1
|
Shah A, Ganguly K, Rauth S, Sheree SS, Khan I, Ganti AK, Ponnusamy MP, Kumar S, Jain M, Batra SK. Unveiling the resistance to therapies in pancreatic ductal adenocarcinoma. Drug Resist Updat 2024; 77:101146. [PMID: 39243602 DOI: 10.1016/j.drup.2024.101146] [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: 06/12/2024] [Revised: 08/15/2024] [Accepted: 08/23/2024] [Indexed: 09/09/2024]
Abstract
Despite the ongoing advances in interventional strategies (surgery, chemotherapy, radiotherapy, and immunotherapy) for managing pancreatic ductal adenocarcinoma (PDAC), the development of therapy refractory phenotypes remains a significant challenge. Resistance to various therapeutic modalities in PDAC emanates from a combination of inherent and acquired factors and is attributable to cancer cell-intrinsic and -extrinsic mechanisms. The critical determinants of therapy resistance include oncogenic signaling and epigenetic modifications that drive cancer cell stemness and metabolic adaptations, CAF-mediated stromagenesis that results in ECM deposition altered mechanotransduction, and secretome and immune evasion. We reviewed the current understanding of these multifaceted mechanisms operating in the PDAC microenvironment, influencing the response to chemotherapy, radiotherapy, and immunotherapy regimens. We then describe how the lessons learned from these studies can guide us to discover novel therapeutic regimens to prevent, delay, or revert resistance and achieve durable clinical responses.
Collapse
Affiliation(s)
- Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Shamema S Sheree
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Imran Khan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Apar K Ganti
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Division of Oncology-hematology, Department of Internal Medicine, VA Nebraska Western Iowa Health Care System and University of Nebraska Medical Center, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha 68198-5870, USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha 68198-5870, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA; Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha 68198-5870, USA.
| |
Collapse
|
2
|
Rajesh R U, Sangeetha D. Therapeutic potentials and targeting strategies of quercetin on cancer cells: Challenges and future prospects. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 133:155902. [PMID: 39059266 DOI: 10.1016/j.phymed.2024.155902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 07/08/2024] [Accepted: 07/19/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Every cell in the human body is vital because it maintains equilibrium and carries out a variety of tasks, including growth and development. These activities are carried out by a set of instructions carried by many different genes and organized into DNA. It is well recognized that some lifestyle decisions, like using tobacco, alcohol, UV, or multiple sexual partners, might increase one's risk of developing cancer. The advantages of natural products for any health issue are well known, and researchers are making attempts to separate flavonoid-containing substances from plants. Various parts of plants contain a phenolic compound called flavonoid. Quercetin, which belongs to the class of compounds known as flavones with chromone skeletal structure, has anti-cancer activity. PURPOSE The study was aimed at investigating the therapeutic action of the flavonoid quercetin on various cancer cells. METHODS The phrases quercetin, anti-cancer, nanoparticles, and cell line were used to search the data using online resources such as PubMed, and Google Scholar. Several critical previous studies have been included. RESULTS Quercetin inhibits various dysregulated signaling pathways that cause cancer cells to undergo apoptosis to exercise its anticancer effects. Numerous signaling pathways are impacted by quercetin, such as the Hedgehog system, Akt, NF-κB pathway, downregulated mutant p53, JAK/STAT, G1 phase arrest, Wnt/β-Catenin, and MAPK. There are downsides to quercetin, like hydrophobicity, first-pass effect, instability in the gastrointestinal tract, etc., because of which it is not well-established in the pharmaceutical industry. The solution to these drawbacks in the future is using bio-nanomaterials like chitosan, PLGA, liposomes, and silk fibroin as carriers, which can enhance the target specificity of quercetin. The first section of this review covers the specifics of flavonoids and quercetin; the second section covers the anti-cancer activity of quercetin; and the third section explains the drawbacks and conjugation of quercetin with nanoparticles for drug delivery by overcoming quercetin's drawback. CONCLUSIONS Overall, this review presented details about quercetin, which is a plant derivative with a promising molecular mechanism of action. They inhibit cancer by various mechanisms with little or no side effects. It is anticipated that plant-based materials will become increasingly relevant in the treatment of cancer.
Collapse
Affiliation(s)
- Udaya Rajesh R
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India
| | - Dhanaraj Sangeetha
- Department of Chemistry, School of Advanced Science, Vellore Institute of Technology, Vellore, 632014 Tamil Nadu, India.
| |
Collapse
|
3
|
Santos VS, Vieira GM, Ruckert MT, Andrade PVD, Nagano LF, Brunaldi MO, Dos Santos JS, Silveira VS. Atypical phosphatase DUSP11 inhibition promotes nc886 expression and potentiates gemcitabine-mediated cell death through NF-kB modulation. Cancer Gene Ther 2024; 31:1402-1411. [PMID: 39048662 DOI: 10.1038/s41417-024-00804-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 06/21/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) represents one of the deadliest cancers among all solid tumors. First-line treatment relies on gemcitabine (Gem) and despite treatment improvements, refractoriness remains a universal challenge. Attempts to decipher how feedback-loops control signaling pathways towards drug resistance have gained attention in recent years, particularly focused on the role of phosphatases. In this study, a CRISPR/Cas9-based phenotypic screen was performed to identify members from the dual-specificity phosphatases (DUSP) family potentially acting on Gem response in PDAC cells. The approach revealed the atypical RNA phosphatase DUSP11 as a potential target, whose inhibition creates vulnerability of PDAC cells to Gem. DUSP11 genetic inhibition impaired cell survival and promoted apoptosis, synergistically enhancing Gem cytotoxicity. In silico transcriptome analysis of RNA-seq data from PDAC human samples identified NF-ĸB signaling pathway highly correlated with DUSP11 upregulation. Consistently, Gem-induced NF-ĸB phosphorylation was blocked upon DUSP11 inhibition in vitro. Mechanistically, we found that DUSP11 directly impacts nc886 expression and modulates PKR-NF-ĸB signaling cascade after Gem exposure in PDAC cells resulting in resistance to Gem-induced cell death. In conclusion, this study provides new insights on DUSP11 role in RNA biology and Gem response in PDAC cells.
Collapse
Affiliation(s)
- Verena Silva Santos
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Gabriela Maciel Vieira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mariana Tannús Ruckert
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Pamela Viani de Andrade
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Luis Fernando Nagano
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Mariângela Ottoboni Brunaldi
- Department of Pathology and Forensic Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - José Sebastião Dos Santos
- Department of Surgery and Anatomy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Vanessa Silva Silveira
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Iosifidou N, Anagnostopoulou E, Botou M, Kalfa E, Tatsaki E, Frillingos S. Elucidation of the Gemcitabine Transporters of Escherichia coli K-12 and Gamma-Proteobacteria Linked to Gemcitabine-Related Chemoresistance. Int J Mol Sci 2024; 25:7012. [PMID: 39000123 PMCID: PMC11241209 DOI: 10.3390/ijms25137012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/22/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Gemcitabine (2',2'-difluoro-2'-deoxycytidine), a widely used anticancer drug, is considered a gold standard in treating aggressive pancreatic cancers. Gamma-proteobacteria that colonize the pancreatic tumors contribute to chemoresistance against gemcitabine by metabolizing the drug to a less active and deaminated form. The gemcitabine transporters of these bacteria are unknown to date. Furthermore, there is no complete knowledge of the gemcitabine transporters in Escherichia coli or any other related proteobacteria. In this study, we investigate the complement of gemcitabine transporters in E. coli K-12 and two common chemoresistance-related bacteria (Klebsiella pneumoniae and Citrobacter freundii). We found that E. coli K-12 has two high-affinity gemcitabine transporters with distinct specificity properties, namely, NupC and NupG, whereas the gemcitabine transporters of C. freundii and K. pneumoniae include the NupC and NupG orthologs, functionally indistinguishable from their counterparts, and, in K. pneumoniae, one additional NupC variant, designated KpNupC2. All these bacterial transporters have a higher affinity for gemcitabine than their human counterparts. The highest affinity (KM 2.5-3.0 μΜ) is exhibited by NupGs of the bacteria-specific nucleoside-H+ symporter (NHS) family followed by NupCs (KM 10-13 μΜ) of the concentrative nucleoside transporter (CNT) family, 15-100 times higher than the affinities reported for the human gemcitabine transporter hENT1/SLC29A1, which is primarily associated with gemcitabine uptake in the pancreatic adenocarcinoma cells. Our results offer a basis for further insight into the role of specific bacteria in drug availability within tumors and for understanding the structure-function differences of bacterial and human drug transporters.
Collapse
Affiliation(s)
- Nikoleta Iosifidou
- Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (N.I.); (E.A.); (M.B.); (E.K.); (E.T.)
| | - Eleni Anagnostopoulou
- Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (N.I.); (E.A.); (M.B.); (E.K.); (E.T.)
| | - Maria Botou
- Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (N.I.); (E.A.); (M.B.); (E.K.); (E.T.)
| | - Eirini Kalfa
- Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (N.I.); (E.A.); (M.B.); (E.K.); (E.T.)
| | - Ekaterini Tatsaki
- Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (N.I.); (E.A.); (M.B.); (E.K.); (E.T.)
| | - Stathis Frillingos
- Laboratory of Biological Chemistry, Department of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (N.I.); (E.A.); (M.B.); (E.K.); (E.T.)
- University Research Center of Ioannina (URCI), Institute of Biosciences, 45110 Ioannina, Greece
| |
Collapse
|
5
|
Chen K, Cheng X, Xue S, Chen J, Zhang X, Qi Y, Chen R, Zhang Y, Wang H, Li W, Cheng G, Huang Y, Xiong Y, Chen L, Mu C, Gu M. Albumin conjugation promotes arsenic trioxide transport through alkaline phosphatase-associated transcytosis in MUC4 wildtype pancreatic cancer cells. Int J Biol Macromol 2024; 257:128756. [PMID: 38092098 DOI: 10.1016/j.ijbiomac.2023.128756] [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: 05/28/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/17/2023]
Abstract
Pancreatic cancer (PC) has a poor prognosis due to chemotherapy resistance and unfavorable drug transportation. Albumin conjugates are commonly used as drug carriers to overcome these obstacles. However, membrane-bound glycoprotein mucin 4 (MUC4) has emerged as a promising biomarker among the genetic mutations affecting albumin conjugates therapeutic window. Human serum albumin-conjugated arsenic trioxide (HSA-ATO) has shown potential in treating solid tumors but is limited in PC therapy due to unclear targets and mechanisms. This study investigated the transport mechanisms and therapeutic efficacy of HSA-ATO in PC cells with different MUC4 mutation statuses. Results revealed improved penetration of ATO into PC tumors through conjugated with HSA. However, MUC4 mutation significantly affected treatment sensitivity and HSA-ATO uptake both in vitro and in vivo. Mutant MUC4 cells exhibited over ten times higher IC50 for HSA-ATO and approximately half the uptake compared to wildtype cells. Further research demonstrated that ALPL activation by HSA-ATO enhanced transcytosis in wildtype MUC4 PC cells but not in mutant MUC4 cells, leading to impaired uptake and weaker antitumor effects. Reprogramming the transport process holds potential for enhancing albumin conjugate efficacy in PC patients with different MUC4 mutation statuses, paving the way for stratified treatment using these delivery vehicles.
Collapse
Affiliation(s)
- Kaidi Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Xiao Cheng
- Huzhou Institute for Food and Drug Control, Huzhou 313000, PR China
| | - Shuai Xue
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Junyan Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Xu Zhang
- Zhejiang Heze Pharmaceutical Technology Co., Ltd., Hangzhou 310018, Zhejiang, PR China
| | - Yuwei Qi
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Rong Chen
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Yan Zhang
- Department of Pharmacy, Hangzhou Red Cross Hospital, Hangzhou 310003, Zhejiang, PR China
| | - Hangjie Wang
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Wei Li
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China
| | - Guilin Cheng
- Department of Pharmacy, Hangzhou Red Cross Hospital, Hangzhou 310003, Zhejiang, PR China
| | - Ye Huang
- Department of Pharmacy, Zhejiang Provincial Dermatology Hospital, Huzhou 313200, Zhejiang, PR China
| | - Yang Xiong
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China; Department of Pharmacy, Hangzhou Red Cross Hospital, Hangzhou 310003, Zhejiang, PR China
| | - Liping Chen
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, PR China; School of Chemistry and Chemical Engineering, Nanjing University, Nanjing 210023, Jiangsu, PR China.
| | - Chaofeng Mu
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China.
| | - Mancang Gu
- School of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China; Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, PR China.
| |
Collapse
|
6
|
Chen Z, Qiao S, Yang L, Sun M, Li B, Lu A, Li F. Mechanistic Insights into the Roles of the IL-17/IL-17R Families in Pancreatic Cancer. Int J Mol Sci 2023; 24:13539. [PMID: 37686343 PMCID: PMC10487659 DOI: 10.3390/ijms241713539] [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: 07/24/2023] [Revised: 08/17/2023] [Accepted: 08/23/2023] [Indexed: 09/10/2023] Open
Abstract
The members of the cytokine interleukin 17 (IL-17) family, along with their receptors (IL-17R), are vital players in a range of inflammatory diseases and cancer. Although generally regarded as proinflammatory, the effects they exhibit on cancer progression are a double-edged sword, with both antitumor and protumor activities being discovered. There is growing evidence that the IL-17 signaling pathways have significant impacts on the tumor microenvironment (TME), immune response, and inflammation in various types of cancer, including pancreatic cancer. However, the detailed mechanistic functions of the IL-17/IL-17R families in pancreatic cancer were rarely systematically elucidated. This review considers the role of the IL-17/IL-17R families in inflammation and tumor immunity and elaborates on the mechanistic functions and correlations of these members with pathogenesis, progression, and chemoresistance in pancreatic cancer. By summarizing the advanced findings on the role of IL-17/IL17R family members and IL-17 signaling pathways at the molecular level, cellular level, and disease level in pancreatic cancer, this review provides an in-depth discussion on the potential of IL-17/IL-17R as prognostic markers and therapeutic targets in pancreatic cancer.
Collapse
Affiliation(s)
- Zheng Chen
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shuangying Qiao
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Liu Yang
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Meiheng Sun
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Boyue Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Aiping Lu
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Fangfei Li
- Shum Yiu Foon Shum Bik Chuen Memorial Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; (Z.C.); (S.Q.); (L.Y.); (M.S.); (B.L.)
