1
|
Reshkin SJ, Cardone RA, Koltai T. Genetic Signature of Human Pancreatic Cancer and Personalized Targeting. Cells 2024; 13:602. [PMID: 38607041 PMCID: PMC11011857 DOI: 10.3390/cells13070602] [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: 02/12/2024] [Revised: 03/27/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Pancreatic cancer is a highly lethal disease with a 5-year survival rate of around 11-12%. Surgery, being the treatment of choice, is only possible in 20% of symptomatic patients. The main reason is that when it becomes symptomatic, IT IS the tumor is usually locally advanced and/or has metastasized to distant organs; thus, early diagnosis is infrequent. The lack of specific early symptoms is an important cause of late diagnosis. Unfortunately, diagnostic tumor markers become positive at a late stage, and there is a lack of early-stage markers. Surgical and non-surgical cases are treated with neoadjuvant and/or adjuvant chemotherapy, and the results are usually poor. However, personalized targeted therapy directed against tumor drivers may improve this situation. Until recently, many pancreatic tumor driver genes/proteins were considered untargetable. Chemical and physical characteristics of mutated KRAS are a formidable challenge to overcome. This situation is slowly changing. For the first time, there are candidate drugs that can target the main driver gene of pancreatic cancer: KRAS. Indeed, KRAS inhibition has been clinically achieved in lung cancer and, at the pre-clinical level, in pancreatic cancer as well. This will probably change the very poor outlook for this disease. This paper reviews the genetic characteristics of sporadic and hereditary predisposition to pancreatic cancer and the possibilities of a personalized treatment according to the genetic signature.
Collapse
Affiliation(s)
- Stephan J. Reshkin
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Environment, University of Bari “Aldo Moro”, 70125 Bari, Italy;
| | - Tomas Koltai
- Oncomed, Via Pier Capponi 6, 50132 Florence, Italy
| |
Collapse
|
2
|
Khabipov A, Trung DN, van der Linde J, Miebach L, Lenz M, Erne F, von Bernstorff W, Schulze T, Kersting S, Bekeschus S, Partecke LI. CCR4 Blockade Diminishes Intratumoral Macrophage Recruitment and Augments Survival of Syngeneic Pancreatic Cancer-Bearing Mice. Biomedicines 2023; 11:1517. [PMID: 37371612 DOI: 10.3390/biomedicines11061517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/29/2023] Open
Abstract
Pancreatic cancer is known for its tumor microenvironment (TME), which is rich in stromal and immune cells supporting cancer growth and therapy resistance. In particular, tumor-associated macrophages (TAMs) are known for their angiogenesis- and metastasis-promoting properties, which lead to the failure of conventional therapies for pancreatic cancer. Hence, treatment options targeting TAMs are needed. The C-C chemokine receptor type 4 (CCR4) is critical for immune cell recruitment into the TME, and in this paper we explore the effects of its genetic or immunotherapeutic blockade in pancreatic-cancer-bearing mice. Murine PDA6606 pancreatic cancer cells and murine peritoneal macrophages were used for in vitro migration assays. In vivo, a syngeneic, orthotropic pancreatic cancer model was established. Tumor growth and survival were monitored under prophylactic and therapeutic application of a CCR4 antagonist (AF-399/420/18025) in wildtype (CCR4wt) and CCR4-knockout (CCR4-/-) mice. Immune infiltration was monitored in tumor tissue sections and via flow cytometry of lysed tumors. PDA6606 cells induced less migration in CCR4-/- than in CCR4wt macrophages in vitro. Pancreatic TAM infiltration was higher, and survival was reduced in CCR4wt mice compared to CCR4-/- mice. Antagonizing CCR4 in wildtype mice revealed similar results as in CCR4-/- mice without antagonization. Prophylactic CCR4 antagonist application in wildtype mice was more efficient than therapeutic antagonization. CCR4 seems to be critically involved in TAM generation and tumor progression in pancreatic cancer. CCR4 blockade may help prolong the relapse-free period after curative surgery in pancreatic cancer and improve prognosis.
