexity of CRC. On the other, such discoveries are leading to newer biomarker-driven strategies that add to existing anti-epidermal growth factor receptor (EGFR) and anti-angiogenic approaches. In addition, the availability of a wide molecular profiling has relevant implications for patient prognosis and treatment benefits. Here, we will review the molecular underpinnings and clinical data supporting novel targeted treatments under development for refractory mCRC harboring BRAF mutations, KRAS G12C mutations, HER2 amplification, and less common molecular alterations, such as the re-arrangements of NTRK, ALK, and ROS1. Additionally, we will discuss novel strategies driving the rechallenge of EGFR antibodies and the incorporation of newer anti-angiogenic agents in the therapeutic armamentarium.
Collapse
Affiliation(s)
- Nicola Personeni
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Milan, Italy; (N.P.); (M.G.P.)
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy;
| | - Valeria Smiroldo
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy;
| | - Emilio Francesco Giunta
- Medical Oncology Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Via Pansini 5, 80131 Naples, Italy;
| | - Maria Giuseppina Prete
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Milan, Italy; (N.P.); (M.G.P.)
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy;
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Milan, Italy; (N.P.); (M.G.P.)
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Milan, Italy;
| | - Giacomo Bregni
- Department of Medical Oncology, Institut Jules Bordet–Université Libre de Bruxelles (ULB), Boulevard de Waterloo 121, 1000 Bruxelles, Belgium; (G.B.); (F.S.)
| | - Francesco Sclafani
- Department of Medical Oncology, Institut Jules Bordet–Université Libre de Bruxelles (ULB), Boulevard de Waterloo 121, 1000 Bruxelles, Belgium; (G.B.); (F.S.)
| |
Collapse
|
48
|
Sefrioui D, Beaussire L, Gillibert A, Blanchard F, Toure E, Bazille C, Perdrix A, Ziegler F, Gangloff A, Hassine M, Elie C, Bignon AL, Parzy A, Gomez P, Thill C, Clatot F, Sabourin JC, Frebourg T, Benichou J, Bouhier-Leporrier K, Gallais MP, Sarafan-Vasseur N, Michel P, Di Fiore F. CEA, CA19-9, circulating DNA and circulating tumour cell kinetics in patients treated for metastatic colorectal cancer (mCRC). Br J Cancer 2021; 125:725-733. [PMID: 34112948 PMCID: PMC8405627 DOI: 10.1038/s41416-021-01431-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 04/17/2021] [Accepted: 04/28/2021] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND We previously reported that CEA kinetics are a marker of progressive disease (PD) in metastatic colorectal cancer (mCRC). This study was specifically designed to confirm CEA kinetics for predicting PD and to evaluate CA19-9, cell-free DNA (cfDNA), circulating tumour DNA (ctDNA) and circulating tumour cell (CTC) kinetics. METHODS Patients starting a chemotherapy (CT) with pre-treatment CEA > 5 ng/mL and/or CA19.9 > 30 UI/mL were prospectively included. Samples were collected from baseline to cycle 4 for CEA and CA19-9 and at baseline and the sixth week for other markers. CEA kinetics were calculated from the first to the third or fourth CT cycle. RESULTS A total of 192 mCRC patients were included. CEA kinetics based on the previously identified >0.05 threshold was significantly associated with PD (p < 0.0001). By dichotomising by the median value, cfDNA, ctDNA and CA19-9 were associated with PD, PFS and OS in multivariate analysis. A circulating scoring system (CSS) combining CEA kinetics and baseline CA19-9 and cfDNA values classified patients based on high (n = 58) and low risk (n = 113) of PD and was independently associated with PD (ORa = 4.6, p < 0.0001), PFS (HRa = 2.07, p < 0.0001) and OS (HRa = 2.55, p < 0.0001). CONCLUSIONS CEA kinetics alone or combined with baseline CA19-9 and cfDNA are clinically relevant for predicting outcomes in mCRC. TRIAL REGISTRATION NUMBER NCT01212510.