- Institute of Precision Medicine and Innovative Drug Discovery (PMID), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| |
Collapse
|
7
|
Wang Y, Liu L, Yu Y. Mucins and mucinous ovarian carcinoma: Development, differential diagnosis, and treatment. Heliyon 2023; 9:e19221. [PMID: 37664708 PMCID: PMC10468386 DOI: 10.1016/j.heliyon.2023.e19221] [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: 07/19/2023] [Revised: 08/11/2023] [Accepted: 08/16/2023] [Indexed: 09/05/2023] Open
Abstract
Mucinous ovarian carcinoma (MOC) is a rare histological type of epithelial ovarian cancer. It has poor response to conventional platinum-based chemotherapy regimens and PARPi-based maintenance treatment, resulting in short survival and poor prognosis in advanced-disease patients. MOC is characterized by mucus that is mainly composed of mucin in the cystic cavity. Our review discusses in detail the role of mucins in MOC. Mucins are correlated with MOC development. Furthermore, they are valuable in the differential diagnosis of primary and secondary ovarian mucinous tumors. Some types of mucins have been studied in the context of chemoresistance and targeted therapy for ovarian cancer. This review may provide a new direction for the diagnosis and treatment of advanced MOC.
Collapse
Affiliation(s)
- Yicong Wang
- Department of Obstetrics and Gynecology, Dalian Municipal Central Hospital, Dalian, China
| | - Lifeng Liu
- Department of Obstetrics and Gynecology, Dalian Municipal Central Hospital, Dalian, China
| | - Yongai Yu
- Department of Obstetrics and Gynecology, Dalian Municipal Central Hospital, Dalian, China
| |
Collapse
|
8
|
Chen L, Wang Y, Hu Q, Liu Y, Qi X, Tang Z, Hu H, Lin N, Zeng S, Yu L. Unveiling tumor immune evasion mechanisms: abnormal expression of transporters on immune cells in the tumor microenvironment. Front Immunol 2023; 14:1225948. [PMID: 37545500 PMCID: PMC10401443 DOI: 10.3389/fimmu.2023.1225948] [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: 05/20/2023] [Accepted: 07/06/2023] [Indexed: 08/08/2023] Open
Abstract
The tumor microenvironment (TME) is a crucial driving factor for tumor progression and it can hinder the body's immune response by altering the metabolic activity of immune cells. Both tumor and immune cells maintain their proliferative characteristics and physiological functions through transporter-mediated regulation of nutrient acquisition and metabolite efflux. Transporters also play an important role in modulating immune responses in the TME. In this review, we outline the metabolic characteristics of the TME and systematically elaborate on the effects of abundant metabolites on immune cell function and transporter expression. We also discuss the mechanism of tumor immune escape due to transporter dysfunction. Finally, we introduce some transporter-targeted antitumor therapeutic strategies, with the aim of providing new insights into the development of antitumor drugs and rational drug usage for clinical cancer therapy.
Collapse
Affiliation(s)
- Lu Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, China
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuchen Wang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qingqing Hu
- The Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Jinhua, China
| | - Yuxi Liu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Haihong Hu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Nengming Lin
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang, Department of Clinical Pharmacy, Affiliated Hangzhou First People’s Hospital, Cancer Center, Zhejiang University School of Medicine, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, China
| | - Su Zeng
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Lushan Yu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
- Department of Pharmacy, Shaoxing People’s Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
- Westlake Laboratory of Life Sciences and Biomedicine of Zhejiang Province, Hangzhou, China
- Department of Pharmacy, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Persaud AK, Bernier MC, Massey MA, Agrawal S, Kaur T, Nayak D, Xie Z, Weadick B, Raj R, Hill K, Abbott N, Joshi A, Anabtawi N, Bryant C, Somogyi A, Cruz-Monserrate Z, Amari F, Coppola V, Sparreboom A, Baker SD, Unadkat JD, Phelps MA, Govindarajan R. Increased renal elimination of endogenous and synthetic pyrimidine nucleosides in concentrative nucleoside transporter 1 deficient mice. Nat Commun 2023; 14:3175. [PMID: 37264059 PMCID: PMC10235067 DOI: 10.1038/s41467-023-38789-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/16/2023] [Indexed: 06/03/2023] Open
Abstract
Concentrative nucleoside transporters (CNTs) are active nucleoside influx systems, but their in vivo roles are poorly defined. By generating CNT1 knockout (KO) mice, here we identify a role of CNT1 in the renal reabsorption of nucleosides. Deletion of CNT1 in mice increases the urinary excretion of endogenous pyrimidine nucleosides with compensatory alterations in purine nucleoside metabolism. In addition, CNT1 KO mice exhibits high urinary excretion of the nucleoside analog gemcitabine (dFdC), which results in poor tumor growth control in CNT1 KO mice harboring syngeneic pancreatic tumors. Interestingly, increasing the dFdC dose to attain an area under the concentration-time curve level equivalent to that achieved by wild-type (WT) mice rescues antitumor efficacy. The findings provide new insights into how CNT1 regulates reabsorption of endogenous and synthetic nucleosides in murine kidneys and suggest that the functional status of CNTs may account for the optimal action of pyrimidine nucleoside analog therapeutics in humans.
Collapse
Affiliation(s)
- Avinash K Persaud
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Matthew C Bernier
- Campus Chemical Instrument Center Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Michael A Massey
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- The Center for Life Sciences Education, College of Arts and Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Shipra Agrawal
- Division of Nephrology & Hypertension, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Tejinder Kaur
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Debasis Nayak
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhiliang Xie
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Brenna Weadick
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Ruchika Raj
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Kasey Hill
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Nicole Abbott
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Arnav Joshi
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Nadeen Anabtawi
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Claire Bryant
- Center for Clinical & Translational Research, Nationwide Children's Hospital, Columbus, OH, 43210, USA
| | - Arpad Somogyi
- Campus Chemical Instrument Center Mass Spectrometry and Proteomics Facility, The Ohio State University, Columbus, OH, 43210, USA
| | - Zobeida Cruz-Monserrate
- Division of Gastroenterology, Hepatology, and Nutrition, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Foued Amari
- Genetically Engineered Mouse Modeling Core, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
| | - Vincenzo Coppola
- Genetically Engineered Mouse Modeling Core, Ohio State University Comprehensive Cancer Center, The Ohio State University, Columbus, OH, 43210, USA
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH, 43210, USA
| | - Alex Sparreboom
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Sharyn D Baker
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
| | - Jashvant D Unadkat
- Department of Pharmaceutics, College of Pharmacy, University of Washington, Seattle, WA, 98195, USA
- Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA
| | - Mitch A Phelps
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA
- Pharmacoanalytic Shared Resource (PhASR), The Ohio State University, Columbus, OH, 43205, USA
| | - Rajgopal Govindarajan
- Division of Pharmaceutics & Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH, 43210, USA.
- Translational Therapeutics, Ohio State University Comprehensive Cancer Center, Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
10
|
Pan YR, Wu CE, Jung SM, Huang SC, Lin SH, Chou WC, Chang YC, Chen MH, Hung TH, Yu AL, Huang WK, Yeh CN. Mucin 4 Confers Gemcitabine Resistance and an Unfavorable Prognosis in Patients with Cholangiocarcinoma via AKT Activation. Int J Biol Sci 2023; 19:2772-2786. [PMID: 37324940 PMCID: PMC10266071 DOI: 10.7150/ijbs.79126] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/24/2023] [Indexed: 06/17/2023] Open
Abstract
Cholangiocarcinoma (CCA) exhibits aggressive biological behavior and a poor prognosis. Gemcitabine (GEM)-based chemotherapy is the first-line chemotherapy for advanced CCA but has a response rate of only 20-30%. Therefore, investigating treatments to overcome GEM resistance in advanced CCA is crucial. Among mucin (MUC) family members, MUC4 showed the greatest increase in the resistant versus parental sublines. MUC4 was upregulated in whole-cell lysates and conditioned media from gemcitabine-resistant (GR) CCA sublines. MUC4 mediated GEM resistance by activating AKT signaling in GR CCA cells. The MUC4-AKT axis induced BAX S184 phosphorylation to inhibit apoptosis and downregulated GEM transporter human equilibrative nucleoside transporter 1 (hENT1) expression. The combination of AKT inhibitors and GEM or afatinib overcame GEM resistance in CCA. In vivo, capivasertib (an AKT inhibitor) increased GEM sensitivity in GR cells. MUC4 promoted EGFR and HER2 activation to mediate GEM resistance. Finally, MUC4 expression in patient plasma correlated with MUC4 expression. Paraffin-embedded specimens from non-responders expressed significantly more MUC4 than did those from responders, and this upregulation was associated with poor progression-free survival and overall survival. In GR CCA, high MUC4 expression promotes sustained EGFR/HER2 signaling and AKT activation. The combination of AKT inhibitors with GEM or afatinib might overcome GEM resistance.
Collapse
Affiliation(s)
- Yi-Ru Pan
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung University, Taoyuan 333, Taiwan
| | - Chiao-En Wu
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taiwan
| | - Shih-Ming Jung
- Department of Pathology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Shih-Chiang Huang
- Department of Pathology, Chang Gung Memorial Hospital, Linkou, Taoyuan 333, Taiwan
| | - Sheng-Hsuan Lin
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung University, Taoyuan 333, Taiwan
| | - Wen-Chi Chou
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ming-Huang Chen
- Center for Immuno-Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Tsai-Hsien Hung
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taiwan
| | - Alice L. Yu
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taiwan
- Department of Pediatrics, University of California in San Diego, San Diego, CA 92103, USA
| | - Wen-Kuan Huang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital, Linkou, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
| | - Chun-Nan Yeh
- Department of Surgery, Chang Gung Memorial Hospital, Linkou, Chang Gung University, Taoyuan 333, Taiwan
- Institute of Stem Cell and Translational Cancer Research, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan
| |
Collapse
|
11
|
Bhoopathi P, Mannangatti P, Das SK, Fisher PB, Emdad L. Chemoresistance in pancreatic ductal adenocarcinoma: Overcoming resistance to therapy. Adv Cancer Res 2023; 159:285-341. [PMID: 37268399 DOI: 10.1016/bs.acr.2023.02.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC), a prominent cause of cancer deaths worldwide, is a highly aggressive cancer most frequently detected at an advanced stage that limits treatment options to systemic chemotherapy, which has provided only marginal positive clinical outcomes. More than 90% of patients with PDAC die within a year of being diagnosed. PDAC is increasing at a rate of 0.5-1.0% per year, and it is expected to be the second leading cause of cancer-related mortality by 2030. The resistance of tumor cells to chemotherapeutic drugs, which can be innate or acquired, is the primary factor contributing to the ineffectiveness of cancer treatments. Although many PDAC patients initially responds to standard of care (SOC) drugs they soon develop resistance caused partly by the substantial cellular heterogeneity seen in PDAC tissue and the tumor microenvironment (TME), which are considered key factors contributing to resistance to therapy. A deeper understanding of molecular mechanisms involved in PDAC progression and metastasis development, and the interplay of the TME in all these processes is essential to better comprehend the etiology and pathobiology of chemoresistance observed in PDAC. Recent research has recognized new therapeutic targets ushering in the development of innovative combinatorial therapies as well as enhancing our comprehension of several different cell death pathways. These approaches facilitate the lowering of the therapeutic threshold; however, the possibility of subsequent resistance development still remains a key issue and concern. Discoveries, that can target PDAC resistance, either alone or in combination, have the potential to serve as the foundation for future treatments that are effective without posing undue health risks. In this chapter, we discuss potential causes of PDAC chemoresistance and approaches for combating chemoresistance by targeting different pathways and different cellular functions associated with and mediating resistance.
Collapse
Affiliation(s)
- Praveen Bhoopathi
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Padmanabhan Mannangatti
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States
| | - Swadesh K Das
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| | - Luni Emdad
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States.
| |
Collapse
|
12
|
Gestrich CK, De Lancy SJ, Kresak A, Sinno MG, Yalley A, Pateva I, Meyerson H, Shetty S, Oduro KA. Mucin 4 (MUC4) Protein is Expressed in B-Acute Lymphoblastic Leukemia (B-ALL) and is restricted to BCR::ABL1 Positive and BCR::ABL-like Subtypes. Hum Pathol 2023; 136:75-83. [PMID: 37023866 DOI: 10.1016/j.humpath.2023.03.018] [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] [Received: 01/30/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
Mucin 4 (MUC4) is a transmembrane mucin that, like most mucins, is not expressed in normal hematopoietic cells but little is known about its expression in malignant hematopoiesis. B-Acute Lymphoblastic Leukemia (B-ALL) consists of genetically distinct disease subtypes with similarities and differences in gene expression most frequently studied at the mRNA level, which is less amenable to widespread routine clinical use. Here, we demonstrate using immunohistochemistry (IHC) that MUC4 protein is expressed in less than 10% of B-ALL with expression restricted to BCR::ABL1+ and BCR::ABL1-like (CRLF2 rearranged) subtypes of B-ALL (4/13, 31%). None (0/36, 0%) of the remaining B-ALL subtypes expressed MUC4. We compare clinical and pathologic features of MUC4+ and MUC4- BCR::ABL1+/like cases and most significantly report a possible shorter time to relapse for MUC4+ BCR::ABL1 B-ALL that would need to be validated in larger studies. In conclusion, MUC4 is a specific, albeit insensitive, marker for these high-risk subtypes of B-ALL. We propose that MUC4 IHC may be employed diagnostically to rapidly identify these B-ALL subtypes particularly in resource limited settings or when an aspirate sample is not available for ancillary genetic studies.
