1
|
Chai X, Zhang Y, Shi Z, Yang R, Liu X, Zhou Y, Li C, Li Z. An Overview of Predictive Biomarkers and Detection Approaches for Immunotherapy Response in GI Malignancies. J Gastroenterol Hepatol 2025; 40:1059-1069. [PMID: 40074558 DOI: 10.1111/jgh.16930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 02/25/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
This review provides an in-depth exploration of the evolving role of immunotherapy in gastrointestinal (GI) cancers, with a particular focus on immune checkpoint inhibitors (ICIs) and their associated predictive biomarkers. We present a detailed analysis of established biomarkers, such as PD-L1, microsatellite instability (MSI), tumor mutational burden (TMB), and the tumor microenvironment (TME), as well as emerging biomarkers, including gut microbiota and Epstein-Barr virus (EBV). The predictive value of these biomarkers in guiding clinical decision-making and optimizing immunotherapy outcomes is thoroughly discussed. Additionally, we highlight recent advancements in biomarker evaluation technologies, including next-generation sequencing (NGS), multiplex immunohistochemistry, and artificial intelligence (AI)-driven models. These technologies are instrumental in advancing precision medicine by enhancing the accuracy and efficiency of biomarker detection and facilitating personalized treatment approaches. The integration of these predictive biomarkers with advanced detection technologies has significantly improved the clinical efficacy of immunotherapy in GI cancers by addressing challenges such as tumor heterogeneity, immune evasion, and variable patient responses. By providing a deeper understanding of tumor biology and patient-specific factors, these tools offer the potential to optimize patient selection, treatment regimens, and, ultimately, clinical outcomes. This review underscores the transformative impact of combining predictive biomarkers with cutting-edge technologies, marking a significant step forward in the field of precision oncology for GI cancer treatment.
Collapse
Affiliation(s)
- Xinyu Chai
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yiwen Zhang
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhihui Shi
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruiling Yang
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Xumin Liu
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yueting Zhou
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Caiyang Li
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhenhui Li
- Department of Radiology, Yunnan Cancer Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
2
|
Chen L, Zhu M, Zhang H, Tang W, Liu J, Tang X, Chang X, Zhu Z, Liu T, Weng L, Zhang Y, Chen X. Remodeling of Effector and Regulatory T Cells by Capture and Utilization of miRNAs Using Nanocomposite Hydrogel for Tumor-Specific Photothermal Immunotherapy. ACS NANO 2025; 19:14873-14892. [PMID: 40223522 DOI: 10.1021/acsnano.4c18801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/15/2025]
Abstract
In immunotherapy for malignant tumors, the dysregulation of the balance between effector T cells and regulatory T cells (Tregs) and the uncertain efficacy due to individual differences have been considered as two critical challenges. In this study, we engineered an injectable nanocomposite hydrogel system (SNAs@M-Gel) capable of suppressing Treg proliferation and blocking PD-1/PD-L1-mediated immune evasion effectively, achieved through the stimulus-responsive modulation of multiple tumor-associated microRNAs. Simultaneously, this system enables microRNA-dependent photothermal immunotherapy, facilitating a highly efficient and personalized approach to tumor treatment. Specifically, oxidized sodium alginate (OSA) and cancer cell membrane (CCM)-encapsulated spherical nucleic acid nanoparticles (SNAs@M) were used to construct the SNAs@M-Gel hydrogel in situ at the tumor site through the formation of pH-sensitive Schiff base bonding and cross-linking using endogenous calcium ions (Ca2+). During treatment, SNAs@M-Gel was retained locally for up to 10 days, and SNAs@M nanoparticles were continuously released into the tumor microenvironment. Through the targeting ability of CCM, SNAs@M precisely entered tumor cells and specifically hybridized with the overexpressed miR-214 and miR-130a, leading to a significant downregulation of PD-L1 expression on tumor cells and the restoration of cytotoxic T lymphocyte (CTL) function suppressed by Tregs, thereby remodeling the immune microenvironment. In addition, miRNAs functioned as cross-linking agents, facilitating the aggregation of SNAs and allowing the localized production of photothermal agents directly inside tumor cells, which, under near-infrared (NIR) irradiation, promoted highly selective photothermal therapy. This cascade of events not only led to the destruction of the primary tumor but also resulted in the release of a substantial number of tumor-related antigens, which triggered the maturation of adjacent dendritic cells (DCs) and subsequent priming of tumor-specific CTLs, while simultaneously depleting Tregs, thereby reversing the tumor-promoting immune microenvironment and enhancing the overall therapeutic efficacy of photothermal immunotherapy.
Collapse
Affiliation(s)
- Li Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Man Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Handan Zhang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Wenjun Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Jie Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Xiaoyu Tang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Xiaowei Chang
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zeren Zhu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Tao Liu
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Lin Weng
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an 710061, P. R. China
| | - Xin Chen
- Department of Chemical Engineering, Shaanxi Key Laboratory of Energy Chemical Process Intensification, Institute of Polymer Science in Chemical Engineering, School of Chemical Engineering and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| |
Collapse
|
3
|
Ding Y, Yu Y. Therapeutic potential of flavonoids in gastrointestinal cancer: Focus on signaling pathways and improvement strategies (Review). Mol Med Rep 2025; 31:109. [PMID: 40017144 PMCID: PMC11884236 DOI: 10.3892/mmr.2025.13474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
Flavonoids are a group of polyphenolic compounds distributed in vegetables, fruits and other plants, which have considerable antioxidant, anti‑tumor and anti‑inflammatory activities. Several types of gastrointestinal (GI) cancer are the most common malignant tumors in the world. A large number of studies have shown that flavonoids have inhibitory effects on cancer, and they are recognized as a class of potential anti‑tumor drugs. Therefore, the present review investigated the molecular mechanisms of flavonoids in the treatment of different types of GI cancer and summarized the drug delivery systems commonly used to improve their bioavailability. First, the classification of flavonoids and the therapeutic effects of various flavonoids on human diseases were briefly introduced. Then, to clarify the mechanism of action of flavonoids on different types of GI cancer in the human body, the metabolic process of flavonoids in the human body and the associated signaling pathways causing five common types of GI cancer were discussed, as well as the corresponding therapeutic targets of flavonoids. Finally, in clinical settings, flavonoids have poor water solubility, low permeability and inferior stability, which lead to low absorption efficiency in vivo. Therefore, the three most widely used drug delivery systems were summarized. Suggestions for improving the bioavailability of flavonoids and the focus of the next stage of research were also put forward.
Collapse
Affiliation(s)
- Ye Ding
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yong Yu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall Medical Research Center, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
- Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
4
|
Piroozkhah M, Zabihi M, Jalali P, Salehi Z. Comprehensive Multi-Omics Analysis Reveals NPC2 and ITGAV Genes as Potential Prognostic Biomarkers in Gastrointestinal Cancers. Cancer Rep (Hoboken) 2024; 7:e70087. [PMID: 39690926 DOI: 10.1002/cnr2.70087] [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: 05/09/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024] Open
Abstract
BACKGROUND Gastrointestinal cancers (GICs) continue to dominate in terms of both incidence and mortality worldwide. Due to the absence of efficient and accurate prognostic biomarkers, the prognosis and treatment outcomes of many GICs are poor. Identifying biomarkers to predict individual clinical outcomes efficiently is a fundamental challenge in clinical oncology. Although several biomarkers have been continually discovered, their predictive accuracy is relatively modest, and their therapeutic use is restricted. In light of this, the discovery of reliable biomarkers for predicting prognosis and outcome in GIC is urgently required. MATERIALS AND METHODS We evaluated the Human Protein Atlas dataset and identified NPC Intracellular Cholesterol Transporter 2 (NPC2) and Integrin Subunit Alpha V (ITGAV) as probable poor predictive genes for these cancers. In addition, we used the GEPIA2, cBioPortal, UALCAN, LinkedOmics, STRING, Enrichr, TISDB, TIMER2.0, hTFTarget, miRTarBase, circBank, and drug-gene interaction database databases to conduct a comprehensive and systematic analysis of the NPC2 and ITGAV genes. RESULT Our results found high expression levels of NPC2 and ITGAV in most GICs. The aforementioned gene expressions were linked to several clinicopathological characteristics of GICs as well as poorer prognosis in LIHC and STAD. The most common alteration type of NPC2 was amplification, and for ITGAV was deep deletion. Significant promotor hypermethylation was also seen in NPC2 and ITGAV in PAAD and COAD, respectively. For the immunologic significance, NPC2 and ITGAV were positively correlated with the abundance of tumor-infiltrating lymphocytes and macrophages. Furthermore, various immunomodulators showed strong correlations with the expression of these genes. There were currently 10 small molecule drugs targeting ITGAV. CONCLUSION Consequently, our bioinformatics analysis showed that NPC2 and ITGAV might be used as potential biomarkers to determine the prognosis of various GICs and are also related to immune infiltration.