Collapse
Affiliation(s)
- David Sefrioui
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Ludivine Beaussire
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - André Gillibert
- grid.41724.34Department of Biostatistics, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - France Blanchard
- grid.41724.34Department of Pathology, Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Emmanuel Toure
- grid.41724.34Department of Pathology, Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Céline Bazille
- grid.411149.80000 0004 0472 0160Department of Pathology, Caen University Hospital, Caen, France
| | - Anne Perdrix
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Biopathology, Henri Becquerel Centre, Rouen, France
| | - Frédéric Ziegler
- grid.7429.80000000121866389Normandie Univ, UNIROUEN, INSERM U1073, Rouen University Hospital and General Biochemistry Laboratory, Institute of Clinical Biology, Rouen, France
| | - Alice Gangloff
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Mélanie Hassine
- Department of Hepatogastroenterology, Elbeuf Hospital, Elbeuf, France
| | - Caroline Elie
- Department of Hepatogastroenterology, Elbeuf Hospital, Elbeuf, France
| | - Anne-Laure Bignon
- grid.411149.80000 0004 0472 0160Department of Hepatogastroenterology, Caen University Hospital, Caen, France
| | - Aurélie Parzy
- Department of Hepatogastroenterology, Francois Baclesse Centre, Caen, France
| | - Philippe Gomez
- Department of Medical Oncology, Frédéric Joliot Centre, Rouen, France
| | - Caroline Thill
- grid.41724.34Department of Biostatistics, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - Florian Clatot
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Medical Oncology, Henri Becquerel Centre, Rouen, France
| | - Jean-Christophe Sabourin
- grid.41724.34Department of Pathology, Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Thierry Frebourg
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Genetics, Rouen, France
| | - Jacques Benichou
- grid.41724.34Department of Biostatistics, Normandie Univ, UNIROUEN, Rouen University Hospital, Rouen, France
| | - Karine Bouhier-Leporrier
- grid.411149.80000 0004 0472 0160Department of Hepatogastroenterology, Caen University Hospital, Caen, France
| | | | - Nasrin Sarafan-Vasseur
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Pierre Michel
- grid.41724.34Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine and Department of Hepatogastroenterology, Rouen, France
| | - Frédéric Di Fiore
- grid.41724.34Department of Hepatogastroenterology and Department of Medical Oncology, Henri Becquerel Centre, Normandie Univ, UNIROUEN, Inserm U1245, IRON Group, Rouen University Hospital, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| |
Collapse
|
49
|
Hsu HC, Huang KC, Chen WS, Jiang JK, Yang SH, Wang HS, Chang SC, Lan YT, Lin CC, Lin HH, Huang SC, Cheng HH, Yang TS, Chen CC, Chao Y, Teng HW. Preference criteria for regorafenib in treating refractory metastatic colorectal cancer are the small tumor burden, slow growth and poor/scanty spread. Sci Rep 2021; 11:15370. [PMID: 34321583 PMCID: PMC8319410 DOI: 10.1038/s41598-021-94968-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 07/19/2021] [Indexed: 02/08/2023] Open
Abstract
Given the unclear preference criteria for regorafenib in treating refractory metastatic colorectal cancer (mCRC), this study aimed to construct an algorithm in selecting right patients for regorafenib. This was a multicenter retrospective cohort study. Patients with pathology confirmed mCRC and administered with regorafenib for > 3 weeks were enrolled. Patients with good response were defined to have progression-free survival (PFS) of ≥ 4 months. The Kaplan–Meier plot was used to analyze survival. A Cox proportional hazards model was used to analyze univariate and multivariate prognostic factors and was visualized using forest plot. A clustering heatmap was used to classify patients according to responses. The decision tree and nomogram were used to construct the approaching algorithm. A total of 613 patients was analyzed. The median PFS and overall survival (OS) were 2.7 and 10.6 months, respectively. The partial response and stable disease rate are 2.4% and 36.4%. The interval between metastasis (M1) and regorafenib, metastatic status (number, liver, and brain), and CEA level were independent prognostics factors of PFS that classifies patients into three groups: good, bad and modest-1/modest-2 group with PFS > = 4 months rates of 51%, 20%, 39% and 30%, respectively. Results were used to develop the decision tree and nomogram for approaching patients indicated with regorafenib. The preference criteria for regorafenib in treating patients with refractory mCRC are small tumor burden (CEA), slow growth (interval between metastasis and regorafenib) and poor/scanty spread (metastatic status: number and sites of metastasis): The 3S rules. TRIAL registration ClinicalTrials.gov Identifier: NCT03829852; Date of first registration (February 11, 2019).