Collapse
Affiliation(s)
- Catherine K Gestrich
- Department of Pathology, University Hospitals Cleveland Medical Center & Rainbow Children's Hospital & Case Western Reserve University, Cleveland, Ohio, 44106, USA; Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, 15213, USA
| | - Shanelle J De Lancy
- Department of Pathology, University Hospitals Cleveland Medical Center & Rainbow Children's Hospital & Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Adam Kresak
- Department of Pathology, University Hospitals Cleveland Medical Center & Rainbow Children's Hospital & Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Mohamad G Sinno
- Department of Pediatrics, Division of Hematology and Oncology, University Hospitals Rainbow Babies and Children's Hospital & Case Western Reserve University, Cleveland, Ohio, 44106, USA; Department of Pediatrics, Center for Cancer and Blood Disorders, Phoenix Children's Hospital, Phoenix AZ, USA
| | - Akua Yalley
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, University of Ghana, Accra, Ghana
| | - Irina Pateva
- Department of Pediatrics, Division of Hematology and Oncology, University Hospitals Rainbow Babies and Children's Hospital & Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Howard Meyerson
- Department of Pathology, University Hospitals Cleveland Medical Center & Rainbow Children's Hospital & Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Shashirekha Shetty
- Department of Pathology, University Hospitals Cleveland Medical Center & Rainbow Children's Hospital & Case Western Reserve University, Cleveland, Ohio, 44106, USA
| | - Kwadwo A Oduro
- Department of Pathology, University Hospitals Cleveland Medical Center & Rainbow Children's Hospital & Case Western Reserve University, Cleveland, Ohio, 44106, USA.
| |
Collapse
|
13
|
Park SJ, Yoo HC, Ahn E, Luo E, Kim Y, Sung Y, Yu YC, Kim K, Min DS, Lee HS, Hwang GS, Ahn T, Choi J, Bang S, Han JM. Enhanced Glutaminolysis Drives Hypoxia-Induced Chemoresistance in Pancreatic Cancer. Cancer Res 2023; 83:735-752. [PMID: 36594876 DOI: 10.1158/0008-5472.can-22-2045] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/19/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) exhibits severe hypoxia, which is associated with chemoresistance and worse patient outcome. It has been reported that hypoxia induces metabolic reprogramming in cancer cells. However, it is not well known whether metabolic reprogramming contributes to hypoxia. Here, we established that increased glutamine catabolism is a fundamental mechanism inducing hypoxia, and thus chemoresistance, in PDAC cells. An extracellular matrix component-based in vitro three-dimensional cell printing model with patient-derived PDAC cells that recapitulate the hypoxic status in PDAC tumors showed that chemoresistant PDAC cells exhibit markedly enhanced glutamine catabolism compared with chemoresponsive PDAC cells. The augmented glutamine metabolic flux increased the oxygen consumption rate via mitochondrial oxidative phosphorylation (OXPHOS), promoting hypoxia and hypoxia-induced chemoresistance. Targeting glutaminolysis relieved hypoxia and improved chemotherapy efficacy in vitro and in vivo. This work suggests that targeting the glutaminolysis-OXPHOS-hypoxia axis is a novel therapeutic target for treating patients with chemoresistant PDAC. SIGNIFICANCE Increased glutaminolysis induces hypoxia via oxidative phosphorylation-mediated oxygen consumption and drives chemoresistance in pancreatic cancer, revealing a potential therapeutic strategy of combining glutaminolysis inhibition and chemotherapy to overcome resistance.
Collapse
Affiliation(s)
- Seung Joon Park
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, South Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Hee Chan Yoo
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Eunyong Ahn
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, South Korea
| | - Enzhi Luo
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Yeabeen Kim
- Department of Life Science, Handong Global University, Pohang, South Korea
| | - Yulseung Sung
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Ya Chun Yu
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Kibum Kim
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Do Sik Min
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Hee Seung Lee
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Geum-Sook Hwang
- Integrated Metabolomics Research Group, Western Seoul Center, Korea Basic Science Institute, Seoul, South Korea
| | - TaeJin Ahn
- Department of Life Science, Handong Global University, Pohang, South Korea
| | - Junjeong Choi
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
| | - Seungmin Bang
- Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jung Min Han
- Interdisciplinary Program of Integrated OMICS for Biomedical Science, Graduate School, Yonsei University, Seoul, South Korea
- Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, South Korea
- POSTECH Biotech Center, Pohang University of Science and Technology, Pohang, South Korea
| |
Collapse
|
14
|
microRNAs Associated with Gemcitabine Resistance via EMT, TME, and Drug Metabolism in Pancreatic Cancer. Cancers (Basel) 2023; 15:cancers15041230. [PMID: 36831572 PMCID: PMC9953943 DOI: 10.3390/cancers15041230] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Despite extensive research, pancreatic cancer remains a lethal disease with an extremely poor prognosis. The difficulty in early detection and chemoresistance to therapeutic agents are major clinical concerns. To improve prognosis, novel biomarkers, and therapeutic strategies for chemoresistance are urgently needed. microRNAs (miRNAs) play important roles in the development, progression, and metastasis of several cancers. During the last few decades, the association between pancreatic cancer and miRNAs has been extensively elucidated, with several miRNAs found to be correlated with patient prognosis. Moreover, recent evidence has revealed that miRNAs are intimately involved in gemcitabine sensitivity and resistance through epithelial-to-mesenchymal transition, the tumor microenvironment, and drug metabolism. Gemcitabine is the gold standard drug for pancreatic cancer treatment, but gemcitabine resistance develops easily after chemotherapy initiation. Therefore, in this review, we summarize the gemcitabine resistance mechanisms associated with aberrantly expressed miRNAs in pancreatic cancer, especially focusing on the mechanisms associated with epithelial-to-mesenchymal transition, the tumor microenvironment, and metabolism. This novel evidence of gemcitabine resistance will drive further research to elucidate the mechanisms of chemoresistance and improve patient outcomes.
Collapse
|
15
|
Zhang C, Atri P, Nallasamy P, Parte S, Rauth S, Nimmakayala RK, Marimuthu S, Chirravuri-Venkata R, Bhatia R, Halder S, Shah A, Cox JL, Smith L, Kumar S, Foster JM, Kukreja RC, Seshacharyulu P, Ponnusamy MP, Batra SK. Small molecule inhibitor against onco-mucins disrupts Src/FosL1 axis to enhance gemcitabine efficacy in pancreatic ductal adenocarcinoma. Cancer Lett 2022; 551:215922. [PMID: 36285687 PMCID: PMC10124158 DOI: 10.1016/j.canlet.2022.215922] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/24/2022]
Abstract
Mucin MUC4 is an aberrantly expressed oncogene in pancreatic ductal adenocarcinoma (PDAC), yet no pharmacological inhibitors have been identified to target MUC4. Here, we adapted an in silico screening method using the Cancer Therapeutic Response Database (CTRD) to Identify Small Molecule Inhibitors against Mucins (SMIMs). We identified Bosutinib as a candidate drug to target oncogenic mucins among 126 FDA-approved drugs from CTRD screening. Functionally, Bosutinib treatment alone/and in combination with gemcitabine (Gem)/5' fluorouracil (5FU) reduced in vitro viability, migration, and colony formation in multiple PDAC cell lines as well as human PDAC organoid prolifertaion and growth and in vivo xenograft growth. Further, biochemical and molecular analyses showed that Bosutinib exhibited these functional effects by downregulating MUC4 mucin at both transcript and translation levels in a dose- and time-dependent manner. Mechanistically, global transcriptome analysis in PDAC cells upon treatment with Bosutinib revealed disruption of the Src-ERK/AKT-FosL1 pathway, leading to decreased expression of MUC4 and MUC5AC mucins. Taken together, Bosutinib is a promising, novel, and highly potent SMIMs to target MUC4/MUC5AC mucins. This mucin-targeting effect of Bosutinib can be exploited in the future with cytotoxic agents to treat mucinous tumors.
Collapse
Affiliation(s)
- Chunmeng Zhang
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Department of Surgical Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Palanisamy Nallasamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Seema Parte
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rama Krishna Nimmakayala
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Saravanakumar Marimuthu
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Rakesh Bhatia
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sushanta Halder
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Ashu Shah
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Lynette Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Jason M Foster
- Department of Surgical Oncology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rakesh C Kukreja
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA, 23298-0204, USA
| | | | - Moorthy P Ponnusamy
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE, USA.
| |
Collapse
|
16
|
Nucleoside transporters and immunosuppressive adenosine signaling in the tumor microenvironment: Potential therapeutic opportunities. Pharmacol Ther 2022; 240:108300. [PMID: 36283452 DOI: 10.1016/j.pharmthera.2022.108300] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/30/2022]
Abstract
Adenosine compartmentalization has a profound impact on immune cell function by regulating adenosine localization and, therefore, extracellular signaling capabilities, which suppresses immune cell function in the tumor microenvironment. Nucleoside transporters, responsible for the translocation and cellular compartmentalization of hydrophilic adenosine, represent an understudied yet crucial component of adenosine disposition in the tumor microenvironment. In this review article, we will summarize what is known regarding nucleoside transporter's function within the purinome in relation to currently devised points of intervention (i.e., ectonucleotidases, adenosine receptors) for cancer immunotherapy, alterations in nucleoside transporter expression reported in cancer, and potential avenues for targeting of nucleoside transporters for the desired modulation of adenosine compartmentalization and action. Further, we put forward that nucleoside transporters are an unexplored therapeutic opportunity, and modulation of nucleoside transport processes could attenuate the pathogenic buildup of immunosuppressive adenosine in solid tumors, particularly those enriched with nucleoside transport proteins.
Collapse
|
17
|
Zhang Z, Chen WQ, Zhang SQ, Bai JX, Liu B, Yung KKL, Ko JKS. Isoliquiritigenin inhibits pancreatic cancer progression through blockade of p38 MAPK-regulated autophagy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154406. [PMID: 36029643 DOI: 10.1016/j.phymed.2022.154406] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/22/2022] [Accepted: 08/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Pancreatic cancer has been characterized by poor prognosis, early metastasis and dissatisfactory treatment outcome. The high basal level of autophagy in tumor cells leads to chemoresistance and tumor progression. Thus, it is imminent to explore novel effective chemotherapeutic adjuvants to increase patients' survival rate. Isoliquiritigenin (ISL) is a bioactive flavonoid obtained from the Traditional Chinese herbal medicine Glycyrrhiza glabra, and it possesses a broad range of pharmacological effects. In this study, the anti-cancer effect of ISL in pancreatic cancer treatment and the underlying mechanism are investigated. METHODS MTT assay, colony formation and EdU analysis were performed to explore the growth inhibition of ISL on pancreatic cancer cells. Apoptosis were analyzed using TUNEL and flow cytometry. The formations of autophagosomes were analyzed by immunofluorescence microscopy and transmission electron microscopy. RFP-GFP-LC3B probe was applied to detect the autophagy flux. To assess the structural interaction of ISL with p38 protein, molecular docking assays were performed. The molecular mechanism was elucidated by using western immunoblotting. Subsequently, the inhibition of ISL on tumor growth was determined in vivo using pancreatic tumor mice model. RESULTS ISL inhibited pancreatic cancer cell growth and induced apoptosis, both in vitro and in vivo. ISL caused accumulation of autophagosome through blockade of late stage autophagic flux. Moreover, autophagy inducer rapamycin enhanced ISL-evoked cell growth inhibition and promoted apoptosis, while inhibition of autophagosome formation by siAtg5 attenuated ISL-induced apoptosis. It is remarkable that ISL synergistically sensitized the cytotoxic effect of gemcitabine and 5-fluorouracil on pancreatic cancer cells as both drugs induced autophagy. Molecular docking analysis has indicated that ISL acted by direct targeting of p38 MAPK, which was confirmed by ISL-induced phosphorylation of p38. The autophagy flux induced by p38 inhibitor SB203580 was blocked by ISL, with further increasing toxicity of ISL in pancreatic cancer cells. CONCLUSION The results have revealed that ISL inhibited pancreatic cancer progression by blockade of autophagy through p38 MAPK signaling.
Collapse
Affiliation(s)
- Zhu Zhang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China; Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wen-Qing Chen
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China
| | - Shi-Qing Zhang
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jing-Xuan Bai
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Bin Liu
- Department of Traditional Chinese Medicine, Institute of Integration of Traditional and Western Medicine of Guangzhou Medical University, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Ken Kin-Lam Yung
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Kowloon, Hong Kong SAR, China; Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China.
| | - Joshua Ka-Shun Ko
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China; Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Hong Kong SAR, China.
| |
Collapse
|
18
|
Rahnama N, Jahangir M, Alesaeid S, Kahrizi MS, Adili A, Mohammed RN, Aslaminabad R, Akbari M, Özgönül AM. Association between microRNAs and chemoresistance in pancreatic cancer: Current knowledge, new insights, and forthcoming perspectives. Pathol Res Pract 2022; 236:153982. [PMID: 35779293 DOI: 10.1016/j.prp.2022.153982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/27/2022] [Accepted: 06/11/2022] [Indexed: 11/25/2022]
Abstract
Pancreatic duct adenocarcinoma, commonly known as pancreatic cancer (PC), is a cancer-related cause of death due to delayed diagnosis, metastasis, and drug resistance. Patients with PC suffer from incorrect responses to chemotherapy due to inherent and acquired chemical resistance. Numerous studies have shown the mechanism of the effect of chemoresistance on PC, such as genetic and epigenetic changes or the elucidation of signaling pathways. In this regard, microRNAs (miRNAs) have been identified as essential modulators of gene expression in various cellular functions, including chemoresistance. Thus, identifying the underlying link between microRNAs and PC chemoresistance helps determine the exact pathogenesis of PC. This study aims to classify miRNAs and signaling pathways related to PC chemoresistance, suggesting new therapeutic approaches to overcome PC chemoresistance.
Collapse
Affiliation(s)
- Negin Rahnama
- Department of Internal Medicine and Health Services, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Samira Alesaeid
- Department of Internal Medicine and Rheumatology, Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ali Adili
- Senior Adult Oncology Department, Moffitt Cancer Center, University of South Florida, FL, USA; Department of Oncology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rebar N Mohammed
- Medical Laboratory Analysis Department, College of Health Sciences, Cihan University of Sulaimaniya, Kurdistan Region, Iraq; College of Veterinary Medicine, University of Sulaimani, Sulaimaniyah, Iraq
| | - Ramin Aslaminabad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ali Mert Özgönül
- Department of Biochemistry, Faculty of Medicine, Ege University, Bornova, Izmir, Turkey.
| |
Collapse
|
19
|
Zhang Z, Chen WQ, Zhang SQ, Bai JX, Lau CL, Sze SCW, Yung KKL, Ko JKS. The human cathelicidin peptide LL-37 inhibits pancreatic cancer growth by suppressing autophagy and reprogramming of the tumor immune microenvironment. Front Pharmacol 2022; 13:906625. [PMID: 35935871 PMCID: PMC9355328 DOI: 10.3389/fphar.2022.906625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/27/2022] [Indexed: 12/28/2022] Open
Abstract
Pancreatic cancer is amongst the most lethal malignancies, while its poor prognosis could be associated with promotion of autophagy and the tumor immune microenvironment. Studies have confirmed the pro-tumorigenic nature of the cathelicidin family of peptide LL-37 in several types of cancer. However, at higher doses, LL-37 exerts significant cytotoxicity against gastrointestinal cancer cells. In our study, we investigated the anti-tumorigenic potential of LL-37 in pancreatic cancer and the underlying mechanisms. Our results have shown that LL-37 inhibited the growth of pancreatic cancer both in vitro and in vivo. Mechanistic studies have demonstrated that LL-37 induced DNA damage and cell cycle arrest through induction of reactive oxygen species (ROS). Further study indicates that LL-37 suppressed autophagy in pancreatic cancer cells through activation of mTOR signaling, leading to more accumulation of ROS production and induction of mitochondrial dysfunctions. With combined treatment of LL-37 with the mTOR inhibitor rapamycin, LL-37-induced ROS production and cancer cell growth inhibition were attenuated. Subsequent in vivo study has shown that LL-37 downregulated the immunosuppressive myeloid-derived suppressor cells and M2 macrophages while upregulated the anti-cancer effectors CD8+ and CD4+ T cells in the tumor microenvironment. By using an in vitro co-culture system, it was shown that promotion of M2 macrophage polarization would be suppressed by LL-37 with inhibition of autophagy, which possessed significant negative impact on cancer growth. Taken together, our findings implicate that LL-37 could attenuate the development of pancreatic cancer by suppressing autophagy and reprogramming of the tumor immune microenvironment.
