1
|
Vaghi C, Mauri G, Agostara AG, Patelli G, Pizzutilo EG, Nakamura Y, Yoshino T, Siena S, Sartore-Bianchi A. The predictive role of ERBB2 point mutations in metastatic colorectal cancer: A systematic review. Cancer Treat Rev 2023; 112:102488. [PMID: 36410093 DOI: 10.1016/j.ctrv.2022.102488] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/03/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
Abstract
ERBB2 amplification is a driver oncogenic alteration in many cancers and it has recently been incorporated among therapeutically actionable biomarkers also in metastatic colorectal cancer (mCRC). In contrast, the role of ERBB2 point mutations, which are detectable in up to 3% of CRC patients, remains to be assessed. In this systematic review, we collected preclinical and clinical data addressing the role of ERBB2 point mutations in mCRC patients as a predictive biomarker for anti-EGFR and anti-HER2 targeted agents, and as mechanism of acquired resistance to ERBB2 amplified mCRC treated with any anti-HER2 regimen. In both preclinical and clinical studies, most ERBB2 point mutations were associated with resistance to anti-EGFR agents, particularly L755S and R784G, which occur in the HER2 protein kinase domain. No ERBB2 mutation was associated with tumor response to HER2-targeted agents in mCRC patients, although signals of activity were observed in preclinical models. Eight ongoing clinical trials are underway to test different anti-HER2 treatments in ERBB2 mutant mCRC. Several reports documented the emergence of ERBB2 mutations in the circulating tumor DNA (ctDNA) of ERBB2 amplified mCRC progressing to anti-HER2 agents, thus hinting a role in acquired resistance.
Collapse
Affiliation(s)
- Caterina Vaghi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Gianluca Mauri
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy; IFOM-Istituto Fondazione di Oncologia Molecolare ETS, Milano, Italy
| | - Alberto Giuseppe Agostara
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Giorgio Patelli
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Elio Gregory Pizzutilo
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Salvatore Siena
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy
| | - Andrea Sartore-Bianchi
- Department of Hematology, Oncology, and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, 20162 Milano, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milano, Italy.
| |
Collapse
|
2
|
Tao H, Shen M, Zhang X, Wang M, Wu Y, Sun H, Ling C, Yang Y, Chen K, Li D. A study of gene variation in All- RAS wild-type metastatic colorectal cancer and its correlation with cetuximab. J Gastrointest Oncol 2022; 13:3009-3024. [PMID: 36636055 PMCID: PMC9830345 DOI: 10.21037/jgo-22-1237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 12/15/2022] [Indexed: 12/28/2022] Open
Abstract
Background This study sought to explore the biological significance of genetic variation in RAS wild-type metastatic colorectal cancer (mCRC) in the real world, the difference in the efficacy of cetuximab in the treatment of mCRC with different genetic variants and identify clinical features and new predictors of efficacy. Methods A retrospective analysis of the data of 60 patients with stage IV mCRC who received cetuximab at The First and Second Affiliated Hospital of Soochow University from 2016 to 2020 was conducted. The patients were divided into the following 3 groups according to the genetic test results: (I) group A (the all-RAS wild-type group); (II) group B (the all-RAS wild-type group with the tumor suppressor gene mutation); and (III) group C (the all-RAS wild-type group with the oncogenic driver gene mutation). A subgroup analysis was conducted to examine left CRC and local intervention, and the progression-free survival (PFS) and overall survival (OS) of the patients were observed. Results The all-RAS wild-type mCRC patients were divided into group A (n=10), group B (including the TP53, APC, PTEN, BRCA2, and SMAD4 variants) (n=42), and group C (including the ERBB2, BRAF, PIK3CA, and RET variants) (n=8). The median PFS of groups A, B, and C were 15.0, 12.0, and 3.0 months, respectively (P=0.007). Fitting sex as a stratified variable to the Cox survival analysis model showed that only the PFS of groups B and C differed significantly (P=0.011). In the left-sided mCRC patients, the median PFS of groups A, B, C were 3.0, 13.0, and 3.0 months, respectively (P=0.009). Among the patients in group B, the median PFS of the metastatic local intervention subgroup was 14.0 months, and the non-local intervention subgroup was 12.0 months (P=0.55). Only the type of combined gene mutation was an independent factor affecting PFS. Conclusions The PFS and OS of mCRC patients with all-RAS wild-type and no combined mutations treated with cetuximab were not better than those of patients with combined mutations. Compared to mCRC patients with all-RAS wild-type and oncogenic driver gene mutations, cetuximab significantly prolonged the PFS of all-RAS wild-type patients with the tumor suppressor gene mutations.
