1
|
Buvaneswari T, Ramkumar M, Venkatesan P, Kumar RS. Leveraging Radiomics and Hybrid Quantum-Classical Convolutional Networks for Non-Invasive Detection of Microsatellite Instability in Colorectal Cancer. Mol Imaging Biol 2025; 27:227-237. [PMID: 39979579 DOI: 10.1007/s11307-025-01990-w] [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: 10/25/2024] [Revised: 01/29/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
PURPOSE The goal of this study is to create a novel framework for identifying MSI status in colorectal cancer using advanced radiomics and deep learning strategies, aiming to enhance clinical decision-making and improve patient outcomes in oncology. PROCEDURES The study utilizes histopathological slide images from the NCT-CRC-HE-100 K and PAIP 2020 databases. Key procedures include self-attentive adversarial stain normalization for data standardization, tumor delineation via a Slimmable Transformer, and radiomics feature extraction using a hybrid quantum-classical neural network. RESULTS The proposed system reaches 99% accuracy when identifying colorectal cancer MSI status. It shows the model is good at telling the difference between MSI and MSS tumors and can be used in real medical care for cancer. CONCLUSIONS Our research shows that the new system improves colorectal cancer MSI status determination better than previous methods. Our optimized processing technology works better than other methods to divide and analyze tissue features making the system good for improving patient care decisions.
Collapse
Affiliation(s)
- T Buvaneswari
- Department of Computer Secience and Engineering, Annapoorana Engineering College (Autonomous), NH_47, Sankari Main Road, Periyaseeragapaddi, Salem, Tamil Nadu, 636 308, India.
| | - M Ramkumar
- Department of Electronics and Communication Engineering, Sri Krishna College of Engineering and Technology, Coimbatore, 641-008, Tamil Nadu, India
| | - Prabhu Venkatesan
- Department of Electronics and Communication Engineering, Vel Tech Multi Tech Dr. Rangarajan Dr. Sakunthala Engineering College, Avadi, Chennai, 600062, Tamil Nadu, India
| | - R Sarath Kumar
- Department of Electronics And Communication Engineering Department, Sri Krishna College Of Engineering And Technology, Coimbatore, 641008, Tamil Nadu, India
| |
Collapse
|
2
|
Fan WX, Su F, Zhang Y, Zhang XL, Du YY, Gao YJ, Li WL, Hu WQ, Zhao J. Oncological characteristics, treatments and prognostic outcomes in MMR-deficient colorectal cancer. Biomark Res 2024; 12:89. [PMID: 39183366 PMCID: PMC11346251 DOI: 10.1186/s40364-024-00640-7] [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: 07/09/2024] [Accepted: 08/14/2024] [Indexed: 08/27/2024] Open
Abstract
Colorectal cancer (CRC) ranks as the third most prevalent cancer globally. It's recognized that the molecular subtype of CRC, characterized by mismatch repair deficiency (dMMR) or microsatellite instability-high (MSI-H), plays a critical role in determining appropriate treatment strategies. This review examines the current molecular classifications, focusing on dMMR/MSI-H CRC and its subtypes: Lynch syndrome (LS), Lynch-like syndrome (LLS), and sporadic cases. Despite advances in understanding of these genetic backgrounds, clinical trials have not conclusively differentiated the efficacy of immune checkpoint inhibitors among these subgroups. Therefore, while this review details the molecular characteristics and their general implications for treatment and prognosis, it also highlights the limitations and the need for more refined clinical studies to ascertain tailored therapeutic strategies for each subtype. Furthermore, this review summarizes completed and ongoing clinical studies, emphasizing the importance of developing treatments aligned more closely with molecular profiles. By discussing these aspects, the review seeks to provide a comprehensive analysis of oncological characteristics, presenting a detailed understanding of their implications for treatment and prognosis in dMMR/MSI-H CRC.
Collapse
Affiliation(s)
- Wen-Xuan Fan
- Graduate School of Shanxi Medical University, Taiyuan, Shanxi, 030607, China
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Fei Su
- Graduate School of Shanxi Medical University, Taiyuan, Shanxi, 030607, China
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yan Zhang
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
- Graduate School of Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Xiao-Ling Zhang
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yun-Yi Du
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Yang-Jun Gao
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Wei-Ling Li
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
- Graduate School of Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Wen-Qing Hu
- Department of Gastrointestinal Surgery, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China
| | - Jun Zhao
- Department of Oncology, Changzhi People's Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, 046000, China.