Collapse
Affiliation(s)
- Zhu Zhang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
- Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Wen-Qing Chen
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shi-Qing Zhang
- Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
- JNU-HKUST Joint Laboratory for Neuroscience and Innovative Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Jing-Xuan Bai
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ching-Lam Lau
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Stephen Cho-Wing Sze
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
- Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ken Kin-Lam Yung
- Department of Biology, Hong Kong Baptist University, Hong Kong SAR, China
- Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong SAR, China
- *Correspondence: Ken Kin-Lam Yung, ; Joshua Ka-Shun Ko,
| | - Joshua Ka-Shun Ko
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Centre for Cancer and Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- *Correspondence: Ken Kin-Lam Yung, ; Joshua Ka-Shun Ko,
| |
Collapse
|
20
|
Ye Z, Yang Y, Wei Y, Li L, Wang X, Zhang J. PCDH1 promotes progression of pancreatic ductal adenocarcinoma via activation of NF-κB signalling by interacting with KPNB1. Cell Death Dis 2022; 13:633. [PMID: 35864095 PMCID: PMC9304345 DOI: 10.1038/s41419-022-05087-y] [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: 01/04/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 01/21/2023]
Abstract
Uncontrolled growth, distant metastasis and chemoresistance are critical characteristics of pancreatic ductal adenocarcinoma (PDAC), and they result in high mortality; however, the mechanisms triggering these effects have not been fully investigated. In this study, we analysed a dataset in the Cancer Genome Atlas (TCGA) and identified PCDH1, a rarely studied transmembrane protein, as a novel prognostic marker in PDAC patients. We demonstrated that PCDH1 expression was upregulated in PDAC tissues, and its expression levels were associated with the depth of tumour invasion and lymph node metastasis. Patients with high PCDH1 levels showed poor overall survival (OS). We also investigated the biological significance of PCDH1 in PDAC cell growth, metastasis, and side population (SP) phenotype acquisition and explored the internal molecular mechanisms of PCDH1 action. Our results demonstrated that PCDH1 enhanced p65 nuclear localization by interacting with KPNB1, a well-characterized nuclear transporter, thereby activating the NF-κB signalling pathway and increasing its functional effects during PDAC progression. Hence, our results indicate that PCDH1 can be used as a negative prognostic marker and may be a potential therapeutic target for PDAC patients.
Collapse
Affiliation(s)
- Zhihua Ye
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Yingyu Yang
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Ying Wei
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Lamei Li
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Xinyi Wang
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China
| | - Junkai Zhang
- Department of Medical Oncology Center, Zhongshan City People's Hospital, 528403, Zhongshan City, Guangdong Province, P. R. China.
| |
Collapse
|
21
|
Emran TB, Shahriar A, Mahmud AR, Rahman T, Abir MH, Siddiquee MFR, Ahmed H, Rahman N, Nainu F, Wahyudin E, Mitra S, Dhama K, Habiballah MM, Haque S, Islam A, Hassan MM. Multidrug Resistance in Cancer: Understanding Molecular Mechanisms, Immunoprevention and Therapeutic Approaches. Front Oncol 2022; 12:891652. [PMID: 35814435 PMCID: PMC9262248 DOI: 10.3389/fonc.2022.891652] [Citation(s) in RCA: 156] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/10/2022] [Indexed: 12/15/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide. Several treatments are available for cancer treatment, but many treatment methods are ineffective against multidrug-resistant cancer. Multidrug resistance (MDR) represents a major obstacle to effective therapeutic interventions against cancer. This review describes the known MDR mechanisms in cancer cells and discusses ongoing laboratory approaches and novel therapeutic strategies that aim to inhibit, circumvent, or reverse MDR development in various cancer types. In this review, we discuss both intrinsic and acquired drug resistance, in addition to highlighting hypoxia- and autophagy-mediated drug resistance mechanisms. Several factors, including individual genetic differences, such as mutations, altered epigenetics, enhanced drug efflux, cell death inhibition, and various other molecular and cellular mechanisms, are responsible for the development of resistance against anticancer agents. Drug resistance can also depend on cellular autophagic and hypoxic status. The expression of drug-resistant genes and the regulatory mechanisms that determine drug resistance are also discussed. Methods to circumvent MDR, including immunoprevention, the use of microparticles and nanomedicine might result in better strategies for fighting cancer.
Collapse
Affiliation(s)
- Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Asif Shahriar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, United States
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, Bangladesh
| | - Tanjilur Rahman
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, University of Chittagong, Chittagong, Bangladesh
| | - Mehedy Hasan Abir
- Faculty of Food Science and Technology, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| | | | - Hossain Ahmed
- Department of Biotechnology and Genetic Engineering, University of Development Alternative, Dhaka, Bangladesh
| | - Nova Rahman
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Dhaka, Bangladesh
| | - Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Elly Wahyudin
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Saikat Mitra
- Department of Pharmacy, Faculty of Pharmacy, University of Dhaka, Dhaka, Bangladesh
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| | - Mahmoud M Habiballah
- Medical Laboratory Technology Department, Jazan University, Jazan, Saudi Arabia
- SMIRES for Consultation in Specialized Medical Laboratories, Jazan University, Jazan, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Bursa Uludağ University Faculty of Medicine, Bursa, Turkey
| | | | - Mohammad Mahmudul Hassan
- Queensland Alliance for One Health Sciences, School of Veterinary Science, The University of Queensland, Gatton, QLD, Australia
- Department of Physiology, Biochemistry and Pharmacology, Faculty of Veterinary Medicine, Chattogram Veterinary and Animal Sciences University, Chattogram, Bangladesh
| |
Collapse
|
22
|
Vaghari-Tabari M, Hassanpour P, Sadeghsoltani F, Malakoti F, Alemi F, Qujeq D, Asemi Z, Yousefi B. CRISPR/Cas9 gene editing: a new approach for overcoming drug resistance in cancer. Cell Mol Biol Lett 2022; 27:49. [PMID: 35715750 PMCID: PMC9204876 DOI: 10.1186/s11658-022-00348-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 05/24/2022] [Indexed: 12/18/2022] Open
Abstract
The CRISPR/Cas9 system is an RNA-based adaptive immune system in bacteria and archaea. Various studies have shown that it is possible to target a wide range of human genes and treat some human diseases, including cancers, by the CRISPR/Cas9 system. In fact, CRISPR/Cas9 gene editing is one of the most efficient genome manipulation techniques. Studies have shown that CRISPR/Cas9 technology, in addition to having the potential to be used as a new therapeutic approach in the treatment of cancers, can also be used to enhance the effectiveness of existing treatments. Undoubtedly, the issue of drug resistance is one of the main obstacles in the treatment of cancers. Cancer cells resist anticancer drugs by a variety of mechanisms, such as enhancing anticancer drugs efflux, enhancing DNA repair, enhancing stemness, and attenuating apoptosis. Mutations in some proteins of different cellular signaling pathways are associated with these events and drug resistance. Recent studies have shown that the CRISPR/Cas9 technique can be used to target important genes involved in these mechanisms, thereby increasing the effectiveness of anticancer drugs. In this review article, studies related to the applications of this technique in overcoming drug resistance in cancer cells will be reviewed. In addition, we will give a brief overview of the limitations of the CRISP/Cas9 gene-editing technique.
Collapse
Affiliation(s)
- Mostafa Vaghari-Tabari
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Hassanpour
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadeghsoltani
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Malakoti
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Forough Alemi
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center (CMBRC), Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Zatollah Asemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Kashan University of Medical Sciences, Kashan, Iran.
| | - Bahman Yousefi
- Department of Clinical Biochemistry and Laboratory Medicine, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran. .,Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
23
|
Koltai T, Reshkin SJ, Carvalho TMA, Di Molfetta D, Greco MR, Alfarouk KO, Cardone RA. Resistance to Gemcitabine in Pancreatic Ductal Adenocarcinoma: A Physiopathologic and Pharmacologic Review. Cancers (Basel) 2022; 14:2486. [PMID: 35626089 PMCID: PMC9139729 DOI: 10.3390/cancers14102486] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a very aggressive tumor with a poor prognosis and inadequate response to treatment. Many factors contribute to this therapeutic failure: lack of symptoms until the tumor reaches an advanced stage, leading to late diagnosis; early lymphatic and hematic spread; advanced age of patients; important development of a pro-tumoral and hyperfibrotic stroma; high genetic and metabolic heterogeneity; poor vascular supply; a highly acidic matrix; extreme hypoxia; and early development of resistance to the available therapeutic options. In most cases, the disease is silent for a long time, andwhen it does become symptomatic, it is too late for ablative surgery; this is one of the major reasons explaining the short survival associated with the disease. Even when surgery is possible, relapsesare frequent, andthe causes of this devastating picture are the low efficacy ofand early resistance to all known chemotherapeutic treatments. Thus, it is imperative to analyze the roots of this resistance in order to improve the benefits of therapy. PDAC chemoresistance is the final product of different, but to some extent, interconnected factors. Surgery, being the most adequate treatment for pancreatic cancer and the only one that in a few selected cases can achieve longer survival, is only possible in less than 20% of patients. Thus, the treatment burden relies on chemotherapy in mostcases. While the FOLFIRINOX scheme has a slightly longer overall survival, it also produces many more adverse eventsso that gemcitabine is still considered the first choice for treatment, especially in combination with other compounds/agents. This review discusses the multiple causes of gemcitabine resistance in PDAC.
Collapse
Affiliation(s)
| | - Stephan Joel Reshkin
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Tiago M. A. Carvalho
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Daria Di Molfetta
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Maria Raffaella Greco
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| | - Khalid Omer Alfarouk
- Zamzam Research Center, Zamzam University College, Khartoum 11123, Sudan;
- Alfarouk Biomedical Research LLC, Temple Terrace, FL 33617, USA
| | - Rosa Angela Cardone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari, 70126 Bari, Italy; (T.M.A.C.); (D.D.M.); (M.R.G.); (R.A.C.)
| |
Collapse
|
24
|
Ganguly K, Bhatia R, Rauth S, Kisling A, Atri P, Thompson C, Vengoji R, Ram Krishn S, Shinde D, Thomas V, Kaur S, Mallya K, Cox JL, Kumar S, Batra SK. Mucin 5AC Serves as the Nexus for β-Catenin/c-Myc Interplay to Promote Glutamine Dependency During Pancreatic Cancer Chemoresistance. Gastroenterology 2022; 162:253-268.e13. [PMID: 34534538 PMCID: PMC8678212 DOI: 10.1053/j.gastro.2021.09.017] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 08/17/2021] [Accepted: 09/08/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND & AIMS A major clinical challenge for patients with pancreatic cancer (PC) is metabolic adaptation. Neoplastic cells harboring molecular perturbations suffice for their increased anabolic demand and nucleotide biosynthesis to acquire chemoresistance. The mucin 5AC expressed de novo in malignant pancreas promotes cancer cell stemness and is significantly associated with poor patient survival. Identification of MUC5AC-associated drivers of chemoresistance through metabolic alterations may facilitate the sculpting of a new combinatorial regimen. METHODS The contributions of MUC5AC to glutaminolysis and gemcitabine resistance were examined by The Cancer Genome Atlas data analysis, RNA sequencing, and immunohistochemistry analysis on pancreatic tissues of KrasG12D;Pdx1-Cre (KC) and KrasG12D;Pdx1-Cre;Muc5ac-/- mice. These were followed by metabolite flux assays as well as biochemical and xenograft studies on MUC5AC-depleted human and murine PC cells. Murine and human pancreatic 3-dimensional tumoroids were used to evaluate the efficacy of gemcitabine in combination with β-catenin and glutaminolysis inhibitors. RESULTS Transcriptional analysis showed that high MUC5AC-expressing human and autochthonous murine PC tumors exhibit higher resistance to gemcitabine because of enhanced glutamine use and nucleotide biosynthesis. Gemcitabine treatment led to MUC5AC overexpression, resulting in disruption of E-cadherin/β-catenin junctions and the nuclear translocation of β-catenin, which increased c-Myc expression, with a concomitant rise in glutamine uptake and glutamate release. MUC5AC depletion and glutamine deprivation sensitized human PC cells to gemcitabine, which was obviated by glutamine replenishment in MUC5AC-expressing cells. Coadministration of β-catenin and glutaminolysis inhibitors with gemcitabine abrogated the MUC5AC-mediated resistance in murine and human tumoroids. CONCLUSIONS The MUC5AC/β-catenin/c-Myc axis increases the uptake and use of glutamine in PC cells, and cotargeting this axis along with gemcitabine may improve therapeutic efficacy in PC.
Collapse
Affiliation(s)
- Koelina Ganguly
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Rakesh Bhatia
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sanchita Rauth
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Andrew Kisling
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Christopher Thompson
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Raghupathy Vengoji
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Shiv Ram Krishn
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Dhananjay Shinde
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Vinai Thomas
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Kavita Mallya
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Jesse L Cox
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
25
|
Quader S, Tanabe S, Cabral H. Abnormal Glycosylation in Cancer Cells and Cancer Stem Cells as a Therapeutic Target. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1393:141-156. [PMID: 36587306 DOI: 10.1007/978-3-031-12974-2_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Tumor resistance and recurrence have been associated with the presence of cancer stem cells (CSCs) in tumors. The functions and survival of the CSCs have been associated with several intracellular and extracellular features. Particularly, the abnormal glycosylation of these signaling pathways and markers of CSCs have been correlated with maintaining survival, self-renewal and extravasation properties. Here, we highlight the importance of glycosylation in promoting the stemness character of CSCs and the current strategies for targeting abnormal glycosylation toward generating effective therapies against the CSC population.
