1
|
Nishida N. Biomarkers and Management of Cholangiocarcinoma: Unveiling New Horizons for Precision Therapy. Cancers (Basel) 2025; 17:1243. [PMID: 40227772 PMCID: PMC11987923 DOI: 10.3390/cancers17071243] [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: 01/22/2025] [Revised: 03/31/2025] [Accepted: 04/03/2025] [Indexed: 04/15/2025] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive malignancy with limited methods for early detection, necessitating the development of reliable biomarkers for diagnosis and management. However, conventional tumor markers, such as CA19-9 and CEA, exhibit insufficient diagnostic accuracy. Recent advancements in molecular genetics have identified several actionable mutations in CCA, enabling molecularly targeted therapies that improve survival in patients harboring these genetic alterations. Cancer panels, which facilitate multiplex genetic profiling, are critical for identifying these mutations. Studies indicate that several actionable mutations are detected in CCA cases, with patients receiving mutation-guided therapies achieving markedly better outcomes. Liquid biopsies, including cell-free DNA and circulating tumor DNA, offer real-time, non-invasive approaches to monitoring tumor dynamics, heterogeneity, and treatment responses. Furthermore, numerous studies have identified non-coding RNAs in serum and bile as promising biomarkers for the diagnosis and management of CCA. On the other hand, immunotherapy, particularly immune checkpoint inhibitors, has shown efficacy in subsets of CCA patients. However, the success of these therapies is often affected by the status of the tumor immune microenvironment (TME), underscoring the need for comprehensive TME analysis to predict responses to immune checkpoint inhibitors. Despite these advances, no single biomarker currently demonstrates sufficient sensitivity or specificity for clinical application. The integration of multi-omics approaches with cutting-edge technologies holds promise for enhancing diagnostic accuracy, optimizing treatment stratification, and advancing precision medicine in CCA. These developments highlight the transformative potential of biomarkers to improve early detection, prognostic assessment, and personalized therapeutic interventions for CCA.
Collapse
Affiliation(s)
- Naoshi Nishida
- Department of Gastroenterology and Hepatology, Faculty of Medicine, Kindai University Osaka 589-8511, Japan
| |
Collapse
|
2
|
Fu S, Du H, Dai Y, Zheng K, Cao G, Xu L, Zhong Y, Niu C, Kong Y, Wang X. Screening and molecular mechanism research on bile microRNAs associated with chemotherapy efficacy in perihilar cholangiocarcinoma. iScience 2024; 27:111437. [PMID: 39717085 PMCID: PMC11664176 DOI: 10.1016/j.isci.2024.111437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 08/22/2024] [Accepted: 11/18/2024] [Indexed: 12/25/2024] Open
Abstract
The efficacy of hepatic arterial infusion chemotherapy (HAIC) with oxaliplatin (OXA) and 5-fluorouracil (5-Fu) for treating advanced perihilar cholangiocarcinoma (pCCA) has been demonstrated, yet the survival benefits of HAIC for pCCA patients vary. Here, we aimed to screen out HAIC resistance-related bile microRNAs (miRNAs) and explore the functions of specific bile miRNAs in pCCA based on high-throughput sequencing. Levels of bile miR-532-3p, miR-1250-5p, and miR-4772-5p were related to the survival of advanced pCCA patients after HAIC. However, only overexpression of miR-532-3p promoted OXA/5-Fu resistance, and downregulation of its expression improved sensitivity to OXA/5-Fu. Mechanistic investigations revealed secreted protein acidic and rich in cysteine (SPARC) as the direct target of miR-532-3p. Our study reveals that bile miR-532-3p, miR-1250-5p, and miR-4772-5p may serve as survival biomarkers in advanced pCCA patients after HAIC and that bile miR-532-3p promotes resistance to HAIC with OXA and 5-Fu via negatively regulating SPARC expression.