| |
Collapse
|
3
|
Xiao G, Li J, Deng L, Gao S, Tan C, He G, Du R. Microsatellite instability evaluation by a novel PCR-based 8-loci test kit in colorectal cancer. Biotechnol Appl Biochem 2024; 71:860-867. [PMID: 38556769 DOI: 10.1002/bab.2582] [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: 11/08/2023] [Accepted: 03/18/2024] [Indexed: 04/02/2024]
Abstract
Microsatellite instability (MSI) assessment is strongly recommended for colorectal cancer patients, as MSI status is crucial in determining optimal treatment and predicting prognosis. This study evaluated the reliability and accuracy of a novel polymerase chain reaction (PCR)-based 8-loci MSI test kit, a rapid test kit designed to detect MSI, by comparing its performance with immunohistochemistry (IHC) and the National Cancer Institute (NCI) 2B3D Panel. MSI status was determined in 186 formalin-fixed paraffin-embedded (FFPE) colorectal cancer tissue samples with known mismatch repair (MMR) status by IHC using the novel PCR-based 8-loci MSI test kit. Additionally, the consistency between the NCI 2B3D Panel and the novel PCR-based 8-loci panel was compared using 69 FFPE tumor tissues paired with adjacent non-cancerous tissue. The novel PCR-based 8-loci MSI test kit and IHC demonstrated high concordance (overall agreement: 97.8%). However, four samples displayed discordant results, exhibiting MMR deficiency using IHC and microsatellite stability using the novel PCR-based 8-loci MSI test kit. Of the 69 samples reanalyzed using the NCI 2B3D Panel, high concordance with the novel PCR-based 8-loci MSI test kit was observed in 67 of 69 cases (overall agreement: 97.1%). The novel PCR-based 8-loci MSI test kit is a rapid and reliable tool for accurately detecting MSI status in colorectal cancer.
Collapse
Affiliation(s)
- Gaofang Xiao
- Department of Pathology, Shantou University Affiliated Yuebei People's Hospital, Shaoguan, Guangdong, China
| | - Jing Li
- Department of Pathology, Shantou University Affiliated Yuebei People's Hospital, Shaoguan, Guangdong, China
| | - Lijun Deng
- Department of Medical Engineering, Shantou University Affiliated Yuebei People's Hospital, Shaoguan, Guangdong, China
| | - Shuangquan Gao
- Department of Pathology, Shantou University Affiliated Yuebei People's Hospital, Shaoguan, Guangdong, China
| | - Caiyun Tan
- Department of Pathology, Shantou University Affiliated Yuebei People's Hospital, Shaoguan, Guangdong, China
| | - Guiqing He
- Department of Pathology, Shantou University Affiliated Yuebei People's Hospital, Shaoguan, Guangdong, China
| | - Richang Du
- Department of Pathology, Shantou University Affiliated Yuebei People's Hospital, Shaoguan, Guangdong, China
| |
Collapse
|
4
|
Aydın E, Tokat ÜM, Adibi A, Özgü E, Bilgiç ŞN, Demiray M. Case report: Precision guided reactive cancer management: molecular complete response in heavily pretreated metastatic CRC by dual immunotherapy and sorafenib. Front Oncol 2024; 14:1405170. [PMID: 39011472 PMCID: PMC11246968 DOI: 10.3389/fonc.2024.1405170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/13/2024] [Indexed: 07/17/2024] Open
Abstract
Background Metastatic colon adenocarcinoma presents significant challenges in treatment, particularly when resistant to standard therapies. Precision oncology, guided by multidisciplinary tumor boards (MTBs), offers a promising way for individualized therapeutic approaches. Integration of comprehensive genomic profiling (CGP) and minimal residual disease (MRD) testing strengthens treatment decision-making, yet challenges persist in identifying and overcoming resistance mechanisms. FLT3 amplification can be one of those resistance/escape mechanisms that needs to be targeted. Case presentation This case report presents a 58-year-old male diagnosed with metastatic colon adenocarcinoma with liver metastasis, resistant to conventional treatments. Utilizing CGP and MRD testing, our multidisciplinary MTB identified a complex mutational profile, including APC, DAXX, TP53 mutations, and CDK8 and FLT3 amplifications. With a tumor mutational burden of 10 muts/mb and TPS, CPS scores of 0, immunotherapy was considered, employing dual immune checkpoint inhibitors alongside mebendazole and Lenvatinib targeting the WNT and VEGF/angiogenesis pathways. MRD testing revealed early treatment failure. Re-evaluation identified high copied FLT3 amplification (62 copies) as a resistance mechanism, prompting modification to incorporate sorafenib and dual immunotherapy with mebendazole. Subsequent MRD assessments and radiological scans demonstrated a remarkable therapeutic response, with sustained efficacy and absence of detectable residual disease. Conclusion This case highlights the successful application of precision oncology principles, facilitated by dynamic MTB-guided treatment strategies. Integration of MRD testing provided early detection of treatment inefficacy, allowing for timely intervention and adaptation of the treatment plan. Additionally, the case highlights the educational value of rare molecular alterations, emphasizing continual learning and refinement of treatment approaches in precision oncology.