Collapse
Affiliation(s)
- Sabina Quader
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
| | - Shihori Tanabe
- Division of Risk Assessment, Center for Biological Safety and Research, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki-ku, Kawasaki, 210-9501, Japan
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| |
Collapse
|
26
|
Touil Y, Latreche-Carton C, Bouazzati HE, Nugues AL, Jouy N, Thuru X, Laine W, Lepretre F, Figeac M, Tardivel M, Kluza J, Idziorek T, Quesnel B. p65/RelA NF-κB fragments generated by RIPK3 activity regulate tumorigenicity, cell metabolism, and stemness characteristics. J Cell Biochem 2021; 123:543-556. [PMID: 34927768 PMCID: PMC9299825 DOI: 10.1002/jcb.30198] [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: 08/09/2021] [Revised: 11/17/2021] [Accepted: 12/10/2021] [Indexed: 12/16/2022]
Abstract
Receptor‐interacting protein kinase 3 (RIPK3) can induce necroptosis, apoptosis, or cell proliferation and is silenced in several hematological malignancies. We previously reported that RIPK3 activity independent of its kinase domain induces caspase‐mediated p65/RelA cleavage, resulting in N‐terminal 1‐361 and C‐terminal 362‐549 fragments. We show here that a noncleavable p65/RelA D361E mutant expressed in DA1‐3b leukemia cells decreases mouse survival times and that coexpression of p65/RelA fragments increases the tumorigenicity of B16F1 melanoma cells. This aggressiveness in vivo did not correlate with NF‐κB activity measured in vitro. The fragments and p65/RelA D361E mutant induced different expression profiles in DA1‐3b and B16F1 cells. Stemness markers were affected: p65/RelA D361E increased ALDH activity in DA1‐3b cells, and fragment expression increased melanoma sphere formation in B16/F1 cells. p65/RelA fragments and the D361E noncleavable mutant decreased oxidative or glycolytic cell metabolism, with differences observed between models. Thus, p65/RelA cleavage initiated by kinase‐independent RIPK3 activity in cancer cells is not neutral and induces pleiotropic effects in vitro and in vivo that may vary across tumor types.
Collapse
Affiliation(s)
- Yasmine Touil
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Céline Latreche-Carton
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Hassiba El Bouazzati
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Anne-Lucie Nugues
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Nathalie Jouy
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Xavier Thuru
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - William Laine
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | | | - Martin Figeac
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Meryem Tardivel
- UMS 2014 CNRS/US 41 Inserm, University of Lille, Lille, France
| | - Jérôme Kluza
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Thierry Idziorek
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France
| | - Bruno Quesnel
- CANTHER, UMR 1277 Inserm - 9020 CNRS, University of Lille, Lille, France.,Institut pour la Recherche sur le Cancer de Lille, UMR 1277 Inserm - 9020 CNRS, Lille, France.,Service des Maladies du Sang, CHU Lille, Lille, France
| |
Collapse
|
27
|
Boukrout N, Souidi M, Lahdaoui F, Duchêne B, Neve B, Coppin L, Leteurtre E, Torrisani J, Van Seuningen I, Jonckheere N. Antagonistic Roles of the Tumor Suppressor miR-210-3p and Oncomucin MUC4 Forming a Negative Feedback Loop in Pancreatic Adenocarcinoma. Cancers (Basel) 2021; 13:cancers13246197. [PMID: 34944818 PMCID: PMC8699468 DOI: 10.3390/cancers13246197] [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: 10/26/2021] [Revised: 12/01/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary We aimed at characterizing microRNAs activated downstream of MUC4-associated signaling in pancreatic adenocarcinoma. We investigated the MUC4-miR-210-3p reciprocal regulation and deciphered miR-210-3p biological roles in vitro and in vivo. We showed a MUC4-miR-210-3p negative feedback loop that involves NF-κB in PDAC-derived cells and the miR-210-3p anti-tumoral functions, suggesting a complex balance between antagonistic pro-oncogenic functions of the oncomucin MUC4 and anti-tumoral roles of the miR-210-3p. Abstract Background: Pancreatic adenocarcinoma (PDAC) is a deadly cancer with an extremely poor prognosis. MUC4 membrane-bound mucin is neoexpressed in early pancreatic neoplastic lesions and is associated with PDAC progression and chemoresistance. In cancers, microRNAs (miRNAs, small noncoding RNAs) are crucial regulators of carcinogenesis, chemotherapy response and even metastatic processes. In this study, we aimed at identifying and characterizing miRNAs activated downstream of MUC4-associated signaling in pancreatic adenocarcinoma. MiRnome analysis comparing MUC4-KD versus Mock cancer cells showed that MUC4 inhibition impaired miR-210-3p expression. Therefore, we aimed to better understand the miR-210-3p biological roles. Methods: miR-210-3p expression level was analyzed by RT-qPCR in PDAC-derived cell lines (PANC89 Mock and MUC4-KD, PANC-1 and MiaPACA-2), as well as in mice and patients tissues. The MUC4-miR-210-3p regulation was investigated using luciferase reporter construct and chromatin immunoprecipitation experiments. Stable cell lines expressing miR-210-3p or anti-miR-210-3p were established using CRISPR/Cas9 technology or lentiviral transduction. We evaluated the biological activity of miR-210-3p in vitro by measuring cell proliferation and migration and in vivo using a model of subcutaneous xenograft. Results: miR-210-3p expression is correlated with MUC4 expression in PDAC-derived cells and human samples, and in pancreatic PanIN lesions of Pdx1-Cre; LstopL-KrasG12D mice. MUC4 enhances miR-210-3p expression levels via alteration of the NF-κB signaling pathway. Chromatin immunoprecipitation experiments showed p50 NF-κB subunit binding on miR-210-3p promoter regions. We established a reciprocal regulation since miR-210-3p repressed MUC4 expression via its 3′-UTR. MiR-210-3p transient transfection of PANC89, PANC-1 and MiaPACA-2 cells led to a decrease in cell proliferation and migration. These biological effects were validated in cells overexpressing or knocked-down for miR-210-3p. Finally, we showed that miR-210-3p inhibits pancreatic tumor growth and proliferation in vivo. Conclusion: We identified a MUC4-miR-210-3p negative feedback loop in early-onset PDAC, but also revealed new functions of miR-210-3p in both in vitro and in vivo proliferation and migration of pancreatic cancer cells, suggesting a complex balance between MUC4 pro-oncogenic roles and miR-210-3p anti-tumoral effects.
Collapse
Affiliation(s)
- Nihad Boukrout
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
| | - Mouloud Souidi
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
| | - Fatima Lahdaoui
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
| | - Belinda Duchêne
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
| | - Bernadette Neve
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
| | - Lucie Coppin
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
| | - Emmanuelle Leteurtre
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
| | - Jérôme Torrisani
- Université de Toulouse, INSERM, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, F-31037 Toulouse, France;
| | - Isabelle Van Seuningen
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
| | - Nicolas Jonckheere
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277-CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.B.); (M.S.); (F.L.); (B.D.); (B.N.); (L.C.); (E.L.); (I.V.S.)
- Correspondence: ; Tel.: +33-3-2029-8865
| |
Collapse
|
28
|
Stoup N, Liberelle M, Schulz C, Cavdarli S, Vasseur R, Magnez R, Lahdaoui F, Skrypek N, Peretti F, Frénois F, Thuru X, Melnyk P, Renault N, Jonckheere N, Lebègue N, Van Seuningen I. The EGF Domains of MUC4 Oncomucin Mediate HER2 Binding Affinity and Promote Pancreatic Cancer Cell Tumorigenesis. Cancers (Basel) 2021; 13:cancers13225746. [PMID: 34830899 PMCID: PMC8616066 DOI: 10.3390/cancers13225746] [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: 10/06/2021] [Revised: 11/11/2021] [Accepted: 11/11/2021] [Indexed: 12/03/2022] Open
Abstract
Simple Summary A feature of pancreatic cancer (PC) is the frequent overexpression of tyrosine kinase membrane receptor HER2 along with its membrane partner the MUC4 oncomucin in the early stages of the pancreatic carcinogenesis. However, therapeutic approaches targeting HER2 in PC are not efficient. MUC4 could indeed represent an alternative therapeutic strategy to target HER2 signaling pathway, but this approach needs to characterize MUC4/HER2 interaction at the molecular level. In this study, we successfully showed the impact of the EGF domains of MUC4 on HER2 binding affinity and demonstrated their “growth factor-like” biological activities in PC cells. Moreover, homology models of the MUC4EGF/HER2 complexes allowed identification of binding hotspots mediating binding affinity with HER2 and PC cell proliferation. These results allow a better understanding of the mechanisms involved in the MUC4/HER2 complex formation and may lead to the design of potential MUC4/HER2 inhibitors. Abstract The HER2 receptor and its MUC4 mucin partner form an oncogenic complex via an extracellular region of MUC4 encompassing three EGF domains that promotes tumor progression of pancreatic cancer (PC) cells. However, the molecular mechanism of interaction remains poorly understood. Herein, we decipher at the molecular level the role and impact of the MUC4EGF domains in the mediation of the binding affinities with HER2 and the PC cell tumorigenicity. We used an integrative approach combining in vitro bioinformatic, biophysical, biochemical, and biological approaches, as well as an in vivo study on a xenograft model of PC. In this study, we specified the binding mode of MUC4EGF domains with HER2 and demonstrate their “growth factor-like” biological activities in PC cells leading to stimulation of several signaling proteins (mTOR pathway, Akt, and β-catenin) contributing to PC progression. Molecular dynamics simulations of the MUC4EGF/HER2 complexes led to 3D homology models and identification of binding hotspots mediating binding affinity with HER2 and PC cell proliferation. These results will pave the way to the design of potential MUC4/HER2 inhibitors targeting the EGF domains of MUC4. This strategy will represent a new efficient alternative to treat cancers associated with MUC4/HER2 overexpression and HER2-targeted therapy failure as a new adapted treatment to patients.
Collapse
Affiliation(s)
- Nicolas Stoup
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Maxime Liberelle
- Univ. Lille, Inserm, CHU Lille, U1172—LilNCog—Lille Neurosciences & Cognition, F-59000 Lille, France; (M.L.); (P.M.)
| | - Céline Schulz
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Sumeyye Cavdarli
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Romain Vasseur
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Romain Magnez
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Fatima Lahdaoui
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Nicolas Skrypek
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Fabien Peretti
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Frédéric Frénois
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Xavier Thuru
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Patricia Melnyk
- Univ. Lille, Inserm, CHU Lille, U1172—LilNCog—Lille Neurosciences & Cognition, F-59000 Lille, France; (M.L.); (P.M.)
| | - Nicolas Renault
- Univ. Lille, Inserm, CHU Lille, U1286—INFINITE—Institute for Translational Research in Inflammation, F-59000 Lille, France;
| | - Nicolas Jonckheere
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
| | - Nicolas Lebègue
- Univ. Lille, Inserm, CHU Lille, U1172—LilNCog—Lille Neurosciences & Cognition, F-59000 Lille, France; (M.L.); (P.M.)
- Correspondence: (N.L.); (I.V.S.); Tel.: +33-32096-4977 (N.L.)
| | - Isabelle Van Seuningen
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020-U1277—CANTHER—Cancer Heterogeneity Plasticity and Resistance to Therapies, F-59000 Lille, France; (N.S.); (C.S.); (S.C.); (R.V.); (R.M.); (F.L.); (N.S.); (F.P.); (F.F.); (X.T.); (N.J.)
- Correspondence: (N.L.); (I.V.S.); Tel.: +33-32096-4977 (N.L.)
| |
Collapse
|
29
|
Singh K, Shishodia G, Koul HK. Pancreatic cancer: genetics, disease progression, therapeutic resistance and treatment strategies. JOURNAL OF CANCER METASTASIS AND TREATMENT 2021; 7:60. [PMID: 38107772 PMCID: PMC10722911 DOI: 10.20517/2394-4722.2021.96] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
Pancreatic cancer is a deadly disease and the third-highest cause of cancer-related deaths in the United States. It has a very low five-year survival rate (< 5%) in the United States as well as in the world (about 9%). The current gemcitabine-based therapy soon becomes ineffective because treatment resistance and surgical resection also provides only selective benefit. Signature mutations in pancreatic cancer confer chemoresistance by deregulating the cell cycle and promoting anti-apoptotic mechanisms. The stroma-rich tumor microenvironment impairs drug delivery and promotes tumor-specific immune escape. All these factors render the current treatment incompetent and prompt an urgent need for new, improved therapy. In this review, we have discussed the genetics of pancreatic cancer and its role in tumor evolution and treatment resistance. We have also evaluated new treatment strategies for pancreatic cancer, like targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Karnika Singh
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Gauri Shishodia
- Department of Otolaryngology/Head & Neck Cancer Surgery, LSU Health Sciences Center, Shreveport, LA 71103, USA
| | - Hari K. Koul
- Department of Biochemistry & Molecular Biology, Urology and Stanley S Scott Cancer Center School of Medicine LSU Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
30
|
Carter CJ, Mekkawy AH, Morris DL. Role of human nucleoside transporters in pancreatic cancer and chemoresistance. World J Gastroenterol 2021; 27:6844-6860. [PMID: 34790010 PMCID: PMC8567477 DOI: 10.3748/wjg.v27.i40.6844] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 04/19/2021] [Accepted: 09/14/2021] [Indexed: 02/06/2023] Open
Abstract
The prognosis of pancreatic cancer is poor with the overall 5-year survival rate of less than 5% changing minimally over the past decades and future projections predicting it developing into the second leading cause of cancer related mortality within the next decade. Investigations into the mechanisms of pancreatic cancer development, progression and acquired chemoresistance have been constant for the past few decades, thus resulting in the identification of human nucleoside transporters and factors affecting cytotoxic uptake via said transporters. This review summaries the aberrant expression and role of human nucleoside transports in pancreatic cancer, more specifically human equilibrative nucleoside transporter 1/2 (hENT1, hENT2), and human concentrative nucleoside transporter 1/3 (hCNT1, hCNT3), while briefly discussing the connection and importance between these nucleoside transporters and mucins that have also been identified as being aberrantly expressed in pancreatic cancer. The review also discusses the incidence, current diagnostic techniques as well as the current therapeutic treatments for pancreatic cancer. Furthermore, we address the importance of chemoresistance in nucleoside analogue drugs, in particular, gemcitabine and we discuss prospective therapeutic treatments and strategies for overcoming acquired chemoresistance in pancreatic cancer by the enhancement of human nucleoside transporters as well as the potential targeting of mucins using a combination of mucolytic compounds with cytotoxic agents.