Collapse
Affiliation(s)
- Huimin Tao
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Meng Shen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaochang Zhang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Minghui Wang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yan Wu
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hui Sun
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chen Ling
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ying Yang
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Kai Chen
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Dapeng Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
3
|
Crisafulli G, Sartore-Bianchi A, Lazzari L, Pietrantonio F, Amatu A, Macagno M, Barault L, Cassingena A, Bartolini A, Luraghi P, Mauri G, Battuello P, Personeni N, Zampino MG, Pessei V, Vitiello PP, Tosi F, Idotta L, Morano F, Valtorta E, Bonoldi E, Germano G, Di Nicolantonio F, Marsoni S, Siena S, Bardelli A. Temozolomide Treatment Alters Mismatch Repair and Boosts Mutational Burden in Tumor and Blood of Colorectal Cancer Patients. Cancer Discov 2022; 12:1656-1675. [PMID: 35522273 PMCID: PMC9394384 DOI: 10.1158/2159-8290.cd-21-1434] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 04/16/2022] [Accepted: 05/04/2022] [Indexed: 01/07/2023]
Abstract
The majority of metastatic colorectal cancers (mCRC) are mismatch repair (MMR) proficient and unresponsive to immunotherapy, whereas MMR-deficient (MMRd) tumors often respond to immune-checkpoint blockade. We previously reported that the treatment of colorectal cancer preclinical models with temozolomide (TMZ) leads to MMR deficiency, increased tumor mutational burden (TMB), and sensitization to immunotherapy. To clinically translate these findings, we designed the ARETHUSA clinical trial whereby O6-methylguanine-DNA-methyltransferase (MGMT)-deficient, MMR-proficient, RAS-mutant mCRC patients received priming therapy with TMZ. Analysis of tissue biopsies and circulating tumor DNA (ctDNA) revealed the emergence of a distinct mutational signature and increased TMB after TMZ treatment. Multiple alterations in the nucleotide context favored by the TMZ signature emerged in MMR genes, and the p.T1219I MSH6 variant was detected in ctDNA and tissue of 94% (16/17) of the cases. A subset of patients whose tumors displayed the MSH6 mutation, the TMZ mutational signature, and increased TMB achieved disease stabilization upon pembrolizumab treatment. SIGNIFICANCE MMR-proficient mCRCs are unresponsive to immunotherapy. We provide the proof of concept that inactivation of MMR genes can be achieved pharmacologically with TMZ and molecularly monitored in the tissue and blood of patients with mCRC. This strategy deserves additional evaluation in mCRC patients whose tumors are no longer responsive to standard-of-care treatments. See related commentary by Willis and Overman, p. 1612. This article is highlighted in the In This Issue feature, p. 1599.