Collapse
Affiliation(s)
- Esranur Aydın
- Precision Oncology Center, Medicana Health Group, Istanbul, Türkiye
| | - Ünal Metin Tokat
- Precision Oncology Center, Medicana Health Group, Istanbul, Türkiye
| | - Ashkan Adibi
- Precision Oncology Center, Medicana Health Group, Istanbul, Türkiye
- Department of Basic Oncology, Division of Cancer Genetics, Institute of Oncology, University of Istanbul, Istanbul, Türkiye
| | - Eylül Özgü
- Precision Oncology Center, Medicana Health Group, Istanbul, Türkiye
| | | | - Mutlu Demiray
- Precision Oncology Center, Medicana Health Group, Istanbul, Türkiye
| |
Collapse
|
5
|
Gustav M, Reitsam NG, Carrero ZI, Loeffler CML, van Treeck M, Yuan T, West NP, Quirke P, Brinker TJ, Brenner H, Favre L, Märkl B, Stenzinger A, Brobeil A, Hoffmeister M, Calderaro J, Pujals A, Kather JN. Deep learning for dual detection of microsatellite instability and POLE mutations in colorectal cancer histopathology. NPJ Precis Oncol 2024; 8:115. [PMID: 38783059 PMCID: PMC11116442 DOI: 10.1038/s41698-024-00592-z] [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: 11/07/2023] [Accepted: 04/14/2024] [Indexed: 05/25/2024] Open
Abstract
In the spectrum of colorectal tumors, microsatellite-stable (MSS) tumors with DNA polymerase ε (POLE) mutations exhibit a hypermutated profile, holding the potential to respond to immunotherapy similarly to their microsatellite-instable (MSI) counterparts. Yet, due to their rarity and the associated testing costs, systematic screening for these mutations is not commonly pursued. Notably, the histopathological phenotype resulting from POLE mutations is theorized to resemble that of MSI. This resemblance not only could facilitate their detection by a transformer-based Deep Learning (DL) system trained on MSI pathology slides, but also indicates the possibility for MSS patients with POLE mutations to access enhanced treatment options, which might otherwise be overlooked. To harness this potential, we trained a Deep Learning classifier on a large dataset with the ground truth for microsatellite status and subsequently validated its capabilities for MSI and POLE detection across three external cohorts. Our model accurately identified MSI status in both the internal and external resection cohorts using pathology images alone. Notably, with a classification threshold of 0.5, over 75% of POLE driver mutant patients in the external resection cohorts were flagged as "positive" by a DL system trained on MSI status. In a clinical setting, deploying this DL model as a preliminary screening tool could facilitate the efficient identification of clinically relevant MSI and POLE mutations in colorectal tumors, in one go.
Collapse
Affiliation(s)
- Marco Gustav
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | | | - Zunamys I Carrero
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Chiara M L Loeffler
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
- Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Marko van Treeck
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tanwei Yuan
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicholas P West
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Philip Quirke
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
| | - Titus J Brinker
- Digital Biomarkers for Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT), Heidelberg, Germany
- German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Loëtitia Favre
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Bruno Märkl
- Pathology, Faculty of Medicine, University of Augsburg, Augsburg, Germany
| | | | - Alexander Brobeil
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- Tissue Bank of the National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Julien Calderaro
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Anaïs Pujals
- Université Paris Est Créteil, INSERM, IMRB, Créteil, France
- Assistance Publique-Hôpitaux de Paris, Henri Mondor-Albert Chenevier University Hospital, Department of Pathology, Créteil, France
- INSERM, U955, Team Oncogenèse des lymphomes et tumeurs de la Neurofibromatose 1, Créteil, France
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
- Department of Medicine I, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.
- Pathology & Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom.
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
6
|
Qin Y, Huo M, Liu X, Li SC. Biomarkers and computational models for predicting efficacy to tumor ICI immunotherapy. Front Immunol 2024; 15:1368749. [PMID: 38524135 PMCID: PMC10957591 DOI: 10.3389/fimmu.2024.1368749] [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: 01/11/2024] [Accepted: 02/27/2024] [Indexed: 03/26/2024] Open
Abstract
Numerous studies have shown that immune checkpoint inhibitor (ICI) immunotherapy has great potential as a cancer treatment, leading to significant clinical improvements in numerous cases. However, it benefits a minority of patients, underscoring the importance of discovering reliable biomarkers that can be used to screen for potential beneficiaries and ultimately reduce the risk of overtreatment. Our comprehensive review focuses on the latest advancements in predictive biomarkers for ICI therapy, particularly emphasizing those that enhance the efficacy of programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1) inhibitors and cytotoxic T-lymphocyte antigen-4 (CTLA-4) inhibitors immunotherapies. We explore biomarkers derived from various sources, including tumor cells, the tumor immune microenvironment (TIME), body fluids, gut microbes, and metabolites. Among them, tumor cells-derived biomarkers include tumor mutational burden (TMB) biomarker, tumor neoantigen burden (TNB) biomarker, microsatellite instability (MSI) biomarker, PD-L1 expression biomarker, mutated gene biomarkers in pathways, and epigenetic biomarkers. TIME-derived biomarkers include immune landscape of TIME biomarkers, inhibitory checkpoints biomarkers, and immune repertoire biomarkers. We also discuss various techniques used to detect and assess these biomarkers, detailing their respective datasets, strengths, weaknesses, and evaluative metrics. Furthermore, we present a comprehensive review of computer models for predicting the response to ICI therapy. The computer models include knowledge-based mechanistic models and data-based machine learning (ML) models. Among the knowledge-based mechanistic models are pharmacokinetic/pharmacodynamic (PK/PD) models, partial differential equation (PDE) models, signal networks-based models, quantitative systems pharmacology (QSP) models, and agent-based models (ABMs). ML models include linear regression models, logistic regression models, support vector machine (SVM)/random forest/extra trees/k-nearest neighbors (KNN) models, artificial neural network (ANN) and deep learning models. Additionally, there are hybrid models of systems biology and ML. We summarized the details of these models, outlining the datasets they utilize, their evaluation methods/metrics, and their respective strengths and limitations. By summarizing the major advances in the research on predictive biomarkers and computer models for the therapeutic effect and clinical utility of tumor ICI, we aim to assist researchers in choosing appropriate biomarkers or computer models for research exploration and help clinicians conduct precision medicine by selecting the best biomarkers.