Collapse
Affiliation(s)
- Carly Jade Carter
- Hepatobiliary and Surgical Oncology Unit, Department of Surgery, St George Hospital, University of New South Wales, Sydney 2217, New South Wales, Australia
- Mucpharm Pty Ltd, Australia
| | - Ahmed H Mekkawy
- Hepatobiliary and Surgical Oncology Unit, Department of Surgery, St George Hospital, University of New South Wales, Sydney 2217, New South Wales, Australia
- Mucpharm Pty Ltd, Australia
| | - David L Morris
- Hepatobiliary and Surgical Oncology Unit, Department of Surgery, St George Hospital, University of New South Wales, Sydney 2217, New South Wales, Australia
- Mucpharm Pty Ltd, Australia
| |
Collapse
|
31
|
Thompson CM, Cannon A, West S, Ghersi D, Atri P, Bhatia R, Smith L, Rachagani S, Wichman C, Kumar S, Batra SK. Mucin Expression and Splicing Determine Novel Subtypes and Patient Mortality in Pancreatic Ductal Adenocarcinoma. Clin Cancer Res 2021; 27:6787-6799. [PMID: 34615717 DOI: 10.1158/1078-0432.ccr-21-1591] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 08/15/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy demonstrating aberrant and progressive expression of mucins. The contribution of individual mucins has been extensively investigated in PDAC; however, comprehensive mucin profiling including splice variants in PDAC tumors has not been reported. EXPERIMENTAL DESIGN Using publicly available RNA sequencing (RNA-seq) datasets, we assess the expression of mucin family members and their splice variants (SV) in PDAC tumor samples for the first time. Mucin SVs that are correlated with PDAC patient survival are validated in a cohort of patient tumor samples. Further, we use computational methods to derive novel pancreatic tumor subtypes using mucin expression signatures and their associated activated pathways. RESULTS Principal component analysis identified four novel mucin-based PDAC subtypes. Pathway analysis implicated specific biological signatures for each subtype, labeled (i) immune activated, (ii) progressive, (iii) pancreatitis-initiated, and (iv) anti-inflammatory/PanIN-initiated. Assessing mucin SVs, significantly longer survival is observed with higher expression of 4 MUC1 and 1 MUC13 SVs, whereas patients expressing 2 MUC4 and 1 MUC16 SVs had shorter survival. Using a whole-transcriptome correlation, a three-gene panel, including ESRP2, PTK6, and MAGEH1, is designated to assess PDAC tumor sample cellularity by PCR. One MUC4 SV and one MUC13 SV are quantified in a separate PDAC patient cohort, and their effects on survival are experimentally validated. CONCLUSIONS Altogether, we demonstrate the unique expression pattern of mucins, four mucin-based PDAC subtypes, and the contribution of MUC1, MUC4, and MUC16 SVs in PDAC patient survival.
Collapse
Affiliation(s)
- Christopher M Thompson
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Andrew Cannon
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sean West
- School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska, Omaha, Nebraska
| | - Dario Ghersi
- School of Interdisciplinary Informatics, College of Information Science and Technology, University of Nebraska, Omaha, Nebraska
| | - Pranita Atri
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Rakesh Bhatia
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Lynette Smith
- Department of Biostatistics, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Satyayanarayana Rachagani
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Christopher Wichman
- Department of Biostatistics, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sushil Kumar
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, Nebraska. .,The Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
32
|
Słotwiński R, Słotwińska SM. Pancreatic cancer and adaptive metabolism in a nutrient-deficient environment. Cent Eur J Immunol 2021; 46:388-394. [PMID: 34764812 PMCID: PMC8574117 DOI: 10.5114/ceji.2021.109693] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 05/05/2021] [Indexed: 12/27/2022] Open
Abstract
Despite tremendous progress in the treatment of many cancer types, leading to a significant increase in survival, pancreatic ductal adenocarcinoma (PDAC) is still burdened with high mortality rates (5-year survival rate < 9%) due to late diagnosis, aggressiveness, and a lack of more effective treatment methods. Early diagnosis and new therapeutic approaches based on the adaptive metabolism of the tumor in a nutrient-deficient environment are expected to improve the future treatment of PDAC patients. It was found that blocking selected metabolic pathways related to the local adaptive metabolic activity of pancreatic cancer cells, improving nutrient acquisition and metabolic crosstalk within the microenvironment to sustain proliferation, may inhibit cancer development, increase cancer cell death, and increase sensitivity to other forms of treatment (e.g., chemotherapy). The present review highlights selected metabolic signaling pathways and their regulators aimed at inhibiting the neoplastic process. Particular attention is paid to the adaptive metabolism of pancreatic cancer, including fatty acids, autophagy, macropinocytosis, and deregulated cell-surface glycoproteins, which promotes cancer cell development in an oxygen-deficient and nutrient-poor environment.
Collapse
Affiliation(s)
- Robert Słotwiński
- Department of Immunology, Biochemistry and Nutrition, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
33
|
Hadj Bachir E, Poiraud C, Paget S, Stoup N, El Moghrabi S, Duchêne B, Jouy N, Bongiovanni A, Tardivel M, Weiswald LB, Vandepeutte M, Beugniez C, Escande F, Leteurtre E, Poulain L, Lagadec C, Pigny P, Jonckheere N, Renaud F, Truant S, Van Seuningen I, Vincent A. A new pancreatic adenocarcinoma-derived organoid model of acquired chemoresistance to FOLFIRINOX: First insight of the underlying mechanisms. Biol Cell 2021; 114:32-55. [PMID: 34561874 DOI: 10.1111/boc.202100003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 09/02/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND INFORMATION Although improvements have been made in the management of pancreatic adenocarcinoma (PDAC) during the past 20 years, the prognosis of this deadly disease remains poor with an overall 5-year survival under 10%. Treatment with FOLFIRINOX, a combined regimen of 5-fluorouracil, irinotecan (SN-38) and oxaliplatin, is nonetheless associated with an excellent initial tumour response and its use has allowed numerous patients to go through surgery while their tumour was initially considered unresectable. These discrepancies between initial tumour response and very low long-term survival are the consequences of rapidly acquired chemoresistance and represent a major therapeutic frontier. To our knowledge, a model of resistance to the combined three drugs has never been described due to the difficulty of modelling the FOLFIRINOX protocol both in vitro and in vivo. Patient-derived tumour organoids (PDO) are the missing link that has long been lacking in the wide range of epithelial cancer models between 2D adherent cultures and in vivo xenografts. In this work we sought to set up a model of PDO with resistance to FOLFIRINOX regimen that we could compare to the paired naive PDO. RESULTS We first extrapolated physiological concentrations of the three drugs using previous pharmacodynamics studies and bi-compartmental elimination models of oxaliplatin and SN-38. We then treated PaTa-1818x naive PDAC organoids with six cycles of 72 h-FOLFIRINOX treatment followed by 96 h interruption. Thereafter, we systematically compared treated organoids to PaTa-1818x naive organoids in terms of growth, proliferation, viability and expression of genes involved in cancer stemness and aggressiveness. CONCLUSIONS We reproductively obtained resistant organoids FoxR that significantly showed less sensitivity to FOLFORINOX treatment than the PaTa-1818x naive organoids from which they were derived. Our resistant model is representative of the sequential steps of chemoresistance observed in patients in terms of growth arrest (proliferation blockade), residual disease (cell quiescence/dormancy) and relapse. SIGNIFICANCE To our knowledge, this is the first genuine in vitro model of resistance to the three drugs in combined therapy. This new PDO model will be a great asset for the discovery of acquired chemoresistance mechanisms, knowledge that is mandatory before offering new therapeutic strategies for pancreatic cancer.
Collapse
Affiliation(s)
- Elsa Hadj Bachir
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Charles Poiraud
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Digestive Surgery and Transplantation, CHU Lille, Lille, France
| | - Sonia Paget
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Nicolas Stoup
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Soumaya El Moghrabi
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Belinda Duchêne
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Nathalie Jouy
- UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, BioImaging Center Lille (BICeL), Univ. Lille, Lille, France
| | - Antonino Bongiovanni
- UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, BioImaging Center Lille (BICeL), Univ. Lille, Lille, France
| | - Meryem Tardivel
- UMS 2014 - US 41 - PLBS - Plateformes Lilloises en Biologie & Santé, BioImaging Center Lille (BICeL), Univ. Lille, Lille, France
| | - Louis-Bastien Weiswald
- UNICAEN, Inserm U1086 ANTICIPE "Interdisciplinary Research Unit for Cancer Prevention and Treatment", Normandie Univ, Caen, France.,Cancer Centre F. Baclesse, UNICANCER, Caen, France
| | - Marie Vandepeutte
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - César Beugniez
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Digestive Surgery and Transplantation, CHU Lille, Lille, France
| | - Fabienne Escande
- Department of Biochemistry and Molecular Biology, CHU Lille, Hormonology Metabolism Nutrition Oncology, Lille, France
| | - Emmanuelle Leteurtre
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Pathology, CHU Lille, Univ. Lille, Lille, France
| | -
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Laurent Poulain
- UNICAEN, Inserm U1086 ANTICIPE "Interdisciplinary Research Unit for Cancer Prevention and Treatment", Normandie Univ, Caen, France.,Cancer Centre F. Baclesse, UNICANCER, Caen, France
| | - Chann Lagadec
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Pascal Pigny
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Nicolas Jonckheere
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Florence Renaud
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Pathology, CHU Lille, Univ. Lille, Lille, France
| | - Stephanie Truant
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France.,Department of Digestive Surgery and Transplantation, CHU Lille, Lille, France
| | - Isabelle Van Seuningen
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| | - Audrey Vincent
- CNRS, Inserm, CHU Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity Plasticity and Resistance to Therapies, Univ. Lille, Lille, France
| |
Collapse
|
34
|
Mekkawy AH, Pillai K, Badar S, Akhter J, Ke K, Valle SJ, Morris DL. Addition of bromelain and acetylcysteine to gemcitabine potentiates tumor inhibition in vivo in human colon cancer cell line LS174T. Am J Cancer Res 2021; 11:2252-2263. [PMID: 34094682 PMCID: PMC8167695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 02/08/2021] [Indexed: 06/12/2023] Open
Abstract
The combinations of Bromelain and Acetylcysteine (BromAc®) with cytotoxics such as Gemcitabine, 5-Fluorouracil or Oxaliplatin have shown a dramatic reduction in IC50 values in a variety of cancers, including colon cancer, suggesting the possibility of effective treatment without undesired side effects. In the current study, we investigated whether a similar effect is present in vivo using the colorectal cell line LS174T. Animals after acclimatization were randomized and allocated equally in the groups for the different studies (safety, dose-escalation, and efficacy). Drugs were delivered by the intraperitoneal route and animals were monitored for wellbeing. Separately, an efficacy study was conducted with intraperitoneal drug delivery after intraperitoneal tumor induction. At the termination of the experiment, tumors and other tissues were collected for evaluation. BromAc® was safe when delivered intraperitoneally in a rat model at the concentrations used. Subsequent investigations of these adjuvants in combination with Gemcitabine, Oxaliplatin, and 5-Fluorouracil in mice were also proven to be safe. Preliminary efficacy studies with Oxaliplatin and 5-Fluorouracil on tumor growth (LS174T) were negative. Gemcitabine was assessed with BromAc® showing an almost 71% tumor inhibition compared to controls. This in vivo study indicates that Gemcitabine at 2 mg/kg in combination with BromAc® 3 mg/300 mg/Kg was effective and safe, supporting its potential for future clinical application.
Collapse
Affiliation(s)
- Ahmed H Mekkawy
- Department of Surgery, St. George HospitalKogarah, NSW 2217, Australia
- Mucpharm Pty LtdAustralia
- University of New South Wales, St. George HospitalKogarah, NSW 2217, Australia
| | - Krishna Pillai
- Department of Surgery, St. George HospitalKogarah, NSW 2217, Australia
- Mucpharm Pty LtdAustralia
| | - Samina Badar
- Department of Surgery, St. George HospitalKogarah, NSW 2217, Australia
- University of New South Wales, St. George HospitalKogarah, NSW 2217, Australia
| | - Javed Akhter
- Department of Surgery, St. George HospitalKogarah, NSW 2217, Australia
- Mucpharm Pty LtdAustralia
| | - Kevin Ke
- Mucpharm Pty LtdAustralia
- University of New South Wales, St. George HospitalKogarah, NSW 2217, Australia
| | - Sarah J Valle
- Department of Surgery, St. George HospitalKogarah, NSW 2217, Australia
- Mucpharm Pty LtdAustralia
- University of New South Wales, St. George HospitalKogarah, NSW 2217, Australia
| | - David L Morris
- Department of Surgery, St. George HospitalKogarah, NSW 2217, Australia
- Mucpharm Pty LtdAustralia
- University of New South Wales, St. George HospitalKogarah, NSW 2217, Australia
| |
Collapse
|
35
|
Abstract
Nucleosides play central roles in all facets of life, from metabolism to cellular signaling. Because of their physiochemical properties, nucleosides are lipid bilayer impermeable and thus rely on dedicated transport systems to cross biological membranes. In humans, two unrelated protein families mediate nucleoside membrane transport: the concentrative and equilibrative nucleoside transporter families. The objective of this review is to provide a broad outlook on the current status of nucleoside transport research. We will discuss the role played by nucleoside transporters in human health and disease, with emphasis placed on recent structural advancements that have revealed detailed molecular principles of these important cellular transport systems and exploitable pharmacological features.
Collapse
Affiliation(s)
- Nicholas J. Wright
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, North Carolina, 27710, USA
| | - Seok-Yong Lee
- Department of Biochemistry, Duke University Medical Center, 303 Research Drive, Durham, North Carolina, 27710, USA
- Correspondence and requests for materials should be addressed to: S.-Y. Lee., , tel: 919-684-1005, fax: 919-684-8885
| |
Collapse
|
36
|
Mollaei M, Hassan ZM, Khorshidi F, Langroudi L. Chemotherapeutic drugs: Cell death- and resistance-related signaling pathways. Are they really as smart as the tumor cells? Transl Oncol 2021; 14:101056. [PMID: 33684837 PMCID: PMC7938256 DOI: 10.1016/j.tranon.2021.101056] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/05/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Chemotherapeutic drugs kill cancer cells or control their progression all over the patient's body, while radiation- and surgery-based treatments perform in a particular site. Based on their mechanisms of action, they are classified into different groups, including alkylating substrates, antimetabolite agents, anti-tumor antibiotics, inhibitors of topoisomerase I and II, mitotic inhibitors, and finally, corticosteroids. Although chemotherapeutic drugs have brought about more life expectancy, two major and severe complications during chemotherapy are chemoresistance and tumor relapse. Therefore, we aimed to review the underlying intracellular signaling pathways involved in cell death and resistance in different chemotherapeutic drug families to clarify the shortcomings in the conventional single chemotherapy applications. Moreover, we have summarized the current combination chemotherapy applications, including numerous combined-, and encapsulated-combined-chemotherapeutic drugs. We further discussed the possibilities and applications of precision medicine, machine learning, next-generation sequencing (NGS), and whole-exome sequencing (WES) in promoting cancer immunotherapies. Finally, some of the recent clinical trials concerning the application of immunotherapies and combination chemotherapies were included as well, in order to provide a practical perspective toward the future of therapies in cancer cases.
