1
|
Zhang S, Liu L, Li S, Sun X. Effect and imaging analysis of cetuximab combined with radiotherapy in patients with rectal carcinoma. Biotechnol Genet Eng Rev 2024; 40:4953-4963. [PMID: 37248703 DOI: 10.1080/02648725.2023.2219944] [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: 02/23/2023] [Accepted: 05/23/2023] [Indexed: 05/31/2023]
Abstract
OBJECTIVE To analyze the effect of cetuximab combined with radiotherapy in patients with rectal carcinoma (RC) by imaging analysis. METHODS The clinical data of 104 RC patients at our hospital from February 2021 to February 2022 were retrospectively analyzed. They were separated into control group (n = 52) and experimental group (n = 52) according to the order of admission, with the former treated with radiotherapy alone and the latter receiving cetuximab and radiotherapy. The clinical efficacy, tumor marker levels and imaging parameters of different treatment regimens were compared, and Quality of Life questionnaire (QLQ-C30) was used to evaluate the quality of life. RESULTS The incidence of tumor regression grade (TRG) downgrade, T stage downgrade and N stage downgrade was remarkably higher in the experimental group than in the control group (P < 0.05). The experimental group had remarkably lower tumor marker levels (P < 0.001) and higher mean score of EORTC Core QLQ-C30 (P < 0.001) than those in the control group. The relative signal intensity of tumor (SIT/M), relative signal intensity reduction rate of tumor (SIT/MRR) and apparent diffusion coefficient (ADC) values were remarkably higher (P < 0.001) and the absolute signal intensity of tumor (SIT) value was remarkably lower (P < 0.001) in the experimental group than the control group. CONCLUSION Treatment with cetuximab and radiotherapy can greatly reduce serum tumor marker levels in RC patients and bring them health benefits, and further studies will help establish a better solution for such patients.
Collapse
Affiliation(s)
- Shuai Zhang
- Gastrointestinal Cancer Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Liangliang Liu
- Department of Pharmacy, Hebei North University, Zhangjiakou, Hebei, China
| | - Shuguang Li
- Gastrointestinal Cancer Surgery, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xin Sun
- Department of Imaging, The Fourth People's Hospital of Jinan, Jinan, Shandong, China
| |
Collapse
|
2
|
Milan N, Navarria F, Cecchin E, De Mattia E. Somatic pharmacogenomics in the treatment prognosis of locally advanced rectal cancer patients: a narrative review of the literature. Expert Rev Clin Pharmacol 2024; 17:683-719. [PMID: 39046146 DOI: 10.1080/17512433.2024.2375449] [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: 01/23/2024] [Accepted: 06/28/2024] [Indexed: 07/25/2024]
Abstract
INTRODUCTION Standard treatment for patients with locally advanced rectal cancer (LARC) includes neoadjuvant chemoradiotherapy (nCRT) with fluoropyrimidines, followed by surgical excision. The newly introduced therapeutic strategies propose intensified regimens or more conservative approaches based on risk stratification algorithms that currently include clinicoradiological criteria but not molecular variables. How to better stratify patients is a burning clinical question, and pharmacogenomics may prove useful in identifying new genetic markers that could be incorporated into clinical algorithms to personalize nCRT. An emerging area could be the evaluation of somatic mutations as potential genetic markers that correlate with patient prognosis. Tumor mutations in the RAS/BRAF genes, as well as microsatellite instability (MSI) status, are currently used in treatment selection for colorectal cancer (CRC); however, their clinical value in LARC is still unclear. AREA COVERED This literature review discusses the relevant findings on the prognostic role of mutations in the key oncogenes RAS, KRAS, BRAF, PIK3CA, SMAD4 and TP53, including MSI status in LARC patients treated with nCRT. EXPERT OPINION KRAS proved to be the most promising marker, consistently associated with poorer disease-free survival and overall survival. Therefore, KRAS could be a good candidate for integration into the risk stratification algorithm to develop a personalized treatment.