Collapse
Affiliation(s)
- Aydar Khabipov
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Dung Nguyen Trung
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Julia van der Linde
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Lea Miebach
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
| | - Maik Lenz
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Felix Erne
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Wolfram von Bernstorff
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Tobias Schulze
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Stephan Kersting
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
| | - Sander Bekeschus
- ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489 Greifswald, Germany
- Clinic and Policlinic for Dermatology and Venerology, Rostock University Medical Center, Strempelstr. 13, 18057 Rostock, Germany
| | - Lars Ivo Partecke
- Department of General, Thoracic, Visceral, and Vascular Surgery, Greifswald University Medical Center, Ferdinand-Sauerbruch-Str., 17475 Greifswald, Germany
- Department of General, Visceral, and Thoracic Surgery, Helios Clinic Schleswig, St. Jurgener Str. 1-3, 24837 Schleswig, Germany
| |
Collapse
|
3
|
Ni L. Advances in Human Dendritic Cell-Based Immunotherapy Against Gastrointestinal Cancer. Front Immunol 2022; 13:887189. [PMID: 35619702 PMCID: PMC9127253 DOI: 10.3389/fimmu.2022.887189] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/08/2022] [Indexed: 11/23/2022] Open
Abstract
Dendritic cells (DCs), the strongest antigen-presenting cells, are a focus for orchestrating the immune system in the fight against cancer. Basic scientific investigations elucidating the cellular biology of the DCs have resulted in new strategies in this fight, including cancer vaccinology, combination therapy, and adoptive cellular therapy. Although immunotherapy is currently becoming an unprecedented bench-to-bedside success, the overall response rate to the current immunotherapy in patients with gastrointestinal (GI) cancers is pretty low. Here, we have carried out a literature search of the studies of DCs in the treatment of GI cancer patients. We provide the advances in DC-based immunotherapy and highlight the clinical trials that indicate the therapeutic efficacies and toxicities related with each vaccine. Moreover, we also offer the yet-to-be-addressed questions about DC-based immunotherapy. This study focuses predominantly on the data derived from human studies to help understand the involvement of DCs in patients with GI cancers.
Collapse
Affiliation(s)
- Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
4
|
The tumor microenvironment in pancreatic ductal adenocarcinoma: current perspectives and future directions. Cancer Metastasis Rev 2021; 40:675-689. [PMID: 34591240 DOI: 10.1007/s10555-021-09988-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is among the most lethal malignancies and is characterized by a unique tumor microenvironment (TME) consisting of an abundant stromal component. Many features contained with the PDAC stroma contribute to resistance to cytotoxic and immunotherapeutic regimens, as well as the propensity for this tumor to metastasize. At the cellular level, PDAC cells crosstalk with a complex mixture of non-neoplastic cell types including fibroblasts, endothelial cells, and immune cells. These intricate interactions fuel the progression and therapeutic resistance of this aggressive cancer. Moreover, data suggest the polarization of these cell types, in particular immune and fibroblast populations, dictate how PDAC tumors grow, metastasize, and respond to therapy. As a result, current research is focused on how to best target these populations to render tumors responsive to treatment. Herein, we summarize the cell populations implicated in providing a supporting role for the development and progression of PDAC. We focus on stromal fibroblasts and immune subsets that have been widely researched. We discuss factors which govern the phenotype of these populations and provide insight on how they have been targeted therapeutically. This review provides an overview of the tumor microenvironment and postulates that cellular and soluble factors within the microenvironment can be specifically targeted to improve patient outcomes.