Collapse
Affiliation(s)
- Mojtaba Mollaei
- Department of Immunology, School of Medicine, Tarbiat Modares University, Tehran, Iran.
| | | | - Fatemeh Khorshidi
- Department of Immunology, School of Medicine, Tarbiat Modares University, Tehran, Iran; Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Ladan Langroudi
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
37
|
Tang M, Lozano Hernandez L, Reginald-Opara JN, Svirskis D, Leung E, Wang H, Wu Z. Zebularine suppressed gemcitabine-induced senescence and improved the cellular and plasma pharmacokinetics of gemcitabine, augmented by liposomal co-delivery. Int J Pharm 2021; 602:120659. [PMID: 33933647 DOI: 10.1016/j.ijpharm.2021.120659] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/17/2021] [Accepted: 04/25/2021] [Indexed: 02/06/2023]
Abstract
Chemoresistance is a major factor driving cancer recurrence. This study investigated the potential of zebularine, a dual cytidine deaminase (CDA)/epigenetic inhibitor, to circumvent gemcitabine-resistance in pancreatic cancer using a nanomedicine co-delivery approach. The mRNA expression of key metabolic enzymes, including CDA for gemcitabine deactivation in a gemcitabine-resistant cell line Gr2000 and its parental MIA PaCa-2 was compared using quantitative reverse transcription polymerase chain reaction. A highly gemcitabine-resistant population (HRP) in Gr2000 were characterised for their growth pattern, β-galactosidase activity (a hallmark of senescence) and chemosensitivity to zebularine after isolation. The CDA inhibition effects of zebularine on the intracellular gemcitabine accumulation and pharmacokinetics in rats when co-delivered with pH-sensitive liposomes (pSL) were investigated. Gr2000 had a 3-time upregulated mRNA expression and enzyme activity for CDA. The HRP (28% of bulk Gr2000) were predominately senescent cells which re-proliferated following a growth arrest for a week. Zebularine suppressed the regrowth of senescent cells, meanwhile enhanced cellular gemcitabine concentration by 2-fold. When co-delivered with pSL, zebularine increased cellular gemcitabine concentration by 4-fold, and extended the half-life of gemcitabine in plasma by 22-fold in rats. In conclusion, multiple mechanisms including therapy-induced senescence were identified with gemcitabine-resistance. Co-delivery of zebularine using liposomes could provide multifaceted benefits in gemcitabine therapy for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Mingtan Tang
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Lina Lozano Hernandez
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Joy N Reginald-Opara
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand
| | - Hongbo Wang
- School of Pharmacy, Yantai University, Yantai 264005, PR China
| | - Zimei Wu
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1023, New Zealand.
| |
Collapse
|
38
|
Sagar S, Leiphrakpam PD, Thomas D, McAndrews KL, Caffrey TC, Swanson BJ, Clausen H, Wandall HH, Hollingsworth MA, Radhakrishnan P. MUC4 enhances gemcitabine resistance and malignant behaviour in pancreatic cancer cells expressing cancer-associated short O-glycans. Cancer Lett 2021; 503:91-102. [PMID: 33485947 PMCID: PMC7981252 DOI: 10.1016/j.canlet.2021.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/18/2022]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is highly lethal. MUC4 (mucin4) is a heavily glycosylated protein aberrantly expressed in PDAC and promotes tumorigenesis via an unknown mechanism. To assess this, we genetically knocked out (KO) MUC4 in PDAC cells that did not express and did express truncated O-glycans (Tn/STn) using CRISPR/Cas9 technology. We found that MUC4 knockout cells possess less tumorigenicity in vitro and in vivo, which was further reduced in PDAC cells that express aberrant overexpression of truncated O-glycans. Also, MUC4KO cells showed a further reduction of epidermal growth factor receptors (ErbB) and their downstream signaling pathways in truncated O-glycan expressing PDAC cells. Tn-MUC4 specific 3B11 antibody inhibited MUC4-induced ErbB receptor and its downstream signaling cascades. MUC4 knockout differentially regulates apoptosis and cell cycle arrest in branched and truncated O-glycan expressing PDAC cells. Additionally, MUC4KO cells were found to be more sensitive to gemcitabine treatment. They possessed the upregulated expression of hENT1 and hCNT3 compared to parental cells, which were further affected in cells with aberrant O-glycosylation. Taken together, our results indicate that MUC4 enhances the malignant properties and gemcitabine resistance in PDAC tumors that aberrantly overexpress truncated O-glycans via altering ErbB/AKT signaling cascades and expression of nucleoside transporters, respectively.
Collapse
Affiliation(s)
- Satish Sagar
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Pramila D Leiphrakpam
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Divya Thomas
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Kyle L McAndrews
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Thomas C Caffrey
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Benjamin J Swanson
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200, Copenhagen N, Denmark
| | - Hans H Wandall
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200, Copenhagen N, Denmark
| | - Michael A Hollingsworth
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Prakash Radhakrishnan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
39
|
Khan T, Cabral H. Abnormal Glycosylation of Cancer Stem Cells and Targeting Strategies. Front Oncol 2021; 11:649338. [PMID: 33889547 PMCID: PMC8056457 DOI: 10.3389/fonc.2021.649338] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer stem cell (CSCs) are deemed as one of the main reasons of tumor relapse due to their resistance to standard therapies. Numerous intracellular signaling pathways along with extracellular features are crucial in regulating CSCs properties, such as heterogeneity, plasticity and differentiation. Aberrant glycosylation of these cellular signaling pathways and markers of CSCs have been directly correlated with maintaining survival, self-renewal and extravasation properties. In this review, we highlight the importance of glycosylation in promoting stemness character of CSCs, and present strategies for targeting abnormal glycosylation to eliminate the resistant CSC population.
Collapse
Affiliation(s)
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
40
|
Mucins reprogram stemness, metabolism and promote chemoresistance during cancer progression. Cancer Metastasis Rev 2021; 40:575-588. [PMID: 33813658 DOI: 10.1007/s10555-021-09959-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 03/15/2021] [Indexed: 02/06/2023]
Abstract
Mucins are high-molecular-weight glycoproteins dysregulated in aggressive cancers. The role of mucins in disease progression, tumor proliferation, and chemotherapy resistance has been studied extensively. This article provides a comprehensive review of mucin's function as a physical barrier and the implication of mucin overexpression in impeded drug delivery to solid tumors. Mucins regulate the epithelial to mesenchymal transition (EMT) of cancer cells via several canonical and non-canonical oncogenic signaling pathways. Furthermore, mucins play an extensive role in enriching and maintaining the cancer stem cell (CSC) population, thereby sustaining the self-renewing and chemoresistant cellular pool in the bulk tumor. It has recently been demonstrated that mucins regulate the metabolic reprogramming during oncogenesis and cancer progression, which account for tumor cell survival, proliferation, and drug-resistance. This review article focuses on delineating mucin's role in oncogenic signaling and aberrant regulation of gene expressions, culminating in CSC maintenance, metabolic rewiring, and development of chemoresistance, tumor progression, and metastasis.
Collapse
|
41
|
Vetvicka D, Sivak L, Jogdeo CM, Kumar R, Khan R, Hang Y, Oupický D. Gene silencing delivery systems for the treatment of pancreatic cancer: Where and what to target next? J Control Release 2021; 331:246-259. [PMID: 33482273 DOI: 10.1016/j.jconrel.2021.01.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 01/11/2021] [Accepted: 01/15/2021] [Indexed: 02/07/2023]
Abstract
Despite intensive research efforts and development of numerous new anticancer drugs and treatment strategies over the past decades, there has been only very limited improvement in overall patient survival and in effective treatment options for pancreatic cancer. Current chemotherapy improves survival in terms of months and death rates in pancreatic cancer patients are almost equivalent to incidence rates. It is imperative to develop new therapeutic approaches. Among them, gene silencing shows promise of effectiveness in both tumor cells and stromal cells by inhibiting tumor-promoting genes. This review summarizes potential targets for gene silencing in both pancreatic cancer cells and abundant stromal cells focusing on non-viral delivery systems for small RNAs and discusses the potential immunological implications. The review concludes with the importance of multifactorial therapy of pancreatic cancer.
Collapse
Affiliation(s)
- David Vetvicka
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States; Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University, Salmovska 1, Prague 2 12000, Czech Republic
| | - Ladislav Sivak
- Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, Brno CZ-61300, Czech Republic
| | - Chinmay M Jogdeo
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Raj Kumar
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Rubayat Khan
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - Yu Hang
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States
| | - David Oupický
- Center for Drug Delivery and Nanomedicine, Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE 68198, United States.
| |
Collapse
|
42
|
Holbrook MC, Goad DW, Grdzelishvili VZ. Expanding the Spectrum of Pancreatic Cancers Responsive to Vesicular Stomatitis Virus-Based Oncolytic Virotherapy: Challenges and Solutions. Cancers (Basel) 2021; 13:1171. [PMID: 33803211 PMCID: PMC7963195 DOI: 10.3390/cancers13051171] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignancy with poor prognosis and a dismal survival rate, expected to become the second leading cause of cancer-related deaths in the United States. Oncolytic virus (OV) is an anticancer approach that utilizes replication-competent viruses to preferentially infect and kill tumor cells. Vesicular stomatitis virus (VSV), one such OV, is already in several phase I clinical trials against different malignancies. VSV-based recombinant viruses are effective OVs against a majority of tested PDAC cell lines. However, some PDAC cell lines are resistant to VSV. Upregulated type I IFN signaling and constitutive expression of a subset of interferon-simulated genes (ISGs) play a major role in such resistance, while other mechanisms, such as inefficient viral attachment and resistance to VSV-mediated apoptosis, also play a role in some PDACs. Several alternative approaches have been shown to break the resistance of PDACs to VSV without compromising VSV oncoselectivity, including (i) combinations of VSV with JAK1/2 inhibitors (such as ruxolitinib); (ii) triple combinations of VSV with ruxolitinib and polycations improving both VSV replication and attachment; (iii) combinations of VSV with chemotherapeutic drugs (such as paclitaxel) arresting cells in the G2/M phase; (iv) arming VSV with p53 transgenes; (v) directed evolution approach producing more effective OVs. The latter study demonstrated impressive long-term genomic stability of complex VSV recombinants encoding large transgenes, supporting further clinical development of VSV as safe therapeutics for PDAC.
Collapse
Affiliation(s)
| | | | - Valery Z. Grdzelishvili
- Department of Biological Sciences, University of North Carolina at Charlotte, Charlotte, NC 28223, USA; (M.C.H.); (D.W.G.)
| |
Collapse
|
43
|
Giannis D, Moris D, Barbas AS. Diagnostic, Predictive and Prognostic Molecular Biomarkers in Pancreatic Cancer: An Overview for Clinicians. Cancers (Basel) 2021; 13:cancers13051071. [PMID: 33802340 PMCID: PMC7959127 DOI: 10.3390/cancers13051071] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/13/2021] [Accepted: 02/27/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Pancreatic cancer is the fourth most common cancer-related cause of death in the United States and is usually asymptomatic in early stages. There is a scarcity of tests that facilitate early diagnosis or accurately predict the disease progression. To this end, biomarkers have been identified as important tools in the diagnosis and management of pancreatic cancer. Despite the increasing number of biomarkers described in the literature, most of them have demonstrated moderate sensitivity and/or specificity and are far from being considered as screening tests. More efficient non-invasive biomarkers are needed to facilitate early-stage diagnosis and interventions. Multi-disciplinary collaboration might be required to facilitate the identification of such markers. Abstract Pancreatic ductal adenocarcinoma (PDAC) is the most common pancreatic malignancy and is associated with aggressive tumor behavior and poor prognosis. Most patients with PDAC present with an advanced disease stage and treatment-resistant tumors. The lack of noninvasive tests for PDAC diagnosis and survival prediction mandates the identification of novel biomarkers. The early identification of high-risk patients and patients with PDAC is of utmost importance. In addition, the identification of molecules that are associated with tumor biology, aggressiveness, and metastatic potential is crucial to predict survival and to provide patients with personalized treatment regimens. In this review, we summarize the current literature and focus on newer biomarkers, which are continuously added to the armamentarium of PDAC screening, predictive tools, and prognostic tools.
Collapse
Affiliation(s)
- Dimitrios Giannis
- Institute of Health Innovations and Outcomes Research, The Feinstein Institutes for Medical Research, Northwell Health, Manhasset, NY 11030, USA;
| | - Dimitrios Moris
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA;
- Correspondence: ; Tel.: +1-21-6571-6614
| | - Andrew S. Barbas
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA;
| |
Collapse
|
44
|
Gu ZT, Li ZZ, Wang CF. Research advances of intracellular mechanisms underlying gemcitabine resistance in pancreatic cancer. Shijie Huaren Xiaohua Zazhi 2020; 28:1150-1161. [DOI: 10.11569/wcjd.v28.i22.1150] [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
Pancreatic cancer is one of the most deadly malignant tumors that endanger human health, and pancreatic ductal adenocarcinoma (PDAC) is the most common histological type. Due to the lack of specific clinical symptoms, physical signs, and effective screening biomarkers for early stage PDAC, only 15%-20% of patients are qualified for surgical resection. Consequently, gemcitabine (GEM)-based monotherapy or combination therapy is still the most important or even the only treatment option. However, the overall response rate of PDAC to GEM is less than 20%, and GEM resistance is one of the most important factors affecting the efficacy of chemotherapy. At present, the mechanism of GEM resistance has not been clarified, which may involve congenital and acquired regulation. The heterogeneity of PDAC further increases its complexity. However, regulation of intracellular signaling pathways is the ultimate event to induce GEM resistance. This article will review the recent advances in research of GEM metabolism and regulation of signaling pathways in PDAC cells, and discuss potential GEM chemosensitization strategies, in order to improve the effective rate of chemotherapy and the outcome.