Collapse
Affiliation(s)
- Moein Piroozkhah
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Zabihi
- Institute of Biochemistry and Biophysics (IBB), Department of Bioinformatics, Laboratory of Complex Biological Systems and Bioinformatics (CBB), University of Tehran, Tehran, Iran
| | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Jin D, Wang YW, Lin ZM, Li C, Li M. Medical dilemma: Programmed death 1 blockade (sintilimab) therapy in patients suffering from tumours combined with psoriasis. World J Cardiol 2024; 16:546-549. [PMID: 39351336 PMCID: PMC11439104 DOI: 10.4330/wjc.v16.i9.546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 08/16/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024] Open
Abstract
Tumour immunotherapy represented by immune checkpoint inhibitors (ICIs) has greatly improved the overall prognosis of patients with malignant tumours, and is regarded as an important breakthrough in the field of medicine in recent years. ICIs have gradually become the core of tumour therapy and are increasingly used in the clinic. In order to achieve early clinical prediction and management of immune-related adverse events (irAEs), it is still necessary to perform further research on the mechanisms, risk factors, and predictors of irAE occurrence in the future. Zhou et al describe the consultation of a patient with advanced gastric cancer combined with chronic plaque psoriasis. This case provides an important reference for the use of programmed cell death protein-1 (PD-1) inhibitors in patients of tumours combined with chronic plaque psoriasis. This case also highlights that screening of high-risk groups for irAEs is critical before applying PD-1 inhibitors to patients with chronic psoriasis combined with tumours. PD-1 inhibitors are new and potent antineoplastic agents that can cause serious immune-related adverse events such as toxic epidermal necrolysis release and psoriasis. Glucocorticosteroids are the first-line agents for irAEs. The incidence of rheumatic irAEs may be higher in reality, which will inevitably become a new challenge for rheumatologists and dermatologists.
Collapse
Affiliation(s)
- Di Jin
- Department of Rheumatology, Weifang People's Hospital, Weifang 261000, Shandong Province, China
| | - Yu-Wei Wang
- Department of Cardiology, Yidu Central Hospital of Weifang, Weifang 261000, Shandong Province, China
| | - Zhi-Min Lin
- Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing 100020, China
| | - Chen Li
- Department of Rheumatology, Fangshan Hospital, Beijing University of Chinese Medicine, Beijing 102400, China
| | - Ming Li
- Department of Rheumatology, Weifang People's Hospital, Weifang 261000, Shandong Province, China.
| |
Collapse
|
6
|
Liu P, Wu J, Chen L, Wu Z, Wu Y, Zhang G, Yu B, Zhang B, Wei N, Shi J, Zhang C, Lei L, Yu S, Lai J, Guo Z, Zheng Y, Jing Z, Jiang H, Wang T, Zhou J, Wu Y, Sun C, Shen J, Zhang J, Wu Z. Water-filtered infrared A radiation hyperthermia combined with immunotherapy for advanced gastrointestinal tumours. Cancer Med 2024; 13:e70024. [PMID: 39049187 PMCID: PMC11269209 DOI: 10.1002/cam4.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Revised: 06/18/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
This study pioneered the use of WIRA whole-body infrared hyperthermia combined with ICI therapy to treat GIT and verified the feasibility and safety of HIT. The final results showed a DCR of 55.6%, with a median PFS of 53.5 days, median OS of 134 days, and an irAE incidence of 22.2%. Therefore, we believe that HIT can exert multiple synergistic sensitisation effects, thereby providing clinical benefits to patients with advanced GITs, increasing overall safety, and improving patients' QOL.
Collapse
Affiliation(s)
- Pengyuan Liu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jing Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Liting Chen
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhenhai Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Yufei Wu
- ACS (International) School of SingaporeSingapore
| | - Ganlu Zhang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Bingqi Yu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Beibei Zhang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Nan Wei
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jinan Shi
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | | | - Lan Lei
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Shuhuan Yu
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Jianjun Lai
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhen Guo
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Yuli Zheng
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Zhao Jing
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | - Hao Jiang
- Department of Oncology, Zhejiang HospitalHangzhouChina
| | | | - Jueyi Zhou
- Department of OncologyLishui People's HospitalLishuiChina
| | - Yajun Wu
- TCM Dispensary, Zhejiang HospitalHangzhouChina
| | - Chuan Sun
- Geriatrics Institute of Zhejiang ProvinceDepartment of Geriatrics, Zhejiang HospitalHangzhouChina
| | - Jie Shen
- Department of Medical Oncology, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Jian Zhang
- Department of Gastrointestinal Surgery, The First Affiliated HospitalZhejiang University School of MedicineHangzhouChina
| | - Zhibing Wu
- Department of Oncology, Zhejiang HospitalHangzhouChina
- Department of Radiation Oncology, Affiliated Zhejiang HospitalZhejiang University School of MedicineHangzhouChina
- Cancer CenterZhejiang UniversityHangzhouChina
| |
Collapse
|
7
|
Chen J, Zhao Y, Wang X, Zang L, Yin D, Tan S. Hyperoside Inhibits RNF8-mediated Nuclear Translocation of β-catenin to Repress PD-L1 Expression and Prostate Cancer. Anticancer Agents Med Chem 2024; 24:464-476. [PMID: 38305391 DOI: 10.2174/0118715206289246240110044931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/30/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024]
Abstract
BACKGROUND Hyperoside is a flavonol glycoside isolated from Hypericum perforatum L. that has inhibitory effects on cancer cells; however, its effects on prostate cancer (PCa) remain unclear. Therefore, we studied the anti-PCa effects of hyperoside and its underlying mechanisms in vitro and in vivo. AIM This study aimed to explore the mechanism of hyperoside in anti-PCa. METHODS 3-(4,5-Dimethyl-2-Thiazolyl)-2,5-Diphenyl Tetrazolium Bromide (MTT), transwell, and flow cytometry assays were used to detect PCa cell growth, invasion, and cell apoptosis. Immunoblot analysis, immunofluorescence, immunoprecipitation, and quantitative real-time PCR (qRT-PCR) were used to analyze the antitumor mechanism of hyperoside. RESULTS Hyperoside inhibited PCa cell growth, invasion, and cell cycle and induced cell apoptosis. Furthermore, RING finger protein 8 (RNF8), an E3 ligase that assembles K63 polyubiquitination chains, was predicted to be a direct target of hyperoside and was downregulated by hyperoside. Downregulation of RNF8 by hyperoside impeded the nuclear translocation of β-catenin and disrupted the Wnt/β-catenin pathway, which reduced the expression of the target genes c-myc, cyclin D1, and programmed death ligand 1 (PD-L1). Decreased PD-L1 levels contributed to induced immunity in Jurkat cells in vitro. Finally, in vivo studies demonstrated that hyperoside significantly reduced tumor size, inhibited PD-L1 and RNF8 expression, and induced apoptosis in tumor tissues of a subcutaneous mouse model. CONCLUSION Hyperoside exerts its anti-PCa effect by reducing RNF8 protein, inhibiting nuclear translocation of β-catenin, and disrupting the Wnt/β-catenin pathway, in turn reducing the expression of PD-L1 and improving Jurkat cell immunity.