Collapse
Affiliation(s)
- Noemi Milan
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Federico Navarria
- Radiation Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Elena De Mattia
- Clinical and Experimental Pharmacology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| |
Collapse
|
3
|
Singh H, Kang A, Bloudek L, Hsu LI, Corinna Palanca-Wessels M, Stecher M, Siadak M, Ng K. Systematic literature review and meta-analysis of HER2 amplification, overexpression, and positivity in colorectal cancer. JNCI Cancer Spectr 2024; 8:pkad082. [PMID: 37815820 PMCID: PMC10868379 DOI: 10.1093/jncics/pkad082] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 08/25/2023] [Accepted: 09/21/2023] [Indexed: 10/11/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the second most common cause of cancer death globally. Recent clinical trials suggest an emerging role for HER2 as a potential clinically relevant biomarker in CRC. Testing for HER2 in CRC is not standard practice; consequently, the prevalence of HER2 positivity (HER2+) in patients with CRC remains uncertain. METHODS A systematic literature review and meta-analysis were conducted to generate estimates of proportions of patients with CRC with HER2 overexpression or HER2 amplification and HER2+ (either overexpression or amplification), overall and in patients with rat sarcoma virus (RAS) wild-type cancer. HER2+ was defined as 1) immunohistochemistry with a score of 3+, 2) immunohistochemistry with a score of 2+ and in situ hybridization+, or 3) next-generation sequencing positive. RESULTS Of 224 studies identified with information on HER2 in CRC, 52 studies used a US Food and Drug Administration-approved assay and were selected for further analysis. Estimated HER2+ rate was 4.1% (95% confidence interval [CI] = 3.4% to 5.0%) overall (n = 17 589). HER2+ rates were statistically higher in RAS wild-type (6.1%, 95% CI = 5.4% to 6.9%) vs RAS mutant CRC (1.1%, 95% CI = 0.3% to 4.4%; P < .0001). Despite limited clinical information, we confirmed enrichment of HER2+ CRC in patients with microsatellite stable and left-sided CRC. CONCLUSION This meta-analysis provides an estimate of HER2+ CRC and confirms enrichment of HER2 in microsatellite stable, left-sided, RAS wild-type CRC tumors. Our work is important given the recently described clinical efficacy of HER2-targeted therapies in HER2+ CRC and informs strategies for incorporation of HER2 testing into standard of care.
Collapse
Affiliation(s)
- Harshabad Singh
- Division of Gastrointestinal Cancers, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | | | | | | - Kimmie Ng
- Division of Gastrointestinal Cancers, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
4
|
Huang J, Chen G, Li H. An Update on the Role of Immunohistochemistry in the Evaluation of Pancreatic/Liver/Gastrointestinal Luminal Tract Disorders. Arch Pathol Lab Med 2023; 147:1374-1382. [PMID: 37134268 DOI: 10.5858/arpa.2022-0462-ra] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/02/2023] [Indexed: 05/05/2023]
Abstract
CONTEXT.— Immunohistochemistry serves as an ancillary diagnostic tool for a wide variety of neoplastic and nonneoplastic disorders, including infections, workup of inflammatory conditions, and subtyping neoplasms of the pancreas/liver/gastrointestinal luminal tract. In addition, immunohistochemistry is also used to detect a variety of prognostic and predictive molecular biomarkers for carcinomas of the pancreas, liver, and gastrointestinal luminal tract. OBJECTIVE.— To highlight an update on the role of immunohistochemistry in the evaluation of pancreatic/liver/gastrointestinal luminal tract disorders. DATA SOURCES.— Literature review and authors' research data and personal practice experience were used. CONCLUSIONS.— Immunohistochemistry is a valuable tool, assisting in the diagnosis of problematic tumors and benign lesions of the pancreas, liver, and gastrointestinal luminal tract, and also in the prediction of prognosis and therapeutic response for carcinomas of the pancreas, liver, and gastrointestinal luminal tract.