Collapse
Affiliation(s)
- Yurong Qin
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Miaozhe Huo
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| | - Xingwu Liu
- School of Mathematical Sciences, Dalian University of Technology, Dalian, Liaoning, China
| | - Shuai Cheng Li
- Department of Computer Science, City University of Hong Kong, Kowloon, China
- City University of Hong Kong Shenzhen Research Institute, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
Wei J, Ge X, Qian Y, Jiang K, Chen X, Lu W, Yang H, Fu D, Fang Y, Zhou X, Xiao Q, Tang Y, Ding K. Development and verification of a combined immune- and cancer-associated fibroblast related prognostic signature for colon adenocarcinoma. Front Immunol 2024; 15:1291938. [PMID: 38312843 PMCID: PMC10834644 DOI: 10.3389/fimmu.2024.1291938] [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: 09/10/2023] [Accepted: 01/04/2024] [Indexed: 02/06/2024] Open
Abstract
INTRODUCTION To better understand the role of immune escape and cancer-associated fibroblasts (CAFs) in colon adenocarcinoma (COAD), an integrative analysis of the tumor microenvironment was performed using a set of 12 immune- and CAF-related genes (ICRGs). METHODS Univariate and least absolute shrinkage and selection operator (LASSO) Cox regression analyses were used to establish a prognostic signature based on the expression of these 12 genes (S1PR5, AEN, IL20RB, FGF9, OSBPL1A, HSF4, PCAT6, FABP4, KIF15, ZNF792, CD1B and GLP2R). This signature was validated in both internal and external cohorts and was found to have a higher C-index than previous COAD signatures, confirming its robustness and reliability. To make use of this signature in clinical settings, a nomogram incorporating ICRG signatures and key clinical parameters, such as age and T stage, was developed. Finally, the role of S1PR5 in the immune response of COAD was validated through in vitro cytotoxicity experiments. RESULTS The developed nomogram exhibited slightly improved predictive accuracy compared to the ICRG signature alone, as indicated by the areas under the receiver operating characteristic curves (AUC, nomogram:0.838; ICRGs:0.807). The study also evaluated the relationships between risk scores (RS) based on the expression of the ICRGs and other key immunotherapy variables, including immune checkpoint expression, immunophenoscore (IPS), and microsatellite instability (MSI). Integration of these variables led to more precise prediction of treatment efficacy, enabling personalized immunotherapy for COAD patients. Knocking down S1PR5 can enhance the efficacy of PD-1 monoclonal antibody, promoting the cytotoxicity of T cells against HCT116 cells ((p<0.05). DISCUSSION These findings indicate that the ICRG signature may be a valuable tool for predicting prognostic risk, evaluating the efficacy of immunotherapy, and tailoring personalized treatment options for patients with COAD.
Collapse
Affiliation(s)
- Jingsun Wei
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaoxu Ge
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yucheng Qian
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kai Jiang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Chen
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Wei Lu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Hang Yang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Dongliang Fu
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yimin Fang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xinyi Zhou
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Qian Xiao
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Yang Tang
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| | - Kefeng Ding
- Department of Colorectal Surgery and Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, Zhejiang, China
- Department of Colorectal Surgery and Oncology, Cancer Center of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
8
|
Chandran EBA, Iannantuono GM, Atiq SO, Akbulut D, Sinaii N, Simon NI, Banday AR, Boudjadi S, Gurram S, Nassar AH, Rosenberg JE, Butera G, Teo MY, Sonpavde G, Coleman JA, Apolo AB. Mismatch repair deficiency and microsatellite instability in urothelial carcinoma: a systematic review and meta-analysis. BMJ ONCOLOGY 2024; 3:e000335. [PMID: 39086924 PMCID: PMC11203074 DOI: 10.1136/bmjonc-2024-000335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/03/2024] [Indexed: 08/02/2024]
Abstract
Background Mismatch repair deficiency (dMMR) and microsatellite instability-high (MSI-H) occur in a subset of cancers and have been shown to confer sensitivity to immune checkpoint inhibition (ICI); however, there is a lack of prospective data in urothelial carcinoma (UC). Methods and analysis We performed a systematic review to estimate the prevalence of dMMR and MSI-H in UC, including survival and clinical outcomes. We searched for studies published up to 26 October 2022 in major scientific databases. We screened 1745 studies and included 110. Meta-analyses were performed if the extracted data were suitable. Results The pooled weighted prevalences of dMMR in bladder cancer (BC) and upper tract UC (UTUC) were 2.30% (95% CI 1.12% to 4.65%) and 8.95% (95% CI 6.81% to 11.67%), respectively. The pooled weighted prevalences of MSI-H in BC and UTUC were 2.11% (95% CI 0.82% to 5.31%) and 8.36% (95% CI 5.50% to 12.53%), respectively. Comparing localised versus metastatic disease, the pooled weighted prevalences for MSI-H in BC were 5.26% (95% CI 0.86% to 26.12%) and 0.86% (95% CI 0.59% to 1.25%), respectively; and in UTUC, they were 18.04% (95% CI 13.36% to 23.91%) and 4.96% (95% CI 2.72% to 8.86%), respectively. Cumulatively, the response rate in dMMR/MSI-H metastatic UC treated with an ICI was 22/34 (64.7%) compared with 1/9 (11.1%) with chemotherapy. Conclusion Both dMMR and MSI-H occur more frequently in UTUC than in BC. In UC, MSI-H occurs more frequently in localised disease than in metastatic disease. These biomarkers may predict sensitivity to ICI in metastatic UC and resistance to cisplatin-based chemotherapy.