Collapse
Affiliation(s)
- Giovanni Crisafulli
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Luca Lazzari
- The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Alessio Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Marco Macagno
- Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Ludovic Barault
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Andrea Cassingena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Paolo Luraghi
- The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Gianluca Mauri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy.,The FIRC Institute of Molecular Oncology, Milan, Italy
| | - Paolo Battuello
- Department of Oncology, University of Torino, Candiolo, Italy
| | - Nicola Personeni
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Maria Giulia Zampino
- Division of Gastrointestinal Medical Oncology and Neuroendocrine Tumors, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Pietro Paolo Vitiello
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Federica Tosi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Laura Idotta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Emanuele Valtorta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Emanuela Bonoldi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giovanni Germano
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | - Federica Di Nicolantonio
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy
| | | | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Alberto Bardelli
- Department of Oncology, University of Torino, Candiolo, Italy.,Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.,Corresponding Author: Alberto Bardelli, University of Turin, Department of Oncology, Candiolo Cancer Institute, FPO - IRCCS, Str.Prov.le 142, km 3.95, 10060, Candiolo, Torino, Italy. Phone/Fax: 39-011-993-3235; E-mail:
| |
Collapse
|
4
|
Boas FE, Kemeny NE, Sofocleous CT, Yeh R, Thompson VR, Hsu M, Moskowitz CS, Ziv E, Yarmohammadi H, Bendet A, Solomon SB. Bronchial or Pulmonary Artery Chemoembolization for Unresectable and Unablatable Lung Metastases: A Phase I Clinical Trial. Radiology 2021; 301:474-484. [PMID: 34463550 DOI: 10.1148/radiol.2021210213] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Background Lung chemoembolization is an emerging treatment option for lung tumors, but the optimal embolic, drug, and technique are unknown. Purpose To determine the technical success rate and safety of bronchial or pulmonary artery chemoembolization of lung metastases using ethiodized oil, mitomycin, and microspheres. Materials and Methods Patients with unresectable and unablatable lung, endobronchial, or mediastinal metastases, who failed systemic chemotherapy, were enrolled in this prospective, single-center, single-arm, phase I clinical trial (December 2019-September 2020). Pulmonary and bronchial angiography was performed to determine the blood supply to the lung metastases. Based on the angiographic findings, bronchial or pulmonary artery chemoembolization was performed using an ethiodized oil and mitomycin emulsion, followed by microspheres. The primary objectives were technical success rate and safety, according to the National Cancer Institute Common Terminology Criteria for Adverse Events. CIs of proportions were estimated with the equal-tailed Jeffreys prior interval, and correlations were evaluated with the Spearman test. Results Ten participants (median age, 60 years; interquartile range, 52-70 years; six women) were evaluated. Nine of the 10 participants (90%) had lung metastases supplied by the bronchial artery, and one of the 10 participants (10%) had lung metastases supplied by the pulmonary artery. The technical success rate of intratumoral drug delivery was 10 of 10 (100%) (95% CI: 78, 100). There were no severe adverse events (95% CI: 0, 22). The response rate of treated tumors was one of 10 (10%) according to the Response Evaluation Criteria in Solid Tumors and four of 10 (40%) according to the PET Response Criteria in Solid Tumors. Ethiodized oil retention at 4-6 weeks was correlated with reduced tumor size (ρ = -0.83, P = .003) and metabolic activity (ρ = -0.71, P = .03). Pharmacokinetics showed that 45% of the mitomycin dose underwent burst release in 2 minutes, and 55% of the dose was retained intratumorally with a half-life of more than 5 hours. The initial tumor-to-plasma ratio of mitomycin concentration was 380. Conclusion Lung chemoembolization was technically successful for the treatment of lung, mediastinal, and endobronchial metastases, with no severe adverse events. Clinical trial registration no. NCT04200417 © RSNA, 2021 Online supplemental material is available for this article. See also the editorial by Georgiades et al in this issue.
Collapse
Affiliation(s)
- F Edward Boas
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Nancy E Kemeny
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Constantinos T Sofocleous
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Randy Yeh
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Vanessa R Thompson
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Meier Hsu
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Chaya S Moskowitz
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Etay Ziv
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Hooman Yarmohammadi
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Achiude Bendet
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| | - Stephen B Solomon
- From the Department of Radiology, City of Hope Cancer Center, 1500 E Duarte Rd, Duarte, CA 91010 (F.E.B.); Interventional Radiology Service, Department of Radiology (F.E.B., C.T.S., E.Z., H.Y., A.B., S.B.S.), Department of Medicine (N.E.K.), Molecular Imaging and Therapy Service (R.Y.), and Department of Epidemiology and Biostatistics (M.H., C.S.M.), Memorial Sloan-Kettering Cancer Center, New York, NY; and Antitumor Assessment Core Facility, Sloan Kettering Institute, New York, NY (V.R.T.)
| |
Collapse
|
5
|
Kong C, Fu T. Value of methylation markers in colorectal cancer (Review). Oncol Rep 2021; 46:177. [PMID: 34212989 DOI: 10.3892/or.2021.8128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/18/2021] [Indexed: 11/05/2022] Open
Abstract
Colorectal cancer (CRC) is a multifactorial and multistage process that occurs due to both genetic and epigenetic variations in normal epithelial cells. Analysis of the CRC epigenome has revealed that almost all CRC types have a large number of abnormally methylated genes. Hypermethylation of cell‑free DNA from CRC in the blood or stool is considered as a potential non‑invasive cancer biomarker, and various methylation markers have shown high sensitivity and specificity. The aim of the present review was to examine potential methylation markers in CRC that have been used or are expected to be used in the clinical setting, focusing on their screening, predictive, prognostic and therapeutic roles in CRC.