Collapse
Affiliation(s)
- Jie Chen
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Yi Zhao
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Xiaoli Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Long Zang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Dengke Yin
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| | - Song Tan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, Anhui, 230012, China
| |
Collapse
|
8
|
Cong S, Bai S, Zhang M, Bi Y, Wang Y, Jin S, He H. A study on metabolic characteristics and metabolic markers of gastrointestinal tumors. Cancer Biol Ther 2023; 24:2255369. [PMID: 37705174 PMCID: PMC10503448 DOI: 10.1080/15384047.2023.2255369] [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: 05/22/2022] [Revised: 09/28/2022] [Accepted: 06/06/2023] [Indexed: 09/15/2023] Open
Abstract
Tumor cells have significant heterogeneity in metabolism and are closely related to prognosis, gene mutation, and subtype. However, this association has not been demonstrated in reports of gastrointestinal tumors. In this study, we constructed four metabolic subtypes and identified four gene signatures using the expression data and clinical information of 252 metabolism-related genes from TCGA and NCBI databases for gastric adenocarcinoma (STAD) and colorectal cancer (COAD and READ). MC1 had the worst prognosis compared to other classifications. GSig1 was mainly related to drug metabolism and was the highest in MC1 with the worst prognosis, while the other subtypes were mainly related to glucose metabolism pathways. This difference also existed in other different malignant tumors. In addition, metabolic typing was associated with chemotherapeutic drug response and tumor heterogeneity, which indicated that monitoring metabolic typing could contribute to drug efficacy and gene-targeted therapy. In conclusion, we identified differences among subtypes in clinical characteristics such as prognosis and revealed the potential function of metabolic subtype in response to chemotherapeutic agents and oncogene mutations. This work highlighted the potential clinical meaning of metabolic subtype and characteristics in drug therapy and prognosis assessment of malignant tumors.
Collapse
Affiliation(s)
- Shan Cong
- Department of Laparoscopic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - shanshan Bai
- Department of Ultrasound, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - Minghao Zhang
- Department of Vascular Interventional, Affiliated Hongqi Hospital of Mudanjiang Medical College, Mudanjiang, China
| | - yanfang Bi
- Department of Nursing, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - yu Wang
- Department of Laparoscopic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - shi Jin
- Department of Laparoscopic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| | - hui He
- Department of Laparoscopic Surgery, the First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, China
| |
Collapse
|
9
|
Zhang J, Hu C, Zhang R, Xu J, Zhang Y, Yuan L, Zhang S, Pan S, Cao M, Qin J, Cheng X, Xu Z. The role of macrophages in gastric cancer. Front Immunol 2023; 14:1282176. [PMID: 38143746 PMCID: PMC10746385 DOI: 10.3389/fimmu.2023.1282176] [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: 08/23/2023] [Accepted: 11/24/2023] [Indexed: 12/26/2023] Open
Abstract
As one of the deadliest cancers of the gastrointestinal tract, there has been limited improvement in long-term survival rates for gastric cancer (GC) in recent decades. The poor prognosis is attributed to difficulties in early detection, minimal opportunity for radical resection and resistance to chemotherapy and radiation. Macrophages are among the most abundant infiltrating immune cells in the GC stroma. These cells engage in crosstalk with cancer cells, adipocytes and other stromal cells to regulate metabolic, inflammatory and immune status, generating an immunosuppressive tumour microenvironment (TME) and ultimately promoting tumour initiation and progression. In this review, we summarise recent advances in our understanding of the origin of macrophages and their types and polarisation in cancer and provide an overview of the role of macrophages in GC carcinogenesis and development and their interaction with the GC immune microenvironment and flora. In addition, we explore the role of macrophages in preclinical and clinical trials on drug resistance and in treatment of GC to assess their potential therapeutic value in this disease.
Collapse
Affiliation(s)
- Jiaqing Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Can Hu
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ruolan Zhang
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Jingli Xu
- Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
| | - Yanqiang Zhang
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Li Yuan
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shengjie Zhang
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Siwei Pan
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Mengxuan Cao
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Jiangjiang Qin
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiangdong Cheng
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhiyuan Xu
- Department of Gastric Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- Zhejiang Provincial Research Center for Upper Gastrointestinal Tract Cancer, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
10
|
Quan Q, Guo L, Huang L, Liu Z, Guo T, Shen Y, Ding S, Liu C, Cao L. Expression and clinical significance of PD-L1 and infiltrated immune cells in the gastric adenocarcinoma microenvironment. Medicine (Baltimore) 2023; 102:e36323. [PMID: 38050283 PMCID: PMC10695517 DOI: 10.1097/md.0000000000036323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 11/03/2023] [Indexed: 12/06/2023] Open
Abstract
Programmed death-ligand 1 (PD-L1) is a crucial negative costimulatory molecule expressed on both tumor and immune cells. It binds to programmed death-1, facilitating tumor escape. Tumor-infiltrating immune cells play a vital role in this process. However, the clinical relationship between PD-L1 expression and tumor-infiltrating immune cells remains uncertain. Immunohistochemistry (IHC) was utilized to assess PD-L1 expression and TIIC markers (CD3, CD4, CD8, CD19, CD31, CD68, CD11c, CD56, and α-smooth muscle actin) in gastric adenocarcinoma tissues from 268 patients. The aim was to explore the prognostic significance of PD-L1 and the infiltration of different immune cell types. The study analyzed overall survival and the correlations between PD-L1 expression, immune cell infiltration, and clinicopathological characteristics. Among the 268 patients, 52 (19.40%) exhibited high PD-L1 expression on tumor cells (TPD-L1), while 167 (62.31%) displayed high PD-L1 expression on immune cells (IPD-L1). Patients with high IPD-L1 expression showed improved survival compared to those with low IPD-L1 expression (P = .028). High TPD-L1 expression associated with various clinicopathological features, such as larger tumor size, poorer differentiation, deeper invasion depth, and higher tumor stage. Conversely, patients with high IPD-L1 expression exhibited shallower tumor invasion and lower mortality rates. Univariate analysis indicated that superficial tumor infiltration, absence of lymph node and distant metastasis, low tumor stage, high IPD-L1 expression, and elevated CD8 and CD19 expression were associated with a reduced risk of tumor progression. Multivariate analysis revealed that patients with high IPD-L1 and CD8 expression or high TPD-L1 and low CD31 expression experienced significantly better overall survival than patients with other combinations. The findings indicate that patients with high PD-L1 expression in immune cells have a substantially improved prognosis. Additionally, the combination of PD-L1 with CD8 or CD31 expression status can serve as an indicator of prognosis in patients with gastric adenocarcinoma.
Collapse
Affiliation(s)
- Qiuying Quan
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lingchuan Guo
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lili Huang
- Department of Clinical Laboratory, Children’s Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Zhiju Liu
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Tianwei Guo
- Department of Pathology, Changshu Hospital of Affiliated to Nanjing University of Chinese Medicine, Changshu, Jiangsu, China
| | - Yu Shen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Sisi Ding
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Cuiping Liu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Lei Cao
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, Jiangsu, China
- Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, Suzhou, Jiangsu, China
| |
Collapse
|
11
|
Li YT, Yuan WZ, Jin WL. Vagus innervation in the gastrointestinal tumor: Current understanding and challenges. Biochim Biophys Acta Rev Cancer 2023; 1878:188884. [PMID: 36990250 DOI: 10.1016/j.bbcan.2023.188884] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/17/2023] [Accepted: 02/28/2023] [Indexed: 03/30/2023]
Abstract
The vagus nerve (VN) is the main parasympathetic nerve of the autonomic nervous system. It is widely distributed in the gastrointestinal tract and maintains gastrointestinal homeostasis with the sympathetic nerve under physiological conditions. The VN communicates with various components of the tumor microenvironment to positively and dynamically affect the progression of gastrointestinal tumors (GITs). The intervention in vagus innervation delays GIT progression. Developments in adeno-associated virus vectors, nanotechnology, and in vivo neurobiological techniques have enabled the creation of precisely regulated "tumor neurotherapies". Furthermore, the combination of neurobiological techniques and single cell sequencing may reveal more insights into VN and GIT. The present review aimed to summarize the mechanisms of communication between the VN and the gastrointestinal TME and to explore the potential and challenges of VN-based tumor neurotherapy in GITs.