Collapse
Affiliation(s)
- Shijie Fu
- Department of Interventional Radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100000, China
| | - Haizhen Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Yuyang Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Kanglian Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Guang Cao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Liang Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yujie Zhong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chuanxin Niu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Yan Kong
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Renal Cancer and Melanoma, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiaodong Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Interventional Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
3
|
Yan M, Wang H, Wei R, Li W. Arsenic trioxide: applications, mechanisms of action, toxicity and rescue strategies to date. Arch Pharm Res 2024; 47:249-271. [PMID: 38147202 DOI: 10.1007/s12272-023-01481-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023]
Abstract
Arsenical medicine has obtained its status in traditional Chinese medicine for more than 2,000 years. In the 1970s, arsenic trioxide was identified to have high efficacy and potency for the treatment of acute promyelocytic leukemia, which promoted many studies on the therapeutic effects of arsenic trioxide. Currently, arsenic trioxide is widely used to treat acute promyelocytic leukemia and various solid tumors through various mechanisms of action in clinical practice; however, it is accompanied by a series of adverse reactions, especially cardiac toxicity. This review presents a comprehensive overview of arsenic trioxide from preclinical and clinical efficacy, potential mechanisms of action, toxicities, and rescue strategies for toxicities to provide guidance or assistance for the clinical application of arsenic trioxide.
Collapse
Affiliation(s)
- Meng Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China.
| | - Hao Wang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Rui Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
- Pharmacy Department, Jinling Hospital, Medical School of Nanjing University, Nanjing, China
| | - Wenwen Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
4
|
Wu Z, Jiang S, Chen Y. Non-coding RNA and Drug resistance in cholangiocarcinoma. Noncoding RNA Res 2024; 9:194-202. [PMID: 38125756 PMCID: PMC10730441 DOI: 10.1016/j.ncrna.2023.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 12/23/2023] Open
Abstract
Cholangiocarcinoma is a highly aggressive cancer with a dismal prognosis and limited resectability. Chemotherapy has demonstrated tremendous benefits for patients with advanced and inoperable cancer, but drug resistance poses a significant obstacle. Despite recent progress in cancer therapy, the mechanisms driving drug resistance are multifaceted and not completely comprehended. Non-coding RNA refers to RNA molecules that are endogenous and do not code for proteins. Particularly microRNAs, long non-coding RNAs, circular RNAs, are widely acknowledged to be involved in cancer initiation, proliferation, and metastasis. Recently, evidences suggests that abnormal expression of non-coding RNAs contributes to resistance to different type of cancer therapies in cholangiocarcinoma. This occurs via the rewiring of signaling pathways including the reduction of anticancer drugs, apoptosis, interaction between cholangiocarcinoma and tumor-infiltrating immune cells, and cancer stemness. Thus, our review aims to demonstrate the potential of targeting non-coding RNA to override drug resistance and summarize the molecular mechanisms of how non-coding RNA contributes to drug resistance in cholangiocarcinoma.
Collapse
Affiliation(s)
- Zhaowei Wu
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, 404100, Chongqing, China
| | - Shiming Jiang
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, 404100, Chongqing, China
| | - Yong Chen
- Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Medical College Street, Yuzhong District, 404100, Chongqing, China
| |
Collapse
|
5
|
Liu Q, Lei Z. The Role of microRNAs in Arsenic-Induced Human Diseases: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37930083 DOI: 10.1021/acs.jafc.3c03721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
MicroRNAs (miRNAs) are noncoding RNAs with 20-22 nucleotides, which are encoded by endogenous genes and are capable of targeting the majority of human mRNAs. Arsenic is regarded as a human carcinogen, which can lead to many adverse health effects including diabetes, skin lesions, kidney disease, neurological impairment, male reproductive injury, and cardiovascular disease (CVD) such as cardiac arrhythmias, ischemic heart failure, and endothelial dysfunction. miRNAs can act as tumor suppressors and oncogenes via directly targeting oncogenes or tumor suppressors. Recently, miRNA dysregulation was considered to be an important mechanism of arsenic-induced human diseases and a potential biomarker to predict the diseases caused by arsenic exposure. Endogenic miRNAs such as miR-21, the miR-200 family, miR-155, and the let-7 family are involved in arsenic-induced human disease by inducing translational repression or RNA degradation and influencing multiple pathways, including mTOR/Arg 1, HIF-1α/VEGF, AKT, c-Myc, MAPK, Wnt, and PI3K pathways. Additionally, exogenous miRNAs derived from plants, such as miR-34a, miR-159, miR-2911, miR-159a, miR-156c, miR-168, etc., among others, can be transported from blood to specific tissue/organ systems in vivo. These exogenous miRNAs might be critical players in the treatment of human diseases by regulating host gene expression. This review summarizes the regulatory mechanisms of miRNAs in arsenic-induced human diseases, including cancers, CVD, and other human diseases. These special miRNAs could serve as potential biomarkers in the management and treatment of human diseases linked to arsenic exposure. Finally, the protective action of exogenous miRNAs, including antitumor, anti-inflammatory, anti-CVD, antioxidant stress, and antivirus are described.