Collapse
|
5
|
Ji YX, Wang CZ, Li X, Li L. KLF5 promotes HpSlyD induced gastric intestinal metaplasia by activating Wnt/β-catenin pathway. Shijie Huaren Xiaohua Zazhi 2021; 29:274-281. [DOI: 10.11569/wcjd.v29.i6.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Krüppel like factor 5 (KLF5) and the Wnt/β-catenin pathway are hot topics in the research of tumorigenesis. Some studies have found that KLF5 is related to the differentiation and proliferation of intestinal epithelial cells, suggesting that KLF5 may be involved in the occurrence of intestinal metaplasia. However, there are few reports on the effect of KLF5 on intestinal metaplasia and the underlying molecular mechanism.
AIM To explore the effect of KLF5 on gastric intestinal metaplasia (GIM) induced by Helicobacter pylori (H. pylori) and the underlying mechanism.
METHODS The mRNA and protein expression of KLF5 and Wnt3a was detected by RT-PCR and Western blot, respectively. Gastric epithelial cell line GES1 was cultured in vitro and divided into a blank control group, HpSlyD group (200 ng/mL HpSlyD + negative sequence), KLF5 interference group (200 ng/mL HpSlyD + KLF5 siRNA), Wnt agonist group (200 ng/mL HpSlyD + KLF5 siRNA + lithium chloride), and Wnt agonist + KLF5 interference group. GES1 cell proliferation and apoptosis were determined by MTT assay and flow cytometry, respectively. RT-PCR was used to detect KLF5, Wnt3a, beta-catenin, wool protein 1 (VIL1), trefoil factor 2 (TFF2), and caudal homeobox factor 2 (CDX2) mRNA expression. The expression of VIL1, TFF2, and CDX2 proteins was detected by Western blot.
RESULTS The expression of KLF5 and Wnt3a mRNA and protein in intestinal metaplasia was significantly higher than that in the normal gastric mucosa (P < 0.01). The cell proliferation rates in the HpSlyD group and interference control group were significantly higher than that of the blank control group, and the apoptosis rates were significantly lower than that of the blank control group (P < 0.05). The cell proliferation rate of the KLF5 interference group was significantly lower than that of the HpSlyD group, and the apoptosis rate was significantly higher than that of the HpSlyD group (P < 0.05). The mRNA and protein expression of VIL1, TFF2, and CDX2 in the HpSlyD group and interference control group was significantly higher than that of the blank control group (P < 0.05), while the mRNA and protein expression of VIL1, TFF2, and CDX2 in the KLF5 interference group were significantly lower than those of the HpSlyD group (P < 0.05). The expression of Wnt3a, β-catenin, and CDX2 mRNA in the KLF5 interference group was significantly lower than that of the HpSlyD group (P < 0.05). The expression of Wnt3a, β-catenin, and CDX2 mRNA in the Wnt agonist + KLF5 interference group was significantly higher than that of the KLF5 interference group (P < 0.05).
CONCLUSION Interference of KLF5 expression can significantly inhibit HpSlyD induced gastric metaplasia. KLF5 may promote HpSlyD induced gastric metaplasia by activating the Wnt/β-catenin pathway, which provides a new target for clinical prevention of gastric cancer.