Collapse
Affiliation(s)
- Elias B A Chandran
- Genitourinary Malignancies Branch, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Saad O Atiq
- Genitourinary Malignancies Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Dilara Akbulut
- Laboratory of Pathology, National Institutes of Health, Bethesda, Maryland, USA
| | - Ninet Sinaii
- Biostatistics and Clinical Epidemiology Service, National Institutes of Health, Bethesda, Maryland, USA
| | - Nicholas I Simon
- Genitourinary Malignancies Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Abdul Rouf Banday
- Genitourinary Malignancies Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Salah Boudjadi
- Genitourinary Malignancies Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Sandeep Gurram
- Urologic Oncology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Amin H Nassar
- Department of Hematology/Oncology, Yale New Haven Hospital, New Haven, Connecticut, USA
| | | | - Gisela Butera
- Division of Library Services, National Institutes of Health, Bethesda, Maryland, USA
| | - Min Yuen Teo
- Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Guru Sonpavde
- Medical Oncology, AdventHealth Central Florida, Orlando, Florida, USA
| | | | - Andrea B Apolo
- Genitourinary Malignancies Branch, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
9
|
He L, Yang J, Zhang B, Wang Y, Wang J, Ye Q. A comparison of performance of 6-mononucleotide site panel and NCI panel for microsatellite instability detection in patients with colorectal adenocarcinoma. Pathol Res Pract 2023; 244:154390. [PMID: 36905693 DOI: 10.1016/j.prp.2023.154390] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/17/2023] [Accepted: 02/26/2023] [Indexed: 03/02/2023]
Abstract
BACKGROUND Microsatellite instability (MSI) represents as a molecular hallmark of deficient MMR system at the genomic level. Increasing clinical significance of MSI status highlights the necessity of simple, accurate markers for detection. Although 2B3D NCI panel is the most widely applied, it has been questioned whether the performance of NCI panel is second to none in MSI detection. METHODS We evaluated the efficacy of the NCI panel versus a 6-mononucleotide site panel (BAT25, BAT26, NR21, NR24, NR27, and MONO-27) in assessing MSI status of 468 Chinese patients with CRC, and compared MSI test results with the results by immunohistochemistry of four MMR proteins (MLH1, PMS2, MSH2, MSH6) in the present study. Clinicopathological variables were also collected, and their associations with MSI or MMR proteins status were analyzed using either the chi-square test or the Fisher's exact test. RESULTS MSI-H/dMMR was significantly associated with right colon involvement, poor differentiation, early stage, mucinous adenocarcinoma, negative lymph node, less neural invasion, and KRAS/NRAS/BRAF wild-type. As to the efficiency of detecting deficient MMR system, both panels had good concordance with MMR proteins expression by IHC, and 6-mononucleotide site panel outperformed NCI panel in sensitivity, specificity, positive predictive value, and negative predictive value numerically despite the lack of statistical significance. The advantage was more obvious in the sensitivity and specificity analyses of each single microsatellite markers from 6-mononucleotide site panel in comparison with NCI panel. Additionally, the rate of MSI-L detected by 6-mononucleotide site panel was much lower than that detected by the NCI panel (0.64% vs. 2.86%, P = 0.0326). CONCLUSION 6-mononucleotide site panel had a greater ability to help resolve cases of MSI-L into either MSI-H or MSS. We propose that 6-mononucleotide site panel may be potentially more suitable than NCI panel for Chinese CRC population. Large-scale studies are warranted to validate our findings.