Collapse
Affiliation(s)
- Qianying Liu
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Zhiqun Lei
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| |
Collapse
|
6
|
Wang G, Gao X, Sun Z, He T, Huang C, Li S, Long H. Circular RNA SMARCA5 inhibits cholangiocarcinoma via microRNA-95-3p/tumor necrosis factor receptor associated factor 3 axis. Anticancer Drugs 2023; 34:1002-1009. [PMID: 36727735 PMCID: PMC10501356 DOI: 10.1097/cad.0000000000001487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 11/16/2022] [Indexed: 02/03/2023]
Abstract
Enhancing research indicatedthat circular RNA (circRNA) acted a critical part in cholangiocarcinoma (CHOL) development. This research aims to discover the role of circRNA SWI/SNF-related, matrix-associated, actin-dependent regulator of chromatin, subfamily a, member 5 (SMARCA5) in CHOL bio-progression, which has been proved to be downregulated in CHOL tissues. In this study, quantitative reverse transcription polymerase chain reaction was used to reveal the level and linkage of circRNA SMARCA5, miRNA-95-3p and TNF receptor-associated factor 3 gene (TRAF3) in CHOL tissues and cancer cells. The target sites of circRNA SMARCA5 and miRNA-95-3p were forecast by Starbase, and Targetscan was conducted to forecast the potential linkage points of TRAF3 and miRNA-95-3p, and which is affirmed by double luciferase reporter assay. CCK-8 and flow cytometry assay was carried to indicate cell viability. And apoptosis-related protein was counted by caspase3 activity and Western blot assay. CircRNA SMARCA5 was downregulated in CHOL cell lines and cancer samples. Besides, over-expression of SMARCA5 inhibited cell growth and promoted apoptotic rate. Dual-luciferase reporter assays presented that miRNA-95-3p could link with circRNA SMARCA5. Moreover, miRNA-95-3p was discovered highly expressed in CHOL. Interference of miRNA-95-3p repressed cell proliferation and raised the apoptosis. Importantly, TRAF3 was validated to be a downstream of miRNA-95-3p. Strengthen of miRNA-95-3p reversed the inhibitory impact of circRNA SMARCA5-plasmid transfection, and the results of miRNA-95-3p inhibitor were reversed by si-TRAF3. CircRNA SMARCA5 is involved in CHOL development by interosculating miRNA-95-3p/TRAF3 axis and may become a novel approach for treating CHOL.
Collapse
Affiliation(s)
- Guangxin Wang
- Department of General Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University)
| | - Xia Gao
- Department of Oncology, Wuhan Asia General Hospital, Wuhan, China
| | - Zhijun Sun
- Department of General Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University)
| | - Tianyou He
- Department of General Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University)
| | - Chaogang Huang
- Department of General Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University)
| | - Shouwei Li
- Department of General Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University)
| | - Haocheng Long
- Department of General Surgery, Wuhan Third Hospital (Tongren Hospital of Wuhan University)
| |
Collapse
|
7
|
Jalil AT, Abdulhadi MA, Al-Ameer LR, Khaleel LA, Abdulameer SJ, Hadi AM, Merza MS, Zabibah RS, Ali A. Small but mighty: How microRNAs drive the deadly progression of cholangiocarcinoma. Pathol Res Pract 2023; 247:154565. [PMID: 37267725 DOI: 10.1016/j.prp.2023.154565] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/17/2023] [Accepted: 05/20/2023] [Indexed: 06/04/2023]
Abstract
Cholangiocarcinoma, also referred to as CCA, is a highly complex epithelial malignancy that can impact various organs and regions of the body, including the perihilar, intrahepatic, and distal organs. This cancer is characterized by the malignant growth of the epithelial lining in the bile ducts, which spans the entire biliary tree and is accountable for disease progression. The current state of affairs concerning CCA is concerning, with poor prognoses, high recurrence rates, and dismal long-term survival rates significantly burden healthcare facilities worldwide. Studies have identified numerous signaling pathways and molecules involved in the development and progression of CCA, including microRNAs, an important class of non-coding RNAs that have the ability to modulate these cellular signaling pathways significantly. In addition, microRNAs may serve as an innovative target for developing novel therapeutic approaches for CCA. In this review, we explore the underlying mechanisms and signaling pathways implicated in the initiation and progression of CCA, focusing on the future direction of utilizing microRNAs as a promising treatment option for this challenging malignancy.