Collapse
Affiliation(s)
- Jialing Huang
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania (Huang, Li)
| | - Guoli Chen
- The Department of Pathology, PennState Health, Hershey, Pennsylvania (Chen)
| | - Hongjie Li
- From the Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania (Huang, Li)
| |
Collapse
|
5
|
Sun Q, Li Q, Gao F, Wu H, Fu Y, Yang J, Fan X, Cui X, Pu X. HER2 overexpression/amplification status in colorectal cancer: a comparison between immunohistochemistry and fluorescence in situ hybridization using five different immunohistochemical scoring criteria. J Cancer Res Clin Oncol 2023; 149:579-592. [PMID: 36018511 PMCID: PMC9931822 DOI: 10.1007/s00432-022-04230-8] [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: 03/29/2022] [Accepted: 07/19/2022] [Indexed: 10/15/2022]
Abstract
OBJECTIVE Although HER2 has gradually become an important therapeutic target for colorectal cancer (CRC), a unified and standard HER2 scoring system was still not established in CRC, and the debatable results of immunohistochemistry and fluorescence in situ hybridization (FISH) in CRC requires further exploration. METHODS In this study, we use five immunohistochemical (IHC) scoring criteria (i.e., IRS-p, IRS-m, GEA-s, GEA-b and HERACLES) and two FISH criteria to evaluate HER2 status, and further evaluate the correlation between HER2 status and clinicopathological features, survival in a large, unselected Chinese cohort of 664 CRCs. RESULTS Finally, we set HER2/CEP17 ratio ≥ 2.0, or an average HER2 copy number ≥ 6.0 as FISH-positive threshold and the amplification rate of HER2 gene was 7.08% (47/664).The HER2 positivity (IHC 3+) was 2.71%, 3.16%, 2.56%, 2.71% and 3.16%, according to the IHC scoring criteria of IRS-p, IRS-m, GEA-s, GEA-b and HERACLES, respectively. Set FISH results as the golden standard; receiver-operating characteristic analysis showed that IRS-p had both high sensitivity and specificity than other IHC scoring systems to evaluate HER2 status. Based on IRS-p criterion, There were significant differences in tumor differentiation (p = 0.038), lymphatic vascular invasion (p = 0.001), pN stage (p value = 0.043), and overall survival (p < 0.001) among IHC score 0-3 + groups. Meanwhile, there were significant differences in pT stage (p = 0.031), pN stage (p = 0.009) and overall survival (p < 0.001) among FISH subgroups. CONCLUSION The IRS-p criterion was more suitable for assessing the HER2 status in CRC patients than other IHC criteria. Whereas for FISH scoring system, only HER2/CEP17 < 2.0, meanwhile HER2cn < 4.0 and HER2cn ≥ 6.0 were subgroups with unique clinicopathological characteristics.
Collapse
Affiliation(s)
- Qi Sun
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
- Center for Digestive Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, Jiangsu, China
| | - Qi Li
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
| | - Fuping Gao
- Department of Pathology, Gaochun People's Hospital, Nanjing, 210008, Jiangsu, China
| | - Hongyan Wu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
| | - Jun Yang
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China
| | - Xiangshan Fan
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China.
| | - Xiaobin Cui
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China.
| | - Xiaohong Pu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhong-Shan Road, Nanjing, 210008, Jiangsu, China.
| |
Collapse
|
6
|
Torres-Jiménez J, Esteban-Villarrubia J, Ferreiro-Monteagudo R. Precision Medicine in Metastatic Colorectal Cancer: Targeting ERBB2 (HER-2) Oncogene. Cancers (Basel) 2022; 14:3718. [PMID: 35954382 PMCID: PMC9367374 DOI: 10.3390/cancers14153718] [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: 06/29/2022] [Revised: 07/27/2022] [Accepted: 07/28/2022] [Indexed: 02/01/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer in terms of incidence rate in adults and the second most common cause of cancer-related death in Europe. The treatment of metastatic CRC (mCRC) is based on the use of chemotherapy, anti-vascular endothelial growth factor (VEGF), and anti-epidermal growth factor receptor (EGFR) for RAS wild-type tumors. Precision medicine tries to identify molecular alterations that could be treated with targeted therapies. ERBB2 amplification (also known as HER-2) has been identified in 2-3% of patients with mCRC, but there are currently no approved ERBB2-targeted therapies for mCRC. The purpose of this review is to describe the molecular structure of ERBB2, clinical features of these patients, diagnosis of ERBB2 alterations, and the most relevant clinical trials with ERBB2-targeted therapies in mCRC.