Collapse
Affiliation(s)
- Hung-Chih Hsu
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Tao-Yuan City, Taiwan.,College of Medicine, Chang Gung University, Tao-Yuan City, Taiwan
| | - Kuo-Cheng Huang
- Department of Hematology and Medical Oncology, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Wei-Shone Chen
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Jeng-Kai Jiang
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Shung-Haur Yang
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan.,Department of Surgery, National Yang Ming Chiao Tung University Hospital, Yilan, Taiwan
| | - Huann-Sheng Wang
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Shih-Ching Chang
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Yuan-Tzu Lan
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Chun-Chi Lin
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Hung-Hsin Lin
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Sheng-Chieh Huang
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Hou-Hsuan Cheng
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Surgery, Division of Colon and Rectal Surgery, Taipei Veterans General Hospital, Taipei, 112, Taiwan
| | - Tsai-Sheng Yang
- Division of Hematology-Oncology, Chang Gung Memorial Hospital at Linkou, Tao-Yuan City, Taiwan.,College of Medicine, Chang Gung University, Tao-Yuan City, Taiwan
| | - Chien-Chih Chen
- Department of Surgery, Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | - Yee Chao
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan.,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan.,Department of Oncology, Division of Medical Oncology, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan
| | - Hao-Wei Teng
- School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan. .,School of Medicine, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 112, Taiwan. .,Department of Oncology, Division of Medical Oncology, Taipei Veterans General Hospital, No. 201, Sec. 2, Shih-Pai Road, Taipei, 112, Taiwan.
| |
Collapse
|
50
|
Lu Z, Nie B, Zhai W, Hu Z. Delineating the longitudinal tumor evolution using organoid models. J Genet Genomics 2021; 48:560-570. [PMID: 34366272 DOI: 10.1016/j.jgg.2021.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 06/11/2021] [Accepted: 06/16/2021] [Indexed: 02/07/2023]
Abstract
Cancer is an evolutionary process fueled by genetic or epigenetic alterations in the genome. Understanding the evolutionary dynamics that are operative at different stages of tumor progression might inform effective strategies in early detection, diagnosis, and treatment of cancer. However, our understanding on the dynamics of tumor evolution through time is very limited since it is usually impossible to sample patient tumors repeatedly. The recent advances in in vitro 3D organoid culture technologies have opened new avenues for the development of more realistic human cancer models that mimic many in vivo biological characteristics in human tumors. Here, we review recent progresses and challenges in cancer genomic evolution studies and advantages of using tumor organoids to study cancer evolution. We propose to establish an experimental evolution model based on continuous passages of patient-derived organoids and longitudinal sampling to study clonal dynamics and evolutionary patterns over time. Development and integration of population genetic theories and computational models into time-course genomic data in tumor organoids will help to pinpoint the key cellular mechanisms underlying cancer evolutionary dynamics, thus providing novel insights on therapeutic strategies for highly dynamic and heterogeneous tumors.
Collapse
Affiliation(s)
- Zhaolian Lu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Beina Nie
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Weiwei Zhai
- CAS Key Laboratory of Zoological Systematics and Evolution, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming 650223, China
| | - Zheng Hu
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|