Collapse
Affiliation(s)
- Lu He
- Department of Pathology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Jun Yang
- Department of Pathology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Biao Zhang
- Department of Pathology, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, China
| | - Yuyang Wang
- Department of Pathology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Jing Wang
- Department of Pathology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Qing Ye
- Department of Pathology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Intelligent Pathology Institute, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
10
|
Mei WJ, Mi M, Qian J, Xiao N, Yuan Y, Ding PR. Clinicopathological characteristics of high microsatellite instability/mismatch repair-deficient colorectal cancer: A narrative review. Front Immunol 2022; 13:1019582. [PMID: 36618386 PMCID: PMC9822542 DOI: 10.3389/fimmu.2022.1019582] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancers (CRCs) with high microsatellite instability (MSI-H) and deficient mismatch repair (dMMR) show molecular and clinicopathological characteristics that differ from those of proficient mismatch repair/microsatellite stable CRCs. Despite the importance of MSI-H/dMMR status in clinical decision making, the testing rates for MSI and MMR in clinical practice remain low, even in high-risk populations. Additionally, the real-world prevalence of MSI-H/dMMR CRC may be lower than that reported in the literature. Insufficient MSI and MMR testing fails to identify patients with MSI-H/dMMR CRC, who could benefit from immunotherapy. In this article, we describe the current knowledge of the clinicopathological features, molecular landscape, and radiomic characteristics of MSI-H/dMMR CRCs. A better understanding of the importance of MMR/MSI status in the clinical characteristics and prognosis of CRC may help increase the rates of MMR/MSI testing and guide the development of more effective therapies based on the unique features of these tumors.
Collapse
Affiliation(s)
- Wei-Jian Mei
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| | - Mi Mi
- Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jing Qian
- Global Medical Affairs, MSD China, Shanghai, China
| | - Nan Xiao
- Global Medical Affairs, MSD China, Shanghai, China
| | - Ying Yuan
- Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Provincial Clinical Research Center for CANCER, Hangzhou, China
- Cancer Center of Zhejiang University, Hangzhou, China
| | - Pei-Rong Ding
- Department of Colorectal Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Qin S, Li J, Zhong H, Jin C, Chen L, Yuan X, Fan Q, Chen K, Cao P, Xiao J, Jiang D, Zhang T, Zhang H, Wang X, Wang W, Han L, Wang Q, Zhu J. Serplulimab, a novel anti-PD-1 antibody, in patients with microsatellite instability-high solid tumours: an open-label, single-arm, multicentre, phase II trial. Br J Cancer 2022; 127:2241-2248. [PMID: 36261583 PMCID: PMC9726893 DOI: 10.1038/s41416-022-02001-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 09/07/2022] [Accepted: 09/23/2022] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND Microsatellite instability-high/mismatch repair-deficient (MSI-H/dMMR) tumours have a high response rate to immunotherapy. Antitumour activity and safety of serplulimab, a novel humanised anti-PD-1 monoclonal antibody, were evaluated in this phase II study. METHODS In this ongoing, single-arm, open-label, phase II trial, patients with previously treated unresectable or metastatic MSI-H/dMMR solid tumours received intravenous serplulimab 3 mg/kg every 2 weeks for up to 52 cycles. The primary endpoint was objective response rate (ORR) assessed by an independent radiological review committee per Response Evaluation Criteria in Solid Tumors v1.1. Secondary endpoints included additional efficacy measures, safety, and tolerability. RESULTS As of 9 January 2021, 108 patients were enrolled, and 68 patients with confirmed MSI-H solid tumours were included in the main efficacy analysis population (MEAP). The median follow-up duration in the MEAP was 7.7 months, with an ORR of 38.2% (95% confidence interval, 26.7-50.8). Of the 108 patients, grade ≥3 treatment-emergent adverse events were reported in 53 (49.1%) patients; immune-related adverse events occurred in 52 (48.1%) patients. CONCLUSIONS Serplulimab demonstrates a durable antitumour effect and a manageable safety profile in previously treated patients with MSI-H solid tumours. Serplulimab is a promising tissue-agnostic treatment for previously treated MSI-H solid tumours. TRIAL REGISTRATION NCT03941574.
Collapse
Affiliation(s)
- Shukui Qin
- Department of Oncology, Qinhuai Medical Area, Eastern Theater General Hospital of PLA China, Nanjing, China
| | - Jin Li
- Department of Oncology, Tongji University Shanghai East Hospital, Shanghai, China.