Collapse
Affiliation(s)
- Javier Torres-Jiménez
- Medical Oncology Department, MD Anderson Cancer Center Madrid, 28033 Madrid, Spain
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
| | - Jorge Esteban-Villarrubia
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
- Medical Oncology Department, University Hospital 12 de Octubre, 28041 Madrid, Spain
| | - Reyes Ferreiro-Monteagudo
- Medical Oncology Department, University Hospital Ramon y Cajal, 28034 Madrid, Spain; (J.E.-V.); (R.F.-M.)
| |
Collapse
|
7
|
Cai G, Sun M, Li X, Zhu J. Construction and characterization of rectal cancer-related lncRNA-mRNA ceRNA network reveals prognostic biomarkers in rectal cancer. IET Syst Biol 2021; 15:192-204. [PMID: 34613665 PMCID: PMC8675822 DOI: 10.1049/syb2.12035] [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: 06/05/2021] [Revised: 08/22/2021] [Accepted: 09/23/2021] [Indexed: 12/26/2022] Open
Abstract
Rectal cancer is an important cause of cancer‐related deaths worldwide. In this study, the differentially expressed (DE) lncRNAs/mRNAs were first identified and the correlation level between DE lncRNAs and mRNAs were calculated. The results showed that genes of highly correlated lncRNA‐mRNA pairs presented strong prognosis effects, such as GPM6A, METTL24, SCN7A, HAND2‐AS1 and PDZRN4. Then, the rectal cancer‐related lncRNA‐mRNA network was constructed based on the ceRNA theory. Topological analysis of the network revealed that the network was maintained by hub nodes and a hub subnetwork was constructed, including the hub lncRNA MIR143HG and MBNL1‐SA1. Further analysis indicated that the hub subnetwork was highly related to cancer pathways, such as ‘Focal adhesion’ and ‘Wnt signalling pathway’. Hub subnetwork also had significant prognosis capability. A closed lncRNA‐mRNA module was identified by bilateral network clustering. Genes in modules also showed high prognosis effects. Finally, a core lncRNA‐TF crosstalk network was identified to uncover the crosstalk and regulatory mechanisms of lncRNAs and TFs by integrating ceRNA crosstalks and TF binding affinities. Some core genes, such as MEIS1, GLI3 and HAND2‐AS1 were considered as the key regulators in tumourigenesis. Based on the authors’ comprehensive analysis, all these lncRNA‐mRNA crosstalks provided promising clues for biological prognosis of rectal cancer.
Collapse
Affiliation(s)
- Guoying Cai
- Department of Integrative Medicine & Medical Oncology, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University, Shengzhou Branch), Shengzhou, Zhejiang, China
| | - Meifei Sun
- Department of Integrative Medicine & Medical Oncology, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University, Shengzhou Branch), Shengzhou, Zhejiang, China
| | - Xinrong Li
- Department of Integrative Medicine & Medical Oncology, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University, Shengzhou Branch), Shengzhou, Zhejiang, China
| | - Junquan Zhu
- Department of Integrative Medicine & Medical Oncology, Shengzhou People's Hospital (the First Affiliated Hospital of Zhejiang University, Shengzhou Branch), Shengzhou, Zhejiang, China
| |
Collapse
|
8
|
Qiu MZ, He CY, Yang XH, Yang LQ, Lin JZ, Zhou DL, Long YK, Guan WL, Jin Y, Li YH, Wang FH, Yang DJ, Xu RH. Relationship of HER2 Alteration and Microsatellite Instability Status in Colorectal Adenocarcinoma. Oncologist 2021; 26:e1161-e1170. [PMID: 33844372 PMCID: PMC8265359 DOI: 10.1002/onco.13786] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 11/13/2020] [Indexed: 12/30/2022] Open
Abstract
Background The impact of HER2 somatic mutations in colorectal carcinoma (CRC) has not been well studied and its relationship with microsatellite instability‐high (MSI‐H) is yet to be fully elucidated. Materials and Methods From February 2017 to February 2020, the data of patients with CRC who underwent next‐generation sequencing and had detailed record of clinicopathological information were investigated. HER2 alteration and its relationship with MSI‐H were analyzed. Results Among 731 patients who underwent sequencing, 55 patients (7.5%) had HER2 alteration, including 29 (4.0%) with HER2 somatic mutations, 24 (3.3%) with HER2 gene amplification, and 2 patients (0.2%) with both HER2 mutations and amplification. R678Q was the most common mutated kinase domain, and no HER2 kinase domain in‐frame insertions/deletions were found in HER2 mutated cases. MSI‐H was found in 5.2% of our cohort and 36.8% of MSI‐H patients had HER2 mutation. For HER2 mutated cases, 48.3% were MSI‐H, whereas none of the HER2 amplification cases were MSI‐H. MSI‐H patients with HER2 mutation had significantly worse median progression‐free survival for programmed death‐1 (PD‐1) antibody than those without HER2 alteration (p = .036). Conclusion High MSI‐H