Collapse
Affiliation(s)
- Yong-Xin Ji
- Department of Emergency Medicine, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, Zhejiang Province, China
| | - Chun-Zi Wang
- Department of Emergency Medicine, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, Zhejiang Province, China
| | - Xue Li
- Department of Emergency Medicine, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, Zhejiang Province, China
| | - Li Li
- Department of Gastroenterology, Huzhou Hospital of Traditional Chinese Medicine Affiliated to Zhejiang University of Traditional Chinese Medicine, Huzhou 313000, Zhejiang Province, China
| |
Collapse
|
6
|
Huan Y, Wei J, Zhou J, Liu M, Yang J, Gao Y. Label-Free Liquid Chromatography-Mass Spectrometry Proteomic Analysis of the Urinary Proteome for Measuring the Escitalopram Treatment Response From Major Depressive Disorder. Front Psychiatry 2021; 12:700149. [PMID: 34658947 PMCID: PMC8514635 DOI: 10.3389/fpsyt.2021.700149] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 09/07/2021] [Indexed: 11/13/2022] Open
Abstract
Major depressive disorder (MDD) is a common mental disorder that can cause substantial impairments in quality of life. Clinical treatment is usually built on a trial-and-error method, which lasts ~12 weeks to evaluate whether the treatment is efficient, thereby leading to some inefficient treatment measures. Therefore, we intended to identify early candidate urine biomarkers to predict efficient treatment response in MDD patients. In this study, urine samples were collected twice from 19 respondent and 10 non-respondent MDD patients receiving 0-, 2-, and 12-week treatments with escitalopram. Differential urinary proteins were subsequently analyzed by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). Our two pilot tests suggested that the urine proteome reflects changes associated with major depressive disorder at the early stage of treatment measures. On week 2, 20 differential proteins were identified in the response group compared with week 0, with 14 of these proteins being associated with the mechanisms of MDD. In the non-response group, 60 differential proteins were identified at week 2, with 28 of these proteins being associated with the mechanisms of MDD. In addition, differential urinary proteins at week 2 between the response and non-response groups can be clearly distinguished by using orthogonal projection on latent structure-discriminant analysis (OPLS-DA). Our small pilot tests indicated that the urine proteome can reflect early effects of escitalopram therapy between the response and non-response groups since at week 2, which may provide potential early candidate urine biomarkers to predict efficient treatment measures in MDD patients.
Collapse
Affiliation(s)
- Yuhang Huan
- Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, China
| | - Jing Wei
- Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, China
| | - Jingjing Zhou
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Min Liu
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Jian Yang
- The National Clinical Research Center for Mental Disorders and Beijing Key Laboratory of Mental Disorders, Beijing Anding Hospital, Capital Medical University, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Youhe Gao
- Department of Biochemistry and Molecular Biology, Gene Engineering Drug and Biotechnology Beijing Key Laboratory, Beijing Normal University, Beijing, China
| |
Collapse
|
7
|
Zhou Y, Zhang Y, Wang J. Trefoil Factor 2 Regulates Proliferation and Apoptosis of Pancreatic Cancer Cells and LPS-Induced Normal Pancreatic Duct Cells by β-Catenin Pathway. Cancer Manag Res 2020; 12:10705-10713. [PMID: 33149677 PMCID: PMC7605628 DOI: 10.2147/cmar.s274578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/11/2020] [Indexed: 01/13/2023] Open
Abstract
Introduction Pancreatic cancer (PC) is a malignant tumor with poor prognosis. This study aimed to determine the role of trefoil factor 2 (TFF2) in the proliferation and apoptosis of LPS-induced normal pancreatic duct cells and pancreatic cancer cells through β-catenin pathway. Methods TFF2 expression in normal pancreatic duct cells, pancreatic cancer cells and LPS-induced normal pancreatic duct cells was detected by RT-qPCR analysis and Western blot analysis. The transfection effects in pancreatic cancer cells and LPS-induced normal pancreatic duct cells were analyzed by RT-qPCR analysis. After indicated transfection, proliferation, apoptosis and inflammation of these cells were respectively detected by CCK-8 assay, TUNEL assay and certain ELISA kits. Expression of β-catenin pathway-related proteins was analyzed by Western blot analysis. Co-immunoprecipitation assay determined the combination of TFF2 and β-catenin. Results TFF2 expression was increased in pancreatic cancer cells and LPS-induced HPDE cells compared with HPDE cells. According to TFF2 expression in these cells, PanC-1 cells and 5 μg/mL LPS were selected. In addition, TFF2 interference decreased the proliferation and promoted the apoptosis of PanC-1 cells and LPS-induced HPDE cells. However, TFF2 interference did not obviously change the levels of TNF-α, IL-1β and IL-6 in PanC-1 cells and LPS-induced HPDE cells. Furthermore, TFF2 interference suppressed the expression of β-catenin, c-Myc, Cyclin D1 and BIRC5 in PanC-1 cells and LPS-induced HPDE cells. TFF2 was demonstrated to combine with β-catenin. Discussion TFF2 interference inhibits proliferation and promotes apoptosis of PanC-1 cells and LPS-induced HPDE cells by suppressing β-catenin pathway.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, People's Republic of China