Collapse
|
12
|
Current Targeted Therapy for Metastatic Colorectal Cancer. Int J Mol Sci 2023; 24:ijms24021702. [PMID: 36675216 PMCID: PMC9864602 DOI: 10.3390/ijms24021702] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/07/2023] [Accepted: 01/13/2023] [Indexed: 01/17/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common type of cancer and the second leading cause of cancer deaths worldwide. Surgery or surgery plus radiotherapy and/or chemotherapy for patients with metastatic CRC (mCRC) were accepted as the main therapeutic strategies until the early 2000s, when targeted drugs, like cetuximab and bevacizumab, were developed. The use of targeted drugs in clinical practice has significantly increased patients' overall survival. To date, the emergence of several types of targeted drugs has opened new possibilities and revealed new prospects for mCRC treatment. Therapeutic strategies are continually being updated to select the most suitable targeted drugs based on the results of clinical trials that are currently underway. This review discusses the up-to date molecular evidence of targeted therapy for mCRC and summarizes the Food and Drug Administration-approved targeted drugs including the results of clinical trials. We also explain their mechanisms of action and how these affect the choice of a suitable targeted therapy.
Collapse
|
13
|
Wang Q, Shen Z, Ge M, Xu J, Zhang X, Zhu W, Liu J, Hua W, Mao Y. Unexpected curative effect of PD-1 inhibitor in gastric cancer with brain metastasis: A case report. Front Oncol 2023; 13:1042417. [PMID: 36874117 PMCID: PMC9978328 DOI: 10.3389/fonc.2023.1042417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 02/02/2023] [Indexed: 02/18/2023] Open
Abstract
Background Gastric cancer (GC) is the third most common cause of cancer-related death in the world. Several clinical trials have proven that the use of PD-1/PD-L1 inhibitors can improve the survival of late-stage GC patients and is suggested in NCCN and CSCO guidelines. However, the correlation between PD-L1 expression and the response to PD-1/PD-L1 inhibitors is still controversial. GC rarely develops brain metastasis (BrM) and currently there is no therapeutic protocol for GC BrMs. Case presentation We report a case of a 46-year-old male suffering from GC with PD-L1 negative BrMs 12 years after GC resection and 5 cycles of chemotherapy. We treated the patient with the immune checkpoint inhibitor (ICI) pembrolizumab and all metastatic tumors achieved a complete response (CR). A durable remission of the tumors is confirmed after 4 years of follow-up. Conclusion We shared a rare case with PD-L1 negative GC BrM responsive to PD-1/PD-L1 inhibitors, the mechanism of which is still unclear. The protocol of therapeutic choice for late-stage GC with BrM is urgently needed. And we are expecting biomarkers other than PD-L1 expressions to predict the efficacy of ICI treatment.
Collapse
Affiliation(s)
- Qijun Wang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Zhewei Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Mengxi Ge
- Department of Oncology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jie Xu
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xin Zhang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Jie Liu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Wei Hua
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Ying Mao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China.,Shanghai Key Laboratory of Brain Function Restoration and Neural Regeneration, Shanghai, China.,Neurosurgical Institute of Fudan University, Shanghai, China.,Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| |
Collapse
|
14
|
Pang K, Shi ZD, Wei LY, Dong Y, Ma YY, Wang W, Wang GY, Cao MY, Dong JJ, Chen YA, Zhang P, Hao L, Xu H, Pan D, Chen ZS, Han CH. Research progress of therapeutic effects and drug resistance of immunotherapy based on PD-1/PD-L1 blockade. Drug Resist Updat 2023; 66:100907. [PMID: 36527888 DOI: 10.1016/j.drup.2022.100907] [Citation(s) in RCA: 105] [Impact Index Per Article: 52.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/12/2022] [Accepted: 11/27/2022] [Indexed: 12/05/2022]
Abstract
The binding of programmed death-1 (PD-1) on the surface of T cells and PD-1 ligand 1 (PD-L1) on tumor cells can prevent the immune-killing effect of T cells on tumor cells and promote the immune escape of tumor cells. Therefore, immune checkpoint blockade targeting PD-1/PD-L1 is a reliable tumor therapy with remarkable efficacy. However, the main challenges of this therapy are low response rate and acquired resistance, so that the outcomes of this therapy are usually unsatisfactory. This review begins with the description of biological structure of the PD-1/PD-L1 immune checkpoint and its role in a variety of cells. Subsequently, the therapeutic effects of immune checkpoint blockers (PD-1 / PD-L1 inhibitors) in various tumors were introduced and analyzed, and the reasons affecting the function of PD-1/PD-L1 were systematically analyzed. Then, we focused on analyzing, sorting out and introducing the possible underlying mechanisms of primary and acquired resistance to PD-1/PD-L1 blockade including abnormal expression of PD-1/PD-L1 and some factors, immune-related pathways, tumor immune microenvironment, and T cell dysfunction and others. Finally, promising therapeutic strategies to sensitize the resistant patients with PD-1/PD-L1 blockade treatment were described. This review is aimed at providing guidance for the treatment of various tumors, and highlighting the drug resistance mechanisms to offer directions for future tumor treatment and improvement of patient prognosis.
Collapse
Affiliation(s)
- Kun Pang
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China; School of Life Sciences, Jiangsu Normal University, Jiangsu, China
| | - Zhen-Duo Shi
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China; School of Life Sciences, Jiangsu Normal University, Jiangsu, China; Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang, China
| | - Liu-Ya Wei
- School of Pharmacy, Weifang Medical University, Weifang, Shandong 261053, China
| | - Yang Dong
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China
| | - Yu-Yang Ma
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Wei Wang
- Department of Medical College, Southeast University, 87 DingjiaQiao, Nanjing, China
| | - Guang-Yue Wang
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Ming-Yang Cao
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China
| | - Jia-Jun Dong
- School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China
| | - Yu-Ang Chen
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China
| | - Peng Zhang
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Lin Hao
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China
| | - Hao Xu
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Deng Pan
- Graduate School, Bengbu Medical College, Building 1, Administration Building, 2600 Donghai Avenue, Bengbu, Anhui, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| | - Cong-Hui Han
- Department of Urology, Xuzhou Clinical School of Xuzhou Medical University, Xuzhou Central Hospital, 199 Jiefang South Road, Xuzhou, Jiangsu, China; School of Life Sciences, Jiangsu Normal University, Jiangsu, China; Department of Urology, Heilongjiang Provincial Hospital, Heilongjiang, China.
| |
Collapse
|
15
|
A Novel m7G-Related Gene Signature Predicts the Prognosis of Colon Cancer. Cancers (Basel) 2022; 14:cancers14225527. [PMID: 36428620 PMCID: PMC9688272 DOI: 10.3390/cancers14225527] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/06/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022] Open
Abstract
Colon cancer (CC), one of the most common malignancies worldwide, lacks an effective prognostic prediction biomarker. N7-methylguanosine (m7G) methylation is a common RNA modification type and has been proven to influence tumorigenesis. However, the correlation between m7G-related genes and CC remains unclear. The gene expression levels and clinical information of CC patients were downloaded from public databases. Twenty-nine m7G-related genes were obtained from the published literature. Via unsupervised clustering based on the expression levels of m7G-related genes, CC patients were divided into three m7G clusters. Based on differentially expressed genes (DEGs) from the above three groups, CC patients were further divided into three gene clusters. The m7G score, a prognostic model, was established using principal component analysis (PCA) based on 15 prognosis-associated m7G genes. KM curve analysis demonstrated that the overall survival rate was remarkably higher in the high-m7G score group, which was much more significant in advanced CC patients as confirmed by subgroup analysis. Correlation analysis indicated that the m7G score was associated with tumor mutational burden (TMB), PD-L1 expression, immune infiltration, and drug sensitivity. The expression level of prognosis-related m7G genes was further confirmed in human CC cell lines and samples. This study established an m7G gene-based prognostic model (m7G score), which demonstrated the important roles of m7G-related genes during CC initiation and progression. The m7G score could be a practical biomarker to predict immunotherapy response and prognosis in CC patients.