Collapse
Affiliation(s)
- Can Kong
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Tao Fu
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
6
|
Chung C. Predictive and prognostic biomarkers with therapeutic targets in colorectal cancer: A 2021 update on current development, evidence, and recommendation. J Oncol Pharm Pract 2021; 28:850-869. [PMID: 33832365 DOI: 10.1177/10781552211005525] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although therapeutically actionable molecular alterations are widely distributed across many cancer types, only a handful of them show evidence of clinical utility and are recommended for routine clinical practice in the management of cancers of colon and rectum (CRC). This 2021 update aims to provide a succinct summary on the use of prognostic and/or predictive biomarkers (expanded RAS, BRAF, microsatellite-high [MSI-H] or deficient mismatch repair [dMMR], neurotrophic tyrosine receptor kinase [NTRK] fusion genes, and human epidermal growth factor receptor type II [HER2] gene amplification) associated with CRC. Therapeutic implications of each relevant predictive or prognostic biomarker for patients with CRC are described, along with discussion on new developments on (1) biomarker-driven therapies such as testing of BRAF, MLH1 promoter methylation and MMR germline genes in differentiating sporadic CRC or hereditary conditions such as Lynch syndrome; (2) first-line use of immune checkpoint inhibitors in metastatic CRC; (3) risk stratification and therapy selection based on primary tumor location (left-sided vs. right-sided colon cancer); (3) atypical BRAF mutations; (4) use of EGFR directed therapy in the perioperative oligometastatic disease setting; (5) re-challenge of EGFR directed therapy and (6) personalizing therapy of fluoropyrimidine and irinotecan based on new evidence in pharmacogenomic testing. Data are collected and analyzed from available systematic reviews and meta-analyses of treatments with known therapeutic targets in CRC, which may be associated with predictive and/or prognostic biomarkers. Discussions are presented in an application-based format, with goal to empower pharmacists or other clinicians to gain awareness and understanding in biomarker-driven cancer therapy issues.
Collapse
Affiliation(s)
- Clement Chung
- 23530Houston Methodist West Hospital, Houston, TX, USA
| |
Collapse
|
7
|
Lau DK, Burge M, Roy A, Chau I, Haller DG, Shapiro JD, Peeters M, Pavlakis N, Karapetis CS, Tebbutt NC, Segelov E, Price TJ. Update on optimal treatment for metastatic colorectal cancer from the AGITG expert meeting: ESMO congress 2019. Expert Rev Anticancer Ther 2020; 20:251-270. [PMID: 32186929 DOI: 10.1080/14737140.2020.1744439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Outcomes in metastatic colorectal cancer are improving, due to the tailoring of therapy enabled by better understanding of clinical behavior according to molecular subtype.Areas covered: A review of the literature and recent conference presentations was undertaken on the topic of systemic treatment of metastatic colorectal cancer. This review summarizes expert discussion of the current evidence for therapies in metastatic colorectal cancer (mCRC) based on molecular subgrouping.Expert opinion: EGFR-targeted and VEGF-targeted antibodies are now routinely incorporated into treatment strategies for mCRC. EGFR-targeted antibodies are restricted to patients with extended RAS wild-type profiles, with evidence that they should be further restricted to patients with left-sided tumors. Clinically distinct treatment pathways based on tumor RAS, BRAF, HER2 and MMR status, are now clinically applicable. Evidence suggests therapy for additional subgroups will soon be defined; the most advanced being for patients with KRAS G12 C mutation and gene TRK fusion defects.