| | - Haijun Zhong
- Department of Abdominal Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Chuan Jin
- Department of Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Lili Chen
- Department of Hematology and Oncology, Taizhou First People's Hospital, Taizhou, China
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Qingxia Fan
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kehe Chen
- Department of Oncology, The People's Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Peiguo Cao
- Department of Oncology, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Jianjun Xiao
- Chemotherapeutic Department, Zhongshan City People's Hospital, Zhongshan, China
| | - Da Jiang
- Department of Oncology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tao Zhang
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hongyu Zhang
- Cancer Center, The Fifth Affiliated Hospital Sun Yat-sen University, Zhuhai, China
| | - Xicheng Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
| | - Wei Wang
- Department of Gastrointestinal Oncology, The First People's Hospital of Foshan, Foshan, China
| | - Lin Han
- Shanghai Henlius Biotech, Inc., Shanghai, China
| | - Qingyu Wang
- Shanghai Henlius Biotech, Inc., Shanghai, China
| | - Jun Zhu
- Shanghai Henlius Biotech, Inc., Shanghai, China
| |
Collapse
|
12
|
Li Y, Xiao J, Zhang T, Zheng Y, Jin H. Analysis of KRAS, NRAS, and BRAF Mutations, Microsatellite Instability, and Relevant Prognosis Effects in Patients With Early Colorectal Cancer: A Cohort Study in East Asia. Front Oncol 2022; 12:897548. [PMID: 35837115 PMCID: PMC9273961 DOI: 10.3389/fonc.2022.897548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Accepted: 05/23/2022] [Indexed: 11/18/2022] Open
Abstract
Background Early colorectal cancer (ECRC) refers to any size of colorectal cancer (CRC) whose depth of invasion is limited to the mucosa and submucosa. About 10% of patients with ECRC die from cancer after surgery. KRAS, NRAS, and BRAF mutations and microsatellite instability (MSI) are considered diagnostic and prognostic markers in CRC. However, their characteristics in ECRC and whether postoperative chemotherapy based on them will benefit ECRC patients or not remain unknown. Patients and Methods Patients with ECRC and 298 patients with advanced colorectal cancer (ACRC) were collected in our hospital from January 2013 to December 2015. The Amplification Refractory Mutation System (ARMS)-PCR was used to perform the KRAS, NRAS, and BRAF mutant tests. Results In ECRC patients, 43 cases of KRAS mutation were found, accounting for 69.35%. Interestingly, among KRAS mutations, there were 10 KRAS multi-site mutation patients (16.13% in 62 ECRC patients). Moreover, the NRAS mutation rate was 3.23% but no BRAF mutation was found and only 1 case of MSI-High was detected. KRAS mutation was only related to the depth of tumor invasion whereas KRAS multi-site mutations were related to mucus components and tumor size. As far as NRAS is concerned, mutations were associated with elevated CEA, mucus components, and the depth of tumor invasion. Notably, compared with 2.35% KRAS multi-site mutation in ACRC, the rate of KRAS multi-site mutation in ECRC was much higher. Furthermore, Cox regression analysis revealed that KRAS mutation could be an independent prognostic factor of ECRC in patients who have undergone endoscopic resection or surgery. Conclusion Patients with ECRC might benefit from KRAS mutation testing but not from postoperative chemotherapy.
Collapse
Affiliation(s)
- Yang Li
- Gastroenterology Endoscopy Center, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
| | - Jun Xiao
- Gastroenterology Endoscopy Center, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
- *Correspondence: Jun Xiao,
| | - Tiancheng Zhang
- Gastroenterology Endoscopy Center, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
| | - Yanying Zheng
- Department of Pathology, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
| | - Hailin Jin
- Gastroenterology Endoscopy Center, Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of Chinese Medicine), Nanjing, China
| |
Collapse
|
13
|
Zhang X, Lian XM, Gu XF, Liu Y, Feng CY, Li L, Xu HF, Du JC, Zhao YQ, Ma L, Liu YY, Huang JX, Cao J, Wang XH, Du LB, Duan SX, Wang WJ, Fan YP, Yu YQ, Zhang SK, Shi JH, Qiao YL. Utilization of genetic biomarkers testing and its associated factors in advanced colorectal cancer patients in China: a nationwide multicenter clinical epidemiological study. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:324. [PMID: 35434030 PMCID: PMC9011275 DOI: 10.21037/atm-22-988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
Abstract
Background Biomarkers are a key tool in early detection, prognostication, survival, and predicting treatment response of colorectal cancer (CRC). However, little is known about biomarker testing for CRC patients in real-life clinical practice in China. This study aimed to address the usage of biomarker testing and analyze factors related to its acceptance among Chinese patients with advanced CRC. Methods A multicenter, cross-sectional, hospital-based clinical epidemiology study was conducted from March 2020 to March 2021. Nineteen hospitals were selected in seven geographical regions of China using stratified, multistage, nonrandomized cluster sampling. Data on demographics and clinical characteristics of each eligible CRC patient in stage III or IV diseases were recorded based on the patients' self-reporting and/or medical records. In addition, information on whether biomarker testing [RAS, BRAF, and microsatellite instability (MSI)] was performed, the results and timing for performing biomarker testing, and the reasons for refusing biomarker testing were also recorded. Univariate and multivariate logistic regression were conducted to explore the potential factors of biomarker testing. Results A total of 4,526 patients were enrolled in the study, of whom 41.4%, 36.1%, and 28.2% underwent RAS, BRAF, and MSI testing, respectively. RAS, BRAF, and high-level MSI (MSI-high) mutation rates in Chinese patients with advanced CRC were 37.0%, 9.9%, and 8.1%, respectively. The logistic regression analysis revealed that the treating hospital, age at diagnosis, education, family income, tumor site, history of chemotherapy and radiotherapy, and metastases were dependent factors affecting the utilization of biomarker testing in advanced CRC in China (P<0.005). Conclusions The biomarker testing rate, especially MSI testing, is less prevalent in clinical practice for patients with advanced CRC in China. Our findings may guide the formulation of biomarker testing of CRC strategies in China and other low-income countries.