Collapse
|
16
|
ATXN2-Mediated PI3K/AKT Activation Confers Gastric Cancer Chemoresistance and Attenuates CD8+ T Cell Cytotoxicity. J Immunol Res 2022; 2022:6863240. [PMID: 36213324 PMCID: PMC9535133 DOI: 10.1155/2022/6863240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/26/2022] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
As one of the primary therapeutic choices, chemotherapy is widely adopted for progressive gastric cancer (GC), but the development of chemoresistance has limited chemotherapy efficacy and partly contributes to poor prognosis. Immunotherapy is increasingly being applied in the clinical treatment of GC and is also benefitting patients. To ascertain whether ATXN2 affects chemotherapy efficacy in GC cells and its role in GC immune escape, we performed high-throughput sequencing to clarify genes differentially expressed between 5-FU-resistant and 5-FU-sensitive GC cells and then conducted qRT–PCR to assess ATXN2 expression in GC tissues. Furthermore, the influence of ATXN2 on resistance was studied in vitro and in vivo, ATXN2 and other protein expression levels were detected using Western blotting and immunohistochemistry (IHC), and the direct association of SP1 and ATXN2 was confirmed through luciferase reporter gene analysis. We found elevated ATXN2 in GC tumors and a negative correlation between ATXN2 levels and the prognosis of GC. Furthermore, by activating the PI3K/AKT pathway, ATXN2 was found to promote chemoresistance in GC, facilitating BCL2L1 expression. In GC cells, ATXN2 further stimulated PD-L1 expression and provided better immunotherapy efficacy. Finally, we demonstrated that SP1 transcriptionally regulated the expression of ATXN2 and prompted GC chemoresistance and immune escape. In conclusion, our study reveals the important roles of the SP1/ATXN2/PI3K-AKT/BCL2L1 signalling pathway in GC chemoresistance and of the SP1/ATXN2/PI3K-AKT/PD-L1 signalling pathway in GC immunotherapy. Our findings provide new theories and experimental references for overcoming chemotherapy resistance in GC and enhancing the efficacy of immunotherapy for GC.
Collapse
|
17
|
He D, Qin Z, Liu Z, Ji X, Gao J, Guo H, Yang F, Fan H, Wei Y, Wang Z, Liu Q, Pang Q. Comprehensive Analysis of the Prognostic Value and Immune Infiltration of Butyrophilin Subfamily 2/3 (BTN2/3) Members in Pan-Glioma. Front Oncol 2022; 12:816760. [PMID: 36033440 PMCID: PMC9399357 DOI: 10.3389/fonc.2022.816760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 04/15/2022] [Indexed: 11/13/2022] Open
Abstract
The BTN2/3 subfamilies are overexpressed in many cancers, including pan-glioma (low- and high-grade gliomas). However, the expression and prognosis of BTN2/3 subfamilies and tumor-infiltrating lymphocytes in pan-glioma remain unknown. In the present study, we systematically explored and validated the expression and prognostic value of BTN2/3 subfamily members in pan-glioma [The Cancer Genome Atlas–glioblastoma and low-grade glioma (TCGA-GBMLGG) merge cohort] using multiple public databases. We used clinical specimens for high-throughput verification and cell lines for qRT-PCR verification, which confirmed the expression profiles of BTN2/3 subfamilies. In addition, the function of the BTN2/3 subfamily members and the correlations between BTN2/3 subfamily expression and pan-glioma immune infiltration levels were investigated. We found that BTN2/3 subfamily members were rarely mutated. BTN2/3 subfamilies were overexpressed in pan-glioma; high expression of BTN2/3 subfamily members was correlated with poor prognosis. In addition, BTN2/3 subfamilies might positively regulate proliferation, and the overexpression of BTN2/3 subfamilies influenced cell cycle, differentiation, and glioma stemness. In terms of immune infiltrating levels, BTN2/3 subfamily expression was positively associated with CD4+ T-cell, B-cell, neutrophil, macrophage, and dendritic cell infiltrating levels. These findings suggest that BTN2/3 subfamily expression is correlated with prognosis and immune infiltration levels in glioma. Therefore, the BTN2/3 subfamilies can be used as biomarkers for pan-glioma and prognostic biomarkers for determining the prognosis and immune infiltration levels in pan-glioma.
Collapse
Affiliation(s)
- Dong He
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Histology and Embryology, Cheeloo College of Medicine, School of Basic Medical Sciences Shandong University, Jinan, China
| | - Zhen Qin
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zihao Liu
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- Department of Histology and Embryology, Cheeloo College of Medicine, School of Basic Medical Sciences Shandong University, Jinan, China
| | - Xiaoshuai Ji
- Department of Histology and Embryology, Cheeloo College of Medicine, School of Basic Medical Sciences Shandong University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jiajia Gao
- Department of Histology and Embryology, Cheeloo College of Medicine, School of Basic Medical Sciences Shandong University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hua Guo
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Fan Yang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haitao Fan
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yanbang Wei
- Department of Histology and Embryology, Cheeloo College of Medicine, School of Basic Medical Sciences Shandong University, Jinan, China
| | - Zixiao Wang
- Department of Histology and Embryology, Cheeloo College of Medicine, School of Basic Medical Sciences Shandong University, Jinan, China
| | - Qian Liu
- Department of Histology and Embryology, Cheeloo College of Medicine, School of Basic Medical Sciences Shandong University, Jinan, China
- *Correspondence: Qian Liu, ; Qi Pang,
| | - Qi Pang
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Neurosurgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Qian Liu, ; Qi Pang,
| |
Collapse
|
18
|
Liao K, Hu J, Huang Y, Yu S, Yang Q, Sun F, Wu C, Cheng Y, Zhang W, Zhang X, Li H, Wang X. m 6A Regulator-Based Methylation Modification Patterns Characterized by Distinct Tumor Microenvironment Immune Profiles in Rectal Cancer. Front Oncol 2022; 12:879405. [PMID: 35875124 PMCID: PMC9299953 DOI: 10.3389/fonc.2022.879405] [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: 03/12/2022] [Accepted: 05/23/2022] [Indexed: 12/24/2022] Open
Abstract
Background Previous studies reported the related role of RNA n6-methyladenosine (m6A) modification in tumorigenesis and development. However, it is not clear whether m6A modification also plays a potential role in the immune regulation of rectal cancer (RC) and the formation of tumor microenvironment. Methods In this study, we screened 23 m6A regulatory factors from 369 rectal cancer specimens, further determined the modification patterns of m6A in RC, and systematically linked these modification patterns with the characteristics of TME cell infiltration. The principal component analysis (PCA) algorithm was used to evaluate the m6A modification pattern of a single tumor related to immune response. Results Three different m6A modification patterns were found in the measurement results, which are related to different clinical results and biological pathways. TME identification results show that the identified m6A pattern is closely related to immune characteristics. According to the m6Ascore extracted from m6A-related signature genes, RC patients were divided into high and low score subgroups combined with tumor mutation burden. Patients with high tumor mutation burden and higher m6Ascore have a significant survival advantage and enhanced immune infiltration. Further analysis showed that patients with higher m6Ascore had higher PD-L1 expression levels and showed better immune response and lasting clinical benefits. Conclusions M6A modification plays a crucial role in the formation of TME diversity and complexity. The evaluation of the m6A modification mode will help us to enhance our understanding of the characteristics of TME infiltration and provide new insights for more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Kaili Liao
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jialing Hu
- Department of Emergency medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yu Huang
- School of Advanced Manufacturing of Nanchang University, Nanchang, China
| | - Siji Yu
- School of Advanced Manufacturing of Nanchang University, Nanchang, China
| | - Qijun Yang
- Queen Mary College of Nanchang University, Xuefu Road, Nanchang, Nanchang, China
| | - Fan Sun
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chengfeng Wu
- Department of Vascular Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yunqi Cheng
- Queen Mary College of Nanchang University, Xuefu Road, Nanchang, Nanchang, China