Collapse
Affiliation(s)
- David K Lau
- GI and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London and Surrey, UK
| | - Matthew Burge
- Medical Oncology, Royal Brisbane Hospital, Brisbane, Australia.,University of Queensland, Brisbane, Australia
| | - Amitesh Roy
- Medical Oncology, Flinders Centre for Innovation in Cancer, Bedford Park, Australia
| | - Ian Chau
- GI and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London and Surrey, UK
| | - Daniel G Haller
- Abramson Cancer Center at the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeremy D Shapiro
- Monash University, Melbourne, Australia.,Medical Oncology, Cabrini Medical Centre, Melbourne, Australia
| | - Marc Peeters
- Medical Oncology, University Hospital Antwerp, Edegem, Belgium
| | - Nick Pavlakis
- Medical Oncology, Royal North Shore Hospital, St Leonards, Australia.,Sydney University, Camperdown, Sydney, Australia
| | | | - Niall C Tebbutt
- Medical Oncology, Austin Health, Heidelberg, Australia.,Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Eva Segelov
- Monash University, Melbourne, Australia.,Medical Oncology, Monash Medical Centre, Clayton, Australia
| | - Timothy J Price
- Medical Oncology, The Queen Elizabeth Hospital, Woodville, Australia
| |
Collapse
|
8
|
Chu P, Batson S, Hodgson M, Mitchell CR, Steenrod A. Systematic review of neurotrophic tropomyosin-related kinase inhibition as a tumor-agnostic management strategy. Future Oncol 2020; 16:61-74. [PMID: 31942815 DOI: 10.2217/fon-2019-0534] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Aim: To conduct a systematic review and meta-analysis feasibility of clinical, quality of life and economic evidence for neurotrophic tropomyosin-related receptor tyrosine kinases (NTRK) inhibitors in patients with NTRK gene fusion-positive tumors. Materials & methods: Databases were searched for studies on NTRK inhibitors in adult and pediatric patients. Results: 27 publications reported clinical data for seven interventions. Efficacy/safety data were available for two interventions only. Four trials each reported data for larotrectinib and entrectinib with pooled analyses reporting objective response rates of 75% (95% CI: 61-85) and 57.4% (43.2-70.8), respectively. No publications reported economic or quality of life evidence. Conclusion: Preliminary data demonstrate that NTRK inhibitors are well tolerated and show impressive clinical benefit; corroboration of existing studies and real-world data are required.
Collapse
Affiliation(s)
- Paula Chu
- F Hoffmann-La Roche Ltd., Global Access, 4070 Basel, Switzerland
| | - Sarah Batson
- Mtech Access Ltd., 30 Murdock Road, Bicester, OX26 4PP, UK
| | - Matthew Hodgson
- Roche Products Ltd., Health Economics and Strategic Pricing, Welwyn Garden City, AL7 1TW, UK
| | | | - Anna Steenrod
- F Hoffmann-La Roche Ltd., Global Access, 4070 Basel, Switzerland
| |
Collapse
|
9
|
Abstract
Background The incorporation of novel biomarkers into therapy selection for patients with metastatic colorectal cancer (mcrc) has significantly improved outcomes. Optimal treatment planning now takes into account diverse characteristics of patients and their tumours to create personalized therapeutic plans. Discussion This review is split into two sections. In the first section, we review the prognostic and predictive significance of expanded RAS mutation testing, BRAF mutations, ERBB2 (her2) amplification, microsatellite instability (msi) and deficient mismatch repair (dmmr) protein, NTRK fusions, PIK3CA mutations, and met amplifications. The therapeutic implication of each of those biomarkers for personalizing therapies for each patient with mcrc is discussed. In the second section, we touch on testing methods and considerations of relevance to clinicians when they interpret companion diagnostics meant to guide therapy selection. The advantages and pitfalls of various methods are evaluated, and we also look at the potential of liquid biopsies and circulating tumour dna (ctdna) to change the landscape of therapeutic choice and biologic understanding of the disease. Summary Routine testing for extended RAS, BRAF, dmmr or high msi, and NTRK fusions is necessary to determine the best sequencing of chemotherapy and biologic agents for patients with mcrc. Although next-generation sequencing and ctdna are increasingly being adopted, other techniques such as immunohistochemistry retain their relevance in detection of her2 amplification, NTRK fusions, and dmmr.