| | - Yan Zhang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, People's Republic of China
| | - Jia Wang
- Department of Clinical Laboratory Medicine, Shanghai Tenth People's Hospital of Tongji University, Shanghai 200072, People's Republic of China
| |
Collapse
|
8
|
Zhu S, Yang N, Wu J, Wang X, Wang W, Liu YJ, Chen J. Tumor microenvironment-related dendritic cell deficiency: a target to enhance tumor immunotherapy. Pharmacol Res 2020; 159:104980. [PMID: 32504832 DOI: 10.1016/j.phrs.2020.104980] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 05/07/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
Dendritic cells (DCs), as specialized antigen-presenting cells, are essential for the initiation of specific T cell responses in innate antitumor immunity and, in certain cases, support humoral responses to inhibit tumor development. Mounting evidence suggests that the DC system displays a broad spectrum of dysfunctional status in the tumor microenvironment (TME), which ultimately affects antitumor immune responses. DC-based therapy can restore the function of DCs in the TME, thus showing a promising potential in tumor therapy. In this review, we provide an overview of the DC deficiency caused by various factors in the TME and discuss proposed strategies to reverse DC deficiency and the applications of novel combinatorial DC-based therapy for immune normalization of the tumor.
Collapse
Affiliation(s)
- Shan Zhu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Ning Yang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Jing Wu
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xue Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | - Wan Wang
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China
| | | | - Jingtao Chen
- Institute of Translational Medicine, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
9
|
Abolarinwa BA, Ibrahim RB, Huang YH. Conceptual Development of Immunotherapeutic Approaches to Gastrointestinal Cancer. Int J Mol Sci 2019; 20:E4624. [PMID: 31540435 PMCID: PMC6769557 DOI: 10.3390/ijms20184624] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/14/2019] [Accepted: 09/16/2019] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) cancer is one of the common causes of cancer-related death worldwide. Chemotherapy and/or immunotherapy are the current treatments, but some patients do not derive clinical benefits. Recently, studies from cancer molecular subtyping have revealed that tumor molecular biomarkers may predict the immunotherapeutic response of GI cancer patients. However, the therapeutic response of patients selected by the predictive biomarkers is suboptimal. The tumor immune-microenvironment apparently plays a key role in modulating these molecular-determinant predictive biomarkers. Therefore, an understanding of the development and recent advances in immunotherapeutic pharmacological intervention targeting tumor immune-microenvironments and their potential predictive biomarkers will be helpful to strengthen patient immunotherapeutic efficacy. The current review focuses on an understanding of how the host-microenvironment interactions and the predictive biomarkers can determine the efficacy of immune checkpoint inhibitors. The contribution of environmental pathogens and host immunity to GI cancer is summarized. A discussion regarding the clinical evidence of predictive biomarkers for clinical trial therapy design, current immunotherapeutic strategies, and the outcomes to GI cancer patients are highlighted. An understanding of the underlying mechanism can predict the immunotherapeutic efficacy and facilitate the future development of personalized therapeutic strategies targeting GI cancers.
Collapse
Affiliation(s)
- Bilikis Aderonke Abolarinwa
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Ridwan Babatunde Ibrahim
- Institute of Brain Science, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan.
- Taiwan International Graduate Program (TIGP) in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei 11529, Taiwan.
| | - Yen-Hua Huang
- International PhD Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center for Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan.
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan.
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan.
- Ph.D. Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|