| | - Wenyige Zhang
- Queen Mary College of Nanchang University, Xuefu Road, Nanchang, Nanchang, China
| | - Xue Zhang
- Queen Mary College of Nanchang University, Xuefu Road, Nanchang, Nanchang, China
| | - Hongyu Li
- Queen Mary College of Nanchang University, Xuefu Road, Nanchang, Nanchang, China
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Laboratory Medicine, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
19
|
Xiao J, Wang X, Liu Y, Liu X, Yi J, Hu J. Lactate Metabolism-Associated lncRNA Pairs: A Prognostic Signature to Reveal the Immunological Landscape and Mediate Therapeutic Response in Patients With Colon Adenocarcinoma. Front Immunol 2022; 13:881359. [PMID: 35911752 PMCID: PMC9328180 DOI: 10.3389/fimmu.2022.881359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 06/16/2022] [Indexed: 11/30/2022] Open
Abstract
Background Lactate metabolism is critically involved in the tumor microenvironment (TME), as well as cancer progression. It is important to note, however, that lactate metabolism-related long non-coding RNAs (laRlncRNAs) remain incredibly understudied in colon adenocarcinoma (COAD). Methods A gene expression profile was obtained from the Cancer Genome Atlas (TCGA) database to identify laRlncRNA expression in COAD patients. A risk signature with prognostic value was identified from TCGA and Gene Expression Omnibus (GEO) cohort based on laRlncRNA pairs by the least absolute shrinkage and selection operator (LASSO) and Cox regression analyses. Quantitative real-time polymerase chain reaction (qRT-PCR) and functional experiments were carried out to verify the expression of laRlncRNAs in COAD. The relationship of laRlncRNA pairs with immune landscape as well as the sensitivity of different therapies was explored. Results In total, 2378 laRlncRNAs were identified, 1,120 pairs of which were studied to determine their prognostic validity, followed by a risk signature established based on the screened 5 laRlncRNA pairs. The laRlncRNA pairs-based signature provided a better overall survival (OS) prediction than other published signatures and functioned as a prognostic marker for COAD patients. According to the calculated optimal cut-off point, patients were divided into high- and low-risk groups. The OS of COAD patients in the high-risk group were significantly shorter than that of those in the low-risk group (P=4.252e-14 in the TCGA cohort and P=2.865-02 in the GEO cohort). Furthermore, it remained an effective predictor of survival in strata of gender, age, TNM stage, and its significance persisted after univariate and multivariate Cox regressions. Additionally, the risk signature was significantly correlated with immune cells infiltration, tumor mutation burden (TMB), microsatellite instability (MSI) as well as immunotherapeutic efficacy and chemotherapy sensitivity. Finally, one of the laRlncRNA, LINC01315, promotes proliferation and migration capacities of colon cancer cells. Conclusion The newly identified laRlncRNAs pairs-based signature exhibits potential effects in predicting prognosis, deciphering patients’ immune landscape, and mediating sensitivity to immunotherapy and chemotherapy. Findings in our study may provide evidence for the role of laRlncRNAs pairs as novel prognostic biomarkers and potentially individualized therapy targets for COAD patients.
Collapse
Affiliation(s)
- Junbo Xiao
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaotong Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yajun Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaowei Liu
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jun Yi
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, China
- Hunan International Scientific and Technological Cooperation Base of Artificial Intelligence Computer Aided Diagnosis and Treatment for Digestive Disease, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- *Correspondence: Jun Yi, ; Jiuye Hu,
| | - Jiuye Hu
- Department of Gastroenterology, Affiliated Hospital of Xiangnan University, Chenzhou, China
- *Correspondence: Jun Yi, ; Jiuye Hu,
| |
Collapse
|
20
|
Liu Y, Huang K, Yang Y, Wu Y, Gao W. Prediction of Tumor Mutation Load in Colorectal Cancer Histopathological Images Based on Deep Learning. Front Oncol 2022; 12:906888. [PMID: 35686098 PMCID: PMC9171017 DOI: 10.3389/fonc.2022.906888] [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: 03/29/2022] [Accepted: 04/18/2022] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignancies, and immunotherapy can be applied to CRC patients of all ages, while its efficacy is uncertain. Tumor mutational burden (TMB) is important for predicting the effect of immunotherapy. Currently, whole-exome sequencing (WES) is a standard method to measure TMB, but it is costly and inefficient. Therefore, it is urgent to explore a method to assess TMB without WES to improve immunotherapy outcomes. In this study, we propose a deep learning method, DeepHE, based on the Residual Network (ResNet) model. On images of tissue, DeepHE can efficiently identify and analyze characteristics of tumor cells in CRC to predict the TMB. In our study, we used ×40 magnification images and grouped them by patients followed by thresholding at the 10th and 20th quantiles, which significantly improves the performance. Also, our model is superior compared with multiple models. In summary, deep learning methods can explore the association between histopathological images and genetic mutations, which will contribute to the precise treatment of CRC patients.
Collapse
Affiliation(s)
- Yongguang Liu
- Department of Anorectal Surgery, Weifang People’s Hospital, Weifang, China
| | - Kaimei Huang
- Genies (Beijing) Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Yachao Yang
- School of Mathematics and Statistics, Hainan Normal University, Haikou, China
| | - Yan Wu
- Genies (Beijing) Co., Ltd., Beijing, China
- Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao, China
| | - Wei Gao
- Department of Internal Medicine-Oncology, Fujian Cancer Hospital and Fujian Medical University Cancer Hospital, Fuzhou, China
| |
Collapse
|
21
|
Zhang Y, Li J, Feng D, Peng X, Wang B, Han T, Zhang Y. Systematic Analysis of Molecular Characterization and Clinical Relevance of Liquid–Liquid Phase Separation Regulators in Digestive System Neoplasms. Front Cell Dev Biol 2022; 9:820174. [PMID: 35252219 PMCID: PMC8891544 DOI: 10.3389/fcell.2021.820174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/21/2021] [Indexed: 01/02/2023] Open
Abstract
Background: The role of liquid–liquid phase separation (LLPS) in cancer has also attracted more and more attention, which is found to affect transcriptional regulation, maintaining genomic stability and signal transduction, and contribute to the occurrence and progression of tumors. However, the role of LLPS in digestive system tumors is still largely unknown. Results: Here, we characterized the expression profiles of LLPS regulators in 3 digestive tract tumor types such as COAD, STAD, and ESCA with The Cancer Genome Atlas (TCGA) data. Our results for the first time showed that LLPS regulatory factors, such as Brd4, FBN1, and TP53, were frequently mutated in all types of digestive system tumors. Variant allele frequency (VAF) and APOBEC analysis demonstrated that genetic alterations of LLPS regulators were related to the progression of digestive system neoplasms (DSNs), such as TP53, NPHS1, TNRC6B, ITSN1, TNPO1, PML, AR, BRD4, DLG4, and PTPN1. KM plotter analysis showed that the mutation status of LLPS regulators was significantly related to the overall survival (OS) time of DSNs, indicating that they may contribute to the progression of DSN. The expression analysis of LLPS regulatory factors showed that a variety of LLPS regulatory factors were significantly dysregulated in digestive system tumors, such as SYN2 and MAPT. It is worth noting that we first found that LLPS regulatory factors were significantly correlated with tumor immune infiltration of B cells, CD4+ T cells, and CD8+ T cells in digestive system tumors. Bioinformatics analysis showed that the LLPS regulators’ expression was closely related to multiple signaling, including the ErbB signaling pathway and T-cell receptor signaling pathway. Finally, several LLPS signatures were constructed and had a strong prognostic stratification ability in different digestive gland tumors. Finally, the results demonstrated the LLPS regulators’ signature score was significantly positively related to the infiltration levels of CD4+ T cells, neutrophil cells, macrophage cells, and CD8+ T cells. Conclusion: Our study for the first time showed the potential roles of LLPS regulators in carcinogenesis and provide novel insights to identify novel biomarkers for the prediction of immune therapy and prognosis of DSNs.