Collapse
|
10
|
Ricciuti B, Genova C, Crinò L, Libra M, Leonardi GC. Antitumor activity of larotrectinib in tumors harboring NTRK gene fusions: a short review on the current evidence. Onco Targets Ther 2019; 12:3171-3179. [PMID: 31118670 PMCID: PMC6503327 DOI: 10.2147/ott.s177051] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 03/20/2019] [Indexed: 12/11/2022] Open
Abstract
The development of deep-sequencing methods is now unveiling a new landscape of previously undetected gene fusion across different tumor types. Chromosomal translocation involving the NTRK gene family occur across a wide range of cancers in both children and adults. Preclinical studies have demonstrated that chimeric proteins encoded by NTRK rearrangements have oncogenic properties and drive constitutive expression and ligand-independent activation. Larotrectinib (ARRY470, LOXO101, Vitrakvi) is a highly and potent inhibitor of TRKA, TRKB, and TRKC, and has demonstrated rema rkable antitumor activity against TRK-fusion-positive cancers with a favorable side-effect profile in phase I/II clinical trials. In November 2018, the US Food and Drug Administration granted accelerated approval to larotrectinib for adult and pediatric patients with solid tumors harboring NTRK gene fusions without known acquired resistance mutation. In this review, we discuss the clinical activity and safety profile of larotrectinib, focusing on the clinical trials that led to its first global approval.
Collapse
Affiliation(s)
- Biagio Ricciuti
- Department of Medical Oncology, Thoracic Oncology Unit, Santa Maria della Misericordia Hospital, University of Perugia, Perugia, Italy
| | - Carlo Genova
- Lung Cancer Unit, Ospedale Policlinico San Martino, Genova16132, Italy
| | - Lucio Crinò
- Department of Medical Oncology, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Meldola, Italy
| | - Massimo Libra
- Department of Biomedical and Biotechnological Sciences, Pathology and Oncology Section, University of Catania, Catania, Italy
| | - Giulia Costanza Leonardi
- Department of Biomedical and Biotechnological Sciences, Pathology and Oncology Section, University of Catania, Catania, Italy
- Department of Pathology, Children’s Hospital, Boston, MA, USA
| |
Collapse
|
11
|
Modest DP, Pant S, Sartore-Bianchi A. Treatment sequencing in metastatic colorectal cancer. Eur J Cancer 2019; 109:70-83. [PMID: 30690295 DOI: 10.1016/j.ejca.2018.12.019] [Citation(s) in RCA: 200] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 12/18/2018] [Indexed: 12/17/2022]
Abstract
Metastatic colorectal cancer (mCRC) remains incurable in most cases, but survival has improved with advances in cytotoxic chemotherapy and targeted agents. However, the optimal use and sequencing of these agents across multiple lines of treatment is unclear. Here, we review current treatment approaches and optimal treatment sequencing across the first-, second- and third-line settings in mCRC, including biological aspects affecting sequencing and rechallenge. Effective first-line therapy is a key determinant of treatment outcomes and should be selected after considering both clinical factors and biological markers, notably RAS and BRAF. The second-line regimen choice depends on the systemic therapies given in first-line. Anti-angiogenic agents (e.g. bevacizumab, ramucirumab and aflibercept) are indicated for most patients, whereas epidermal growth factor receptor (EGFR) inhibitors do not improve survival in the second-line setting. Molecular profiling is important in third-line treatment, with options in RAS wild-type patients including EGFR inhibitors (cetuximab or panitumumab), regorafenib and trifluridine/tipiracil. Immunotherapy with pembrolizumab or nivolumab ± ipilimumab may be considered for patients with high microsatellite instability disease. Targeting HER2/neu amplification shows promise for the subset of CRC tumours displaying this abnormality. Sequencing decisions are complicated by the potential for any treatment break or de-escalation to evoke a distinct clinical progression type. Ongoing trials are investigating the optimal sequencing and timing of therapies for mCRC. Molecular profiling has established new targets, and increasing knowledge of tumour evolution under drug pressure will possibly impact on sequencing.