rate was found in HER2 mutated cases, but no MSI‐H was found in HER2 amplification cases. MSI‐H patients with HER2 mutated had worse progression‐free survival for PD‐1 antibody than those without. Implications for Practice This study highlights the high microsatellite instability‐high (MSI‐H) rate in HER2 mutated cases but no MSI‐H in HER2 amplification cases. Moreover MSI‐H patients with HER2 mutated had worse progression‐free survival for programmed death‐1 antibody than those without. Further research to explore the internal relationship between HER2 alteration and MSI‐H is needed. The clinical significance of HER2 somatic mutations and its relationship with high microsatellite instability in colorectal cancer is not fully understood. This study investigated the relationship by analyzing data of patients with colorectal cancer who underwent next‐generation sequencing.
Collapse
Affiliation(s)
- Miao-Zhen Qiu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Cai-Yun He
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Xin-Hua Yang
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Li-Qiong Yang
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Jun-Zhong Lin
- Colorectal Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Da-Lei Zhou
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Ya-Kang Long
- Department of Molecular Diagnostics, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Wen-Long Guan
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Ying Jin
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Yu-Hong Li
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Feng-Hua Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Da-Jun Yang
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| | - Rui-Hua Xu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, People's Republic of China
| |
Collapse
|
9
|
Abstract
HER2 (ERBB2) is a member of the ERBB family of receptor tyrosine kinases and functions to drive signaling in the RAS/RAF/MEK/ERK and PI3K/AKT/mTOR pathways. Overall, approximately 2-3% of CRCs exhibit ERBB2 amplification. Multiple phase II clinical trials have now shown that ERBB2 amplification can be predictive of response to anti-ERBB2 targeted therapy. Consequently, recently released guidelines from the National Comprehensive Cancer Network recommend treatment with anti-ERBB2 targeted therapy for RAS wild-type, ERBB2-amplified metastatic CRC. While circumspection is still needed, ERBB2 amplification has now emerged as the next standard-of-care biomarker for metastatic CRC, expanding targeted therapy options for these patients.
Collapse
Affiliation(s)
- Jonathan A Nowak
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA 02115, USA.
| |
Collapse
|
10
|
Lai E, Liscia N, Donisi C, Mariani S, Tolu S, Pretta A, Persano M, Pinna G, Balconi F, Pireddu A, Impera V, Dubois M, Migliari M, Spanu D, Saba G, Camera S, Musio F, Ziranu P, Puzzoni M, Demurtas L, Pusceddu V, Dettori M, Massa E, Atzori F, Dessì M, Astara G, Madeddu C, Scartozzi M. Molecular-Biology-Driven Treatment for Metastatic Colorectal Cancer. Cancers (Basel) 2020; 12:E1214. [PMID: 32413973 PMCID: PMC7281737 DOI: 10.3390/cancers12051214] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/03/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Metastatic CRC (mCRC) is a molecular heterogeneous disease. The aim of this review is to give an overview of molecular-driven treatment of mCRC patients. METHODS A review of clinical trials, retrospective studies and case reports was performed regarding molecular biomarkers with therapeutic implications. RESULTS RAS wild-type status was confirmed as being crucial for anti-epidermal growth factor receptor (EGFR) monoclonal antibodies and for rechallenge strategy. Antiangiogenic therapies improve survival in first- and second-line settings, irrespective of RAS status, while tyrosine kinase inhibitors (TKIs) remain promising in refractory mCRC. Promising results emerged from anti-HER2 drugs trials in HER2-positive mCRC. Target inhibitors were successful for BRAFV600E mutant mCRC patients, while immunotherapy was successful for microsatellite instability-high/defective mismatch repair (MSI-H/dMMR) or DNA polymerase epsilon catalytic subunit (POLE-1) mutant patients. Data are still lacking on NTRK, RET, MGMT, and TGF-β, which require further research. CONCLUSION Several molecular biomarkers have been identified for the tailored treatment of mCRC patients and multiple efforts are currently ongoing to increase the therapeutic options. In the era of precision medicine, molecular-biology-driven treatment is the key to impro patient selection and patient outcomes. Further research and large phase III trials are required to ameliorate the therapeutic management of these patients.