Collapse
Affiliation(s)
- Yaxin Zhang
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Jie Li
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Dan Feng
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Xiaobo Peng
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Bin Wang
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Bin Wang, ; Ting Han, ; Yingyi Zhang,
| | - Ting Han
- Departments of General Surgery, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Bin Wang, ; Ting Han, ; Yingyi Zhang,
| | - Yingyi Zhang
- Department of Oncology, Changhai Hospital, Naval Medical University, Shanghai, China
- *Correspondence: Bin Wang, ; Ting Han, ; Yingyi Zhang,
| |
Collapse
|
22
|
Luo L, Zhong A, Wang Q, Zheng T. Structure-Based Pharmacophore Modeling, Virtual Screening, Molecular Docking, ADMET, and Molecular Dynamics (MD) Simulation of Potential Inhibitors of PD-L1 from the Library of Marine Natural Products. Mar Drugs 2021; 20:29. [PMID: 35049884 PMCID: PMC8777599 DOI: 10.3390/md20010029] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND In the past decade, several antibodies directed against the PD-1/PD-L1 interaction have been approved. However, therapeutic antibodies also exhibit some shortcomings. Using small molecules to regulate the PD-1/PD-L1 pathway may be another way to mobilize the immune system to fight cancer. METHOD 52,765 marine natural products were screened against PD-L1(PDBID: 6R3K). To identify natural compounds, a structure-based pharmacophore model was generated, following by virtual screening and molecular docking. Then, the absorption, distribution, metabolism, and excretion (ADME) test was carried out to select the most suitable compounds. Finally, molecular dynamics simulation was also performed to validate the binding property of the top compound. RESULTS Initially, 12 small marine molecules were screened based on the pharmacophore model. Then, two compounds were selected for further evaluation based on the molecular docking scores. After ADME and toxicity studies, molecule 51320 was selected for further verification. By molecular dynamics analysis, molecule 51320 maintains a stable conformation with the target protein, so it has the chance to become an inhibitor of PD-L1. CONCLUSIONS Through structure-based pharmacophore modeling, virtual screening, molecular docking, ADMET approaches, and molecular dynamics (MD) simulation, the marine natural compound 51320 can be used as a small molecule inhibitor of PD-L1.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang 524023, China
| | - Ai Zhong
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (A.Z.); (Q.W.); (T.Z.)
| | - Qu Wang
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (A.Z.); (Q.W.); (T.Z.)
| | - Tongyu Zheng
- The First Clinical College, Guangdong Medical University, Zhanjiang 524023, China; (A.Z.); (Q.W.); (T.Z.)
| |
Collapse
|
23
|
Xie P, Zheng H, Chen H, Wei K, Pan X, Xu Q, Wang Y, Tang C, Gevaert O, Meng X. Tumor response as defined by iRECIST in gastrointestinal malignancies treated with PD-1 and PD-L1 inhibitors and correlation with survival. BMC Cancer 2021; 21:1246. [PMID: 34798858 PMCID: PMC8605503 DOI: 10.1186/s12885-021-08944-9] [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] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 10/28/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Atypical tumor response patterns during immune checkpoint inhibitor therapy pose a challenge to clinicians and investigators in immuno-oncology practice. This study evaluated tumor burden dynamics to identify imaging biomarkers for treatment response and overall survival (OS) in advanced gastrointestinal malignancies treated with PD-1/PD-L1 inhibitors. METHODS This retrospective study enrolled a total of 198 target lesions in 75 patients with advanced gastrointestinal malignancies treated with PD-1/PD-L1 inhibitors between January 2017 and March 2021. Tumor diameter changes as defined by immunotherapy Response Evaluation Criteria in Solid Tumors (iRECIST) were studied to determine treatment response and association with OS. RESULTS Based on the best overall response, the tumor diameter ranged from - 100 to + 135.3% (median: - 9.6%). The overall response rate was 32.0% (24/75), and the rate of durable disease control for at least 6 months was 30.7% (23/75, one (iCR, immune complete response) or 20 iPR (immune partial response), or 2iSD (immune stable disease). Using univariate analysis, patients with a tumor diameter maintaining a < 20% increase (48/75, 64.0%) from baseline had longer OS than those with ≥20% increase (27/75, 36.0%) and, a reduced risk of death (median OS: 80 months vs. 48 months, HR = 0.22, P = 0.034). The differences in age (HR = 1.09, P = 0.01), combined surgery (HR = 0.15, P = 0.01) and cancer type (HR = 0.23, P = 0.001) were significant. In multivariable analysis, patients with a tumor diameter with a < 20% increase had notably reduced hazards of death (HR = 0.15, P = 0.01) after adjusting for age, combined surgery, KRAS status, cancer type, mismatch repair (MMR) status, treatment course and cancer differentiation. Two patients (2.7%) showed pseudoprogression. CONCLUSIONS Tumor diameter with a < 20% increase from baseline during therapy in gastrointestinal malignancies was associated with therapeutic benefit and longer OS and may serve as a practical imaging marker for treatment response, clinical outcome and treatment decision making.
Collapse
Affiliation(s)
- Peiyi Xie
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, No.26 Yuancunerheng Road, Guangzhou, 510655, Guangdong, China
- Department of Medicine and Department of Biomedical Data Science, The Stanford Center for Biomedical Informatics Research (BMIR), 1265 Welch Rd, Stanford, CA, 94305, USA
| | - Hong Zheng
- Department of Medicine and Department of Biomedical Data Science, The Stanford Center for Biomedical Informatics Research (BMIR), 1265 Welch Rd, Stanford, CA, 94305, USA
| | - Haiyang Chen
- Department of Radiation Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, No.26 Yuancunerheng Road, Guangzhou, 510655, Guangdong, China
| | - Kaikai Wei
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, No.26 Yuancunerheng Road, Guangzhou, 510655, Guangdong, China
| | - Ximin Pan
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, No.26 Yuancunerheng Road, Guangzhou, 510655, Guangdong, China
| | - Qinmei Xu
- Department of Medicine and Department of Biomedical Data Science, The Stanford Center for Biomedical Informatics Research (BMIR), 1265 Welch Rd, Stanford, CA, 94305, USA
- Department of Medical Imaging, Jinling Hospital, Nanjing University School of Medicine, No.305, Zhongshan East Road, Nanjing, 210002, China
| | - Yongchen Wang
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, No.26 Yuancunerheng Road, Guangzhou, 510655, Guangdong, China
| | - Changguan Tang
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, No.26 Yuancunerheng Road, Guangzhou, 510655, Guangdong, China
| | - Olivier Gevaert
- Department of Medicine and Department of Biomedical Data Science, The Stanford Center for Biomedical Informatics Research (BMIR), 1265 Welch Rd, Stanford, CA, 94305, USA.
| | - Xiaochun Meng
- Department of Radiology, The Sixth Affiliated Hospital of Sun Yat-sen University, No.26 Yuancunerheng Road, Guangzhou, 510655, Guangdong, China.
| |
Collapse
|
24
|
Li Z, Zhang X, Liu C, Ma J. Non-immune Cell Components in the Gastrointestinal Tumor Microenvironment Influencing Tumor Immunotherapy. Front Cell Dev Biol 2021; 9:729941. [PMID: 34722510 PMCID: PMC8549829 DOI: 10.3389/fcell.2021.729941] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 09/20/2021] [Indexed: 12/15/2022] Open
Abstract
Interactions of genetic susceptibility factors, immune microenvironment, and microbial factors contribute to gastrointestinal tumorigenesis. The suppressive immune microenvironment reshaped by the tumors during gastrointestinal tumorigenesis directly contributes to T-cell depletion in tumor immunotherapy. Soluble factors secreted by tumor cells or stromal cells collectively shape the suppressive immune environment. Here, we reviewed the key factors in the gastrointestinal tumor microenvironment that influence tumor immunotherapy, focusing on the effects of fibroblasts, neuronal cells, soluble cytokines, exosomes, and the microbiome in tumor microenvironment. Research in this field has helped to identify more precise and effective biomarkers and therapeutic targets in the era of tumor immunotherapy.