Collapse
Affiliation(s)
- D P Modest
- Department of Medicine III, University Hospital, LMU Munich, Germany.
| | - S Pant
- Department of Investigational Cancer Therapeutics, The University of Texas M.D. Anderson Cancer Center, Houston, TX, USA
| | - A Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Piazza Ospedale Maggiore, 3, 20162, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| |
Collapse
|
12
|
Sartore-Bianchi A, Siena S. The right chance for temozolomide in metastatic colorectal cancer? Ann Oncol 2018; 29:1618-1619. [DOI: 10.1093/annonc/mdy223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
13
|
Kheder ES, Hong DS. Emerging Targeted Therapy for Tumors with NTRK Fusion Proteins. Clin Cancer Res 2018; 24:5807-5814. [PMID: 29986850 DOI: 10.1158/1078-0432.ccr-18-1156] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/18/2018] [Accepted: 07/05/2018] [Indexed: 11/16/2022]
Abstract
The oncogenesis-promoting role of chromosomal rearrangements for several hematologic and solid malignancies is well recognized. However, identifying targetable, actionable, and druggable chromosomal rearrangements remains a challenge. Targeting gene fusions and chromosomal rearrangements is an effective strategy in treating gene rearrangement-driven tumors. The NTRK (Neurotrophic Tyrosine Receptor Kinase) gene family encodes three tropomyosin-related kinase (TRK) receptors that preserve central and peripheral nervous system development and function. NTRK genes, similar to other genes, are subject to alterations, including fusions. Preclinical studies have demonstrated that TRK fusion proteins promote oncogenesis by mediating constitutive cell proliferation and survival. Several clinical trials have estimated the safety and efficacy of TRK fusion kinase receptor inhibitors and have demonstrated encouraging antitumor activity in patients with NTRK-rearranged malignancies. Specifically, larotrectinib and entrectinib have emerged as potent, safe, and promising TRK inhibitors. Herein, we discuss the potential oncogenic characteristics of TRK fusion proteins in various malignancies and highlight ongoing clinical trials of kinase inhibitors targeting them.
Collapse
Affiliation(s)
- Ed S Kheder
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
14
|
Lam M, Loree JM, Pereira AAL, Chun YS, Kopetz S. Accelerating Therapeutic Development through Innovative Trial Design in Colorectal Cancer. Curr Treat Options Oncol 2018; 19:11. [PMID: 29488033 DOI: 10.1007/s11864-018-0524-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OPINION STATEMENT Current trial design is challenged by the advancement of technologies that have enabled deeper understanding of the molecular drivers of colorectal cancer (CRC). The speed of trial testing and the ability to test larger volumes of promising novel agents in the face of smaller populations identified by molecular profiling are challenges posed to clinical studies. Master protocols that utilize umbrella designs are equipped to deal with potential biomarker and matched treatments simultaneously. Although complex in nature, they increase trial efficiency by utilizing shared screening platforms, test multiple treatments together, and simplify regulatory submission and reporting under a common protocol. Emerging technologies such as circulating tumor DNA (ctDNA) may help speed up adjuvant trials. These studies have been traditionally slow to complete due to low event rates and the high numbers needed to recruit. ctDNA used as a surrogate for minimal residual disease (MRD) and as an early marker of relapse may help counter some of these factors that deter innovation in this setting. Finally, in the era of precision medicine, surgery should not be forgotten as the only potentially curative option to date in metastatic disease. Five-year overall survival following resection of liver metastasis exceeds what can be achieved with chemotherapy alone in selected cases. Surgical advances have lowered morbidity and allow for greater resection volumes and repeated interventions. Although historically challenging, a well-designed randomized surgical intervention trial would greatly facilitate moving single-institution guidelines reported by case series into wider clinical practice.
Collapse
Affiliation(s)
- Michael Lam
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard-Unit 0426, Houston, TX, 77030, USA
| | - Jonathan M Loree
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard-Unit 0426, Houston, TX, 77030, USA
| | - Allan Anderson Lima Pereira
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard-Unit 0426, Houston, TX, 77030, USA
| | - Yun Shin Chun
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Scott Kopetz
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard-Unit 0426, Houston, TX, 77030, USA.
| |
Collapse
|