Collapse
Affiliation(s)
| | - Mohanad Ali Abdulhadi
- Department of Medical Laboratory Techniques, Al-maarif University College, Al-Anbar, Iraq
| | | | - Luay Ali Khaleel
- College of Dentistry, National University of Science and Technology, Dhi Qar, Iraq
| | - Sada Jasim Abdulameer
- Biology Department, College of Education for Pure Science, Wasit University, Kut, Wasit, Iraq
| | | | - Muna S Merza
- Prosthetic dental Techniques Department, Al-Mustaqbal university College, Babylon, 51001, Iraq
| | - Rahman S Zabibah
- Medical Laboratory Technology Department, College of Medical Technology, The Islamic University, Najaf, Iraq
| | - Ahmed Ali
- Medical technical college, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
8
|
Targeting prolyl isomerase Pin1 as a promising strategy to overcome resistance to cancer therapies. Pharmacol Res 2022; 184:106456. [PMID: 36116709 DOI: 10.1016/j.phrs.2022.106456] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/10/2022] [Accepted: 09/14/2022] [Indexed: 11/22/2022]
Abstract
The development of tumor therapeutic resistance is one of the important reasons for the failure of antitumor therapy. Starting with multiple targets and multiple signaling pathways is helpful in understanding the mechanism of tumor resistance. The overexpression of prolyl isomerase Pin1 is highly correlated with the malignancy of cancer, since Pin1 controls many oncogenes and tumor suppressors, as well as a variety of cancer-driving signaling pathways. Strikingly, numerous studies have shown that Pin1 is directly involved in therapeutic resistance. In this review, we mainly summarize the functions and mechanisms of Pin1 in therapeutic resistance of multifarious cancers, such as breast, liver, and pancreatic carcinomas. Furtherly, from the perspective of Pin1-driven cancer signaling pathways including Raf/MEK/ERK, PI3K/Akt, Wnt/β-catenin, NF-κB, as well as Pin1 inhibitors containing juglone, epigallocatechin-3-gallate (EGCG), all-trans retinoic acid (ATRA) and arsenic trioxide (ATO), it is better to demonstrate the important potential role and mechanism of Pin1 in resistance and sensitization to cancer therapies. It will provide new therapeutic approaches for clinical reversal and prevention of tumor resistance by employing synergistic administration of Pin1 inhibitors and chemotherapeutics, implementing combination therapy of Pin1-related cancer signaling pathway inhibitors and Pin1 inhibitors, and exploiting novel Pin1-specific inhibitors.
Collapse
|
9
|
Cholangiopathies and the noncoding revolution. Curr Opin Gastroenterol 2022; 38:128-135. [PMID: 35098934 DOI: 10.1097/mog.0000000000000806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
PURPOSE OF REVIEW Noncoding RNAs (ncRNAs), including microRNAs (miRNAs) and long noncoding RNAs (lncRNAs) among others, have attracted a great deal of attention for their potential role as master regulators of gene expression and as therapeutic targets. This review focuses on recent advances on the role of ncRNAs in the pathogenesis, diagnosis and treatment of diseases of the cholangiocytes (i.e. cholangiopathies). RECENT FINDINGS In the recent years, there has been an exponential growth in the knowledge on ncRNAs and their role in cholangiopathies, particularly cholangiocarcinoma. SUMMARY Although several studies focused on miRNAs as noninvasive biomarkers for diagnosis and staging, several studies also highlighted their functions and provided new insights into disease mechanisms.
Collapse
|
10
|
Zhao Y, Yao H, Yang K, Han S, Chen S, Li Y, Chen S, Huang K, Lian G, Li J. Arsenic Trioxide-loaded nanoparticles Enhance the Chemosensitivity of Gemcitabine in Pancreatic Cancer via Reversal of Pancreatic Stellate Cells Desmoplasia through Targeting AP4/Galectin-1 Pathway. Biomater Sci 2022; 10:5989-6002. [DOI: 10.1039/d2bm01039a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Pancreatic stellate cell (PSCs) constitutes the fibrotic tumor microenvironment composed of the stroma matrix, which blocks the penetration of Gemcitabine (GEM) in pancreatic adenocarcinoma (PDAC) and results in chemoresistance. We...
Collapse
|