Collapse
Affiliation(s)
- Zhengshuo Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Changsha, China.,NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Hunan Key Laboratory of Translational Radiation Oncology, Changsha, China
| | - Xiaoyue Zhang
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Changsha, China.,NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Hunan Key Laboratory of Translational Radiation Oncology, Changsha, China
| | - Can Liu
- Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Changsha, China.,NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Hunan Key Laboratory of Translational Radiation Oncology, Changsha, China
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Cancer Research Institute and School of Basic Medical Science, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Changsha, China.,NHC Key Laboratory of Carcinogenesis and Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Hunan Key Laboratory of Translational Radiation Oncology, Changsha, China
| |
Collapse
|
25
|
de Fátima Aquino Moreira-Nunes C, de Souza Almeida Titan Martins CN, Feio D, Lima IK, Lamarão LM, de Souza CRT, Costa IB, da Silva Maués JH, Soares PC, de Assumpção PP, Burbano RMR. PD-L1 Expression Associated with Epstein-Barr Virus Status and Patients' Survival in a Large Cohort of Gastric Cancer Patients in Northern Brazil. Cancers (Basel) 2021; 13:3107. [PMID: 34206307 PMCID: PMC8268941 DOI: 10.3390/cancers13133107] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/31/2021] [Accepted: 06/03/2021] [Indexed: 12/15/2022] Open
Abstract
Gastric cancer (GC) is a worldwide health problem, making it one of the most common types of cancer, in fifth place of all tumor types, and the third highest cause of cancer deaths in the world. There is a subgroup of GC that consists of tumors infected with the Epstein-Barr virus (EBV) and is characterized mainly by the overexpression of programmed cell death protein-ligand-1 (PD-L1). In the present study, we present histopathological and survival data of a thousand GC patients, associated with EBV status and PD-L1 expression. Of the thousand tumors analyzed, 190 were EBV-positive and the vast majority (86.8%) had a high relative expression of mRNA and PD-L1 protein (p < 0.0001) in relation to non-neoplastic control. On the other hand, in EBV-negative samples, the majority had a low PD-L1 expression of RNA and protein (p < 0.0001). In the Kaplan-Meier analysis, the probability of survival and increased overall survival of EBV-positive GC patients was impacted by the PD-L1 overexpression (p < 0.0001 and p = 0.004, respectively). However, the PD-L1 low expression was correlated with low overall survival in those patients. Patients with GC positive for EBV, presenting PD-L1 overexpression can benefit from immunotherapy treatments and performing the quantification of PD-L1 in gastric neoplasms should be adopted as routine.
Collapse
Affiliation(s)
- Caroline de Fátima Aquino Moreira-Nunes
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
- Laboratory of Pharmacogenetics, Department of Medicine, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, 60430-275 CE, Brazil
| | | | - Danielle Feio
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
| | - Isamu Komatsu Lima
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
| | - Leticia Martins Lamarão
- Foundation Center for Hemotherapy and Hematology of Pará (HEMOPA), Department of Sorology, Belém, 66033-000 PA, Brazil;
| | | | - Igor Brasil Costa
- Department of Virology, Evandro Chagas Institute, Ananindeua, 67030-000 PA, Brazil;
| | - Jersey Heitor da Silva Maués
- Hematology and Transfusion Medicine Center, Laboratory of Molecular and Cell Biology, Department of Medicine, University of Campinas, Campinas, 13083-970 SP, Brazil;
| | - Paulo Cardoso Soares
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
| | - Paulo Pimentel de Assumpção
- Oncology Research Center, Department of Biological Sciences, Federal University of Pará, Belém, 66073-005 PA, Brazil;
| | - Rommel Mário Rodríguez Burbano
- Laboratory of Molecular Biology, Department of Clinical Medicine, Ophir Loyola Hospital, Belém, 66063-240 PA, Brazil; (C.N.d.S.A.T.M.); (D.F.); (I.K.L.); (P.C.S.)
- Oncology Research Center, Department of Biological Sciences, Federal University of Pará, Belém, 66073-005 PA, Brazil;
| |
Collapse
|
26
|
Ahmad R, Singh JK, Wunnava A, Al-Obeed O, Abdulla M, Srivastava SK. Emerging trends in colorectal cancer: Dysregulated signaling pathways (Review). Int J Mol Med 2021; 47:14. [PMID: 33655327 PMCID: PMC7834960 DOI: 10.3892/ijmm.2021.4847] [Citation(s) in RCA: 93] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC) is the third most frequently detected type of cancer, and the second most common cause of cancer‑related mortality globally. The American Cancer Society predicted that approximately 147,950 individuals would be diagnosed with CRC, out of which 53,200 individuals would succumb to the disease in the USA alone in 2020. CRC‑related mortality ranks third among both males and females in the USA. CRC arises from 3 major pathways: i) The adenoma‑carcinoma sequence; ii) serrated pathway; and iii) the inflammatory pathway. The majority of cases of CRC are sporadic and result from risk factors, such as a sedentary lifestyle, obesity, processed diets, alcohol consumption and smoking. CRC is also a common preventable cancer. With widespread CRC screening, the incidence and mortality from CRC have decreased in developed countries. However, over the past few decades, CRC cases and mortality have been on the rise in young adults (age, <50 years). In addition, CRC cases are increasing in developing countries with a low gross domestic product (GDP) due to lifestyle changes. CRC is an etiologically heterogeneous disease classified by tumor location and alterations in global gene expression. Accumulating genetic and epigenetic perturbations and aberrations over time in tumor suppressor genes, oncogenes and DNA mismatch repair genes could be a precursor to the onset of colorectal cancer. CRC can be divided as sporadic, familial, and inherited depending on the origin of the mutation. Germline mutations in APC and MLH1 have been proven to play an etiological role, resulting in the predisposition of individuals to CRC. Genetic alterations cause the dysregulation of signaling pathways leading to drug resistance, the inhibition of apoptosis and the induction of proliferation, invasion and migration, resulting in CRC development and metastasis. Timely detection and effective precision therapies based on the present knowledge of CRC is essential for successful treatment and patient survival. The present review presents the CRC incidence, risk factors, dysregulated signaling pathways and targeted therapies.
Collapse
Affiliation(s)
- Rehan Ahmad
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | - Jaikee Kumar Singh
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan 303007, India
| | - Amoolya Wunnava
- Department of Biosciences, Manipal University Jaipur, Jaipur, Rajasthan 303007, India
| | - Omar Al-Obeed
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | - Maha Abdulla
- Colorectal Research Chair, Department of Surgery, King Saud University College of Medicine, Riyadh 11472, Saudi Arabia
| | | |
Collapse
|
27
|
Inhibition of T-cell-mediated immune response via the PD-1/ PD-L1 axis in cholangiocarcinoma cells. Eur J Pharmacol 2021; 897:173960. [PMID: 33617828 DOI: 10.1016/j.ejphar.2021.173960] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/05/2021] [Accepted: 02/15/2021] [Indexed: 12/14/2022]
Abstract
Cholangiocarcinoma (CCA) is a malignant biliary tract epithelium tumor. The programmed death-1 (PD-1)/programmed receptor-ligand 1 (PD-L1) signaling pathway has been implicated as an immune escape mechanism in several cancers. The present study aimed to assess the expression of PD-L1 on human CCA cell lines and its potential role in suppressing CD8+ T- cell function. A panel of intrahepatic CCA cell lines was evaluated for immune regulatory checkpoint ligands and inflammation markers. Effects of pro-inflammatory cytokine, interferon gamma (IFN-γ), on the expression of immune regulatory checkpoint ligands and inflammation markers were determined. The PD-L1 function was measured by co-culturing CCA cells with lymphocytes. Most of the selected Thai CCA cell lines, including HuCCA-1, RMCCA-1, KKU-100, and KKU-213, expressed higher PD-L1 than normal cholangiocyte MMNK-1 and ANK-1 cells. Both PD-L1 and cyclooxygenase-2 (COX-2) expressions were highest in HuCCA-1 cells. A 48 h treatment with IFN-γ increased the expression of PD-L1 and COX-2 in CCA cells. The expression of CTLA-4 ligands, including H7-1 and H7-2, did not change after IFN-γ treatment. Rofecoxib, a specific COX-2 inhibitor, mitigated IFN-γ-induced PD-L1 expression. After 48 h co-incubation, CD8+ T-cell apoptosis was increased as compared to the control group. Pretreatment of CCA cells with IFN-γ further increased CD8+ T-cell apoptosis. Pembrolizumab, an anti-PD-1 antibody, mitigated CCA cell escape phenomenon. The inhibition of T-cell-mediated immune response via the PD-L1/PD-1 axis are evidenced in intrahepatic CCA. Immunotherapy with checkpoint inhibitor offers a potentially therapeutic strategy for CCA patients; however, further in vivo and clinical studies are required.
Collapse
|