Collapse
Affiliation(s)
- Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Nicole Liscia
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Clelia Donisi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Simona Tolu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Mara Persano
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Giovanna Pinna
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Francesca Balconi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Annagrazia Pireddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Valentino Impera
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Marco Dubois
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Marco Migliari
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Silvia Camera
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
- Medical Oncology Unit, Sapienza University of Rome, 00161 Rome, Italy
| | - Francesca Musio
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Laura Demurtas
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Manuela Dettori
- Medical Oncology Unit, Azienda Ospedaliera Brotzu, Ospedale Businco, 09134 Cagliari, Italy
| | - Elena Massa
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Francesco Atzori
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Mariele Dessì
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Giorgio Astara
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Clelia Madeddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (N.L.); (C.D.); (S.M.); (S.T.); (A.P.); (M.P.); (G.P.); (F.B.); (A.P.); (V.I.); (M.D.); (M.M.); (D.S.); (G.S.); (S.C.); (F.M.); (P.Z.); (M.P.); (L.D.); (V.P.); (E.M.); (F.A.); (M.D.); (G.A.); (C.M.)
| |
Collapse
|
11
|
Liu F, Ren C, Jin Y, Xi S, He C, Wang F, Wang Z, Xu RH, Wang F. Assessment of two different HER2 scoring systems and clinical relevance for colorectal cancer. Virchows Arch 2019; 476:391-398. [PMID: 31720832 PMCID: PMC7085476 DOI: 10.1007/s00428-019-02668-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/28/2019] [Accepted: 09/10/2019] [Indexed: 12/24/2022]
Abstract
Although the positivity of human epidermal growth factor receptor 2 (HER2) is low in colorectal cancer (CRC), anti-HER2 is becoming a new target therapy in metastatic colorectal cancer (mCRC). However, assessment of the HER2 scoring system was still not established in CRC. The purpose of our study was to evaluate HER2 status and its correlation with clinicopathological characteristics and survival according to the HER2 diagnostic criteria for gastroesophageal adenocarcinoma (GEA criteria) and the HERACLES diagnostic criteria (HERACLES criteria) in a large cohort of Chinese CRC patients. The HER2 positivity was 2.9% (43/1490) and 2.6% (39/1490) in CRCs based on the GEA criteria and the HERACLES criteria, and 3.7% (9/243) in mCRC according to both criteria. HER2 status was associated with primary tumor location (P = 0.037), regional lymph node metastasis (P = 0.035), and TNM stage (P = 0.022) in CRCs based on the HERACLES criteria. No such association was found based on the GEA criteria. Furthermore, HER2 positive only presented in patients with RAS gene wild type (P = 0.001). Significant difference was only observed between the HER2-positive and HER2-negative groups in terms of disease-free survival for stage II-III CRCs (P = 0.048) according to the HERACLES criteria, but not based on the GEA criteria. Our findings suggest that the frequency of HER2 overexpression or amplification was low in Chinese CRC patients, and provide a rationale for further evaluation of HER2 in CRC based on the HERACLES criteria and the HER2 diagnostic criteria for gastroesophageal adenocarcinoma.
Collapse
Affiliation(s)
- Furong Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Chao Ren
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Ying Jin
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Shaoyan Xi
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Department of Pathology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Caiyun He
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Fang Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Department of Molecular Diagnostics, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Zixian Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Rui-Hua Xu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.,Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China
| | - Feng Wang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China. .,Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, People's Republic of China.
| |
Collapse
|