Collapse
Affiliation(s)
- Xi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xue-Mei Lian
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
| | - Xiao-Fen Gu
- Department of Student Affairs, Affiliated Tumor Hospital, Xinjiang Medical University, Ürümqi, China
| | - Yin Liu
- Department of Cancer Epidemiology, Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Chang-Yan Feng
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Li Li
- Department of Clinical Research, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Hui-Fang Xu
- Department of Cancer Epidemiology, Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Jing-Chang Du
- School of Public Health, Chengdu Medical College, Chengdu, China
| | - Yu-Qian Zhao
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Li Ma
- Public Health School, Dalian Medical University, Dalian, China
| | - Yun-Yong Liu
- Liaoning Office for Cancer Control and Research, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
| | - Juan-Xiu Huang
- Department of Gastroenterology, Wuzhou Red Cross Hospital, Wuzhou, China
| | - Ji Cao
- Department of Cancer Prevention and Control Office, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiao-Hui Wang
- Department of Public Health, Gansu Provincial Cancer Hospital, Lanzhou, China
| | - Ling-Bin Du
- Department of Cancer Prevention, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shuang-Xia Duan
- Department of Preventive Health, Xinxiang Central Hospital, Xinxiang, China
| | - Wen-Jun Wang
- School of Nursing, Jining Medical University, Jining, China
| | - Yan-Ping Fan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yan-Qin Yu
- The Clinical Epidemiology of Research Center, Department of Public Health and Preventive Medicine, Baotou Medical College, Baotou, China
| | - Shao-Kai Zhang
- Department of Cancer Epidemiology, Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
| | - Ji-Hai Shi
- The Clinical Epidemiology of Research Center, Department of Dermatology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
| | - You-Lin Qiao
- Department of Cancer Epidemiology, Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
- Center for Global Health, School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - China Working Group on Colorectal Cancer Survey
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Beijing Office for Cancer Prevention and Control, Peking University Cancer Hospital & Institute, Beijing, China
- School of Public Health and Management, Chongqing Medical University, Chongqing, China
- Department of Student Affairs, Affiliated Tumor Hospital, Xinjiang Medical University, Ürümqi, China
- Department of Cancer Epidemiology, Affiliated Cancer Hospital of Zhengzhou University/Henan Cancer Hospital, Henan Engineering Research Center of Cancer Prevention and Control, Henan International Joint Laboratory of Cancer Prevention, Zhengzhou, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
- Department of Clinical Research, The First Affiliated Hospital, Jinan University, Guangzhou, China
- School of Public Health, Chengdu Medical College, Chengdu, China
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- Public Health School, Dalian Medical University, Dalian, China
- Liaoning Office for Cancer Control and Research, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, China
- Department of Gastroenterology, Wuzhou Red Cross Hospital, Wuzhou, China
- Department of Cancer Prevention and Control Office, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of Public Health, Gansu Provincial Cancer Hospital, Lanzhou, China
- Department of Cancer Prevention, The Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou, China
- Department of Preventive Health, Xinxiang Central Hospital, Xinxiang, China
- School of Nursing, Jining Medical University, Jining, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
- The Clinical Epidemiology of Research Center, Department of Public Health and Preventive Medicine, Baotou Medical College, Baotou, China
- The Clinical Epidemiology of Research Center, Department of Dermatology, The First Affiliated Hospital of Baotou Medical College, Baotou, China
- Center for Global Health, School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
Yao D, Yu L, He W, Hu Y, Xu H, Yuan Y, Dai H. Antineoplastic prescription among patients with colorectal cancer in eight major cities of China, 2015-2019: an observational retrospective database analysis. BMJ Open 2021; 11:e046166. [PMID: 34706945 PMCID: PMC8552170 DOI: 10.1136/bmjopen-2020-046166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES It is unclear what is driving rising colorectal cancer (CRC) treatment costs in China, whether an adjustment in drug prices changes use and total cost. This study aims to estimate trends in drug use, prescribing patterns and spending for antineoplastic drug therapies for CRC in major cities of China. METHODS Information from 128 811 antineoplastic drug prescriptions in CRC was retrospectively collected from the Hospital Prescription Analysis Cooperative Project. The prescriptions extracted included demographic information of patients, the generic name and the price of antineoplastic drugs. The Mann-Kendall and Cochran-Armitage trend test was used to estimate the trends of antineoplastic agent usage. RESULTS The number of antineoplastic prescriptions ranged from 18 966 in 2015 to 34 219 in 2019. Among the prescriptions collected in this study, the annual cost of antineoplastic drugs increased by 117.2%, and average prescription cost increased by 20%. Throughout the study period, the most prescribed antineoplastic drugs were capecitabine, oxaliplatin, fluorouracil and irinotecan, representing 49%, 27%, 21% and 9% of (per cent of visits (PV)). The PV of bevacizumab and cetuximab increased by 494% and 338% (from 1.8% and 1.3% in 2015 to 10.7% and 5.7% in 2019). In prescribing patterns of antineoplastic agents, monotherapy gradually decreased, while combination therapy, especially three-drug combination, increased significantly from 1.35% to 7.31%. CONCLUSION This study estimated recent trends of antineoplastic drug use and expenditure for Chinese patients with CRC. These results would inform CRC treatment decisions, including health insurance negotiation, precision therapy access, allocation of research funding and evaluation of the financial burden of CRC drug treatment.
Collapse
Affiliation(s)
- Difei Yao
- Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingyan Yu
- Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Wei He
- Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yangmin Hu
- Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Huimin Xu
- Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ying Yuan
- Medical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Cancer Institute (Key Laboratory of Cancer Prevention and Intervention, Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Haibin Dai
- Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|