Collapse
Affiliation(s)
- Zong-Ting Gu
- Cheng-Feng Wang, State Key Lab of Molecular Oncology & Department of Pancreatic and Gastric Surgery, National Cancer Center/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zong-Ze Li
- Cheng-Feng Wang, State Key Lab of Molecular Oncology & Department of Pancreatic and Gastric Surgery, National Cancer Center/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | | |
Collapse
|
45
|
Unsupervised Hierarchical Clustering of Pancreatic Adenocarcinoma Dataset from TCGA Defines a Mucin Expression Profile that Impacts Overall Survival. Cancers (Basel) 2020; 12:cancers12113309. [PMID: 33182511 PMCID: PMC7697168 DOI: 10.3390/cancers12113309] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/02/2020] [Accepted: 11/04/2020] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Pancreatic cancer has a dramatic outcome (survival curve < 6 months) that is the consequence of late diagnosis and the lack of efficient therapy. We investigated the relationship between the 22 mucin gene expression and the patient survival in pancreatic cancer datasets that provide a comprehensive mapping of transcriptomic alterations occurring during carcinogenesis. Using unsupervised hierarchical clustering analysis of mucin gene expression patterns, we identified two major clusters of patients: atypical mucin signature (#1; MUC15, MUC14/EMCN, and MUC18/MCAM) and membrane-bound mucin signature (#2; MUC1, -4, -16, -17, -20, and -21). The signature #2 is associated with shorter overall survival, suggesting that the pattern of membrane-bound mucin expression could be a new prognostic marker for PDAC patients. Abstract Mucins are commonly associated with pancreatic ductal adenocarcinoma (PDAC) that is a deadly disease because of the lack of early diagnosis and efficient therapies. There are 22 mucin genes encoding large O-glycoproteins divided into two major subgroups: membrane-bound and secreted mucins. We investigated mucin expression and their impact on patient survival in the PDAC dataset from The Cancer Genome Atlas (PAAD-TCGA). We observed a statistically significant increased messenger RNA (mRNA) relative level of most of the membrane-bound mucins (MUC1/3A/4/12/13/16/17/20), secreted mucins (MUC5AC/5B), and atypical mucins (MUC14/18) compared to normal pancreas. We show that MUC1/4/5B/14/17/20/21 mRNA levels are associated with poorer survival in the high-expression group compared to the low-expression group. Using unsupervised clustering analysis of mucin gene expression patterns, we identified two major clusters of patients. Cluster #1 harbors a higher expression of MUC15 and atypical MUC14/MUC18, whereas cluster #2 is characterized by a global overexpression of membrane-bound mucins (MUC1/4/16/17/20/21). Cluster #2 is associated with shorter overall survival. The patient stratification appears to be independent of usual clinical features (tumor stage, differentiation grade, lymph node invasion) suggesting that the pattern of membrane-bound mucin expression could be a new prognostic marker for PDAC patients.
Collapse
|
46
|
Tsai LH, Hsu KW, Chiang CM, Yang HJ, Liu YH, Yang SF, Peng PH, Cheng WC, Wu HH. Targeting interleukin-17 receptor B enhances gemcitabine sensitivity through downregulation of mucins in pancreatic cancer. Sci Rep 2020; 10:17817. [PMID: 33082357 PMCID: PMC7576602 DOI: 10.1038/s41598-020-73659-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Pancreatic cancer is the fourth leading cause of death worldwide due to its poorest prognoses with a 7% 5-year survival rate. Eighty percent of pancreatic cancer patients relapse after chemotherapy and develop early metastasis and drug resistance. Resistance to nucleoside analog gemcitabine frequently used in first-line therapy is an urgent issue in pancreatic cancer treatment. Expression of mucin (MUC) glycoproteins has been shown to enhance chemoresistance via increased cell stemness. Here we show interlukine-17 receptor B (IL-17RB) expression is positively correlated with MUC1 and MUC4 expression in pancreatic cancer cells and tumor tissue. Moreover, IL-17RB transcriptionally up-regulates expression of MUC1 and MUC4 to enhance cancer stem-like properties and resistance to gemcitabine. These results suggest IL-17RB can be a potential target for pancreatic cancer therapy. Indeed, treatment with IL-17RB-neutralizing antibody has a synergistic effect in combination with gemcitabine for killing pancreatic cancer cells. Altogether, these findings provide feasible applications for IL-17RB-targeting therapy in pancreatic cancer treatment.
Collapse
Affiliation(s)
- Lung-Hung Tsai
- Research Center for Cancer Biology, China Medical University, No. 91, Hsueh-Shih Road, North District, Taichung, Taiwan.,Drug Development Center, China Medical University, Taichung, Taiwan
| | - Kai-Wen Hsu
- Research Center for Cancer Biology, China Medical University, No. 91, Hsueh-Shih Road, North District, Taichung, Taiwan.,Drug Development Center, China Medical University, Taichung, Taiwan.,Institute of New Drug Development, China Medical University, Taichung, Taiwan
| | - Cheng-Ming Chiang
- Department of Pharmacology, and Department of Biochemistry, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
| | - Hsiu-Ju Yang
- Research Center for Cancer Biology, China Medical University, No. 91, Hsueh-Shih Road, North District, Taichung, Taiwan
| | - Yu-Huei Liu
- Graduate Institute of Integrated Medicine, China Medical University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Hua Peng
- Cancer Genome Research Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Wei-Chung Cheng
- Research Center for Cancer Biology, China Medical University, No. 91, Hsueh-Shih Road, North District, Taichung, Taiwan.,Drug Development Center, China Medical University, Taichung, Taiwan.,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Heng-Hsiung Wu
- Research Center for Cancer Biology, China Medical University, No. 91, Hsueh-Shih Road, North District, Taichung, Taiwan. .,Drug Development Center, China Medical University, Taichung, Taiwan. .,Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
| |
Collapse
|
47
|
Webster P, Saito K, Cortez J, Ramirez C, Baum MM. Concentrative Nucleoside Transporter 3 Is Located on Microvilli of Vaginal Epithelial Cells. ACS OMEGA 2020; 5:20882-20889. [PMID: 32875223 PMCID: PMC7450627 DOI: 10.1021/acsomega.0c02329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/29/2020] [Indexed: 06/11/2023]
Abstract
Transporters are specialized integral membrane proteins, which mediate the passage of virtually all molecules through cell membranes. They are expressed in a broad range of human and animal tissues and play important roles in both normal and disease states. For these reasons, they are evaluated when developing and testing drugs. Two major families of drug transporters, the adenosine 5'-triphosphate-binding cassette and solute carrier transporters (SLC), have critical roles in the absorption, distribution, metabolism, and elimination of drugs. The SLC family contains known nucleoside transporters and therefore are important when nucleoside analogs are used as drugs to prevent or treat viral infections. In this study, we wanted to determine if it was possible to locate one member of the SLC family, the human concentrative nucleoside transporter 3 (CNT3) in human vaginal epithelial cells. The CNT3 protein has important roles in drug delivery, subsequent drug tissue distribution, and, hence, efficacy. Vaginal epithelial cells, taken from two human volunteers (one Caucasian and one African American), were labeled for light and electron microscopy, with a commercial antibody to a cytoplasmic domain of CNT3, the protein product of the SLC28A3 gene. Fluorescent secondary antibodies or protein A-gold were used to detect antibody binding. By electron microscopy, gold particle binding was quantified to determine labeling specificity. By light microscopy, positive labeling with anti-CNT3 antibodies was detected on human vaginal epithelial cells, but specificity to any intracellular structure was not easily determined, most likely a result of specimen preparation. Electron microscopy revealed that the CNT3 transporter protein was present predominantly on microvilli located on one side of some human vaginal epithelial cells. Quantification confirmed specific anti-CNT3 labeling over human vaginal epithelial cell microvilli. The CNT3 protein, present in the microvilli of human vaginal epithelial cells, may have a role in redistributing nucleoside homologues delivered to the vaginal tract. Transporter proteins such as CNT3 could shuttle nucleosides and their analogs through the vaginal epithelium to immune cells located in lower cell layers. Outer layers of cells, which are eventually shed from the epithelium, may remove accumulated nucleoside drug analogs from the vaginal tract.
Collapse
Affiliation(s)
- Paul Webster
- Oak
Crest Institute of Science, 132 W. Chestnut Avenue, Monrovia, California 91016, United States
| | - Kaori Saito
- Oak
Crest Institute of Science, 132 W. Chestnut Avenue, Monrovia, California 91016, United States
| | - John Cortez
- Oak
Crest Institute of Science, 132 W. Chestnut Avenue, Monrovia, California 91016, United States
| | - Christina Ramirez
- Los
Angeles (UCLA) Fielding School of Public Health, University of California, Los Angeles, 650 Charles E. Young Dr. South, 16-035 Center for
Health Sciences, Los Angeles, California 90095-1772, United States
| | - Marc M. Baum
- Oak
Crest Institute of Science, 132 W. Chestnut Avenue, Monrovia, California 91016, United States
| |
Collapse
|
48
|
Pseudopterosin and O-Methyltylophorinidine Suppress Cell Growth in a 3D Spheroid Co-Culture Model of Pancreatic Ductal Adenocarcinoma. Bioengineering (Basel) 2020; 7:bioengineering7020057. [PMID: 32545910 PMCID: PMC7357459 DOI: 10.3390/bioengineering7020057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 12/24/2022] Open
Abstract
Current therapies for treating pancreatic ductal adenocarcinoma (PDAC) are largely ineffective, with the desmoplastic environment established within these tumors being considered a central issue. We established a 3D spheroid co-culture in vitro model using a PDAC cell line (either PANC-1 or Capan-2), combined with stellate cells freshly isolated from pancreatic tumors (PSC) or hepatic lesions (HSC), and human type I collagen to analyze the efficiency of the chemotherapeutic gemcitabine (GEM) as well as two novel drug candidates derived from natural products: pseudopterosin (PsA-D) and O-methyltylophorinidine (TYLO). Traditional 2D in vitro testing of these agents for cytotoxicity on PANC-1 demonstrated IC50 values of 4.6 (±0.47) nM, 34.02 (±1.35) µM, and 1.99 (±0.13) µM for Tylo, PsA-D, and GEM, respectively; these values were comparable for Capan-2: 5.58 (±1.74) nM, 33.94 (±1.02) µM, and 0.41 (±0.06) µM for Tylo, PsA-D, and GEM, respectively. Importantly, by assessing the extent of viable cells within 3D co-culture spheroids of PANC-1 with PSC or HSC, we could demonstrate a significant lack of efficacy for GEM, while TYLO remained active and PsA-D showed slightly reduced efficacy: GEM in PANC-1/PSC (IC50 = >100 µM) or PANC-1/HSC (IC50 = >100 µM) spheroids, TYLO in PANC-1/PSC (IC50 = 3.57 ± 1.30 nM) or PANC-1/HSC (IC50 = 6.39 ± 2.28 nM) spheroids, and to PsA-D in PANC-1/PSC (IC50 = 54.42 ± 12.79 µM) or PANC-1/HSC (IC50 = 51.75 ± 0.60 µM). Microscopic 3D rendering supported these cytotoxicity outcomes, showing little or no morphological spheroid structure change during this period of rapid cell death. Our results support the use of this 3D spheroid co-culture in vitro model having a desmoplastic microenvironment for the identification of possible novel chemotherapeutic drug candidates for PDAC, such as TYLO and PsA-D.
Collapse
|
49
|
Abdkarimi S, Razi Soofiyani S, Elham G, Mashhadi Abdolahi H, Safarzadeh E, Baradaran B. Targeting immune checkpoints: Building better therapeutic puzzle in pancreatic cancer combination therapy. Eur J Cancer Care (Engl) 2020; 29:e13268. [PMID: 32459388 DOI: 10.1111/ecc.13268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/30/2020] [Accepted: 04/30/2020] [Indexed: 12/24/2022]
Abstract
Pancreatic cancer is related to a very weak diagnosis; the close parallel between disease incidence and mortality rates from pancreatic cancer reflects the fatal nature of this disease. Although early detection procedures are growing, they are not applicable yet for pancreatic cancer. The majority of cancer patients suffer from advanced disease, in which surgery has no potential effect. Based on the growing evidence, it is predicated that cancer immunotherapy alone or in combination will probably be an essential section of different cancer treatment methods. There are different kinds of immune processes, including various antitumour and tumour-promoting leukocytes. Moreover, tumour cells utilise numerous approaches to overwhelm the immune response. Use of antibody in the therapeutic protocols is proving significant success and is probably a key element of cancer treatment. This method is directed against numerous negative immunologic regulators and immune checkpoints. In the present review, the clinical outlines of immune checkpoint inhibition are discussed in pancreatic cancer.
Collapse
Affiliation(s)
- Sina Abdkarimi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saiedeh Razi Soofiyani
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Goli Elham
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hossein Mashhadi Abdolahi
- Tabriz Health Services Management Research Centre, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Safarzadeh
- Department of Immunology and Microbiology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
50
|
Abdelhady AS, Abdel Hamid FF, Hassan NM, Ibrahim DM. Prognostic value of bone marrow MUC4 expression in acute myeloid leukaemia. Br J Biomed Sci 2020; 77:202-207. [PMID: 32270747 DOI: 10.1080/09674845.2020.1754583] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Background: Aberrant expression of mucin-4 (MUC4) is present in a variety of solid cancers, but the expression pattern of MUC4 and its clinical relevance in acute myeloid leukaemia (AML) is unknown. We aimed to evaluate the expression level of MUC4 and explore its prognostic value in newly diagnosed adult patients with AML. Methods: Bone marrow from 70 AML patients and 26 healthy donors was obtained. MUC4 levels were quantified by quantitative real-time PCR. Routine blood indices were measured by standard techniques. Results: Bone marrow MUC4 expression levels were significantly elevated in AML patients compared to controls at median (range) 2.77 (0.7-16.6) and 1.14 (0.5-1.99) respectively (p = 0.005). Moreover, lower MUC4 expression was strongly associated with persistent remission (p = 0.001) while higher MUC4 levels were associated with worse overall as well as disease-free survival (p = 0.011 and p = 0.006, respectively). Thus, its level may act as an indicator of disease progression. High MUC4 expression was identified as an independent prognostic predictor for both overall survival and disease-free survival. Conclusion: MUC4 over-expression is an independent predictor of a poor prognosis in AML patients.
Collapse
Affiliation(s)
- A S Abdelhady
- Biochemistry Department, Faculty of Science, Ain Shams University , Cairo, Egypt
| | - F F Abdel Hamid
- Biochemistry Department, Faculty of Science, Ain Shams University , Cairo, Egypt
| | - N M Hassan
- Clinical Pathology Department, National Cancer Institute, Cairo University , Cairo, Egypt
| | - D M Ibrahim
- Biochemistry Department, Faculty of Science, Ain Shams University , Cairo, Egypt
| |
Collapse
|